Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 476
Filter
1.
J Med Chem ; 66(2): 1112-1136, 2023 01 26.
Article in English | MEDLINE | ID: mdl-36645394

ABSTRACT

The death-associated protein kinase (DAPK) family is a member of the calcium/calmodulin-regulated serine/threonine protein kinase family, and studies have shown that its role, as its name suggests, is mainly to regulate cell death. The DAPK family comprises five members, including DAPK1, DAPK2, DAPK3, DRAK1 and DRAK2, which show high homology in the common N-terminal kinase domain but differ in the extra-catalytic domain. Notably, previous research has suggested that the DAPK family plays an essential role in both the development and regulation of human diseases. However, only a few small-molecule inhibitors have been reported. In this Perspective, we mainly discuss the structure, biological function, and role of DAPKs in diseases and the currently discovered small-molecule inhibitors, providing valuable information for the development of the DAPK field.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases , Protein Serine-Threonine Kinases , Humans , Death-Associated Protein Kinases/chemistry , Death-Associated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Catalytic Domain , Calcium-Calmodulin-Dependent Protein Kinases/chemistry
2.
J Med Chem ; 64(19): 14358-14376, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34543009

ABSTRACT

CASK (Ca2+/calmodulin-dependent Ser/Thr kinase) is a member of the MAGUK (membrane-associated guanylate kinase) family that functions as neurexin kinases with roles implicated in neuronal synapses and trafficking. The lack of a canonical DFG motif, which is altered to GFG in CASK, led to the classification as a pseudokinase. However, functional studies revealed that CASK can still phosphorylate substrates in the absence of divalent metals. CASK dysfunction has been linked to many diseases, including colorectal cancer, Parkinson's disease, and X-linked mental retardation, suggesting CASK as a potential drug target. Here, we exploited structure-based design for the development of highly potent and selective CASK inhibitors based on 2,4-diaminopyrimidine-5-carboxamides targeting an unusual pocket created by the GFG motif. The presented inhibitor design offers a more general strategy for the development of pseudokinase ligands that harbor unusual sequence motifs. It also provides a first chemical probe for studying the biological roles of CASK.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Molecular Probes/chemistry , Protein Kinase Inhibitors/chemistry , Serine/chemistry , Threonine/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Drug Design , Humans , Molecular Probes/pharmacology , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship , Substrate Specificity
3.
Biochem Biophys Res Commun ; 525(3): 537-542, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32113680

ABSTRACT

Calcium/calmodulin-dependent protein kinase (CCaMK) has been shown to play important roles in brassinosteroid (BR)-induced antioxidant defense and enhancing the tolerance of plants to drought stress. The autophosphorylation of CCaMK is a key step for the activation of CCaMK, thus promoting substrate phosphorylation. However, how CCaMK autophosphorylation function in BR-induced antioxidant defense is not known yet. Here, seven potential autophosphorylation sites of ZmCCaMK were identified using mass spectroscopy (liquid chromatography-tandem mass spectrometry [LC-MS/MS]) analysis. The transient gene expression analysis in maize protoplasts showed that Thr420 and Ser454 of ZmCCaMK were important for BR-induced antioxidant defense. Furthermore, Thr420 and Ser454 of ZmCCaMK were crucial for improving drought tolerance and alleviating drought induced oxidative damage of plants via overexpressing various mutant versions of ZmCCaMK in tobacco (Nicotiana tabacum). Mutations of Thr420 and Ser454 in ZmCCaMK substantially blocked the autophosphorylation and substrate phosphorylation of ZmCCaMK in vitro. Taken together, our results demonstrate that Thr420 and Ser454 of ZmCCaMK are crucial for BR-induced antioxidant defense and drought tolerance through modulating the autophosphorylation and substrate phosphorylation activities of ZmCCaMK.


Subject(s)
Antioxidants/metabolism , Brassinosteroids/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Serine/metabolism , Threonine/metabolism , Zea mays/enzymology , Adaptation, Physiological/drug effects , Droughts , Mutant Proteins/metabolism , Phosphorylation/drug effects , Plant Proteins/chemistry , Plant Proteins/metabolism , Plants, Genetically Modified , Structure-Activity Relationship , Substrate Specificity/drug effects , Nicotiana/genetics , Zea mays/drug effects
4.
Int J Biol Macromol ; 123: 704-712, 2019 Feb 15.
Article in English | MEDLINE | ID: mdl-30414416

ABSTRACT

Calmodulin binding receptor like cytoplasmic kinase 2 (CRCK2) belongs to the family of receptor like kinases (RLKs) which is mainly implicated in pathways associated with the stress responses in plants. The protein from the stem of Oroxylum indicum was isolated and purified using anion-exchange followed by gel filtration chromatography. The purity of protein was checked using SDS-PAGE, which showed a single band of 50 kDa. The purified protein was identified as CRCK2 using MALDI-TOF. Using I-TASSER, a bioinformatics tools, the model of protein was constructed and its secondary structure was predicted using VADAR. The secondary structure content was also determined by far-UV CD, which indicated that the CRCK2 is mainly ß-sheet dominating protein (43% ß-sheet). The secondary structural content predication from computational method is in close agreement with the result obtained by CD spectropolarimeter. This study validates I-TASSER model for determination of structure of a protein. Moreover, stability of CRCK2 was monitored against heat- and guanidinium chloride (GdmCl)-induced denaturation by using circular dichroism (CD) and fluorescence spectroscopy. Denaturation curve analysis gave values of 2.88 ±â€¯0.12 kcal mol-1and 4.11 ±â€¯0.09 M for ∆°GD (Gibbs free energy change at 25 °C) and Cm (midpoint of denaturation), respectively. It has been observed that purified CRCK2 is quite stable protein against both heat-induced as well as GdmCl-induced denaturation. This is very first report of purification and biophysical characterization of CRCK2 protein from medicinal plant O. indicum.


Subject(s)
Bignoniaceae/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Protein Conformation, beta-Strand , Protein Structure, Secondary , Bignoniaceae/chemistry , Biophysical Phenomena , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Circular Dichroism , Models, Chemical , Protein Binding , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
5.
Biosci Biotechnol Biochem ; 82(8): 1335-1343, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29673297

ABSTRACT

We surveyed genome sequences from the basidiomycetous mushroom Coprinopsis cinerea and isolated a cDNA homologous to CMKA, a calmodulin-dependent protein kinase (CaMK) in Aspergillus nidulans. We designated this sequence, encoding 580 amino acids with a molecular weight of 63,987, as CoPK02. CoPK02 possessed twelve subdomains specific to protein kinases and exhibited 43, 35, 40% identity with rat CaMKI, CaMKII, CaMKIV, respectively, and 40% identity with CoPK12, one of the CaMK orthologs in C. cinerea. CoPK02 showed significant autophosphorylation activity and phosphorylated exogenous proteins in the presence of Ca2+/CaM. By the CaM-overlay assay we confirmed that the C-terminal sequence (Trp346-Arg358) was the calmodulin-binding site, and that the binding of Ca2+/CaM to CoPK02 was reduced by the autophosphorylation of CoPK02. Since CoPK02 evolved in a different clade from CoPK12, and showed different gene expression compared to that of CoPK32, which is homologous to mitogen-activated protein kinase-activated protein kinase, CoPK02 and CoPK12 might cooperatively regulate Ca2+-signaling in C. cinerea.


Subject(s)
Basidiomycota/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Amino Acid Sequence , Animals , Basidiomycota/genetics , Basidiomycota/growth & development , Binding Sites , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calmodulin/metabolism , Catalysis , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Gene Expression Profiling , Genes, Fungal , Phosphorylation , Phylogeny , Rats , Sequence Homology, Amino Acid
6.
Comput Biol Chem ; 72: 164-169, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29191749

ABSTRACT

Calmodulin-dependent protein kinase (CAMK) is physiologically activated in fertilized human oocytes and is involved in the Ca2+ response pathways that link the fertilization calmodulin signal to meiosis resumption and cortical granule exocytosis. The kinase has an unstructured C-terminal tail that can be recognized and bound by the PDZ5 domain of its cognate partner, the multi-PDZ domain protein (MUP). In the current study, we reported a rational biomolecular design of halogen-bonding system at the complex interface of CAMK's C-terminal peptide with MUP PDZ5 domain by using high-level computational approaches. Four organic halogens were employed as atom probes to explore the structural geometry and energetic property of designed halogen bonds in the PDZ5-peptide complex. It was found that the heavier halogen elements such as bromine Br and iodine I can confer stronger halogen bond but would cause bad atomic contacts and overlaps at the complex interface, while fluorine F cannot form effective halogen bond in the complex. In addition, the halogen substitution at different positions of peptide's aromatic ring would result in distinct effects on the halogen-bonding system. The computational findings were then verified by using fluorescence analysis; it is indicated that the halogen type and substitution position play critical role in the interaction strength of halogen bonds, and thus the PDZ5-peptide binding affinity can be improved considerably by optimizing their combination.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carrier Proteins/metabolism , Amino Acid Sequence , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Carrier Proteins/chemistry , Halogenation , Humans , Hydrogen Bonding , Membrane Proteins , Molecular Structure , Protein Binding , Protein Domains , Quantum Theory
7.
Biochem Biophys Res Commun ; 490(2): 441-446, 2017 08 19.
Article in English | MEDLINE | ID: mdl-28623136

ABSTRACT

Arabidopsis inositol polyphosphate kinase 2ß (AtIpk2ß) has multiple functions in plant development and in responding to abiotic stress. Although some related clues suggested a potential role of AtIpk2ß in ABA signaling, the defined evidence was still lack. Here we discovered that a key ABA signaling component calcium-dependent protein kinase 4 (CPK4) can interact with AtIpk2ß under ABA treated conditions through affinity purification and mass spectrometry detection. The interaction between CPK4 and AtIpk2ß were further confirmed by yeast two hybrid and bimolecular fluorescence complementation assays. Expression of AtIpk2ß also can be rapidly induced by ABA. In addition, we found that CPK4 can phosphorylate AtIpk2ß in vitro and identified five novel phosphorylation sites of AtIpk2ß by CPK4 kinase, including Tyr46, Ser48, Ser51, Thr128, Ser147. Overexpression of AtIpk2ß in Arabidopsis was more sensitive to ABA in seed germination, primary root inhibition, ABA-responsive gene expression than wild type plants, whereas knockout mutant atipk2ß exhibited no significant difference. The AtIpk2ß variants containing Tyr46, Thr128, Ser147 mutated to Ala cannot complement the yeast mutant ipk2 growth in high temperature, suggesting that those three amino acid residues are critical for AtIpk2ß. These findings provide insight into the modulation of ABA signaling by AtIpk2ß.


Subject(s)
Abscisic Acid/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction , Amino Acid Sequence , Arabidopsis/chemistry , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Gene Expression Regulation, Plant , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Interaction Maps , Up-Regulation
8.
Nature ; 545(7654): 311-316, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28489820

ABSTRACT

Nutrient signalling integrates and coordinates gene expression, metabolism and growth. However, its primary molecular mechanisms remain incompletely understood in plants and animals. Here we report unique Ca2+ signalling triggered by nitrate with live imaging of an ultrasensitive biosensor in Arabidopsis leaves and roots. A nitrate-sensitized and targeted functional genomic screen identifies subgroup III Ca2+-sensor protein kinases (CPKs) as master regulators that orchestrate primary nitrate responses. A chemical switch with the engineered mutant CPK10(M141G) circumvents embryo lethality and enables conditional analyses of cpk10 cpk30 cpk32 triple mutants to define comprehensive nitrate-associated regulatory and developmental programs. Nitrate-coupled CPK signalling phosphorylates conserved NIN-LIKE PROTEIN (NLP) transcription factors to specify the reprogramming of gene sets for downstream transcription factors, transporters, nitrogen assimilation, carbon/nitrogen metabolism, redox, signalling, hormones and proliferation. Conditional cpk10 cpk30 cpk32 and nlp7 mutants similarly impair nitrate-stimulated system-wide shoot growth and root establishment. The nutrient-coupled Ca2+ signalling network integrates transcriptome and cellular metabolism with shoot-root coordination and developmental plasticity in shaping organ biomass and architecture.


Subject(s)
Amidohydrolases/metabolism , Arabidopsis/growth & development , Arabidopsis/metabolism , Calcium/metabolism , Nitrates/metabolism , Protein Kinases/metabolism , Signal Transduction , Amidohydrolases/genetics , Amino Acid Sequence , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Biomass , Calcium Signaling , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carbon/metabolism , Cellular Reprogramming , Food , Gene Expression Regulation, Plant , Nitrogen/metabolism , Oxidation-Reduction , Phosphorylation , Plant Roots/growth & development , Plant Roots/metabolism , Plant Shoots/growth & development , Plant Shoots/metabolism , Plants, Genetically Modified , Protein Kinases/chemistry , Protein Kinases/genetics , Transcription, Genetic , Transcriptome
9.
Sci Rep ; 6: 26634, 2016 05 23.
Article in English | MEDLINE | ID: mdl-27211275

ABSTRACT

The α-kinases are a family of a typical protein kinases present in organisms ranging from protozoa to mammals. Here we report an autoinhibited conformation for the α-kinase domain of Dictyostelium myosin-II heavy chain kinase A (MHCK-A) in which nucleotide binding to the catalytic cleft, located at the interface between an N-terminal and C-terminal lobe, is sterically blocked by the side chain of a conserved arginine residue (Arg592). Previous α-kinase structures have shown that an invariant catalytic aspartic acid residue (Asp766) is phosphorylated. Unexpectedly, in the autoinhibited conformation the phosphoryl group is transferred to the adjacent Asp663, creating an interaction network that stabilizes the autoinhibited state. The results suggest that Asp766 phosphorylation may play both catalytic and regulatory roles. The autoinhibited structure also provides the first view of a phosphothreonine residue docked into the phospho-specific allosteric binding site (Pi-pocket) in the C-lobe of the α-kinase domain.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Dictyostelium/enzymology , Protozoan Proteins/chemistry , Apoenzymes/chemistry , Protein Domains
10.
J Biol Chem ; 290(39): 23935-46, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26260792

ABSTRACT

The α-kinases are a widely expressed family of serine/threonine protein kinases that exhibit no sequence identity with conventional eukaryotic protein kinases. In this report, we provide new information on the catalytic properties of the α-kinase domain of Dictyostelium myosin-II heavy chain kinase-A (termed A-CAT). Crystallization of A-CAT in the presence of MgATP yielded structures with AMP or adenosine in the catalytic cleft together with a phosphorylated Asp-766 residue. The results show that the ß- and α-phosphoryl groups are transferred either directly or indirectly to the catalytically essential Asp-766. Biochemical assays confirmed that A-CAT hydrolyzed ATP, ADP, and AMP with kcat values of 1.9, 0.6, and 0.32 min(-1), respectively, and showed that A-CAT can use ADP to phosphorylate peptides and proteins. Binding assays using fluorescent 2'/3'-O-(N-methylanthraniloyl) analogs of ATP and ADP yielded Kd values for ATP, ADP, AMP, and adenosine of 20 ± 3, 60 ± 20, 160 ± 60, and 45 ± 15 µM, respectively. Site-directed mutagenesis showed that Glu-713, Leu-716, and Lys-645, all of which interact with the adenine base, were critical for nucleotide binding. Mutation of the highly conserved Gln-758, which chelates a nucleotide-associated Mg(2+) ion, eliminated catalytic activity, whereas loss of the highly conserved Lys-722 and Arg-592 decreased kcat values for kinase and ATPase activities by 3-6-fold. Mutation of Asp-663 impaired kinase activity to a much greater extent than ATPase, indicating a specific role in peptide substrate binding, whereas mutation of Gln-768 doubled ATPase activity, suggesting that it may act to exclude water from the active site.


Subject(s)
Adenine Nucleotides/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Dictyostelium/enzymology , Protozoan Proteins/chemistry , Adenine Nucleotides/genetics , Adenine Nucleotides/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Crystallography, X-Ray , Dictyostelium/genetics , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
11.
Bioorg Med Chem ; 23(12): 2749-60, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25963826

ABSTRACT

Many human protein kinases are regulated by the calcium-sensor protein calmodulin, which binds to a short flexible segment C-terminal to the enzyme's catalytic kinase domain. Our understanding of the molecular mechanism of kinase activity regulation by calcium/calmodulin has been advanced by the structures of two protein kinases-calmodulin kinase II and death-associated protein kinase 1-bound to calcium/calmodulin. Comparison of these two structures reveals a surprising level of diversity in the overall kinase-calcium/calmodulin arrangement and functional readout of activity, as well as complementary mechanisms of kinase regulation such as phosphorylation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Amino Acid Sequence , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Death-Associated Protein Kinases/chemistry , Death-Associated Protein Kinases/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Alignment
12.
Mol Cell Biol ; 34(12): 2294-307, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24732796

ABSTRACT

Eukaryotic elongation factor 2 kinase (eEF2K) is the best-characterized member of the α-kinase family. Within this group, only eEF2K and myosin heavy chain kinases (MHCKs) have known substrates. Here we have studied the roles of specific residues, selected on the basis of structural data for MHCK A and TRPM7, in the function of eEF2K. Our data provide the first information regarding the basis of the substrate specificity of α-kinases, in particular the roles of residues in the so-called N/D loop, which appears to occupy a position in the structure of α-kinases similar to that of the activation loop in other kinases. Several mutations in the EEF2K gene occur in tumors, one of which (Arg303Cys) is at a highly conserved residue in the N/D loop. This mutation greatly enhances eEF2K activity and may be cytoprotective. Our data support the concept that the major autophosphorylation site (Thr348 in eEF2K) docks into a binding pocket to help create the kinase-competent conformation. This is similar to the situation for MHCK A and is consistent with this being a common feature of α-kinases.


Subject(s)
Catalytic Domain , Conserved Sequence , Elongation Factor 2 Kinase/chemistry , Elongation Factor 2 Kinase/metabolism , Amino Acid Sequence , Amino Acids/genetics , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , HEK293 Cells , Humans , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation , Phosphothreonine/metabolism , Protein Binding , Protein Structure, Secondary , Protozoan Proteins/chemistry , Structural Homology, Protein , Structure-Activity Relationship , Substrate Specificity
13.
Biomed Res Int ; 2013: 134813, 2013.
Article in English | MEDLINE | ID: mdl-23991411

ABSTRACT

Ca(2+)/calmodulin-dependent protein kinase phosphatase (CaMKP/PPM1F) and its nuclear homolog CaMKP-N (PPM1E) are Ser/Thr protein phosphatases that belong to the PPM family. CaMKP-N is expressed in the brain and undergoes proteolytic processing to yield a C-terminally truncated form. The physiological significance of this processing, however, is not fully understood. Using a wheat-embryo cell-free protein expression system, we prepared human CaMKP-N (hCaMKP-N(WT)) and the truncated form, hCaMKP-N(1-559), to compare their enzymatic properties using a phosphopeptide substrate. The hCaMKP-N(1-559) exhibited a much higher V(max) value than the hCaMKP-N(WT) did, suggesting that the processing may be a regulatory mechanism to generate a more active species. The active form, hCaMKP-N(1-559), showed Mn(2+) or Mg(2+)-dependent phosphatase activity with a strong preference for phospho-Thr residues and was severely inhibited by NaF, but not by okadaic acid, calyculin A, or 1-amino-8-naphthol-2,4-disulfonic acid, a specific inhibitor of CaMKP. It could bind to postsynaptic density and dephosphorylate the autophosphorylated Ca(2+)/calmodulin-dependent protein kinase II. Furthermore, it was inactivated by H2O2 treatment, and the inactivation was completely reversed by treatment with DTT, implying that this process is reversibly regulated by oxidation/reduction. The truncated CaMKP-N may play an important physiological role in neuronal cells.


Subject(s)
Brain/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Phosphopeptides/chemistry , Phosphoprotein Phosphatases/chemistry , Animals , Enzyme Activation , Enzyme Stability , Rats , Structure-Activity Relationship
14.
Am J Pathol ; 182(3): 1005-20, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23438478

ABSTRACT

The TNF-IL-6-STAT3 pathway plays a crucial role in promoting ulcerative colitis-associated carcinoma (UCC). To date, the negative regulation of STAT3 is poorly understood. Interestingly, intestinal epithelial cells of UCC in comparison to ulcerative colitis show high expression levels of anti-inflammatory death-associated protein kinase (DAPK) and low levels of pSTAT3. Accordingly, epithelial DAPK expression was enhanced in STAT3(IEC-KO) mice. To unravel a possible regulatory mechanism, we used an in vitro TNF-treated intestinal epithelial cell model. We identified a new function of DAPK in suppressing TNF-induced STAT3 activation as DAPK siRNA knockdown and treatment with a DAPK inhibitor potentiated STAT3 activation, IL-6 mRNA expression, and secretion. DAPK attenuated STAT3 activity directly by physical interaction shown in three-dimensional structural modeling. This model suggests that DAPK-induced conformational changes in the STAT3 dimer masked its nuclear localization signal. Alternatively, pharmacological inactivation of STAT3 led to an increase in DAPK mRNA and protein levels. Chromatin immunoprecipitation showed that STAT3 restricted DAPK expression by promoter binding, thereby reinforcing its own activation by inducing IL-6. This novel negative regulation principle might balance TNF-induced inflammation and seems to play an important role in the inflammation-associated transformation process as confirmed in an AOM+DSS colon carcinogenesis mouse model. DAPK as a negative regulator of STAT3 emerges as therapeutic option in the treatment of ulcerative colitis and UCC.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Epithelial Cells/enzymology , Intestines/pathology , STAT3 Transcription Factor/metabolism , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/pathology , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Death-Associated Protein Kinases , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Humans , Hydrogen Bonding/drug effects , Hydrophobic and Hydrophilic Interactions/drug effects , Inflammation/pathology , Interleukin-6/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mutant Proteins/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Multimerization/drug effects , STAT3 Transcription Factor/chemistry , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
15.
Mol Plant ; 6(4): 1274-1289, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23253603

ABSTRACT

14-3-3 proteins play an important role in the regulation of many cellular processes. The Arabidopsis vacuolar two-pore K(+) channel 1 (TPK1) interacts with the 14-3-3 protein GRF6 (GF14-λ). Upon phosphorylation of the putative binding motif in the N-terminus of TPK1, GRF6 binds to TPK1 and activates the potassium channel. In order to gain a deeper understanding of this 14-3-3-mediated signal transduction, we set out to identify the respective kinases, which regulate the phosphorylation status of the 14-3-3 binding motif in TPK1. Here, we report that the calcium-dependent protein kinases (CDPKs) can phosphorylate and thereby activate the 14-3-3 binding motif in TPK1. Focusing on the stress-activated kinase CPK3, we visualized direct and specific interaction of TPK1 with the kinase at the tonoplast in vivo. In line with its proposed role in K(+) homeostasis, TPK1 phosphorylation was found to be induced by salt stress in planta, and both cpk3 and tpk1 mutants displayed salt-sensitive phenotypes. Molecular modeling of the TPK1-CPK3 interaction domain provided mechanistic insights into TPK1 stress-regulated phosphorylation responses and pinpointed two arginine residues in the N-terminal 14-3-3 binding motif in TPK1 critical for kinase interaction. Taken together, our studies provide evidence for an essential role of the vacuolar potassium channel TPK1 in salt-stress adaptation as a target of calcium-regulated stress signaling pathways involving Ca(2+), Ca(2+)-dependent kinases, and 14-3-3 proteins.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/cytology , Arabidopsis/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Potassium Channels/metabolism , Salts/pharmacology , Stress, Physiological/drug effects , Vacuoles/metabolism , 14-3-3 Proteins/metabolism , Amino Acid Motifs , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/deficiency , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cytosol/metabolism , Gene Expression Regulation, Plant , Gene Knockout Techniques , Germination , Homeostasis , Models, Molecular , Mutation , Phosphorylation , Potassium/metabolism , Potassium Channels/chemistry , Potassium Channels/genetics , Protein Conformation , Protein Stability , Protein Transport , Signal Transduction
16.
Sci Rep ; 2: 695, 2012.
Article in English | MEDLINE | ID: mdl-23019516

ABSTRACT

The ROCO proteins are a family of large, multidomain proteins characterised by the presence of a Ras of complex proteins (ROC) domain followed by a COR, or C-terminal of ROC, domain. It has previously been shown that the ROC domain of the human ROCO protein Leucine Rich Repeat Kinase 2 (LRRK2) controls its kinase activity. Here, the ability of the ROC domain of another human ROCO protein, Death Associated Protein Kinase 1 (DAPK1), to bind GTP and control its kinase activity has been evaluated. In contrast to LRRK2, loss of GTP binding by DAPK1 does not result in loss of kinase activity, instead acting to modulate this activity. These data highlight the ROC domain of DAPK1 as a target for modifiers of this proteins function, and casts light on the role of ROC domains as intramolecular regulators in complex proteins with implications for a broad range of human diseases.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Guanosine Triphosphate/metabolism , Protein Interaction Domains and Motifs , Animals , Apoptosis Regulatory Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Line , Chlorobium/chemistry , Death-Associated Protein Kinases , Guanosine Triphosphate/chemistry , HEK293 Cells , Humans , Molecular Docking Simulation , Mutation , Protein Binding , Protein Conformation , Protein Transport
17.
Biochem Soc Trans ; 40(5): 1052-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22988864

ABSTRACT

DAPK (death-associated protein kinase) is a newly recognized member of the mammalian family of ROCO proteins, characterized by common ROC (Ras of complex proteins) and COR (C-terminal of ROC) domains. In the present paper, we review our recent work showing that DAPK is functionally a ROCO protein; its ROC domain binds and hydrolyses GTP. Furthermore, GTP binding regulates DAPK catalytic activity in a novel manner by enhancing autophosphorylation on inhibitory Ser308, thereby promoting the kinase 'off' state. This is a novel mechanism for in cis regulation of kinase activity by the distal ROC domain. The functional similarities between DAPK and the Parkinson's disease-associated protein LRRK2 (leucine-rich repeat protein kinase 2), another member of the ROCO family, are also discussed.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Guanosine Triphosphate/metabolism , Death-Associated Protein Kinases , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism
18.
Biochemistry ; 51(35): 6895-907, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22889004

ABSTRACT

A calcium/calmodulin-dependent protein kinase (CCaMK) is essential in the interpretation of calcium oscillations in plant root cells for the establishment of symbiotic relationships with rhizobia and mycorrhizal fungi. Some of its properties have been studied in detail, but its calcium ion binding properties and subsequent conformational change have not. A biophysical approach was taken with constructs comprising either the visinin-like domain of Medicago truncatula CCaMK, which contains EF-hand motifs, or this domain together with the autoinhibitory domain. The visinin-like domain binds three calcium ions, leading to a conformational change involving the exposure of hydrophobic surfaces and a change in tertiary but not net secondary or quaternary structure. The affinity for calcium ions of visinin-like domain EF-hands 1 and 2 (K(d) = 200 ± 50 nM) was appropriate for the interpretation of calcium oscillations (~125-850 nM), while that of EF-hand 3 (K(d) ≤ 20 nM) implied occupancy at basal calcium ion levels. Calcium dissociation rate constants were determined for the visinin-like domain of CCaMK, M. truncatula calmodulin 1, and the complex between these two proteins (the slowest of which was 0.123 ± 0.002 s(-1)), suggesting the corresponding calcium association rate constants were at or near the diffusion-limited rate. In addition, the dissociation of calmodulin from the protein complex was shown to be on the same time scale as the dissociation of calcium ions. These observations suggest that the formation and dissociation of the complex between calmodulin and CCaMK would substantially mirror calcium oscillations, which typically have a 90 s periodicity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium/metabolism , Medicago truncatula/enzymology , Amino Acid Sequence , Binding Sites , Calcium Signaling , Calmodulin/metabolism , EF Hand Motifs , Hydrophobic and Hydrophilic Interactions , Medicago truncatula/chemistry , Medicago truncatula/metabolism , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Structure, Tertiary
19.
PLoS One ; 7(5): e38209, 2012.
Article in English | MEDLINE | ID: mdl-22666486

ABSTRACT

Förster resonance energy transfer (FRET) microscopy is frequently used to study protein interactions and conformational changes in living cells. The utility of FRET is limited by false positive and negative signals. To overcome these limitations we have developed Fluorescence Polarization and Fluctuation Analysis (FPFA), a hybrid single-molecule based method combining time-resolved fluorescence anisotropy (homo-FRET) and fluorescence correlation spectroscopy. Using FPFA, homo-FRET (a 1-10 nm proximity gauge), brightness (a measure of the number of fluorescent subunits in a complex), and correlation time (an attribute sensitive to the mass and shape of a protein complex) can be simultaneously measured. These measurements together rigorously constrain the interpretation of FRET signals. Venus based control-constructs were used to validate FPFA. The utility of FPFA was demonstrated by measuring in living cells the number of subunits in the α-isoform of Venus-tagged calcium-calmodulin dependent protein kinase-II (CaMKIIα) holoenzyme. Brightness analysis revealed that the holoenzyme has, on average, 11.9 ± 1.2 subunit, but values ranged from 10-14 in individual cells. Homo-FRET analysis simultaneously detected that catalytic domains were arranged as dimers in the dodecameric holoenzyme, and this paired organization was confirmed by quantitative hetero-FRET analysis. In freshly prepared cell homogenates FPFA detected only 10.2 ± 1.3 subunits in the holoenzyme with values ranging from 9-12. Despite the reduction in subunit number, catalytic domains were still arranged as pairs in homogenates. Thus, FPFA suggests that while the absolute number of subunits in an auto-inhibited holoenzyme might vary from cell to cell, the organization of catalytic domains into pairs is preserved.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Fluorescence Polarization/methods , Hydrodynamics , Protein Subunits/chemistry , Protein Subunits/metabolism , Catalytic Domain , Cell Survival , HEK293 Cells , Holoenzymes/chemistry , Holoenzymes/metabolism , Humans , Reproducibility of Results
20.
J Biomol Struct Dyn ; 30(1): 45-61, 2012.
Article in English | MEDLINE | ID: mdl-22571432

ABSTRACT

We have studied the interactions between calmodulin (CaM) and three target peptides from the death-associated protein kinase (DAPK) protein family using both experimental and modeling methods, aimed at determining the details of the underlying biological regulation mechanisms. Experimentally, calorimetric binding free energies were determined for the complexes of CaM with peptides representing the DAPK2 wild-type and S308D mutant, as well as DAPK1. The observed affinity of CaM was very similar for all three studied peptides. The DAPK2 and DAPK1 peptides differ significantly in sequence and total charge, while the DAPK2 S308D mutant is designed to model the effects of DAPK2 Ser308 phosphorylation. The crystal structure of the CaM-DAPK2 S308D mutant peptide is also reported. The structures of CaM-DAPK peptide complexes present a mode of CaM-kinase interaction, in which bulky hydrophobic residues at positions 10 and 14 are both bound to the same hydrophobic cleft. To explain the microscopic effects underlying these interactions, we performed free energy calculations based on the approximate MM-PBSA approach. For these highly charged systems, standard MM-PBSA calculations did not yield satisfactory results. We proposed a rational modification of the approach which led to reasonable predictions of binding free energies. All three complexes are strongly stabilized by two effects: electrostatic interactions and buried surface area. The strong favorable interactions are to a large part compensated by unfavorable entropic terms, in which vibrational entropy is the largest contributor. The electrostatic component of the binding free energy followed the trend of the overall peptide charge, with strongest interactions for DAPK1 and weakest for the DAPK2 mutant. The electrostatics was dominated by interactions of the positively charged residues of the peptide with the negatively charged residues of CaM. The nonpolar binding free energy was comparable for all three peptides, the largest contribution coming from the Trp305. About two-thirds of the buried surface area corresponds to nonpolar residues, showing that hydrophobic interactions play an important role in these CaM-peptide complexes. The simulation results agree with the experimental data in predicting a small effect of the S308D mutation on CaM interactions with DAPK2, suggesting that this mutation is not a good model for the S308 phosphorylation.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calmodulin/chemistry , Models, Molecular , Amino Acid Sequence , Apoptosis Regulatory Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calmodulin/metabolism , Calorimetry , Computer Simulation , Death-Associated Protein Kinases , Molecular Sequence Data , Protein Binding , Protein Conformation , Static Electricity
SELECTION OF CITATIONS
SEARCH DETAIL
...