Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Proc Natl Acad Sci U S A ; 120(28): e2302143120, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37399380

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal and incurable neurodegenerative disease affecting motor neurons and characterized by microglia-mediated neurotoxic inflammation whose underlying mechanisms remain incompletely understood. In this work, we reveal that MAPK/MAK/MRK overlapping kinase (MOK), with an unknown physiological substrate, displays an immune function by controlling inflammatory and type-I interferon (IFN) responses in microglia which are detrimental to primary motor neurons. Moreover, we uncover the epigenetic reader bromodomain-containing protein 4 (Brd4) as an effector protein regulated by MOK, by promoting Ser492-phospho-Brd4 levels. We further demonstrate that MOK regulates Brd4 functions by supporting its binding to cytokine gene promoters, therefore enabling innate immune responses. Remarkably, we show that MOK levels are increased in the ALS spinal cord, particularly in microglial cells, and that administration of a chemical MOK inhibitor to ALS model mice can modulate Ser492-phospho-Brd4 levels, suppress microglial activation, and modify the disease course, indicating a pathophysiological role of MOK kinase in ALS and neuroinflammation.


Subject(s)
Amyotrophic Lateral Sclerosis , Bromodomain Containing Proteins , Mitogen-Activated Protein Kinases , Neurodegenerative Diseases , Animals , Mice , Amyotrophic Lateral Sclerosis/metabolism , Disease Models, Animal , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Bromodomain Containing Proteins/genetics , Bromodomain Containing Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism
2.
Mol Cell ; 47(3): 434-43, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22727666

ABSTRACT

Many critical protein kinases rely on the Hsp90 chaperone machinery for stability and function. After initially forming a ternary complex with kinase client and the cochaperone p50(Cdc37), Hsp90 proceeds through a cycle of conformational changes facilitated by ATP binding and hydrolysis. Progression through the chaperone cycle requires release of p50(Cdc37) and recruitment of the ATPase activating cochaperone AHA1, but the molecular regulation of this complex process at the cellular level is poorly understood. We demonstrate that a series of tyrosine phosphorylation events, involving both p50(Cdc37) and Hsp90, are minimally sufficient to provide directionality to the chaperone cycle. p50(Cdc37) phosphorylation on Y4 and Y298 disrupts client-p50(Cdc37) association, while Hsp90 phosphorylation on Y197 dissociates p50(Cdc37) from Hsp90. Hsp90 phosphorylation on Y313 promotes recruitment of AHA1, which stimulates Hsp90 ATPase activity, furthering the chaperoning process. Finally, at completion of the chaperone cycle, Hsp90 Y627 phosphorylation induces dissociation of the client and remaining cochaperones.


Subject(s)
Cell Cycle Proteins/metabolism , Chaperonins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Tyrosine/metabolism , Animals , COS Cells , Cell Cycle Proteins/genetics , Chaperonins/genetics , Chlorocebus aethiops , Humans , Mice , Molecular Chaperones/genetics , NIH 3T3 Cells , Phosphorylation/physiology
3.
Biochem J ; 475(15): 2559-2576, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30045873

ABSTRACT

Hsp90 is an ATP-dependent molecular chaperone that assists folding and conformational maturation/maintenance of many proteins. It is a potential cancer drug target because it chaperones oncoproteins. A prokaryotic homolog of Hsp90 (HtpG) is essential for thermo-tolerance in some bacteria and virulence of zoonotic pathogens. To identify a new class of small molecules which target prokaryotic and eukaryotic Hsp90s, we studied the effects of a naturally occurring cyclic sesquiterpene, zerumbone, which inhibits proliferation of a wide variety of tumor cells, on the activity of Hsp90. Zerumbone enhanced the ATPase activity of cyanobacterial Hsp90 (Hsp90SE), yeast Hsp90, and human Hsp90α. It also enhanced the catalytic efficiency of Hsp90SE by greatly increasing kcat Mass analysis showed that zerumbone binds to cysteine side chains of Hsp90SE covalently. Mutational studies identified 3 cysteine residues (one per each domain of Hsp90SE) that are involved in the enhancement, suggesting the presence of allosteric sites in the middle and C-terminal domains of Hsp90SE Treatment of cyanobacterial cells with zerumbone caused them to become very temperature-sensitive, a phenotype reminiscent of cyanobacterial Hsp90 mutants, and also decreased the cellular level of linker polypeptides that are clients for Hsp90SE Zerumbone showed cellular toxicity on cancer-derived mammalian cells by inducing apoptosis. In addition, zerumbone inhibited the binding of Hsp90/Cdc37 to client kinases. Altogether, we conclude that modification of cysteine residues of Hsp90 by zerumbone enhances its ATPase activity and inhibits physiological Hsp90 function. The activation of Hsp90 may provide new strategies to inhibit its chaperone function in cells.


Subject(s)
Adenosine Triphosphatases , Bacterial Proteins , Cyanobacteria/enzymology , Cytotoxins , HSP90 Heat-Shock Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/enzymology , Sesquiterpenes , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cysteine/chemistry , Cysteine/metabolism , Enzyme Stability/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology
4.
J Biol Chem ; 289(48): 33320-32, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25342745

ABSTRACT

Trp-Asp (WD) repeat protein 68 (WDR68) is an evolutionarily conserved WD40 repeat protein that binds to several proteins, including dual specificity tyrosine phosphorylation-regulated protein kinase (DYRK1A), MAPK/ERK kinase kinase 1 (MEKK1), and Cullin4-damage-specific DNA-binding protein 1 (CUL4-DDB1). WDR68 affects multiple and diverse physiological functions, such as controlling anthocyanin synthesis in plants, tissue growth in insects, and craniofacial development in vertebrates. However, the biochemical basis and the regulatory mechanism of WDR68 activity remain largely unknown. To better understand the cellular function of WDR68, here we have isolated and identified cellular WDR68 binding partners using a phosphoproteomic approach. More than 200 cellular proteins with wide varieties of biochemical functions were identified as WDR68-binding protein candidates. Eight T-complex protein 1 (TCP1) subunits comprising the molecular chaperone TCP1 ring complex/chaperonin-containing TCP1 (TRiC/CCT) were identified as major WDR68-binding proteins, and phosphorylation sites in both WDR68 and TRiC/CCT were identified. Co-immunoprecipitation experiments confirmed the binding between TRiC/CCT and WDR68. Computer-aided structural analysis suggested that WDR68 forms a seven-bladed ß-propeller ring. Experiments with a series of deletion mutants in combination with the structural modeling showed that three of the seven ß-propeller blades of WDR68 are essential and sufficient for TRiC/CCT binding. Knockdown of cellular TRiC/CCT by siRNA caused an abnormal WDR68 structure and led to reduction of its DYRK1A-binding activity. Concomitantly, nuclear accumulation of WDR68 was suppressed by the knockdown of TRiC/CCT, and WDR68 formed cellular aggregates when overexpressed in the TRiC/CCT-deficient cells. Altogether, our results demonstrate that the molecular chaperone TRiC/CCT is essential for correct protein folding, DYRK1A binding, and nuclear accumulation of WDR68.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/metabolism , Chaperonin Containing TCP-1/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Active Transport, Cell Nucleus/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , COS Cells , Cell Nucleus/genetics , Chaperonin Containing TCP-1/genetics , Chlorocebus aethiops , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/metabolism , Models, Molecular , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Structure, Secondary , Protein-Tyrosine Kinases/genetics , Structure-Activity Relationship , Dyrk Kinases
5.
Life Sci Alliance ; 6(12)2023 12.
Article in English | MEDLINE | ID: mdl-37802655

ABSTRACT

The protein kinase DYRK1A encoded in human chromosome 21 is the major contributor to the multiple symptoms observed in Down syndrome patients. In addition, DYRK1A malfunction is associated with various other neurodevelopmental disorders such as autism spectrum disorder. Here, we identified FAM53C with no hitherto known biological function as a novel suppressive binding partner of DYRK1A. FAM53C is bound to the catalytic protein kinase domain of DYRK1A, whereas DCAF7/WDR68, the major DYRK1A-binding protein, binds to the N-terminal domain of DYRK1A. The binding of FAM53C inhibited autophosphorylation activity of DYRK1A and its kinase activity to an exogenous substrate, MAPT/Tau. FAM53C did not bind directly to DCAF7/WDR68, whereas DYRK1A tethered FAM53C and DCAF7/WDR68 by binding concurrently to both of them, forming a tri-protein complex. DYRK1A possesses an NLS and accumulates in the nucleus when overexpressed in cells. Co-expression of FAM53C induced cytoplasmic re-localization of DYRK1A, revealing the cytoplasmic anchoring function of FAM53C to DYRK1A. Moreover, the binding of FAM53C to DYRK1A suppressed the DYRK1A-dependent nuclear localization of DCAF7/WDR68. All the results show that FAM53C binds to DYRK1A, suppresses its kinase activity, and anchors it in the cytoplasm. In addition, FAM53C is bound to the DYRK1A-related kinase DYRK1B with an Hsp90/Cdc37-independent manner. The results explain for the first time why endogenous DYRK1A is distributed in the cytoplasm in normal brain tissue. FAM53C-dependent regulation of the kinase activity and intracellular localization of DYRK1A may play a significant role in gene expression regulation caused by normal and aberrant levels of DYRK1A.


Subject(s)
Brain , Carrier Proteins , Protein Kinases , Humans , Autism Spectrum Disorder/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation , Phosphorylation , Transcription Factors , Down Syndrome/metabolism , Brain/metabolism , Protein Kinases/metabolism , Dyrk Kinases
6.
Biochim Biophys Acta ; 1813(10): 1728-39, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21777625

ABSTRACT

DYRK1A is encoded in the Down's syndrome critical region on human chromosome 21, and plays an important role in the functional and developmental regulation of many types of cells, including neuronal cells. Here we have identified WDR68, an evolutionarily conserved protein with WD40-repeat domains, as a cellular binding partner of DYRK1A. WDR68 was originally identified in petunia as AN11 that controls the pigmentation of flowers by stimulating the transcription of anthocyanin biosynthetic genes. Experiments with RNA interference showed that WDR68 was indispensable for the optimal proliferation and survival of mammalian cultured cell, and WDR68 depletion induced cell apoptosis. DYRK1A and DYRK1B, but not DYRK2, DYRK3, or DYRK4, bound to endogenous and expressed WDR68. The N-terminal domain, but not the catalytic kinase domain or the C-terminal domain of DYRK1A, was responsible for the WDR68 binding. Deletions in the N-terminal or C-terminal region outside of the central WD40-repeats of WDR68 abolished its binding to DYRK1A, suggesting that WD40 repeats are not sufficient for the association with DYRK1A. Immunofluorescent staining revealed that WDR68 was distributed throughout the cell. Importantly, nuclear accumulation of WDR68 was observed upon co-expression of the wild type and a kinase-dead mutant of DYRK1A. Taken together, these results suggest that DYRK1A binds specifically to WDR68 in cells, and that the binding, but not the phosphorylation event, induces the nuclear translocation of WDR68.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/physiology , Active Transport, Cell Nucleus/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cells, Cultured , Chlorocebus aethiops , Conserved Sequence/genetics , Evolution, Molecular , Humans , Molecular Sequence Data , Protein Binding/genetics , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary/genetics , Protein Transport/genetics , Protein Transport/physiology , Protein-Tyrosine Kinases/genetics , Transfection , Dyrk Kinases
7.
Mol Cell Biochem ; 356(1-2): 191-200, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21739154

ABSTRACT

The nuclear localization signal sequence (NLS) of SV40 Large T antigen is essential and sufficient for the nuclear translocation of the protein. Phosphorylation often modulates the intracellular distribution of signaling proteins. In this study, we investigated effects of the NLS-peptide of Large T antigen on protein phosphorylation. When crude cell lysates were incubated with [γ-(32)P]ATP, phosphorylation of several endogenous substrates with molecular masses of 100, 80, 50, and 45 kDa by an endogenous kinase was stimulated by the addition of the wild type NLS-peptide (CPKKKRKVEDP). The mutated NLS-peptide (CPKTKRKVEDP) and the reversed NLS-peptide (PDEVKRKKKPC) are weak in the nuclear localization activity, and they only weakly stimulated phosphorylation of these substrates. The mobility of the 100 kDa phosphoprotein was indistinguishable with that of an endoplasmic reticulum (ER)-resident molecular chaperone glucose-regulated protein 94 (Grp94) belonging to the Hsp90 family, and purified Grp94 was phosphorylated by a kinase in cell lysates in an NLS-dependent fashion. The 100 kDa protein was identified as Grp94 by immunoprecipitation and reconstitution experiments. Purification of the NLS-dependent Grp94 kinase by sequential biochemical column chromatography steps resulted in isolation of two polypeptides with molecular masses of 42 and 27 kDa, which were identified as α and ß subunit of protein kinase CK2, respectively, by western blotting analysis and biochemical characterization. Moreover, effect of an excess amount of GTP and V8 peptide mapping showed that the NLS-dependent Grp94 kinase in the cell lysate is identical with CK2. Surprisingly purified CK2 did phosphorylate Grp94 even without the NLS-peptide, suggesting that an additional suppressive factor is required for NLS-dependent phosphorylation of Grp94 by CK2. We suggest a possible general role for CK2-catalyzed phosphorylation in the regulation of NLS-dependent protein nuclear translocation.


Subject(s)
Casein Kinase II/metabolism , HSP70 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Nuclear Localization Signals/metabolism , Amino Acid Sequence , Animals , Cell Extracts , Chromatography , HSP90 Heat-Shock Proteins/metabolism , Humans , L Cells , Mice , Molecular Sequence Data , Molecular Weight , Nuclear Localization Signals/chemistry , Phosphorylation , Substrate Specificity
8.
Biochim Biophys Acta Mol Cell Res ; 1868(10): 119081, 2021 09.
Article in English | MEDLINE | ID: mdl-34147560

ABSTRACT

The DYRK (Dual-specificity tYrosine-phosphorylation Regulated protein Kinase) family consists of five related protein kinases (DYRK1A, DYRK1B, DYRK2, DYRK3, DYRK4). DYRKs show homology to Drosophila Minibrain, and DYRK1A in human chromosome 21 is responsible for various neuronal disorders including human Down syndrome. Here we report identification of cellular proteins that associate with specific members of DYRKs. Cellular proteins with molecular masses of 90, 70, and 50-kDa associated with DYRK1B and DYRK4. These proteins were identified as molecular chaperones Hsp90, Hsp70, and Cdc37, respectively. Microscopic analysis of GFP-DYRKs showed that DYRK1A and DYRK1B were nuclear, while DYRK2, DYRK3, and DYRK4 were mostly cytoplasmic in COS7 cells. Overexpression of DYRK1B induced nuclear re-localization of these chaperones with DYRK1B. Treatment of cells with specific Hsp90 inhibitors, geldanamycin and 17-AAG, abolished the association of Hsp90 and Cdc37 with DYRK1B and DYRK4, but not of Hsp70. Inhibition of Hsp90 chaperone activity affected intracellular dynamics of DYRK1B and DYRK4. DYRK1B and DYRK4 underwent rapid formation of cytoplasmic punctate dots after the geldanamycin treatment, suggesting that the chaperone function of Hsp90 is required for prevention of protein aggregation of the target kinases. Prolonged inhibition of Hsp90 by geldanamycin, 17-AAG, or ganetespib, decreased cellular levels of DYRK1B and DYRK4. Finally, DYRK1B and DYRK4 were ubiquitinated in cells, and ubiquitinated DYRK1B and DYRK4 further increased by Hsp90 inhibition with geldanamycin. Taken together, these results indicate that Hsp90 and Cdc37 discriminate specific members of the DYRK kinase family and play an important role in quality control of these client kinases in cells.


Subject(s)
Cell Cycle Proteins/metabolism , Chaperonins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Benzoquinones/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lactams, Macrocyclic/pharmacology , Dyrk Kinases
9.
J Biochem ; 170(2): 255-264, 2021 Oct 11.
Article in English | MEDLINE | ID: mdl-33768253

ABSTRACT

Heat shock protein 90 (Hsp90) is essential for eukaryotic cells, whereas bacterial homologs play a role under stresses and in pathogenesis. Identifying species-specific Hsp90 inhibitors is challenging because Hsp90 is evolutionarily conserved. We found that a cyclic lipopeptide surfactin inhibits the ATPase activity of Hsp90 from the cyanobacterium Synechococcus elongatus (S.elongatus) PCC 7942 but does not inhibit Escherichia coli (E.coli), yeast and human Hsp90s. Molecular docking simulations indicated that surfactin could bind to the N-terminal dimerization interface of the cyanobacterial Hsp90 in the ATP- and ADP-bound states, which provided molecular insights into the species-selective inhibition. The data suggest that surfactin inhibits a rate-limiting conformational change of S.elongatus Hsp90 in the ATP hydrolysis. Surfactin also inhibited the interaction of the cyanobacterial Hsp90 with a model substrate, and suppressed S.elongatus growth under heat stress, but not that of E.coli. Surfactin did not show significant cellular toxicity towards mammalian cells. These results indicate that surfactin inhibits the cellular function of Hsp90 specifically in the cyanobacterium. The present study shows that a cyclic peptide has a great specificity to interact with a specific homolog of a highly conserved protein family.


Subject(s)
Anti-Bacterial Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lipopeptides/pharmacology , Peptides, Cyclic/pharmacology , Synechococcus/drug effects , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Animals , Anti-Bacterial Agents/metabolism , COS Cells , Chlorocebus aethiops , Colistin/pharmacology , Dimerization , Escherichia coli/drug effects , HSP90 Heat-Shock Proteins/metabolism , Humans , Hydrolysis , Lipopeptides/metabolism , Mice , Molecular Docking Simulation/methods , NIH 3T3 Cells , Peptides, Cyclic/metabolism , Saccharomyces cerevisiae/drug effects
10.
Appl Microbiol Biotechnol ; 87(6): 2147-56, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20496148

ABSTRACT

Baculovirus expression systems (BES) are widely used for recombinant protein production in lepidopteran cells or larvae. However, even in BES, the insolubility of recombinant proteins sometimes makes their expression difficult. In this study, to improve the solubility and yield of foreign proteins, we constructed transgenic silkworms using silkworm heat-shock proteins, Hsp70 and Hsp40, or Hsc70 and Hsp90 co-chaperone Hop. In these transgenic silkworms, the expression levels of the transgenes were under the control of a UAS.hsp mini-promoter driven by a Gal4NFkBp65 activator. When the transgenic silkworm with HSP70 and 40 (TGS-HSP70/40) was infected with BmNPV carrying mC3d and Gal4NFkBp65 under the control of baculovirus polyhedrin or p10 promoters, respectively, the soluble fraction of the His- or His.GST-tagged mC3d increased significantly. Similarly, the transgenic silkworm with HSC70 and HOP (TGS-HOP7) was effective for the expression of a steroid hormone receptor, USP2. In conclusion, the His-tagged baculovirus expression system featuring the chaperone effect TGS-HSP70/40 and TGS-HOP7 silkworms is effective for increasing the yields of soluble and functional foreign gene products.


Subject(s)
Animals, Genetically Modified/genetics , Bombyx/genetics , Cytoplasm/metabolism , Gene Expression , Heat-Shock Proteins/genetics , Insect Proteins/genetics , Recombinant Proteins/metabolism , Animals , Animals, Genetically Modified/metabolism , Animals, Genetically Modified/virology , Baculoviridae/genetics , Baculoviridae/physiology , Bombyx/metabolism , Bombyx/virology , Complement C3d/chemistry , Complement C3d/genetics , Complement C3d/metabolism , Cytoplasm/genetics , Genetic Vectors/genetics , Genetic Vectors/physiology , Heat-Shock Proteins/metabolism , Insect Proteins/chemistry , Insect Proteins/metabolism , Mice , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Solubility
11.
Biochim Biophys Acta Proteins Proteom ; 1867(9): 813-820, 2019 09.
Article in English | MEDLINE | ID: mdl-31226489

ABSTRACT

Cdc37 is a protein kinase-targeting molecular chaperone, which cooperates with Hsp90 to assist the folding, assembly and maturation of various signaling kinases. It consists of three distinct domains: the N-terminal, middle, and C-terminal domain. While the middle domain is an Hsp90-binding domain, the N-terminal domain is recognized as a kinase-interacting domain. The N-terminal domain contains a well-conserved Ser residue at position 13, and the phosphorylation at this site has been shown to be a prerequisite for the interaction between Cdc37 and signaling kinases. Although the phosphorylation of Ser13 might induce some conformational change in Cdc37 molecule, little is known about the structure of the N-terminal domain of Cdc37. We examined the structural and dynamic properties of several fragment proteins corresponding to the N-terminal region of Cdc37 by circular dichroism and solution NMR spectroscopy. We found that the N-terminal domain of Cdc37 exhibits highly dynamic structure, and it exists in the equilibrium between α-helical and more disordered structures. We also found that phosphorylation at Ser13 did not significantly change the overall structure of N-terminal fragment protein of Cdc37. The results suggested that more complicated mechanisms might be necessary to explain the phosphorylation-activated interaction of Cdc37 with various kinases.


Subject(s)
Cell Cycle Proteins/chemistry , Chaperonins/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chaperonins/genetics , Chaperonins/metabolism , Circular Dichroism , Humans , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Protein Domains , Protein Structure, Secondary
12.
FEBS J ; 274(21): 5690-703, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17922836

ABSTRACT

The CK2-dependent phosphorylation of Ser13 in cell division cycle protein 37 (Cdc37), a kinase-specific heat shock protein 90 (Hsp90) cochaperone, has previously been reported to be essential for the association of Cdc37 with signaling protein kinases [Bandhakavi S, McCann RO, Hanna DE & Glover CVC (2003) J Biol Chem278, 2829-2836; Shao J, Prince T, Hartson SD & Matts RL (2003) J Biol Chem278, 38117-38220; Miyata Y & Nishida E (2004) Mol Cell Biol24, 4065-4074]. Here we describe a new phospho-specific antibody against Cdc37 that recognizes recombinant purified Cdc37 only when incubated with CK2 in the presence of Mg(2+) and ATP. The replacement of Ser13 in Cdc37 by nonphosphorylatable amino acids abolished binding to this antibody. The antibody was specific for phosphorylated Cdc37 and did not crossreact with other CK2 substrates such as Hsp90 and FK506-binding protein 52. Using this antibody, we showed that complexes of Hsp90 with its client signaling kinases, Cdk4, MOK, v-Src, and Raf1, contained the CK2-phosphorylated form of Cdc37 in vivo. Immunofluorescent staining showed that Hsp90 and the phosphorylated form of Cdc37 accumulated in epidermal growth factor-induced membrane ruffles. We further characterized the phosphorylation of Cdc37 using phospho-affinity gel electrophoresis. Our analyses demonstrated that the CK2-dependent phosphorylation of Cdc37 on Ser13 caused a specific gel mobility shift, and that Cdc37 in the complexes between Hsp90 and its client signaling protein kinases was in the phosphorylated form. Our results show the physiological importance of CK2-dependent Cdc37 phosphorylation and the usefulness of phospho-affinity gel electrophoresis in protein phosphorylation analysis.


Subject(s)
Antibodies, Phospho-Specific , Carrier Proteins/immunology , Carrier Proteins/metabolism , Casein Kinase II/metabolism , Cell Cycle Proteins/immunology , Cell Cycle Proteins/metabolism , Serine/metabolism , Amino Acid Sequence , Animals , COS Cells , Carrier Proteins/analysis , Cell Cycle Proteins/analysis , Chlorocebus aethiops , Chromatography, Affinity , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Humans , Molecular Sequence Data , Phosphorylation , Serine/genetics , Signal Transduction , Tumor Cells, Cultured
13.
Mol Cell Biol ; 24(9): 4065-74, 2004 May.
Article in English | MEDLINE | ID: mdl-15082798

ABSTRACT

Cdc37 is a kinase-associated molecular chaperone whose function in concert with Hsp90 is essential for many signaling protein kinases. Here, we report that mammalian Cdc37 is a pivotal substrate of CK2 (casein kinase II). Purified Cdc37 was phosphorylated in vitro on a conserved serine residue, Ser13, by CK2. Moreover, Ser13 was the unique phosphorylation site of Cdc37 in vivo. Crucially, the CK2 phosphorylation of Cdc37 on Ser13 was essential for the optimal binding activity of Cdc37 toward various kinases examined, including Raf1, Akt, Aurora-B, Cdk4, Src, MOK, MAK, and MRK. In addition, nonphosphorylatable mutants of Cdc37 significantly suppressed the association of Hsp90 with protein kinases, while the Hsp90-binding activity of the mutants was unchanged. The treatment of cells with a specific CK2 inhibitor suppressed the phosphorylation of Cdc37 in vivo and reduced the levels of Cdc37 target kinases. These results unveil a regulatory mechanism of Cdc37, identify a novel molecular link between CK2 and many crucial protein kinases via Cdc37, and reveal the molecular basis for the ability of CK2 to regulate pleiotropic cellular functions.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Molecular Chaperones/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Carrier Proteins , Casein Kinase II , Cell Cycle Proteins/genetics , Chaperonins , Drosophila Proteins/genetics , Humans , Male , Molecular Chaperones/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Rats , Sequence Alignment , Serine/metabolism , Swine
14.
Cancer Lett ; 356(2 Pt B): 751-61, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25449433

ABSTRACT

Screening for protein kinase CK2 inhibitors of the structural diversity compound library (DTP NCI/NIH) led to the discovery of 4-[(E)-(fluoren-9-ylidenehydrazinylidene)-methyl]benzoic acid (E9). E9 induces apoptotic cell death in various cancer cell lines and upon hypoxia, the compound suppresses CK2-catalyzed HSP90/Cdc37 phosphorylation and induces HIF-1α degradation. Furthermore, E9 exerts a strong anti-tumour activity by inducing necrosis in murine xenograft models underlining its potential to be used for cancer treatment in future clinical studies. Crystal structure analysis of human and maize CK2α in complex with E9 reveals unique binding properties of the inhibitor to the enzyme, accounting for its affinity and selectivity.


Subject(s)
Benzoates/pharmacology , Casein Kinase II/antagonists & inhibitors , Cell Proliferation/drug effects , Fluorenes/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Neoplasms/drug therapy , Protein Conformation/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Animals , Blotting, Western , Cell Cycle/drug effects , Combinatorial Chemistry Techniques , Female , Fluorescent Antibody Technique , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoenzyme Techniques , Luciferases/metabolism , Mice , Mice, Nude , Neoplasms/enzymology , Neoplasms/pathology , Phosphorylation/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Ann N Y Acad Sci ; 1030: 150-7, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15659792

ABSTRACT

Overexpression of a pleiotropic Ser/Thr kinase CK2 (casein kinase II), or of a kinase-targeting molecular chaperone Cdc37, induces neoplastic cell growth in animals. Recent genetic and biochemical evidence from several laboratories has revealed an unexpected direct link between CK2 and Cdc37. In this short review, we describe the basic characteristics of CK2 and Cdc37 and introduce recent findings on the interaction between CK2 and Cdc37. Cdc37 was identified as a multicopy suppressor of a temperature-sensitive allele of CK2 in Saccharomyces cerevisiae. CK2 phosphorylates a conserved serine residue in the N-terminal extremity of Cdc37 in vitro and in yeast as well as mammalian cells, and this is the unique phosphorylation site of Cdc37 under normal conditions. Mutations in the CK2-mediated phosphorylation site abolish the association of Cdc37 with various protein kinases. The same mutations in yeast cause severe growth and morphological defects. Specific inhibition of CK2 activity decreases intracellular levels of Cdc37-dependent protein kinases. Altogether, this evidence clearly indicates that the CK2-dependent phosphorylation is essential for the proper function of Cdc37 to bind and stabilize signaling protein kinases. In contrast, CK2 activity is enhanced by Cdc37 both in vitro and in vivo; thus, the CK2-Cdc37 couple seems to constitute a positive feedback control mechanism that may govern the activity of multiple protein kinases. Cdc37-dependent protein kinases include important signaling molecules whose disregulations are intimately related to neoplastic cell growth; hence, inhibition of the CK2-Cdc37 system may simultaneously suppress various cancer-promoting signal cascades. We propose that the CK2-Cdc37 couple can be a novel and efficient pharmacological target for cancer chemotherapy.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Kinases/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Chaperonins , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Sequence Data , Phosphorylation , Sequence Homology, Amino Acid
16.
Masui ; 53(11): 1234-42, 2004 Nov.
Article in Japanese | MEDLINE | ID: mdl-15587172

ABSTRACT

BACKGROUND: Upper airway obstruction and inadequate ventilation often arise during sedation and anesthesia by propofol. To estimate the influence of propofol (PP) on respiratory control, we studied its effect on the neural activity and the respiratory response caused by a brief (60 sec) respiratory arrest (RA) manifesting in the hypoglossal nerve (HG) and the phrenic nerve (PN) in rabbits. METHODS: Experiments were performed on adult rabbits vagotomized, paralyzed and ventilated artificially with 50% N2O, 50% oxygen and 0.5% sevoflurane. We evaluated and compared the effects of PP on the peak amplitude (AMP) and the root mean square (RMS) of HG and PH, and respiratory cycle (Tc). RESULTS: PP depressed HG activity more than PH activity, and increased Tc in a dose related manner, with 0.25 mg x kg(-1) x min(-1) continuous infusion, propofol soon began to reduce both AMPs without any remarkable changing in Tc. AMP&RMS-HG were reduced to about 35% and AMP&RMS-PN to 80% of control. Administration of propofol 1.5 mg x kg(-1) x min(-1) vanished the activity of HG in all animals. RA made a mixed hypercapnic and hypoxic condition and induced RA response which was characterized by raised AMPs, augmented RMSs (deltaAMPs, deltaRMSs) in activity of both nerves activity and lengthened Tc (deltaTc). PP depressed RA response in HG dose-dependently, but did not do so in PN. Significant depressions in cardiovascular effects with tested dosage of PP occurred, but the values were kept in physiological ranges. CONCLUSIONS: These results suggest that propofol induces respiratory depression by its inhibitory effect on the neural regulation of respiration, especially on the maintenance system of upper airway patency and the reflex related to the chemosensitive upper airway patency control.


Subject(s)
Anesthetics, Intravenous/pharmacology , Hypercapnia/physiopathology , Hypoglossal Nerve/physiology , Hypoxia/physiopathology , Phrenic Nerve/physiology , Propofol/pharmacology , Animals , Male , Rabbits , Respiration
17.
Masui ; 52(12): 1286-92, 2003 Dec.
Article in Japanese | MEDLINE | ID: mdl-14733078

ABSTRACT

BACKGROUND: Upper airway obstruction and inadequate ventilation often arise during sedation and anesthesia by benzodiazepines. To estimate the influence of benzodiazepines on the respiratory control, we studied the effect of diazepam and flumazenil on the neural activity and the respiratory response caused by a brief (60 sec) respiratory arrest (RA) observed in the hypoglossal nerve (HG) and phrenic nerve (PH) in rabbits. METHODS: Experiments were preformed on adult rabbits vagotomized, paralyzed and ventilated artificially with 50% N2O, 50% oxygen and 0.3-0.5% sevoflurane. We evaluated and compared the effects of diazepam and flumazenil on the peak amplitude (AMP-HG&PH) and the root mean square (RMS-HG&PH) of HG and PH, and respiratory cycle (Tc). RESULTS: Diazepam depressed HG activity more than PH activity with no influence on Tc. But it did not cause dose-related depression. Flumazenil 0.2 mg.kg-1 completely reversed the respiratory depressions caused by diazepam with the increased Tc. In addition to augmentation of the hypoglossal activity in inspiration, flumazenil caused a rise in its activity in pan-expiratory period in some cases. Additional administration of diazepam 6 mg.kg-1 following flumazenil depressed PH activity again, but did not affect HG activity any more. There was no significant depression in cardiovascular parameters with tested dosages of diazepam and flumazenil. RA response was characterized by raised AMPs and augmented RMSs (delta AMPs, delta RMSs) with marked prolongation in Tc (delta Tc). Diazepam depressed RA response dose dependently, but flumazenil did not seem to antagonize this depression. CONCLUSIONS: These results suggest that 1) flumazenil is not only a specific antagonist of benzodiazepines but also a potential excitatory agent of hypoglossal nerve activity, and that 2) there is some functional diversity in disposition of benzodiazepine-receptor binding GABAA-receptor responsible for neural respiratory control system.


Subject(s)
Diazepam/pharmacology , Flumazenil/pharmacology , Hypoglossal Nerve/drug effects , Phrenic Nerve/drug effects , Respiration/drug effects , Action Potentials/drug effects , Animals , Diazepam/adverse effects , Diazepam/antagonists & inhibitors , Rabbits , Receptors, GABA-A/physiology
18.
Masui ; 53(7): 753-60, 2004 Jul.
Article in Japanese | MEDLINE | ID: mdl-15298241

ABSTRACT

BACKGROUND: Upper airway obstruction and inadequate ventilation often arise during sedation and anesthesia by benzodiazepines (Bz). Flumazenil antagonizes these effects of active benzodiazepines on the central nervous system. To estimate the influence of flumazenil on the endogenous Bz system related respiratory control, we studied the effect of flumazenil and diazepam on the neural activity and the respiratory response caused by a brief (60 sec) respiratory arrest (RA) manifested in the hypoglossal nerve (HG) and the phrenic nerve (PH) activities in rabbits. METHODS: Experiments were performed on adult rabbits which were vagotomized, paralyzed and artificially ventilated with 50% N2O, 50% oxygen and 0.5% sevoflurane. We evaluated and compared the effects of the sequential administrations of flumazenil and diazepam on the peak amplitude (AMP) as well as the root mean square (RMS) of HG and PH, and respiratory cycle (Tc). RESULTS: Flumazenil by itself increased HG activity more than PH activity with no influence on Tc. But it was not dose-related. Previous administration of flumazenil in total dose of 0.25 mg x kg(-1) could not prevent the anticipated respiratory depression caused by diazepam 2.0 mg x kg(-1). These depressions are greater in HG activity than in PH activity. Additional flumazenil 0.15 mg x kg(-1) following the administration of diazepam promptly reversed these inhibitory effects on HG activity beyond the control level. The same dose of flumazenil, however, did not reverse PH activity sufficiently. RA response was characterized by raised AMPs and augmented RMSs (deltaAMPs, deltaRMSs) with marked prolongation in Tc (deltaTc). Flumazenil and diazepam did not seem to have any influence upon these RA responses. There was a significant change in cardiovascular parameters with the tested dosages of flumazenil and diazepam, but the change was in the normal physiological range. CONCLUSIONS: These results suggest the possibility that the endogenous benzodiazepine system is likely to play an inhibitory role in the regulation of respiration, especially in the maintenance of upper airway patency but the system is unrelated to the chemosensitive-respiratory control.


Subject(s)
Flumazenil/pharmacology , Hypoglossal Nerve/drug effects , Phrenic Nerve/drug effects , Respiration/drug effects , Action Potentials/drug effects , Animals , Benzodiazepines/antagonists & inhibitors , Diazepam/pharmacology , Male , Rabbits
19.
Masui ; 52(2): 135-42, 2003 Feb.
Article in Japanese | MEDLINE | ID: mdl-12649867

ABSTRACT

BACKGROUND: Respiratory depression, such as obstruction of upper airway and inadequate ventilation, often appears during sedation and anesthesia. We studied the effect of propofol on the upper airway patency and the inspiratory drives. METHODS: Experiments were performed on the adult rabbits vagotomized, paralyzed and ventilated with nitrous oxide-oxygen-sevoflurane. We evaluated and compared depressant effect of propofol on the peak amplitude of both hypoglossal nerve (AMP-HG) and phrenic nerve (AMP-PH) activities, inspiratory time (Ti), expiratory time (Te) and respiratory cycle (Tc). RESULTS: Bolus injections of propofol transiently reduced AMP-HG more than AMP-PH (18 and 70% of control, respectively). But AMPs returned to the control levels about 10-15 min after the injection. For 0.5 mg.kg-1.min-1 continuous infusion, propofol soon began to reduce both AMPs. AMP-HG was reduced to about 20% and AMP-PH to 60% of control. Administration of propofol with 1.0 mg.kg-1.min-1 caused more reduction in AMPs with respiratory slowing and AMP-HG disappeared in some animals. CONCLUSION: During the sedation with low dose of propofol, we need to pay attention to potential upper airway obstruction. In addition to the above, high doses of propofol could reduce spontaneous inspiratory drive, and we need to keep both upper airway patency and sufficient ventilation.


Subject(s)
Anesthetics, Intravenous/pharmacology , Hypoglossal Nerve/drug effects , Phrenic Nerve/drug effects , Propofol/pharmacology , Animals , Depression, Chemical , Rabbits , Respiration/drug effects
20.
Curr Pharm Des ; 19(3): 347-65, 2013.
Article in English | MEDLINE | ID: mdl-22920906

ABSTRACT

Hsp90 is a major molecular chaperone that is expressed abundantly and plays a pivotal role in assisting correct folding and functionality of its client proteins in cells. The Hsp90 client proteins include a wide variety of signal transducing molecules such as protein kinases and steroid hormone receptors. Cancer is a complex disease, but most types of human cancer share common hallmarks, including self-sufficiency in growth signals, insensitivity to growth-inhibitory mechanism, evasion of programmed cell death, limitless replicative potential, sustained angiogenesis, and tissue invasion and metastasis. A surprisingly large number of Hsp90-client proteins play crucial roles in establishing cancer cell hallmarks. We start the review by describing the structure and function of Hsp90 since conformational changes during the ATPase cycle of Hsp90 are closely related to its function. Many co-chaperones, including Hop, p23, Cdc37, Aha1, and PP5, work together with Hsp90 by modulating the chaperone machinery. Post-translational modifications of Hsp90 and its cochaperones are vital for their function. Many tumor-related Hsp90-client proteins, including signaling kinases, steroid hormone receptors, p53, and telomerase, are described. Hsp90 and its co-chaperones are required for the function of these tumor-promoting client proteins; therefore, inhibition of Hsp90 by specific inhibitors such as geldanamycin and its derivatives attenuates the tumor progression. Hsp90 inhibitors can be potential and effective cancer chemotherapeutic drugs with a unique profile and have been examined in clinical trials. We describe possible mechanisms why Hsp90 inhibitors show selectivity to cancer cells even though Hsp90 is essential also for normal cells. Finally, we discuss the "Hsp90-addiction" of cancer cells, and suggest a role for Hsp90 in tumor evolution.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Drug Delivery Systems/methods , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Drug Delivery Systems/trends , Humans , Protein Binding/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL