515 results on '"Turk, Benjamin E."'
Search Results
152. Specific and high affinity peptide‐based inhibitors of the proto‐oncogenic Pim kinases
- Author
-
Wubah, Vera, primary and Turk, Benjamin E, additional
- Published
- 2006
- Full Text
- View/download PDF
153. Measuring kinase activity: finding needles in a haystack
- Author
-
Turk, Benjamin E, primary
- Published
- 2005
- Full Text
- View/download PDF
154. A rapid method for determining protein kinase phosphorylation specificity
- Author
-
Hutti, Jessica E, primary, Jarrell, Emily T, additional, Chang, James D, additional, Abbott, Derek W, additional, Storz, Peter, additional, Toker, Alex, additional, Cantley, Lewis C, additional, and Turk, Benjamin E, additional
- Published
- 2004
- Full Text
- View/download PDF
155. The structural basis for substrate and inhibitor selectivity of the anthrax lethal factor
- Author
-
Turk, Benjamin E, primary, Wong, Thiang Yian, additional, Schwarzenbacher, Robert, additional, Jarrell, Emily T, additional, Leppla, Stephen H, additional, Collier, R John, additional, Liddington, Robert C, additional, and Cantley, Lewis C, additional
- Published
- 2003
- Full Text
- View/download PDF
156. Identification of small molecule inhibitors of anthrax lethal factor
- Author
-
Panchal, Rekha G, primary, Hermone, Ann R, additional, Nguyen, Tam Luong, additional, Wong, Thiang Yian, additional, Schwarzenbacher, Robert, additional, Schmidt, James, additional, Lane, Douglas, additional, McGrath, Connor, additional, Turk, Benjamin E, additional, Burnett, James, additional, Aman, M Javad, additional, Little, Stephen, additional, Sausville, Edward A, additional, Zaharevitz, Daniel W, additional, Cantley, Lewis C, additional, Liddington, Robert C, additional, Gussio, Rick, additional, and Bavari, Sina, additional
- Published
- 2003
- Full Text
- View/download PDF
157. Mixture-Based Peptide Libraries for Identifying Protease Cleavage Motifs.
- Author
-
Turk, Benjamin E.
- Published
- 2009
- Full Text
- View/download PDF
158. Determination of protease cleavage site motifs using mixture-based oriented peptide libraries
- Author
-
Turk, Benjamin E., primary, Huang, Lisa L., additional, Piro, Elizabeth T., additional, and Cantley, Lewis C., additional
- Published
- 2001
- Full Text
- View/download PDF
159. Global Analysis of Human Nonreceptor Tyrosine Kinase Specificity Using High-Density Peptide Microarrays.
- Author
-
Yang Deng, Alicea-Velázquez, Nilda L., Bannwarth, Ludovic, Lehtonen, Soili I., Boggon, Titus J., Heung-Chin Cheng, Hytönen, Vesa P., and Turk, Benjamin E.
- Published
- 2014
- Full Text
- View/download PDF
160. Identification of Cleavage Sites Leading to the Shed Form of the Anti-Aging Protein Klotho.
- Author
-
Ci-Di Chen, Tze Yu Tung, Jennifer Liang, Zeldich, Elia, Tucker Zhou, Tracey B., Turk, Benjamin E., and Abraham, Carmela R.
- Published
- 2014
- Full Text
- View/download PDF
161. Selective inhibition of amino-terminal methionine processing by TNP-470 and ovalicin in endothelial cells
- Author
-
Turk, Benjamin E, primary, Griffith, Eric C, additional, Wolf, Susan, additional, Biemann, Klaus, additional, Chang, Yie-Hwa, additional, and Liu, Jun O, additional
- Published
- 1999
- Full Text
- View/download PDF
162. Synthetic analogues of TNP-470 and ovalicin reveal a common molecular basis for inhibition of angiogenesis and immunosuppression1In honor of Professor Stuart Schreiber for his pioneering contributions to the interface between chemistry and biology.1
- Author
-
Turk, Benjamin E., primary, Su, Zhuang, additional, and Liu, Jun O., additional
- Published
- 1998
- Full Text
- View/download PDF
163. Enhanced potency of perfluorinated thalidomide derivatives for inhibition of LPS-induced tumor necrosis factor-α production is associated with a change of mechanism of action
- Author
-
Niwayama, Satomi, primary, Loh, Christine, additional, Turk, Benjamin E., additional, Liu, Jun O., additional, Miyachi, Hiroyuki, additional, and Hashimoto, Yuichi, additional
- Published
- 1998
- Full Text
- View/download PDF
164. Methionine aminopeptidase (type 2) is the common target for angiogenesis inhibitors AGM-1470 and ovalicin
- Author
-
Griffith, Eric C., primary, Su, Zhuang, additional, Turk, Benjamin E., additional, Chen, Shaoping, additional, Chang, Yie-Hwa, additional, Wu, Zhuchun, additional, Biemann, Klaus, additional, and Liu, Jun O., additional
- Published
- 1997
- Full Text
- View/download PDF
165. Potent Inhibition of Tumor Necrosis Factor-α Production by Tetrafluorothalidomide and Tetrafluorophthalimides
- Author
-
Niwayama, Satomi, primary, Turk, Benjamin E., additional, and Liu, Jun O., additional
- Published
- 1996
- Full Text
- View/download PDF
166. Reciprocal Phosphorylation of Yeast Glycerol-3-Phosphate Dehydrogenases in Adaptation to Distinct Types of Stress.
- Author
-
Yong Jae Lee, Jeschke, Grace R., Roelants, Françoise M., Thorner, Jeremy, and Turk, Benjamin E.
- Subjects
PHOSPHORYLATION ,EUKARYOTIC cells ,PROTEIN kinases ,DEHYDROGENASES ,GLYCERIN ,SACCHAROMYCES cerevisiae - Abstract
Eukaryotic cells have evolved mechanisms for ensuring growth and survival in the face of stress caused by a fluctuating environment. Saccharomyces cerevisiae has two homologous glycerol-3-phosphate dehydrogenases, Gpd1 and Gpd2, that are required to endure various stresses, including hyperosmotic shock and hypoxia. These enzymes are only partially redundant, and their unique functions were attributed previously to differential transcriptional regulation and localization. We find that Gpd1 and Gpd2 are negatively regulated through phosphorylation by distinct kinases under reciprocal conditions. Gpd2 is phosphorylated by the AMP-activated protein kinase Snf1 to curtail glycerol production when nutrients are limiting. Gpd1, in contrast, is a target of TORC2-dependent kinases Ypk1 and Ypk2. Inactivation of Ypk1 by hyperosmotic shock results in dephosphorylation and activation of Gpd1, accelerating recovery through increased glycerol production. Gpd1 dephosphorylation acts synergistically with its transcriptional upregulation, enabling long-term growth at high osmolarity. Phosphorylation of Gpd1 and Gpd2 by distinct kinases thereby enables rapid adaptation to specific stress conditions. Introduction of phosphorylation motifs targeted by distinct kinases provides a general mechanism for functional specialization of duplicated genes during evolution. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
167. Type II p21 -activated kinases (PAKs) are regulated by an autoinhibitory pseudosubstrate.
- Author
-
Ha, Byung Hak, Davis, Matthew J., Chen, Catherine, Lou, Hua Jane, Gao, Jia, Rong Zhang, Krauthammer, Michael, Halaban, Ruth, Schlessinge, Joseph, Turk, Benjamin E., and Boggon, Titus J.
- Subjects
GUANOSINE triphosphatase ,P21 gene ,CELL death ,PHOSPHORYLATION ,CRYSTAL structure ,PROLINE - Abstract
The type II p21-activated kinases (PAKs) are key effectors of RHO-family GTPases involved in cell motility, survival, and proliferation. Using a structure-guided approach, we discovered that type II PAKs are regulated by an N-terminal autoinhibitory pseudosubstrate motif centered on a critical proline residue, and that this regulation occurs independently of activation loop phosphorylation. We determined six X-ray crystal structures of either full-length PAK4 or its catalytic domain, that demonstrate the molecular basis for pseudosubstrate binding to the active state with phosphorylated activation loop. We show that full-length PAK4 is constitutively autoinhibited, but mutation of the pseudosubstrate releases this inhibition and causes increased phosphorylation of the apoptotic regulation protein Bcl-2/Bcl-X
L antagonist causing cell death and cellular morphological changes. We also find that PAK6 is regulated by the pseudosubstrate region, indicating a common type II PAK autoregulatory mechanism. Finally, we find Src SH3, but not ß-PIX SH3, can activate PAK4. We provide a unique understanding for type II PAK regulation. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
168. Identification of neuronal substrates implicates Pak5 in synaptic vesicle trafficking.
- Author
-
Strochlic, Todd I., Concilio, Susanna, Viaud, Julien, Eberwine, Ryan A., Wong, Lisa Epstein, Minden, Audrey, Turk, Benjamin E., Plomann, Markus, and Peterson, Jeffrey R.
- Subjects
NEURAL transmission ,KINASE genetics ,PLANT growing media ,CENTRAL nervous system ,PHOSPHORYLATION ,ENDOCYTOSIS - Abstract
Synaptic transmission is mediated by a complex set of molecular events that must be coordinated in time and space. While many proteins that function at the synapse have been identified, the signaling pathways regulating these molecules are poorly understood. Pak5 (p21-activated kinase 5) is a brain-specific isoform of the group II Pak kinases whose substrates and roles within the central nervous system are largely unknown. To gain insight into the physiological roles of Pak5, we engineered a Pak5 mutant to selectively radiolabel its substrates in murine brain extract. Using this approach, we identified two novel Pak5 substrates, Pacsin1 and Synaptojanin1, proteins that directly interact with one another to regulate synaptic vesicle endocytosis and recycling. Pacsin1 and Synaptojanin1 were phosphorylated by Pak5 and the other group II Paks in vitro, and Pak5 phosphorylation promoted Pacsin1-Synaptojanin1 binding both in vitro and in vivo. These results implicate Pak5 in Pacsin1- and Synaptojanin1-mediated synaptic vesicle trafficking and may partially account for the cognitive and behavioral deficits observed in group II Pak-deficient mice. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
169. Phosphorylation of Immunity-Related GTPases by a Toxoplasma gondii-Secreted Kinase Promotes Macrophage Survival and Virulence.
- Author
-
Fentress, Sarah J., Behnke, Michael S., Dunay, Ildiko R., Mashayekhi, Mona, Rommereim, Leah M., Fox, Barbara A., Bzik, David J., Taylor, Gregory A., Turk, Benjamin E., Lichti, Cheryl F., Townsend, R. Reid, Qiu, Wei, Hui, Raymond, Beatty, Wandy L., and Sibley, L. David
- Subjects
PHOSPHORYLATION ,GUANOSINE triphosphatase ,TOXOPLASMA gondii ,MACROPHAGES ,PROTEIN kinases ,MICROBIAL virulence ,INTRACELLULAR pathogens ,GENE expression - Abstract
Summary: Macrophages are specialized to detect and destroy intracellular microbes and yet a number of pathogens have evolved to exploit this hostile niche. Here we demonstrate that the obligate intracellular parasite Toxoplasma gondii disarms macrophage innate clearance mechanisms by secreting a serine threonine kinase called ROP18, which binds to and phosphorylates immunity-related GTPases (IRGs). Substrate profiling of ROP18 revealed a preference for a conserved motif within switch region I of the GTPase domain, a modification predicted to disrupt IRG function. Consistent with this, expression of ROP18 was both necessary and sufficient to block recruitment of Irgb6, which was in turn required for parasite destruction. ROP18 phosphorylation of IRGs prevented clearance within inflammatory monocytes and IFN-γ-activated macrophages, conferring parasite survival in vivo and promoting virulence. IRGs are implicated in clearance of a variety of intracellular pathogens, suggesting that other virulence factors may similarly thwart this innate cellular defense mechanism. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
170. The structural basis for substrate and inhibitor selectivity of the anthrax lethal factor.
- Author
-
Turk, Benjamin E., Thiang Yian Wong, Schwarzenbacher, RObert, Jarrell, Emily T., Leppla, Stephen H., Collier, R. John, Liddington, Robert C., and Cantley, Lewis C.
- Subjects
- *
METALLOPROTEINASES , *ANTHRAX , *PEPTIDES , *PATHOLOGY , *CHEMICAL inhibitors , *MOLECULAR biology - Abstract
Recent events have created an urgent need for new therapeutic strategies to treat anthrax. We have applied a mixture-based peptide library approach to rapidly determine the optimal peptide substrate for the anthrax lethal factor (LF), a metalloproteinase with an important role in the pathogenesis of the disease. Using this approach we have identified peptide analogs that inhibit the enzyme in vitro and that protect cultured macrophages from LF-mediated cytolysis. The crystal structures of LF bound to an optimized peptide substrate and to peptide-based inhibitors provide a rationale for the observed selectivity and may be exploited in the design of future generations of LF inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
171. Identification of small molecule inhibitors of anthrax lethal factor.
- Author
-
Panchal, Rekha G., Hermone, Ann R., Tam Luong Nguyen, Tiang Yian Wong, Ann R., Schwarzenbacher, Robert, Schmidt, James, Lane, Douglas, McGrath, Connor, Turk, Benjamin E., Burnett, James, Aman, M. Javad, Little, Stephen, Sausville, Edward A., Zaharevitz, Daniel W., Cantley, Lewis C., Liddington, Robert C., Gussio, Rick, and Bavari, Sina
- Subjects
METALLOPROTEINASES ,BACILLUS anthracis ,CHEMICAL inhibitors ,ANTHRAX ,ZINC ,MOLECULAR biology - Abstract
The virulent spore-forming bacterium Bacillus anthracis secretes anthrax toxin composed of protective antigen (PA), lethal factor (LF) and edema factor (EF). LF is a Zn-dependent metalloprotease that inactivates key signaling molecules, such as mitogen-activated protein kinase kinases (MAPKK), to ultimately cause cell death. We report here the identification of small molecule (nonpeptidic) inhibitors of LF. Using a two-stage screening assay, we determined the LF inhibitory properties of 19 compounds. Here, we describe six inhibitors on the basis of a pharmacophoric relationship determined using X-ray crystallographic data, molecular docking studies and three-dimensional (3D) database mining from the US National Cancer Institute (NCI) chemical repository. Three of these compounds have K
i values in the 0.5-5 µM range and show competitive inhibition. These molecular scaffolds may be used to develop therapeutically viable inhibitors of LF. [ABSTRACT FROM AUTHOR]- Published
- 2004
- Full Text
- View/download PDF
172. Modulation of the Chromatin Phosphoproteome by the Haspin Protein Kinase*
- Author
-
Maiolica, Alessio, de Medina-Redondo, Maria, Schoof, Erwin M., Chaikuad, Apirat, Villa, Fabrizio, Gatti, Marco, Jeganathan, Siva, Lou, Hua Jane, Novy, Karel, Hauri, Simon, Toprak, Umut H., Herzog, Franz, Meraldi, Patrick, Penengo, Lorenza, Turk, Benjamin E., Knapp, Stefan, Linding, Rune, and Aebersold, Ruedi
- Abstract
Recent discoveries have highlighted the importance of Haspin kinase activity for the correct positioning of the kinase Aurora B at the centromere. Haspin phosphorylates Thr3of the histone H3 (H3), which provides a signal for Aurora B to localize to the centromere of mitotic chromosomes. To date, histone H3 is the only confirmed Haspin substrate. We used a combination of biochemical, pharmacological, and mass spectrometric approaches to study the consequences of Haspin inhibition in mitotic cells. We quantified 3964 phosphorylation sites on chromatin-associated proteins and identified a Haspin protein-protein interaction network. We determined the Haspin consensus motif and the co-crystal structure of the kinase with the histone H3 tail. The structure revealed a unique bent substrate binding mode positioning the histone H3 residues Arg2and Lys4adjacent to the Haspin phosphorylated threonine into acidic binding pockets. This unique conformation of the kinase-substrate complex explains the reported modulation of Haspin activity by methylation of Lys4of the histone H3. In addition, the identification of the structural basis of substrate recognition and the amino acid sequence preferences of Haspin aided the identification of novel candidate Haspin substrates. In particular, we validated the phosphorylation of Ser137of the histone variant macroH2A as a target of Haspin kinase activity. MacroH2A Ser137resides in a basic stretch of about 40 amino acids that is required to stabilize extranucleosomal DNA, suggesting that phosphorylation of Ser137might regulate the interactions of macroH2A and DNA. Overall, our data suggest that Haspin activity affects the phosphorylation state of proteins involved in gene expression regulation and splicing.
- Published
- 2014
- Full Text
- View/download PDF
173. Exploiting the Unique ATP-Binding Pocket of ToxoplasmaCalcium-Dependent Protein Kinase 1 To Identify Its Substrates
- Author
-
Lourido, Sebastian, Jeschke, Grace R., Turk, Benjamin E., and Sibley, L. David
- Abstract
Apicomplexan parasites rely on calcium as a second messenger to regulate a variety of essential cellular processes. Calcium-dependent protein kinases (CDPK), which transduce these signals, are conserved among apicomplexans but absent from mammalian hosts, making them attractive targets for therapeutic intervention. Despite their importance, the signaling pathways CDPK regulate remain poorly characterized, and their protein substrates are completely unknown. In Toxoplasma gondii, CDPK1 is required for calcium-regulated secretion from micronemes, thereby controlling motility, invasion, and egress from host cells. CDPK1 is unique among parasite and mammalian kinases in containing glycine at the key “gatekeeper” residue, which results in an expanded ATP-binding pocket. In the present study, we use a synthetic ATPγS analogue that displays steric complementarity to the ATP-binding pocket and hence allows identification of protein substrates based on selective thiophosphorylation. The specificity of this approach was validated by the concordance between the identified phosphorylation sites and the in vitrosubstrate preference of CDPK1. We further demonstrate that the phosphorylation of predicted substrates is dependent on CDPK1 both in vivoand in vitro. This combined strategy for identifying the targets of specific protein kinases provides a platform for defining the roles of CDPKs in apicomplexans.
- Published
- 2013
- Full Text
- View/download PDF
174. Discovery and Development of Anthrax Lethal Factor Metalloproteinase Inhibitors
- Author
-
Turk, Benjamin E.
- Abstract
Anthrax is caused by infection with Bacillus anthracis, a spore forming, rod-shaped, encapsulated gram positive bacteria. The disease manifests itself in distinct ways depending on the route of entry of infective bacterial spores: cutaneous, inhalational, and gastrointestinal. Though rare in humans, inhalational anthrax has become a major concern due to the capacity for spores to be weaponized. The limited success of antibiotic therapy has motivated investigation of complementary therapeutic strategies that target the bacterias secreted toxin. The zinc-dependent metalloproteinase lethal factor (LF) is a critical component of anthrax toxin and an important potential target for small molecule drugs. In the past few years, a number of approaches have been taken to identify LF inhibitors, from generating conventional metal chelating substrate analogs to random screening of diverse compound libraries. These efforts have produced several different classes of specific nanomolar range inhibitors. Some compounds have fared well in animal models for anthrax toxemia and infection, and these inhibitors and their derivatives may form the basis for future therapies to treat the disease in humans.
- Published
- 2008
175. Phosphorylated WNK kinase networks in recoded bacteria recapitulate physiological function.
- Author
-
Schiapparelli, Paula, Pirman, Natasha L., Mohler, Kyle, Miranda-Herrera, Pierre A., Zarco, Natanael, Kilic, Onur, Miller, Chad, Shah, Sagar R., Rogulina, Svetlana, Hungerford, William, Abriola, Laura, Hoyer, Denton, Turk, Benjamin E., Guerrero-Cázares, Hugo, Isaacs, Farren J., Quiñones-Hinojosa, Alfredo, Levchenko, Andre, and Rinehart, Jesse
- Abstract
Advances in genetic code expansion have enabled the production of proteins containing site-specific, authentic post-translational modifications. Here, we use a recoded bacterial strain with an expanded genetic code to encode phosphoserine into a human kinase protein. We directly encode phosphoserine into WNK1 (with-no-lysine [K] 1) or WNK4 kinases at multiple, distinct sites, which produced activated, phosphorylated WNK that phosphorylated and activated SPAK/OSR kinases, thereby synthetically activating this human kinase network in recoded bacteria. We used this approach to identify biochemical properties of WNK kinases, a motif for SPAK substrates, and small-molecule kinase inhibitors for phosphorylated SPAK. We show that the kinase inhibitors modulate SPAK substrates in cells, alter cell volume, and reduce migration of glioblastoma cells. Our work establishes a protein-engineering platform technology that demonstrates that synthetically active WNK kinase networks can accurately model cellular systems and can be used more broadly to target networks of phosphorylated proteins for research and discovery. [Display omitted] • Bacterial strain with expanded genetic code incorporates phosphoserine into WNK1 • Synthetically phosphorylated WNK1 activates SPAK and enables substrate discovery • Active WNK1/SPAK pathway yields a screen for small-molecule kinase inhibitors • Inhibitors modulate WNK/SPAK control of cell volume and reduce GBM migration Schiapparelli et al. describe a protein-engineering platform technology to synthetically activate the WNK/SPAK/OSR1 kinase network. Using this approach, they identify biochemical properties of WNK and SPAK kinases along with small-molecule inhibitors for SPAK. Cellular systems, both in vitro and in vivo , translate findings from the engineered kinase network. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
176. PPP6C negatively regulates oncogenic ERK signaling through dephosphorylation of MEK.
- Author
-
Cho, Eunice, Lou, Hua Jane, Kuruvilla, Leena, Calderwood, David A., and Turk, Benjamin E.
- Abstract
Flux through the RAF-MEK-ERK protein kinase cascade is shaped by phosphatases acting on the core components of the pathway. Despite being an established drug target and a hub for crosstalk regulation, little is known about dephosphorylation of MEK, the central kinase within the cascade. Here, we identify PPP6C, a phosphatase frequently mutated or downregulated in melanoma, as a major MEK phosphatase in cells exhibiting oncogenic ERK pathway activation. Recruitment of MEK to PPP6C occurs through an interaction with its associated regulatory subunits. Loss of PPP6C causes hyperphosphorylation of MEK at activating and crosstalk phosphorylation sites, promoting signaling through the ERK pathway and decreasing sensitivity to MEK inhibitors. Recurrent melanoma-associated PPP6C mutations cause MEK hyperphosphorylation, suggesting that they promote disease at least in part by activating the core oncogenic pathway driving melanoma. Collectively, our studies identify a key negative regulator of ERK signaling that may influence susceptibility to targeted cancer therapies. [Display omitted] • The phosphatase PPP6C promotes sensitivity of melanoma cells to MEK inhibitors • PPP6C loss causes ERK pathway hyperactivation in cells harboring BRAF and RAS mutations • PP6 regulatory subunits recruit MEK for dephosphorylation by PPP6C • Recurrent melanoma-associated PPP6C mutants lead to elevated MEK and ERK activity Through an shRNA screen, Cho et al. identify PPP6C as a phosphatase that inactivates the kinase MEK, sensitizing tumor cells to clinical MEK inhibitors. This study suggests that cancer-associated loss-of-function PPP6C mutations prevalent in melanoma serve to activate the core oncogenic RAF-MEK-ERK pathway that drives the disease. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
177. Aurora B controls anaphase onset and error-free chromosome segregation in trypanosomes.
- Author
-
Ballmer, Daniel, Hua Jane Lou, Ishii, Midori, Turk, Benjamin E., and Akiyoshi, Bungo
- Subjects
- *
CHROMOSOME segregation , *AURORA kinases , *ANAPHASE , *CELL cycle , *KINETOCHORE , *TRYPANOSOMA brucei , *NUCLEAR membranes - Abstract
Kinetochores form the interface between chromosomes and spindle microtubules and are thus under tight control by a complex regulatory circuitry. The Aurora B kinase plays a central role within this circuitry by destabilizing improper kinetochore-microtubule attachments and relaying the attachment status to the spindle assembly checkpoint. Intriguingly, Aurora B is conserved even in kinetoplastids, a group of early-branching eukaryotes which possess a unique set of kinetochore proteins. It remains unclear how their kinetochores are regulated to ensure faithful chromosome segregation. Here, we show in Trypanosoma brucei that Aurora B activity controls the metaphase-to-anaphase transition through phosphorylation of the divergent Bub1-like protein KKT14. Depletion of KKT14 overrides the metaphase arrest resulting from Aurora B inhibition, while expression of non-phosphorylatable KKT14 delays anaphase onset. Finally, we demonstrate that re-targeting Aurora B to the outer kinetochore suffices to promote mitotic exit but causes extensive chromosome missegregation in anaphase. Our results indicate that Aurora B and KKT14 are involved in an unconventional circuitry controlling cell cycle progression in trypanosomes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
178. Phosphorylation of Pal2 by the protein kinases Kin1 and Kin2 modulates HAC1mRNA splicing in the unfolded protein response in yeast
- Author
-
Ghosh, Chandrima, Uppala, Jagadeesh Kumar, Sathe, Leena, Hammond, Charlotte I., Anshu, Ashish, Pokkuluri, P. Raj, Turk, Benjamin E., and Dey, Madhusudan
- Abstract
Phosphorylated Pal2 promotes the cytosolic splicing of HAC1to restore homeostasis in yeast undergoing ER stress.
- Published
- 2021
- Full Text
- View/download PDF
179. Glycogen synthase kinase‐3β regulation: another kinase gets in on the AKT.
- Author
-
Turk, Benjamin E.
- Subjects
- *
GLYCOGEN synthase kinase-3 , *KINASE inhibitors , *PROTEIN kinase inhibitors , *GLYCOGEN synthesis , *PROTEIN kinase genetics - Published
- 2018
- Full Text
- View/download PDF
180. Global view of the RAF-MEK-ERK module and its immediate downstream effectors.
- Author
-
Santini, Cristina C., Longden, James, Schoof, Erwin M., Simpson, Craig D., Jeschke, Grace R., Creixell, Pau, Kim, Jinho, Wu, Xuewei, Turk, Benjamin E., Rosen, Neal, Poulikakos, Poulikos I., and Linding, Rune
- Abstract
Small molecule inhibitors of BRAF and MEK have proven effective at inhibiting tumor growth in melanoma patients, however this efficacy is limited due to the almost universal development of drug resistance. To provide advanced insight into the signaling responses that occur following kinase inhibition we have performed quantitative (phospho)-proteomics of human melanoma cells treated with either dabrafenib, a BRAF inhibitor; trametinib, a MEK inhibitor or SCH772984, an ERK inhibitor. Over nine experiments we identified 7827 class I phosphorylation sites on 4960 proteins. This included 54 phosphorylation sites that were significantly down-modulated after exposure to all three inhibitors, 34 of which have not been previously reported. Functional analysis of these novel ERK targets identified roles for them in GTPase activity and regulation, apoptosis and cell-cell adhesion. Comparison of the results presented here with previously reported phosphorylation sites downstream of ERK showed a limited degree of overlap suggesting that ERK signaling responses may be highly cell line and cue specific. In addition we identified 26 phosphorylation sites that were only responsive to dabrafenib. We provide further orthogonal experimental evidence for 3 of these sites in human embryonic kidney cells over-expressing BRAF as well as further computational insights using KinomeXplorer. The validated phosphorylation sites were found to be involved in actin regulation, which has been proposed as a novel mechanism for inhibiting resistance development. These results would suggest that the linearity of the BRAF-MEK-ERK module is at least context dependent. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
181. Early Steps in Autophagy Depend on Direct Phosphorylation of Atg9 by the Atg1 Kinase
- Author
-
Papinski, Daniel, Schuschnig, Martina, Reiter, Wolfgang, Wilhelm, Larissa, Barnes, Christopher A., Maiolica, Alessio, Hansmann, Isabella, Pfaffenwimmer, Thaddäus, Kijanska, Monika, Stoffel, Ingrid, Lee, Sung Sik, Brezovich, Andrea, Lou, Jane Hua, Turk, Benjamin E., Aebersold, Ruedi, Ammerer, Gustav, Peter, Matthias, and Kraft, Claudine
- Subjects
3. Good health - Abstract
Bulk degradation of cytoplasmic material is mediated by a highly conserved intracellular trafficking pathway termed autophagy. This pathway is characterized by the formation of double-membrane vesicles termed autophagosomes engulfing the substrate and transporting it to the vacuole/lysosome for breakdown and recycling. The Atg1/ULK1 kinase is essential for this process; however, little is known about its targets and the means by which it controls autophagy. Here we have screened for Atg1 kinase substrates using consensus peptide arrays and identified three components of the autophagy machinery. The multimembrane-spanning protein Atg9 is a direct target of this kinase essential for autophagy. Phosphorylated Atg9 is then required for the efficient recruitment of Atg8 and Atg18 to the site of autophagosome formation and subsequent expansion of the isolation membrane, a prerequisite for a functioning autophagy pathway. These findings show that the Atg1 kinase acts early in autophagy by regulating the outgrowth of autophagosomal membranes., Molecular Cell, 53 (3), ISSN:1097-2765, ISSN:1097-4164
182. Deciphering protein kinase specificity through large-scale analysis of yeast phosphorylation site motifs
- Author
-
Mok, Janine, Kim, Philip M., Lam, Hugo Y. K., Piccirillo, Stacy, Zhou, Xiuqiong, Jeschke, Grace R., Sheridan, Douglas L., Parker, Sirlester A., Desai, Ved, Jwa, Miri, Cameroni, Elisabetta, Niu, Hengyao, Good, Matthew, Remenyi, Attila, Ma, Jia-Lin Nianhan, Sheu, Yi-Jun, Sassi, Holly E., Sopko, Richelle, Chan, Clarence S. M., De Virgilio, Claudio, Hollingsworth, Nancy M., Lim, Wendell A., Stern, David F., Stillman, Bruce, Andrews, Brenda J., Gerstein, Mark B., Snyder, Michael, Turk, Benjamin E., Mok, Janine, Kim, Philip M., Lam, Hugo Y. K., Piccirillo, Stacy, Zhou, Xiuqiong, Jeschke, Grace R., Sheridan, Douglas L., Parker, Sirlester A., Desai, Ved, Jwa, Miri, Cameroni, Elisabetta, Niu, Hengyao, Good, Matthew, Remenyi, Attila, Ma, Jia-Lin Nianhan, Sheu, Yi-Jun, Sassi, Holly E., Sopko, Richelle, Chan, Clarence S. M., De Virgilio, Claudio, Hollingsworth, Nancy M., Lim, Wendell A., Stern, David F., Stillman, Bruce, Andrews, Brenda J., Gerstein, Mark B., Snyder, Michael, and Turk, Benjamin E.
- Abstract
Phosphorylation is a universal mechanism for regulating cell behavior in eukaryotes. Although protein kinases target short linear sequence motifs on their substrates, the rules for kinase substrate recognition are not completely understood. We used a rapid peptide screening approach to determine consensus phosphorylation site motifs targeted by 61 of the 122 kinases in Saccharomyces cerevisiae. By correlating these motifs with kinase primary sequence, we uncovered previously unappreciated rules for determining specificity within the kinase family, including a residue determining P–3 arginine specificity among members of the CMGC [CDK (cyclin-dependent kinase), MAPK (mitogen-activated protein kinase), GSK (glycogen synthase kinase), and CDK-like] group of kinases. Furthermore, computational scanning of the yeast proteome enabled the prediction of thousands of new kinase-substrate relationships. We experimentally verified several candidate substrates of the Prk1 family of kinases in vitro and in vivo and identified a protein substrate of the kinase Vhs1. Together, these results elucidate how kinase catalytic domains recognize their phosphorylation targets and suggest general avenues for the identification of previously unknown kinase substrates across eukaryotes.
183. Linear motif specificity in signaling through p38α and ERK2 mitogen-activated protein kinases.
- Author
-
Robles, Jaylissa Torres, Hua Jane Lou, Guangda Shi, Pan, Pauline Lining, and Turk, Benjamin E.
- Subjects
- *
MITOGEN-activated protein kinases , *EUKARYOTIC cells , *SIGNAL integrity (Electronics) , *SIGNAL recognition particle receptor - Abstract
Mitogen-activated protein kinase (MAPK) cascades are essential for eukaryotic cells to integrate and respond to diverse stimuli. Maintaining specificity in signaling through MAPK networks is key to coupling distinct inputs to appropriate cellular responses. Docking sites--short linear motifs found in MAPK substrates, regulators, and scaffolds--can promote signaling specificity through selective interactions, but how they do so remains unresolved. Here, we screened a proteomic library for sequences interacting with the MAPKs extracellular signal-regulated kinase 2 (ERK2) and p38α, identifying selective and promiscuous docking motifs. Sequences specific for p38α had high net charge and lysine content, and selective binding depended on a pair of acidic residues unique to the p38α docking interface. Finally, we validated a set of full-length proteins harboring docking sites selected in our screens to be authentic MAPK interactors and substrates. This study identifies features that help define MAPK signaling networks and explains how specific docking motifs promote signaling integrity. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
184. A high throughput assay to identify substrate-selective inhibitors of the ERK protein kinases.
- Author
-
Miller, Chad J., Muftuoglu, Yagmur, and Turk, Benjamin E.
- Subjects
- *
EXTRACELLULAR signal-regulated kinases , *SELECTIVE inhibition (Chemistry) , *BIOCHEMICAL substrates , *BIOLOGICAL assay , *CANCER cell proliferation - Abstract
Extracellular signal-regulated kinases 1 and 2 (ERK1/2) phosphorylate a variety of substrates important for survival and proliferation, and their activity is frequently deregulated in tumors. ERK pathway inhibitors have shown clinical efficacy as anti-cancer drugs, but most patients eventually relapse due to reactivation of the pathway. One factor limiting the efficacy of current therapeutics is the difficulty in reaching clinically effective inhibition of the ERK pathway in the absence of on-target toxicities. Here, we describe an assay suitable for high throughput screening to discover substrate selective ERK1/2 inhibitors, which may have a larger therapeutic window than conventional inhibitors. Specifically, we aim to target a substrate-binding pocket within the ERK1/2 catalytic domain outside of the catalytic cleft. The assay uses an AlphaScreen format to detect phosphorylation of a high-efficiency substrate harboring an essential docking site motif. Pilot screening established that the assay is suitably robust for high-throughput screening. Importantly, the assay can be conducted at high ATP concentrations, which we show reduces the discovery of conventional ATP-competitive inhibitors. These studies provide the basis for high-throughput screens to discover new classes of non-conventional ERK1/2 inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
185. Identification of Yin-Yang Regulators and a Phosphorylation Consensus for Male Germ Cell-Associated Kinase (MAK)-Related Kinase.
- Author
-
Zheng Fu, Larson, Katherine A., Chitta, Raghu K., Parker, Sirlester A., Turk, Benjamin E., Lawrence, Matthew W., Kaldis, Philipp, Galaktionov, Konstantin, Cohn, Steven M., Shabanowitz, Jeffrey, Hunt, Donald F., and Sturgill, Thomas W.
- Subjects
PROTEIN kinases ,HOMOLOGY (Biology) ,SACCHAROMYCES cerevisiae ,CYCLIN-dependent kinases ,PHOSPHORYLATION - Abstract
MAK (male germ cell-associated protein kinase) and MRK/ICK (MAK-related kinase/intestinal cell kinase) are human homologs of Ime2p in Saccharomyces cerevisiae and of Mde3 and Pit1 in Schizosaccharomyces pombe and are similar to human cyclin-dependent kinase 2 (CDK2) and extracellular signal-regulated kinase 2 (ERK2). MAK and MRK require dual phosphorylation in a TDY motif catalyzed by an unidentified human threonine kinase and tyrosine autophosphorylation. Herein, we establish that human CDK-related kinase CCRK (cell cycle-related kinase) is an activating T157 kinase for MRK, whereas active CDK7/cyclin H/MAT1 complexes phosphorylate CDK2 but not MRK. Protein phosphatase 5 (PP5) interacts with MRK in a complex and dephosphorylates MRK at T157 in vitro and in situ. Thus, CCRK and PP5 are yin-yang regulators of T157 phosphorylation. To determine a substrate consensus, we screened a combinatorial peptide library with active MRK. MRK preferentially phosphorylates R-P-X-S/T-P sites, with the preference for arginine at position -3 (P-3) being more stringent than for prolines at P-2 and P+1. Using the consensus, we identified a putative phosphorylation site (RPLT
1080 S) for MRK in human Scythe, an antiapoptotic protein that interacts with MRK. MRK phosphorylates Scythe at T1080 in vitro as determined by site-directed mutagenesis and mass spectrometry, supporting the consensus and suggesting Scythe as a physiological substrate for MRK. [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
186. Signaling, Regulation, and Specificity of the Type II p21-activated Kinases.
- Author
-
Ha, Byung Hak, Morse, Elizabeth M., Turk, Benjamin E., and Boggon, Titus J.
- Subjects
- *
SERINE/THREONINE kinase regulation , *CELLULAR signal transduction , *P21 gene , *RHO GTPases , *PROTEIN kinases , *G proteins , *PHOSPHORYLATION - Abstract
The p21-activated kinases (PAKs) are a family of six serine/threonine kinases that act as key effectors of RHO family GTPases in mammalian cells. PAKs are subdivided into two groups: type I PAKs (PAK1, PAK2, and PAK3) and type II PAKs (PAK4, PAK5, and PAK6). Although these groups are involved in common signaling pathways, recent work indicates that the two groups have distinct modes of regulation and have both unique and common substrates. Here, we review recent insights into the molecular level details that govern regulation of type II PAK signaling. We also consider mechanisms by which signal transduction is regulated at the level of substrate specificity. Finally, we discuss the implications of these studies for clinical targeting of these kinases. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
187. Substrate priming enhances phosphorylation by the budding yeast kinases Kin1 and Kin2.
- Author
-
Jeschke, Grace R., Hua Jane Lou, Weise, Keith, Hammond, Charlotte I., Demonch, Mallory, Brennwald, Patrick, and Turk, Benjamin E.
- Subjects
- *
PHOSPHORYLATION , *BUDDING (Zoology) , *BIOCHEMICAL substrates , *FUNGAL enzymes , *DNA primers - Abstract
Multisite phosphorylation of proteins is a common mechanism for signal integration and amplification in eukaryotic signaling networks. Proteins are commonly phosphorylated at multiple sites in an ordered manner, whereby phosphorylation by one kinase primes the substrate by generating a recognition motif for a second kinase. Here we show that substrate priming promotes phosphorylation by Saccharomyces cerevisiae Kin1 and Kin2, kinases that regulate cell polarity, exocytosis, and the endoplasmic reticulum (ER) stress response. Kin1/Kin2 phosphorylated substrates within the context of a sequence motif distinct from those of their most closely related kinases. In particular, the rate of phosphorylation of a peptide substrate by Kin1/Kin2 increased>30-fold with incorporation of a phosphoserine residue two residues downstream of the phosphorylation site. Recognition of phosphorylated substrates by Kin1/Kin2 was mediated by a patch of basic residues located in the region of the kinase αC helix. We identified a set of candidate Kin1/Kin2 substrates reported to be dually phosphorylated at sites conforming to the Kin1/Kin2 consensus sequence. One of these proteins, the t-SNARE protein Sec9, was confirmed to be a Kin1/ Kin2 substrate both in vitro and in vivo. Sec9 phosphorylation by Kin1 in vitro was enhanced by prior phosphorylation at the +2 position. Recognition of primed substrates was not required for the ability of Kin2 to suppress the growth defect of secretory pathway mutants but was necessary for optimal growth under conditions of ER stress. These results suggest that at least some endogenous protein substrates of Kin1/Kin2 are phosphorylated in a priming-dependent manner. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
188. Identification of a Substrate-selective Exosite within the Metalloproteinase Anthrax Lethal Factor.
- Author
-
Goldberg, Allison B., Cho, Eunice, Miller, Chad J., Lou, Hua Jane, and Turk, Benjamin E.
- Subjects
- *
BACILLUS anthracis , *METALLOPROTEINASES , *CELLULAR signal transduction , *PROTEOLYTIC enzymes , *MITOGEN-activated protein kinases - Abstract
The metalloproteinase anthrax lethal factor (LF) is secreted by Bacillus anthracis to promote disease virulence through disruption of host signaling pathways. LF is a highly specific protease, exclusively cleaving mitogen-activated protein kinase kinases (MKKs) and rodent NLRP1B (NACHT leucine-rich repeat and pyrin domain-containing protein 1B). How LF achieves such restricted substrate specificity is not understood. Previous studies have suggested the existence of an exosite interaction between LF and MKKs that promotes cleavage efficiency and specificity. Through a combination of in silico prediction and site-directed mutagenesis, we have mapped an exosite to a non-catalytic region of LF. Mutations within this site selectively impair proteolysis of full-length MKKs yet have no impact on cleavage of short peptide substrates. Although this region appears important for cleaving all LF protein substrates, we found that mutation of specific residues within the exosite differentially affects MKK and NLRP1B cleavage in vitro and in cultured cells. One residue in particular, Trp-271, is essential for cleavage of MKK3, MKK4, and MKK6 but dispensable for targeting of MEK1, MEK2, and NLRP1B. Analysis of chimeric substrates suggests that this residue interacts with the MKK catalytic domain. We found that LF-W271A blocked ERK phosphorylation and growth in a melanoma cell line, suggesting that it may provide a highly selective inhibitor of MEK1/2 for use as a cancer therapeutic. These findings provide insight into how a bacterial toxin functions to specifically impair host signaling pathways and suggest a general strategy for mapping protease exosite interactions. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
189. Kinome-wide Decoding of Network-Attacking Mutations Rewiring Cancer Signaling.
- Author
-
Creixell, Pau, Schoof, Erwin M., Simpson, Craig D., Longden, James, Miller, Chad J., Lou, Hua Jane, Perryman, Lara, Cox, Thomas R., Zivanovic, Nevena, Palmeri, Antonio, Wesolowska-Andersen, Agata, Helmer-Citterich, Manuela, Ferkinghoff-Borg, Jesper, Itamochi, Hiroaki, Bodenmiller, Bernd, Erler, Janine T., Turk, Benjamin E., and Linding, Rune
- Subjects
- *
CANCER genetics , *GENETIC code , *CELLULAR signal transduction , *GENETIC mutation , *PROTEIN kinases - Abstract
Summary Cancer cells acquire pathological phenotypes through accumulation of mutations that perturb signaling networks. However, global analysis of these events is currently limited. Here, we identify six types of network-attacking mutations (NAMs), including changes in kinase and SH2 modulation, network rewiring, and the genesis and extinction of phosphorylation sites. We developed a computational platform (ReKINect) to identify NAMs and systematically interpreted the exomes and quantitative (phospho-)proteomes of five ovarian cancer cell lines and the global cancer genome repository. We identified and experimentally validated several NAMs, including PKCγ M501I and PKD1 D665N, which encode specificity switches analogous to the appearance of kinases de novo within the kinome. We discover mutant molecular logic gates, a drift toward phospho-threonine signaling, weakening of phosphorylation motifs, and kinase-inactivating hotspots in cancer. Our method pinpoints functional NAMs, scales with the complexity of cancer genomes and cell signaling, and may enhance our capability to therapeutically target tumor-specific networks. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
190. Unmasking Determinants of Specificity in the Human Kinome.
- Author
-
Creixell, Pau, Palmeri, Antonio, Miller, Chad J., Lou, Hua Jane, Santini, Cristina C., Nielsen, Morten, Turk, Benjamin E., and Linding, Rune
- Subjects
- *
CANCER patients , *PROTEIN kinases , *AMINO acids , *BIOLOGICAL evolution , *CELLULAR signal transduction - Abstract
Summary Protein kinases control cellular responses to environmental cues by swift and accurate signal processing. Breakdowns in this high-fidelity capability are a driving force in cancer and other diseases. Thus, our limited understanding of which amino acids in the kinase domain encode substrate specificity, the so-called determinants of specificity (DoS), constitutes a major obstacle in cancer signaling. Here, we systematically discover several DoS and experimentally validate three of them, named the αC1, αC3, and APE-7 residues. We demonstrate that DoS form sparse networks of non-conserved residues spanning distant regions. Our results reveal a likely role for inter-residue allostery in specificity and an evolutionary decoupling of kinase activity and specificity, which appear loaded on independent groups of residues. Finally, we uncover similar properties driving SH2 domain specificity and demonstrate how the identification of DoS can be utilized to elucidate a greater understanding of the role of signaling networks in cancer ( Creixell et al., 2015 [this issue of Cell ]). [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
191. Small Molecule Inhibition of the Autophagy Kinase ULK1 and Identification of ULK1 Substrates.
- Author
-
Egan, Daniel F., Chun, Matthew G.H., Vamos, Mitchell, Zou, Haixia, Rong, Juan, Miller, Chad J., Lou, Hua Jane, Raveendra-Panickar, Dhanya, Yang, Chih-Cheng, Sheffler, Douglas J., Teriete, Peter, Asara, John M., Turk, Benjamin E., Cosford, Nicholas D.P., and Shaw, Reuben J.
- Subjects
- *
PROTEIN kinase inhibitors , *AUTOPHAGY , *CANCER treatment , *PHOSPHORYLATION , *TARGETED drug delivery , *MEDICAL screening - Abstract
Summary Many tumors become addicted to autophagy for survival, suggesting inhibition of autophagy as a potential broadly applicable cancer therapy. ULK1/Atg1 is the only serine/threonine kinase in the core autophagy pathway and thus represents an excellent drug target. Despite recent advances in the understanding of ULK1 activation by nutrient deprivation, how ULK1 promotes autophagy remains poorly understood. Here, we screened degenerate peptide libraries to deduce the optimal ULK1 substrate motif and discovered 15 phosphorylation sites in core autophagy proteins that were verified as in vivo ULK1 targets. We utilized these ULK1 substrates to perform a cell-based screen to identify and characterize a potent ULK1 small molecule inhibitor. The compound SBI-0206965 is a highly selective ULK1 kinase inhibitor in vitro and suppressed ULK1-mediated phosphorylation events in cells, regulating autophagy and cell survival. SBI-0206965 greatly synergized with mechanistic target of rapamycin (mTOR) inhibitors to kill tumor cells, providing a strong rationale for their combined use in the clinic. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
192. An AMPK-Independent Signaling Pathway Downstream of the LKB1 Tumor Suppressor Controls Snail1 and Metastatic Potential.
- Author
-
Goodwin, Jonathan M., Svensson, Robert U., Lou, Hua Jane, Winslow, Monte M., Turk, Benjamin E., and Shaw, Reuben J.
- Subjects
- *
CELLULAR signal transduction , *TUMOR suppressor genes , *METASTASIS , *PROTEIN kinases , *MESENCHYMAL stem cells , *GENETIC transcription - Abstract
The serine/threonine kinase LKB1 is a tumor suppressor whose loss is associated with increased metastatic potential. In an effort to define biochemical signatures of metastasis associated with LKB1 loss, we discovered that the epithelial-to-mesenchymal transition transcription factor Snail1 was uniquely upregulated upon LKB1 deficiency across cell types. The ability of LKB1 to suppress Snail1 levels was independent of AMPK but required the related kinases MARK1 and MARK4. In a screen for substrates of these kinases involved in Snail regulation, we identified the scaffolding protein DIXDC1. Similar to loss of LKB1, DIXDC1 depletion results in upregulation of Snail1 in a FAK-dependent manner, leading to increased cell invasion. MARK1 phosphorylation of DIXDC1 is required for its localization to focal adhesions and ability to suppress metastasis in mice. DIXDC1 is frequently downregulated in human cancers, which correlates with poor survival. This study defines an AMPK-independent phosphorylation cascade essential for LKB1-dependent control of metastatic behavior. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
193. Early Steps in Autophagy Depend on Direct Phosphorylation of Atg9 by the Atg1 Kinase.
- Author
-
Papinski, Daniel, Schuschnig, Martina, Reiter, Wolfgang, Wilhelm, Larissa, Barnes, Christopher?A., Maiolica, Alessio, Hansmann, Isabella, Pfaffenwimmer, Thaddaeus, Kijanska, Monika, Stoffel, Ingrid, Lee, Sung?Sik, Brezovich, Andrea, Lou, Jane?Hua, Turk, Benjamin?E., Aebersold, Ruedi, Ammerer, Gustav, Peter, Matthias, and Kraft, Claudine
- Subjects
- *
AUTOPHAGY , *PHOSPHORYLATION , *BIODEGRADATION , *PROTEIN kinases , *INTRACELLULAR membranes , *MEMBRANE proteins - Abstract
Summary: Bulk degradation of cytoplasmic material is mediated by a highly conserved intracellular trafficking pathway termed autophagy. This pathway is characterized by the formation of double-membrane vesicles termed autophagosomes engulfing the substrate and transporting it to the vacuole/lysosome for breakdown and recycling. The Atg1/ULK1 kinase is essential for this process; however, little is known about its targets and the means by which it controls autophagy. Here we have screened for Atg1 kinase substrates using consensus peptide arrays and identified three components of the autophagy machinery. The multimembrane-spanning protein Atg9 is a direct target of this kinase essential for autophagy. Phosphorylated Atg9 is then required for the efficient recruitment of Atg8 and Atg18 to the site of autophagosome formation and subsequent expansion of the isolation membrane, a prerequisite for a functioning autophagy pathway. These findings show that the Atg1 kinase acts early in autophagy by regulating the outgrowth of autophagosomal membranes. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
194. Analysis of substrate specificity and cyclin Y binding of PCTAIRE-1 kinase
- Author
-
Shehata, Saifeldin N., Hunter, Roger W., Ohta, Eriko, Peggie, Mark W., Lou, Hua Jane, Sicheri, Frank, Zeqiraj, Elton, Turk, Benjamin E., and Sakamoto, Kei
- Subjects
- *
CYCLINS , *PROTEIN binding , *MYELIN basic protein , *GLUTATHIONE transferase , *CYCLIN-dependent kinases , *REGULATION of cell growth , *HEMAGGLUTININ - Abstract
Abstract: PCTAIRE-1 (cyclin-dependent kinase [CDK] 16) is a highly conserved serine/threonine kinase that belongs to the CDK family of protein kinases. Little is known regarding PCTAIRE-1 regulation and function and no robust assay exists to assess PCTAIRE-1 activity mainly due to a lack of information regarding its preferred consensus motif and the lack of bona fide substrates. We used positional scanning peptide library technology and identified the substrate-specificity requirements of PCTAIRE-1 and subsequently elaborated a peptide substrate termed PCTAIRE-tide. Recombinant PCTAIRE-1 displayed vastly improved enzyme kinetics on PCTAIRE-tide compared to a widely used generic CDK substrate peptide. PCTAIRE-tide also greatly improved detection of endogenous PCTAIRE-1 activity. Similar to other CDKs, PCTAIRE-1 requires a proline residue immediately C-terminal to the phosphoacceptor site (+1) for optimal activity. PCTAIRE-1 has a unique preference for a basic residue at +4, but not at +3 position (a key characteristic for CDKs). We also demonstrate that PCTAIRE-1 binds to a novel cyclin family member, cyclin Y, which increased PCTAIRE-1 activity towards PCTAIRE-tide >100-fold. We hypothesised that cyclin Y binds and activates PCTAIRE-1 in a way similar to which cyclin A2 binds and activates CDK2. Point mutants of cyclin Y predicted to disrupt PCTAIRE-1-cyclin Y binding severely prevented complex formation and activation of PCTAIRE-1. We have identified PCTAIRE-tide as a powerful tool to study the regulation of PCTAIRE-1. Our understanding of the molecular interaction between PCTAIRE-1 and cyclin Y further facilitates future investigation of the functions of PCTAIRE-1 kinase. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
195. Cyclic GMP-dependent Stimulation of Serotonin Transport Does Not Involve Direct Transporter Phosphorylation by cGMP-dependent Protein Kinase.
- Author
-
Wong, Albert, Yuan-Wei Zhang, Jeschke, Grace R., Turk, Benjamin E., and Rudnick, Gary
- Subjects
- *
SEROTONIN , *NEURONS , *EXOCYTOSIS , *ANTIDEPRESSANTS , *ECSTASY (Drug) , *NITRIC-oxide synthases , *GUANYLATE cyclase , *PROTEIN kinases - Abstract
The serotonin transporter (SERT) is responsible for reuptake of serotonin (5-hydroxytryptamine) after its exocytotic release from neurons. It is the primary target for antidepressants and stimulants, including "ecstasy" (3,4-methylenedioxymethamphetamine). SERT is regulated by several processes, including a cyclic GMP signaling pathway involving nitric oxide synthase, guanylyl cyclase, and cGMP-dependent protein kinase (PKG). Here, we show that SERT was phosphorylated in a PKG Iα-dependent manner in vitro, but that SERT was not a direct substrate of PKG.Wegenerated an analog-sensitive gatekeeper residue mutant of PKG Iα (M438G) that efficiently used the ATP analogN6-benzyl-ATP. This mutant, but not the wild type (WT) kinase, used theATPanalog to phosphorylate both a model peptide substrate as well as an established protein substrate of PKG (vasodilator-stimulated phosphoprotein). PKG Iα M438G effectively substituted for the WT kinase in stimulating SERTmediated 5-hydroxytryptamine transport in cultured cells. Addition of either WT or mutant PKGIα M438G to membranes containing SERT in vitro led to radiolabel incorporation from [γ-33P]ATP but not from similarly labeledN6-benzyl-ATP, indicating that SERT was phosphorylated by another kinase that could not utilize the ATP analog. These results are consistent with the proposed SERT phosphorylation site, Thr-276, being highly divergent from the consensus PKG phosphorylation site sequence, which we verified through peptide library screening. Another proposed SERT kinase, the p38 mitogen-activated protein kinase, could not substitute for PKG in this assay, and p38 inhibitors did not block PKG-dependent phosphorylation of SERT. The results suggest that PKG initiates a kinase cascade that leads to phosphorylation of SERT by an as yet unidentified protein kinase. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
196. Identification of Exosite-Targeting Inhibitors of Anthrax Lethal Factor by High-Throughput Screening
- Author
-
Bannwarth, Ludovic, Goldberg, Allison B., Chen, Catherine, and Turk, Benjamin E.
- Subjects
- *
ANTHRAX , *HIGH throughput screening (Drug development) , *PROTEASE inhibitors , *METALLOPROTEINASES , *MITOGEN-activated protein kinase kinase , *PEPTIDES - Abstract
Summary: Protease inhibitor discovery has focused almost exclusively on compounds that bind to the active site. Inhibitors targeting protease exosites, regions outside of the active site that influence catalysis, offer potential advantages of increased specificity but are difficult to systematically discover. Here, we describe an assay suitable for detecting exosite-targeting inhibitors of the metalloproteinase anthrax lethal factor (LF) based on cleavage of a full-length mitogen-activated protein kinase kinase (MKK) substrate. We used this assay to screen a small-molecule library and then subjected hits to a secondary screen to exclude compounds that efficiently blocked cleavage of a peptide substrate. We identified a compound that preferentially inhibited cleavage of MKKs compared with peptide substrates and could suppress LF-induced macrophage cytolysis. This approach should be generally applicable to the discovery of exosite-targeting inhibitors of many additional proteases. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
197. Purification and characterization of tagless recombinant human elongation factor 2 kinase (eEF-2K) expressed in Escherichia coli
- Author
-
Abramczyk, Olga, Tavares, Clint D.J., Devkota, Ashwini K., Ryazanov, Alexey G., Turk, Benjamin E., Riggs, Austen F., Ozpolat, Bulent, and Dalby, Kevin N.
- Subjects
- *
ESCHERICHIA coli , *PROTEIN kinases , *GENE expression , *CALMODULIN , *PROTEIN synthesis , *PHOSPHORYLATION , *BIOCHEMISTRY , *LIGHT scattering - Abstract
Abstract: The eukaryotic elongation factor 2 kinase (eEF-2K) modulates the rate of protein synthesis by impeding the elongation phase of translation by inactivating the eukaryotic elongation factor 2 (eEF-2) via phosphorylation. eEF-2K is known to be activated by calcium and calmodulin, whereas the mTOR and MAPK pathways are suggested to negatively regulate kinase activity. Despite its pivotal role in translation regulation and potential role in tumor survival, the structure, function, and regulation of eEF-2K have not been described in detail. This deficiency may result from the difficulty of obtaining the recombinant kinase in a form suitable for biochemical analysis. Here we report the purification and characterization of recombinant human eEF-2K expressed in the Escherichia coli strain Rosetta-gami 2(DE3). Successive chromatography steps utilizing Ni–NTA affinity, anion-exchange, and gel filtration columns accomplished purification. Cleavage of the thioredoxin-His6-tag from the N-terminus of the expressed kinase with TEV protease yielded 9mg of recombinant (G-D-I)-eEF-2K per liter of culture. Light scattering shows that eEF-2K is a monomer of ∼85kDa. In vitro kinetic analysis confirmed that recombinant human eEF-2K is able to phosphorylate wheat germ eEF-2 with kinetic parameters comparable to the mammalian enzyme. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
198. Structural Bases of PAS Domain-regulated Kinase (PASK) Activation in the Absence of Activation Loop Phosphorylation.
- Author
-
Kikani, Chintan K., Antonysamy, Stephen A., Bonann, Jeffrey B., Romero, Rich, Zhang, Feiyu Fred, Russe, Marijane, Gheyi, Tarun, Iizuka, Miyo, Emtage, Spencer, Sauder, J. Michael, Turk, Benjamin E., Burley, Stephen K., and Rutter, Jared
- Subjects
- *
CHEMICAL reactions , *PHOSPHORYLATION , *PROTEIN kinases , *GENETIC mutation , *BIOCHEMISTRY - Abstract
Per-Arnt-Sim (PAS) domain-containing protein kinase (PASK) is an evolutionary conserved protein kinase that coordinates cellular metabolism with metabolic demand in yeast and mammals. The molecular mechanisms underlying PASK regulation, however, remain unknown. Herein, we describe a crystal structure of the kinase domain of human PASK, which provides insights into the regulatory mechanisms governing catalysis. We show that the kinase domain adopts an active conformation and has catalytic activity in vivo and in vitro in the absence of activation loop phosphorylation. Using site-directed mutagenesis and structural comparison with active and inactive kinases, we identified several key structural features in PASK that enable activation loop phosphorylation-independent activity. Finally, we used combinatorial peptide library screening to determine that PASK prefers basic residues at the P-3 and P-5 positions in substrate peptides. Our results describe the key features of the PASK structure and how those features are important for PASK activity and substrate selection. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
199. Kinase Domain Insertions Define Distinct Roles of CLK Kinases in SR Protein Phosphorylation
- Author
-
Bullock, Alex N., Das, Sanjan, Debreczeni, Judit É., Rellos, Peter, Fedorov, Oleg, Niesen, Frank H., Guo, Kunde, Papagrigoriou, Evangelos, Amos, Ann L., Cho, Suhyung, Turk, Benjamin E., Ghosh, Gourisankar, and Knapp, Stefan
- Subjects
- *
PROTEIN kinases , *PHOSPHORYLATION , *MESSENGER RNA , *SERINE , *ARGININE , *PROTEINS , *MOLECULAR structure , *CHEMICAL inhibitors - Abstract
Summary: Splicing requires reversible phosphorylation of serine/arginine-rich (SR) proteins, which direct splice site selection in eukaryotic mRNA. These phosphorylation events are dependent on SR protein (SRPK) and cdc2-like kinase (CLK) families. SRPK1 phosphorylation of splicing factors is restricted by a specific docking interaction whereas CLK activity is less constrained. To understand functional differences between splicing factor targeting kinases, we determined crystal structures of CLK1 and CLK3. Intriguingly, in CLKs the SRPK1 docking site is blocked by insertion of a previously unseen helix αH. In addition, substrate docking grooves present in related mitogen activating protein kinases (MAPKs) are inaccessible due to a CLK specific β7/8-hairpin insert. Thus, the unconstrained substrate interaction together with the determined active-site mediated substrate specificity allows CLKs to complete the functionally important hyperphosphorylation of splicing factors like ASF/SF2. In addition, despite high sequence conservation, we identified inhibitors with surprising isoform specificity for CLK1 over CLK3. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
200. Structural insights into the inhibited states of the Mer receptor tyrosine kinase
- Author
-
Huang, Xudong, Finerty, Patrick, Walker, John R., Butler-Cole, Christine, Vedadi, Masoud, Schapira, Matthieu, Parker, Sirlester A., Turk, Benjamin E., Thompson, Debra A., and Dhe-Paganon, Sirano
- Subjects
- *
THROMBOSIS , *TYROSINE , *PROTEIN S deficiency , *PROTEIN-tyrosine kinases - Abstract
Abstract: The mammalian ortholog of the retroviral oncogene v-Eyk, and a receptor tyrosine kinase upstream of antiapoptotic and transforming signals, Mer (MerTK) is a mediator of the phagocytic process, being involved in retinal and immune cell clearance and platelet aggregation. Mer knockout mice are viable and are protected from epinephrine-induced pulmonary thromboembolism and ferric chloride-induced thrombosis. Mer overexpression, on the other hand, is associated with numerous carcinomas. Although Mer adaptor proteins and signaling pathways have been identified, it remains unclear how Mer initiates phagocytosis. When bound to its nucleotide cofactor, the high-resolution structure of Mer shows an autoinhibited αC-Glu-out conformation with insertion of an activation loop residue into the active site. Mer complexed with compound-52 (C52: 2-(2-hydroxyethylamino)-6-(3-chloroanilino)-9-isopropylpurine), a ligand identified from a focused library, retains its DFG-Asp-in and αC-Glu-out conformation, but acquires other conformational changes. The αC helix and DFGL region is closer to the hinge region and the ethanolamine moiety of C52 binds in the groove formed between Leu593 and Val601 of the P-loop, causing a compression of the active site pocket. These conformational states reveal the mechanisms of autoinhibition, the pathophysiological basis of disease-causing mutations, and a platform for the development of chemical probes. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.