52 results on '"Igor Kurinov"'
Search Results
2. Discovery and Structural Characterization of Small Molecule Binders of the Human CTLH E3 Ligase Subunit GID4
- Author
-
Chetan K. Chana, Pierre Maisonneuve, Ganna Posternak, Nicolas G.A. Grinberg, Juline Poirson, Samara M. Ona, Derek F. Ceccarelli, Pavel Mader, Daniel J. St-Cyr, Victor Pau, Igor Kurinov, Xiaojing Tang, Dongjing Deng, Weiren Cui, Wenji Su, Letian Kuai, Richard Soll, Mike Tyers, Hannes L. Röst, Robert A. Batey, Mikko Taipale, Anne-Claude Gingras, and Frank Sicheri
- Subjects
Ubiquitin-Protein Ligases ,Proteolysis ,Drug Discovery ,Humans ,Molecular Medicine ,DNA - Abstract
Targeted protein degradation (TPD) strategies exploit bivalent small molecules to bridge substrate proteins to an E3 ubiquitin ligase to induce substrate degradation. Few E3s have been explored as degradation effectors due to a dearth of E3-binding small molecules. We show that genetically induced recruitment to the GID4 subunit of the CTLH E3 complex induces protein degradation. An NMR-based fragment screen followed by structure-guided analog elaboration identified two binders of GID4
- Published
- 2022
- Full Text
- View/download PDF
3. Engineered SH2 Domains for Targeted Phosphoproteomics
- Author
-
Gregory D. Martyn, Gianluca Veggiani, Ulrike Kusebauch, Seamus R. Morrone, Bradley P. Yates, Alex U. Singer, Jiefei Tong, Noah Manczyk, Gerald Gish, Zhi Sun, Igor Kurinov, Frank Sicheri, Michael F. Moran, Robert L. Moritz, and Sachdev S. Sidhu
- Subjects
src Homology Domains ,Proteome ,Humans ,Molecular Medicine ,General Medicine ,Phosphotyrosine ,Biochemistry ,Article ,Mass Spectrometry ,Protein Binding - Abstract
A comprehensive analysis of the phosphoproteome is essential for understanding molecular mechanisms of human diseases. However, current tools used to enrich phosphotyrosine (pTyr) are limited in their applicability and scope. Here, we engineered new superbinder Src-Homology 2 (SH2) domains that enrich diverse sets of pTyr-peptides. We used phage display to select a Fes-SH2 domain variant (superFes; sFes(1)) with high affinity for pTyr and solved its structure bound to a pTyr-peptide. We performed systematic structure–function analyses of the superbinding mechanisms of sFes(1) and superSrc-SH2 (sSrc(1)), another SH2 superbinder. We grafted the superbinder motifs from sFes(1) and sSrc(1) into 17 additional SH2 domains and confirmed increased binding affinity for specific pTyr-peptides. Using mass spectrometry (MS), we demonstrated that SH2 superbinders have distinct specificity profiles and superior capabilities to enrich pTyr-peptides. Finally, using combinations of SH2 superbinders as affinity purification (AP) tools we showed that unique subsets of pTyr-peptides can be enriched with unparalleled depth and coverage.
- Published
- 2022
- Full Text
- View/download PDF
4. Supplemental Table 1 from Caffeic Acid Directly Targets ERK1/2 to Attenuate Solar UV-Induced Skin Carcinogenesis
- Author
-
Zigang Dong, Ziming Dong, Ann M. Bode, Yuqiao Sheng, Do Young Lim, Wei Li, Hanyong Chen, Haitao Li, Ke Yao, Feng Zhu, Igor Kurinov, Margarita Malakhova, Yang Fu, and Ge Yang
- Abstract
X-ray data collection and refinement statistics.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplemental Figure 3 from Caffeic Acid Directly Targets ERK1/2 to Attenuate Solar UV-Induced Skin Carcinogenesis
- Author
-
Zigang Dong, Ziming Dong, Ann M. Bode, Yuqiao Sheng, Do Young Lim, Wei Li, Hanyong Chen, Haitao Li, Ke Yao, Feng Zhu, Igor Kurinov, Margarita Malakhova, Yang Fu, and Ge Yang
- Abstract
Knockdown of ERK2 in HaCaT cells decreases sensitivity to caffeic acid. A, efficiency of ERK2 shRNA in HaCaT cells. B, neoplastic transformation of HaCaT cells transfected with mock shRNA, ERK2 shRNA#1 or ERK2 shRNA#2. Cell transformation dramatically decreased after ERK2 shRNA transfection compared with sh-mock group. Data are shown as means {plus minus} S.D. The asterisk indicates a significant decrease in colony number compared with mock group (*p < 0.05). C, sensitivity of HaCaT cells transfected with sh-mock or ERK2 shRNA#1 or ERK2 shRNA#2 to treatment with caffeic acid (40 µmol/L). Data are shown as means {plus minus} S.D. The asterisk indicates a significant decrease in sensitivity to caffeic acid (*p < 0.05).
- Published
- 2023
- Full Text
- View/download PDF
6. Supplemental Figure Legend from Caffeic Acid Directly Targets ERK1/2 to Attenuate Solar UV-Induced Skin Carcinogenesis
- Author
-
Zigang Dong, Ziming Dong, Ann M. Bode, Yuqiao Sheng, Do Young Lim, Wei Li, Hanyong Chen, Haitao Li, Ke Yao, Feng Zhu, Igor Kurinov, Margarita Malakhova, Yang Fu, and Ge Yang
- Abstract
Supplemental Figure Legend
- Published
- 2023
- Full Text
- View/download PDF
7. Supplemental Figure 2 from Caffeic Acid Directly Targets ERK1/2 to Attenuate Solar UV-Induced Skin Carcinogenesis
- Author
-
Zigang Dong, Ziming Dong, Ann M. Bode, Yuqiao Sheng, Do Young Lim, Wei Li, Hanyong Chen, Haitao Li, Ke Yao, Feng Zhu, Igor Kurinov, Margarita Malakhova, Yang Fu, and Ge Yang
- Abstract
Caffeic acid attenuates MAP kinase signal transduction in HaCaT cells After starvation in serum-free medium for 24 h, cells were treated with caffeic acid at the indicated concentration for 1 h and then were exposed to solar UV (60 kJ UVA/m2, 3.6 UVB kJ/M2). They were next incubated at 37{degree sign}C in a 5% CO2 incubator for 15 min and then harvested and protein levels were determined by Western blotting.
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Figure 1 from Caffeic Acid Directly Targets ERK1/2 to Attenuate Solar UV-Induced Skin Carcinogenesis
- Author
-
Zigang Dong, Ziming Dong, Ann M. Bode, Yuqiao Sheng, Do Young Lim, Wei Li, Hanyong Chen, Haitao Li, Ke Yao, Feng Zhu, Igor Kurinov, Margarita Malakhova, Yang Fu, and Ge Yang
- Abstract
Caffeic acid attenuates EGF-induced cell transformation. A, caffeic acid is not toxic to HaCaT cells. Cells were treated with caffeic acid (0-80 µmol/L) or its vehicle, DMSO (as a negative control) in 10% FBS/DMEM for 24 or 48 h. Cell viability was determined by MTS assay and data are represented as means {plus minus} S.D. No significant difference was observed between any groups. B, caffeic acid inhibits EGF-induced neoplastic transformation of HaCaT cells. Data are shown as means {plus minus} S.D from 3 independent experiments and the asterisk indicates a significant (*p < 0.05) decrease in colony formation in cells treated with caffeic acid compared with the DMSO-treated group.
- Published
- 2023
- Full Text
- View/download PDF
9. Identification of
- Author
-
Monica, Bubenik, Pavel, Mader, Philippe, Mochirian, Fréderic, Vallée, Jillian, Clark, Jean-François, Truchon, Alexander L, Perryman, Victor, Pau, Igor, Kurinov, Karl E, Zahn, Marie-Eve, Leclaire, Robert, Papp, Marie-Claude, Mathieu, Martine, Hamel, Nicole M, Duffy, Claude, Godbout, Matias, Casas-Selves, Jean-Pierre, Falgueyret, Prasamit S, Baruah, Olivier, Nicolas, Rino, Stocco, Hugo, Poirier, Giovanni, Martino, Alexanne Bonneau, Fortin, Anne, Roulston, Amandine, Chefson, Stéphane, Dorich, Miguel, St-Onge, Purvish, Patel, Charles, Pellerin, Stéphane, Ciblat, Thomas, Pinter, Francis, Barabé, Majida, El Bakkouri, Paranjay, Parikh, Christian, Gervais, Agnel, Sfeir, Yael, Mamane, Stephen J, Morris, W Cameron, Black, Frank, Sicheri, and Michel, Gallant
- Subjects
DNA Replication ,Ovarian Neoplasms ,Mice ,Drug Design ,Drug Discovery ,Animals ,Humans ,Female ,DNA-Directed DNA Polymerase - Abstract
DNA polymerase theta (Polθ) is an attractive synthetic lethal target for drug discovery, predicted to be efficacious against breast and ovarian cancers harboring BRCA-mutant alleles. Here, we describe our hit-to-lead efforts in search of a selective inhibitor of human Polθ (encoded by POLQ). A high-throughput screening campaign of 350,000 compounds identified an 11 micromolar hit, giving rise to the N2-substituted fused pyrazolo series, which was validated by biophysical methods. Structure-based drug design efforts along with optimization of cellular potency and ADME ultimately led to the identification of
- Published
- 2022
10. Native SAD phasing at room temperature
- Author
-
Jack B. Greisman, Kevin M. Dalton, Candice J. Sheehan, Margaret A. Klureza, Igor Kurinov, and Doeke R. Hekstra
- Subjects
Models, Molecular ,Structural Biology ,Protein Conformation ,Temperature ,Proteins ,Crystallization ,Crystallography, X-Ray - Abstract
Single-wavelength anomalous diffraction (SAD) is a routine method for overcoming the phase problem when solving macromolecular structures. This technique requires the accurate measurement of intensities to determine differences between Bijvoet pairs. Although SAD experiments are commonly conducted at cryogenic temperatures to mitigate the effects of radiation damage, such temperatures can alter the conformational ensemble of the protein and may impede the merging of data from multiple crystals due to non-uniform freezing. Here, a strategy is presented to obtain high-quality data from room-temperature, single-crystal experiments. To illustrate the strengths of this approach, native SAD phasing at 6.55 keV was used to solve four structures of three model systems at 295 K. The resulting data sets allow automatic phasing and model building, and reveal alternate conformations that reflect the structure of proteins at room temperature.
- Published
- 2021
11. Crystal structure of the CDK11 kinase domain bound to the small-molecule inhibitor OTS964
- Author
-
Susan Kelso, Siobhan O’Brien, Igor Kurinov, Stephane Angers, and Frank Sicheri
- Subjects
Structural Biology ,Molecular Biology - Published
- 2022
- Full Text
- View/download PDF
12. Structural basis for auxiliary subunit KCTD16 regulation of the GABA B receptor
- Author
-
Ming Zhou, Emmanuel Sturchler, Jonathan Liu, Ian W. Glaaser, Haonan Wang, Aurel Frangaj, Yong Geng, Lidia Mosyak, Hao Zuo, Qing R. Fan, Jinseo Park, Yulin Zhao, Paul A. Slesinger, Patricia McDonald, and Igor Kurinov
- Subjects
Models, Molecular ,Binding Sites ,Crystallography ,Multidisciplinary ,Chemistry ,Protein subunit ,Intracellular Signaling Peptides and Proteins ,Nerve Tissue Proteins ,Regulatory site ,GABAB receptor ,Neurotransmission ,Potassium channel ,Metabotropic receptor ,PNAS Plus ,Receptors, GABA-B ,nervous system ,Biophysics ,Humans ,G protein-coupled inwardly-rectifying potassium channel ,Receptor ,Protein Binding ,Signal Transduction - Abstract
Metabotropic GABA B receptors mediate a significant fraction of inhibitory neurotransmission in the brain. Native GABA B receptor complexes contain the principal subunits GABA B1 and GABA B2 , which form an obligate heterodimer, and auxiliary subunits, known as potassium channel tetramerization domain-containing proteins (KCTDs). KCTDs interact with GABA B receptors and modify the kinetics of GABA B receptor signaling. Little is known about the molecular mechanism governing the direct association and functional coupling of GABA B receptors with these auxiliary proteins. Here, we describe the high-resolution structure of the KCTD16 oligomerization domain in complex with part of the GABA B2 receptor. A single GABA B2 C-terminal peptide is bound to the interior of an open pentamer formed by the oligomerization domain of five KCTD16 subunits. Mutation of specific amino acids identified in the structure of the GABA B2 –KCTD16 interface disrupted both the biochemical association and functional modulation of GABA B receptors and G protein-activated inwardly rectifying K + channel (GIRK) channels. These interfacial residues are conserved among KCTDs, suggesting a common mode of KCTD interaction with GABA B receptors. Defining the binding interface of GABA B receptor and KCTD reveals a potential regulatory site for modulating GABA B -receptor function in the brain.
- Published
- 2019
- Full Text
- View/download PDF
13. Bipartite binding of the N terminus of Skp2 to cyclin A
- Author
-
Gerald D. Gish, Stephen Orlicky, Derek F. Ceccarelli, Igor Kurinov, Frank Sicheri, Susan Kelso, and Jonah Beenstock
- Subjects
0303 health sciences ,Binding Sites ,biology ,Kinase ,Chemistry ,030302 biochemistry & molecular biology ,Cyclin A ,Cyclin-dependent kinase 2 ,Cell cycle ,Article ,Ubiquitin ligase ,Cell biology ,Molecular Docking Simulation ,03 medical and health sciences ,Structural Biology ,Cyclin-dependent kinase ,biology.protein ,SKP2 ,Humans ,Molecular Biology ,S-Phase Kinase-Associated Proteins ,030304 developmental biology ,Cyclin ,Protein Binding - Abstract
Summary Skp2 and cyclin A are cell-cycle regulators that control the activity of CDK2. Cyclin A acts as an activator and substrate recruitment factor of CDK2, while Skp2 mediates the ubiquitination and subsequent destruction of the CDK inhibitor protein p27. The N terminus of Skp2 can interact directly with cyclin A but is not required for p27 ubiquitination. To gain insight into this poorly understood interaction, we have solved the 3.2 A X-ray crystal structure of the N terminus of Skp2 bound to cyclin A. The structure reveals a bipartite mode of interaction with two motifs in Skp2 recognizing two discrete surfaces on cyclin A. The uncovered binding mechanism allows for a rationalization of the inhibitory effect of Skp2 on CDK2-cyclin A kinase activity toward the RxL motif containing substrates and raises the possibility that other intermolecular regulators and substrates may use similar non-canonical modes of interaction for cyclin targeting.
- Published
- 2021
14. Structural Insights into the Induced-fit Inhibition of Fascin by a Small-Molecule Inhibitor
- Author
-
Xin-Yun Huang, Jianyun Huang, Igor Kurinov, Jean Jakoncic, Raja Dey, and Yufeng Wang
- Subjects
Models, Molecular ,0301 basic medicine ,Protein Conformation ,macromolecular substances ,Plasma protein binding ,Cell Surface Extension ,Crystallography, X-Ray ,Article ,Metastasis ,Small Molecule Libraries ,03 medical and health sciences ,Structural Biology ,medicine ,Humans ,Molecular Biology ,Fascin ,Binding Sites ,biology ,Chemistry ,Microfilament Proteins ,Actin cytoskeleton ,medicine.disease ,Small molecule ,Cell biology ,030104 developmental biology ,Mechanism of action ,Mutation ,biology.protein ,medicine.symptom ,Carrier Proteins ,Filopodia ,Protein Binding - Abstract
Tumor metastasis is responsible for ~90% of all cancer deaths. One of the key steps of tumor metastasis is tumor cell migration and invasion. Filopodia are cell surface extensions that are critical for tumor cell migration. Fascin protein is the main actin-bundling protein in filopodia. Small-molecule fascin inhibitors block tumor cell migration, invasion, and metastasis. Here we present the structural basis for the mechanism of action of these small-molecule fascin inhibitors. X-ray crystal structural analysis of a complex of fascin and a fascin inhibitor shows that binding of the fascin inhibitor to the hydrophobic cleft between the domains 1 and 2 of fascin induces a ~35o rotation of domain 1, leading to the distortion of both the actin-binding sites 1 and 2 on fascin. Furthermore, the crystal structures of an inhibitor alone indicate that the conformations of the small-molecule inhibitors are dynamic. Mutations of the inhibitor-interacting residues decrease the sensitivity of fascin to the inhibitors. Our studies provide structural insights into the molecular mechanism of fascin protein function as well as the action of small-molecule fascin inhibitors.
- Published
- 2018
- Full Text
- View/download PDF
15. MEK drives BRAF activation through allosteric control of KSR proteins
- Author
-
Frank Sicheri, Ting Jin, Hugo Lavoie, Sara A. Marullo, Pierre Maisonneuve, Neroshan Thevakumaran, Marc Therrien, Malha Sahmi, and Igor Kurinov
- Subjects
Models, Molecular ,Proto-Oncogene Proteins B-raf ,0301 basic medicine ,MAP Kinase Kinase 2 ,Protein domain ,MAP Kinase Kinase 1 ,Protein Serine-Threonine Kinases ,Mitogen-activated protein kinase kinase ,Crystallography, X-Ray ,KSR1 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Allosteric Regulation ,Protein Domains ,Humans ,Phosphorylation ,Protein kinase A ,Mitogen-Activated Protein Kinase Kinases ,Multidisciplinary ,Kinase ,Chemistry ,Cell biology ,Enzyme Activation ,030104 developmental biology ,Protein kinase domain ,030220 oncology & carcinogenesis ,Protein Multimerization ,ARAF ,Protein Kinases ,Protein Binding ,Signal Transduction - Abstract
RAF family kinases have prominent roles in cancer. Their activation is dependent on dimerization of their kinase domains, which has emerged as a hindrance for drug development. In mammals, RAF family kinases include three catalytically competent enzymes (ARAF, BRAF and CRAF) and two pseudokinases (KSR1 and KSR2) that have been described as scaffolds owing to their apparent ability to bridge RAF isoforms and their substrate, mitogen-activated protein kinase kinase (MEK). Kinase suppressor of Ras (KSR) pseudokinases were also shown to dimerize with kinase-competent RAFs to stimulate catalysis allosterically. Although GTP-bound RAS can modulate the dimerization of RAF isoforms by engaging their RAS-binding domains, KSR1 and KSR2 lack an RAS-binding domain and therefore the regulatory principles underlying their dimerization with other RAF family members remain unknown. Here we show that the selective heterodimerization of BRAF with KSR1 is specified by direct contacts between the amino-terminal regulatory regions of each protein, comprising in part a novel domain called BRS in BRAF and the coiled-coil-sterile α motif (CC-SAM) domain in KSR1. We also discovered that MEK binding to the kinase domain of KSR1 asymmetrically drives BRAF-KSR1 heterodimerization, resulting in the concomitant stimulation of BRAF catalytic activity towards free MEK molecules. These findings demonstrate that KSR-MEK complexes allosterically activate BRAF through the action of N-terminal regulatory region and kinase domain contacts and challenge the accepted role of KSR as a scaffold for MEK recruitment to RAF.
- Published
- 2018
- Full Text
- View/download PDF
16. Ultra-High Resolution and Charge-Density Studies on the Type-I Copper Protein Amicyanin, from Paracoccus Denitrificans
- Author
-
Sahana L. Sukumar, James Withrow, Igor Kurinov, Victor L. Davidson, Narayanasami Sukumar, and Malcolm Capel
- Subjects
Crystallography ,Amicyanin ,biology ,Chemistry ,Copper protein ,Biophysics ,biology.protein ,Charge density ,Paracoccus denitrificans ,biology.organism_classification ,Ultra high resolution - Published
- 2021
- Full Text
- View/download PDF
17. Crystal structure of the human Polϵ B-subunit in complex with the C-terminal domain of the catalytic subunit
- Author
-
Andrey G. Baranovskiy, Jianyou Gu, Nigar D. Babayeva, Igor Kurinov, Youri I. Pavlov, and Tahir H. Tahirov
- Subjects
Models, Molecular ,0301 basic medicine ,030102 biochemistry & molecular biology ,DNA-Directed DNA Polymerase ,Cell Biology ,DNA and Chromosomes ,Crystallography, X-Ray ,Biochemistry ,Protein Subunits ,03 medical and health sciences ,030104 developmental biology ,Catalytic Domain ,Humans ,Amino Acid Sequence ,Molecular Biology ,Protein Binding - Abstract
The eukaryotic B-family DNA polymerases include four members: Polα, Polδ, Polϵ, and Polζ, which share common architectural features, such as the exonuclease/polymerase and C-terminal domains (CTDs) of catalytic subunits bound to indispensable B-subunits, which serve as scaffolds that mediate interactions with other components of the replication machinery. Crystal structures for the B-subunits of Polα and Polδ/Polζ have been reported: the former within the primosome and separately with CTD and the latter with the N-terminal domain of the C-subunit. Here we present the crystal structure of the human Polϵ B-subunit (p59) in complex with CTD of the catalytic subunit (p261C). The structure revealed a well defined electron density for p261C and the phosphodiesterase and oligonucleotide/oligosaccharide-binding domains of p59. However, electron density was missing for the p59 N-terminal domain and for the linker connecting it to the phosphodiesterase domain. Similar to Polα, p261C of Polϵ contains a three-helix bundle in the middle and zinc-binding modules on each side. Intersubunit interactions involving 11 hydrogen bonds and numerous hydrophobic contacts account for stable complex formation with a buried surface area of 3094 Å2. Comparative structural analysis of p59–p261C with the corresponding Polα complex revealed significant differences between the B-subunits and CTDs, as well as their interaction interfaces. The B-subunit of Polδ/Polζ also substantially differs from B-subunits of either Polα or Polϵ. This work provides a structural basis to explain biochemical and genetic data on the importance of B-subunit integrity in replisome function in vivo.
- Published
- 2017
- Full Text
- View/download PDF
18. Regulation of Protein Interactions by Mps One Binder (MOB1) Phosphorylation
- Author
-
Anne-Claude Gingras, Amber L. Couzens, Frank Sicheri, Daniel Y. Mao, Michelle J. Kean, Shawn Xiong, Sebastian Guettler, and Igor Kurinov
- Subjects
0301 basic medicine ,Protein-Serine-Threonine Kinases ,030102 biochemistry & molecular biology ,Kinase ,Upstream and downstream (transduction) ,Phosphatase ,Signal transducing adaptor protein ,Biology ,Biochemistry ,Analytical Chemistry ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Mitotic exit ,Phosphorylation ,Signal transduction ,Molecular Biology - Abstract
MOB1 is a multifunctional protein best characterized for its integrative role in regulating Hippo and NDR pathway signaling in metazoans and the Mitotic Exit Network in yeast. Human MOB1 binds both the upstream kinases MST1 and MST2 and the downstream AGC group kinases LATS1, LATS2, NDR1, and NDR2. Binding of MOB1 to MST1 and MST2 is mediated by its phosphopeptide-binding infrastructure, the specificity of which matches the phosphorylation consensus of MST1 and MST2. On the other hand, binding of MOB1 to the LATS and NDR kinases is mediated by a distinct interaction surface on MOB1. By assembling both upstream and downstream kinases into a single complex, MOB1 facilitates the activation of the latter by the former through a trans-phosphorylation event. Binding of MOB1 to its upstream partners also renders MOB1 a substrate, which serves to differentially regulate its two protein interaction activities (at least in vitro). Our previous interaction proteomics analysis revealed that beyond associating with MST1 (and MST2), MOB1A and MOB1B can associate in a phosphorylation-dependent manner with at least two other signaling complexes, one containing the Rho guanine exchange factors (DOCK6-8) and the other containing the serine/threonine phosphatase PP6. Whether these complexes are recruited through the same mode of interaction as MST1 and MST2 remains unknown. Here, through a comprehensive set of biochemical, biophysical, mutational and structural studies, we quantitatively assess how phosphorylation of MOB1A regulates its interaction with both MST kinases and LATS/NDR family kinases in vitro. Using interaction proteomics, we validate the significance of our in vitro studies and also discover that the phosphorylation-dependent recruitment of PP6 phosphatase and Rho guanine exchange factor protein complexes differ in key respects from that elucidated for MST1 and MST2. Together our studies confirm and extend previous work to delineate the intricate regulatory steps in key signaling pathways.
- Published
- 2017
- Full Text
- View/download PDF
19. Rigidification Dramatically Improves Inhibitor Selectivity for RAF Kinases
- Author
-
Frank Sicheri, Caiqun Yu, Pierre Maisonneuve, Chao Zhang, Igor Kurinov, and Amir Assadieskandar
- Subjects
Pyrimidine ,010405 organic chemistry ,Chemistry ,Kinase ,Organic Chemistry ,Dabrafenib ,01 natural sciences ,Biochemistry ,Pyrazolopyrimidine ,0104 chemical sciences ,Cell biology ,010404 medicinal & biomolecular chemistry ,chemistry.chemical_compound ,Protein kinase domain ,Cell culture ,Drug Discovery ,medicine ,Kinome ,Vemurafenib ,medicine.drug - Abstract
[Image: see text] One effective means to achieve inhibitor specificity for RAF kinases, an important family of cancer drug targets, has been to target the monomeric inactive state conformation of the kinase domain, which, unlike most other kinases, can accommodate sulfonamide-containing drugs such as vemurafenib and dabrafenib because of the presence of a unique pocket specific to inactive RAF kinases. We previously reported an alternate strategy whereby rigidification of a nonselective pyrazolo[3,4-d]pyrimidine-based inhibitor through ring closure afforded moderate but appreciable increases in selectivity for RAF kinases. Here, we show that a further application of the rigidification strategy to a different pyrazolopyrimidine-based scaffold dramatically improved selectivity for RAF kinases. Crystal structure analysis confirmed our inhibitor design hypothesis revealing that 2l engages an active-like state conformation of BRAF normally associated with poorly discriminating inhibitors. When screened against a panel of distinct cancer cell lines, the optimized inhibitor 2l primarily inhibited the proliferation of the expected BRAF(V600E)-harboring cell lines consistent with its kinome selectivity profile. These results suggest that rigidification could be a general and powerful strategy for enhancing inhibitor selectivity against protein kinases, which may open up therapeutic opportunities not afforded by other approaches.
- Published
- 2019
20. Crystal structure of human PACRG in complex with MEIG1 reveals roles in axoneme formation and tubulin binding
- Author
-
Dylan Pelletier, Khanh Huy Bui, Corbin Black, Andrew N. Bayne, Muneyoshi Ichikawa, Nimra Khan, Simon Veyron, Ahmad Abdelzaher Zaki Khalifa, Igor Kurinov, Susanne Bechstedt, Thomas S. McAlear, Jean-François Trempe, Sami Chaaban, Gary J. Brouhard, and Nathalie Croteau
- Subjects
Axoneme ,Protein Conformation ,Cell Cycle Proteins ,Flagellum ,Crystallography, X-Ray ,Article ,Tubulin binding ,03 medical and health sciences ,Protein Domains ,Tubulin ,Structural Biology ,Microtubule ,Humans ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Binding Sites ,biology ,Chemistry ,Cilium ,Microfilament Proteins ,030302 biochemistry & molecular biology ,Nuclear Proteins ,Signal transducing adaptor protein ,Phosphoproteins ,Single Molecule Imaging ,Axoneme assembly ,3. Good health ,Cell biology ,Microscopy, Fluorescence ,Multiprotein Complexes ,Mutation ,biology.protein ,Chlamydomonas reinhardtii ,Molecular Chaperones ,Protein Binding - Abstract
The Parkin co-regulated gene protein (PACRG) binds at the inner junction between doublet microtubules of the axoneme, a structure found in flagella and cilia. PACRG binds to the adaptor protein meiosis expressed gene 1 (MEIG1), but how they bind to microtubules is unknown. Here, we report the crystal structure of human PACRG in complex with MEIG1. PACRG adopts a helical repeat fold with a loop that interacts with MEIG1. Using the structure of the axonemal doublet microtubule from the protozoan Chlamydomonas reinhardtii and single-molecule fluorescence microscopy, we propose that PACRG binds to microtubules while simultaneously recruiting free tubulin to catalyze formation of the inner junction. We show that the homologous PACRG-like protein also mediates dual tubulin interactions but does not bind MEIG1. Our findings establish a framework to assess the function of the PACRG family of proteins and MEIG1 in regulating axoneme assembly.
- Published
- 2021
- Full Text
- View/download PDF
21. Structural and functional characterization of KEOPS dimerization by Pcc1 and its role in t6A biosynthesis
- Author
-
Daniel Durocher, Leo C. K. Wan, Neroshan Thevakumaran, Frank Sicheri, Monica C. Pillon, Avi Chakrabartty, Yulong Sun, Alba Guarné, and Igor Kurinov
- Subjects
0301 basic medicine ,TRNA modification ,Adenosine ,Archaeal Proteins ,Protein subunit ,Crystallography, X-Ray ,Biophysical Phenomena ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,X-Ray Diffraction ,Biosynthesis ,Structural Biology ,Scattering, Small Angle ,Genetics ,Scattering, Radiation ,Transferase ,Structure–activity relationship ,biology ,biology.organism_classification ,Pyrococcus furiosus ,Solutions ,030104 developmental biology ,Biochemistry ,chemistry ,Multiprotein Complexes ,Transfer RNA ,Chromatography, Gel ,Protein Multimerization ,Function (biology) - Abstract
KEOPS is an ancient protein complex required for the biosynthesis of N6-threonylcarbamoyladenosine (t(6)A), a universally conserved tRNA modification found on all ANN-codon recognizing tRNAs. KEOPS consist minimally of four essential subunits, namely the proteins Kae1, Bud32, Cgi121 and Pcc1, with yeast possessing the fifth essential subunit Gon7. Bud32, Cgi121, Pcc1 and Gon7 appear to have evolved to regulate the central t(6)A biosynthesis function of Kae1, but their precise function and mechanism of action remains unclear. Pcc1, in particular, binds directly to Kae1 and by virtue of its ability to form dimers in solution and in crystals, Pcc1 was inferred to function as a dimerization module for Kae1 and therefore KEOPS. We now present a 3.4 Å crystal structure of a dimeric Kae1-Pcc1 complex providing direct evidence that Pcc1 can bind and dimerize Kae1. Further biophysical analysis of a complete archaeal KEOPS complex reveals that Pcc1 facilitates KEOPS dimerization in vitro Interestingly, while Pcc1-mediated dimerization of KEOPS is required to support the growth of yeast, it is dispensable for t(6)A biosynthesis by archaeal KEOPS in vitro, raising the question of how precisely Pcc1-mediated dimerization impacts cellular biology.
- Published
- 2016
- Full Text
- View/download PDF
22. Inhibition of SCF ubiquitin ligases by engineered ubiquitin variants that target the Cul1 binding site on the Skp1–F-box interface
- Author
-
Jason Moffat, Maria A. Sartori, Igor Kurinov, Jack Greenblatt, Edyta Marcon, Maryna Gorelik, Xiaojing Tang, Mike Tyers, Frank Sicheri, Sachdev S. Sidhu, and Stephen Orlicky
- Subjects
Models, Molecular ,0301 basic medicine ,F-Box-WD Repeat-Containing Protein 7 ,Phage display ,Ubiquitin-Protein Ligases ,Molecular Sequence Data ,Cell Cycle Proteins ,Protein Engineering ,Bioinformatics ,03 medical and health sciences ,Ubiquitin ,Peptide Library ,Basic research ,Skp1 ,Humans ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Ligase activity ,Enzyme Inhibitors ,Binding site ,Ubiquitins ,Binding Sites ,SKP Cullin F-Box Protein Ligases ,Multidisciplinary ,Sequence Homology, Amino Acid ,biology ,Chemistry ,F-Box Proteins ,Genetic Variation ,Biological Sciences ,Cullin Proteins ,beta-Transducin Repeat-Containing Proteins ,3. Good health ,Ubiquitin ligase ,Cell biology ,030104 developmental biology ,Drug Design ,biology.protein ,CUL1 - Abstract
Skp1-Cul1-F-box (SCF) E3 ligases play key roles in multiple cellular processes through ubiquitination and subsequent degradation of substrate proteins. Although Skp1 and Cul1 are invariant components of all SCF complexes, the 69 different human F-box proteins are variable substrate binding modules that determine specificity. SCF E3 ligases are activated in many cancers and inhibitors could have therapeutic potential. Here, we used phage display to develop specific ubiquitin-based inhibitors against two F-box proteins, Fbw7 and Fbw11. Unexpectedly, the ubiquitin variants bind at the interface of Skp1 and F-box proteins and inhibit ligase activity by preventing Cul1 binding to the same surface. Using structure-based design and phage display, we modified the initial inhibitors to generate broad-spectrum inhibitors that targeted many SCF ligases, or conversely, a highly specific inhibitor that discriminated between even the close homologs Fbw11 and Fbw1. We propose that most F-box proteins can be targeted by this approach for basic research and for potential cancer therapies.
- Published
- 2016
- Full Text
- View/download PDF
23. Structural and Functional Analysis of Ubiquitin-based Inhibitors That Target the Backsides of E2 Enzymes
- Author
-
Alexander F.A. Keszei, Pankaj Garg, Frank Sicheri, Sachdev S. Sidhu, Derek F. Ceccarelli, and Igor Kurinov
- Subjects
Phage display ,Protein Engineering ,Protein Structure, Secondary ,Article ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Structural Biology ,Peptide Library ,Transferase ,Humans ,Amino Acid Sequence ,Enzyme Inhibitors ,Molecular Biology ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Molecular interactions ,biology ,Functional analysis ,Ubiquitination ,Active site ,Protein engineering ,3. Good health ,Cell biology ,Enzyme ,chemistry ,Ubiquitin-Conjugating Enzymes ,biology.protein ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Ubiquitin-conjugating E2 enzymes are central to the ubiquitination cascade and have been implicated in cancer and other diseases. Despite strong interest in developing specific E2 inhibitors, the shallow and exposed active site has proven recalcitrant to targeting with reversible small-molecule inhibitors. Here, we used phage display to generate highly potent and selective ubiquitin variants (UbVs) that target the E2 backside, which is located opposite to the active site. A UbV targeting Ube2D1 did not affect charging but greatly attenuated chain elongation. Likewise, a UbV targeting the E2 variant Ube2V1 did not interfere with the charging of its partner E2 enzyme but inhibited formation of diubiquitin. In contrast, a UbV that bound to the backside of Ube2G1 impeded the generation of thioester-linked ubiquitin to the active site cysteine of Ube2G1 by the E1 enzyme. Crystal structures of UbVs in complex with three E2 proteins revealed distinctive molecular interactions in each case, but they also highlighted a common backside pocket that the UbVs used for enhanced affinity and specificity. These findings validate the E2 backside as a target for inhibition and provide structural insights to aid inhibitor design and screening efforts.
- Published
- 2018
24. Structure of human ADP-ribosyl-acceptor hydrolase 3 bound to ADP-ribose reveals a conformational switch that enables specific substrate recognition
- Author
-
Joel Moss, In-Kwon Kim, Yasin Pourfarjam, Igor Kurinov, Jessica Ventura, and Ahra Cho
- Subjects
0301 basic medicine ,Models, Molecular ,Conformational change ,Glycoside Hydrolases ,Stereochemistry ,Protein Conformation ,Sequence Homology ,Crystallography, X-Ray ,Biochemistry ,Catalysis ,Substrate Specificity ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Protein structure ,Catalytic Domain ,Hydrolase ,Ribose ,Humans ,Amino Acid Sequence ,Molecular Biology ,Adenosine Diphosphate Ribose ,biology ,Adenosine diphosphate ribose ,Hydrolysis ,Active site ,Substrate (chemistry) ,Cell Biology ,030104 developmental biology ,Structural biology ,chemistry ,030220 oncology & carcinogenesis ,Protein Structure and Folding ,biology.protein - Abstract
ADP-ribosyl-acceptor hydrolase 3 (ARH3) plays important roles in regulation of poly(ADP-ribosyl)ation, a reversible post-translational modification, and in maintenance of genomic integrity. ARH3 degrades poly(ADP-ribose) to protect cells from poly(ADP-ribose)–dependent cell death, reverses serine mono(ADP-ribosyl)ation, and hydrolyzes O-acetyl-ADP-ribose, a product of Sirtuin-catalyzed histone deacetylation. ARH3 preferentially hydrolyzes O-linkages attached to the anomeric C1″ of ADP-ribose; however, how ARH3 specifically recognizes and cleaves structurally diverse substrates remains unknown. Here, structures of full-length human ARH3 bound to ADP-ribose and Mg(2+), coupled with computational modeling, reveal a dramatic conformational switch from closed to open states that enables specific substrate recognition. The glutamate flap, which blocks substrate entrance to Mg(2+) in the unliganded closed state, is ejected from the active site when substrate is bound. This closed-to-open transition significantly widens the substrate-binding channel and precisely positions the scissile 1″-O-linkage for cleavage while securing tightly 2″- and 3″-hydroxyls of ADP-ribose. Our collective data uncover an unprecedented structural plasticity of ARH3 that supports its specificity for the 1″-O-linkage in substrates and Mg(2+)-dependent catalysis.
- Published
- 2018
25. Structural Basis for Auto-Inhibition of the NDR1 Kinase Domain by an Atypically Long Activation Segment
- Author
-
Shawn, Xiong, Kristina, Lorenzen, Amber L, Couzens, Catherine M, Templeton, Dushyandi, Rajendran, Daniel Y L, Mao, Yu-Chi, Juang, David, Chiovitti, Igor, Kurinov, Sebastian, Guettler, Anne-Claude, Gingras, and Frank, Sicheri
- Subjects
Models, Molecular ,Protein Conformation, alpha-Helical ,Hippo tumor suppressor pathway ,Genetic Vectors ,Gene Expression ,Cell Cycle Proteins ,Protein Serine-Threonine Kinases ,Crystallography, X-Ray ,Serine-Threonine Kinase 3 ,Article ,Substrate Specificity ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Escherichia coli ,Humans ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Cloning, Molecular ,FRYL ,Adaptor Proteins, Signal Transducing ,X-ray crystallography ,Binding Sites ,Sequence Homology, Amino Acid ,Hepatocyte Growth Factor ,Epithelial Cells ,protein kinase ,Furry-like ,MOB1 ,Recombinant Proteins ,Kinetics ,HEK293 Cells ,kinases ,Gene Expression Regulation ,Mutation ,NDR family ,Protein Conformation, beta-Strand ,Microtubule-Associated Proteins ,Sequence Alignment ,Protein Binding ,Signal Transduction - Abstract
Summary The human NDR family kinases control diverse aspects of cell growth, and are regulated through phosphorylation and association with scaffolds such as MOB1. Here, we report the crystal structure of the human NDR1 kinase domain in its non-phosphorylated state, revealing a fully resolved atypically long activation segment that blocks substrate binding and stabilizes a non-productive position of helix αC. Consistent with an auto-inhibitory function, mutations within the activation segment of NDR1 dramatically enhance in vitro kinase activity. Interestingly, NDR1 catalytic activity is further potentiated by MOB1 binding, suggesting that regulation through modulation of the activation segment and by MOB1 binding are mechanistically distinct. Lastly, deleting the auto-inhibitory activation segment of NDR1 causes a marked increase in the association with upstream Hippo pathway components and the Furry scaffold. These findings provide a point of departure for future efforts to explore the cellular functions and the mechanism of NDR1., Graphical Abstract, Highlights • 2.2 Å crystal structure of an inactive human NDR1 kinase domain is determined • An atypically long activation segment auto-inhibits the NDR1 kinase domain • MOB1 and the activation segment of NDR1 act through independent mechanisms • The activation segment of NDR1 influences interaction with MST1/2 and Furry, NDR family kinases play important roles in regulating cell cycle and growth from yeast to human. To understand how NDR family kinases are regulated, Xiong et al. determine the crystal structure of human NDR1 kinase domain in its non-phosphorylated inactive state.
- Published
- 2017
26. Effects of rigidity on the selectivity of protein kinase inhibitors
- Author
-
Amir Assadieskandar, Pierre Maisonneuve, Igor Kurinov, Caiqun Yu, Chao Zhang, Ying-Chu Chen, Xu Liu, G. K. Surya Prakash, and Frank Sicheri
- Subjects
0301 basic medicine ,Sequence analysis ,01 natural sciences ,Article ,03 medical and health sciences ,Structure-Activity Relationship ,Signaling proteins ,Drug Discovery ,Humans ,Protein kinase A ,Protein Kinase Inhibitors ,Pharmacology ,ABL ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Kinase ,Chemistry ,Organic Chemistry ,General Medicine ,Kinase inhibition ,Isoquinolines ,0104 chemical sciences ,3. Good health ,030104 developmental biology ,Biochemistry ,Selectivity ,Protein Kinases - Abstract
Established strategies for discovering selective kinase inhibitors are target-centric as they often target certain structural or reactive features in the target kinase. In the absence of such prominent features, there is a lack of general methods for discovering selective inhibitors. Here we describe a new strategy that exploits conformational flexibility of kinases for achieving selective kinase inhibition. Through ring closure, we designed and synthesized a panel of isoquinoline-containing compounds as rigidified analogs of an amidophenyl-containing parent compound. These analogs potently inhibit kinases including Abl and BRAF but have diminished inhibition against some other kinases compared to the parent compound. Sequence analysis reveals that many of the kinases that are potently inhibited by the isoquonoline-containing compounds contain a long insertion within their catalytic domains. A crystal structure of one rigid compound bound to BRAF confirmed its binding mode. Our findings highlight the potential of a novel strategy of rigidification for improving the selectivity of kinase inhibitors.
- Published
- 2017
27. Regulation of Protein Interactions by
- Author
-
Shawn, Xiong, Amber L, Couzens, Michelle J, Kean, Daniel Y, Mao, Sebastian, Guettler, Igor, Kurinov, Anne-Claude, Gingras, and Frank, Sicheri
- Subjects
Proteomics ,Research ,Intracellular Signaling Peptides and Proteins ,Phosphoprotein Phosphatases ,Phosphorylation ,Protein Serine-Threonine Kinases ,Serine-Threonine Kinase 3 ,Rho Guanine Nucleotide Exchange Factors ,Adaptor Proteins, Signal Transducing - Abstract
MOB1 is a multifunctional protein best characterized for its integrative role in regulating Hippo and NDR pathway signaling in metazoans and the Mitotic Exit Network in yeast. Human MOB1 binds both the upstream kinases MST1 and MST2 and the downstream AGC group kinases LATS1, LATS2, NDR1, and NDR2. Binding of MOB1 to MST1 and MST2 is mediated by its phosphopeptide-binding infrastructure, the specificity of which matches the phosphorylation consensus of MST1 and MST2. On the other hand, binding of MOB1 to the LATS and NDR kinases is mediated by a distinct interaction surface on MOB1. By assembling both upstream and downstream kinases into a single complex, MOB1 facilitates the activation of the latter by the former through a trans-phosphorylation event. Binding of MOB1 to its upstream partners also renders MOB1 a substrate, which serves to differentially regulate its two protein interaction activities (at least in vitro). Our previous interaction proteomics analysis revealed that beyond associating with MST1 (and MST2), MOB1A and MOB1B can associate in a phosphorylation-dependent manner with at least two other signaling complexes, one containing the Rho guanine exchange factors (DOCK6-8) and the other containing the serine/threonine phosphatase PP6. Whether these complexes are recruited through the same mode of interaction as MST1 and MST2 remains unknown. Here, through a comprehensive set of biochemical, biophysical, mutational and structural studies, we quantitatively assess how phosphorylation of MOB1A regulates its interaction with both MST kinases and LATS/NDR family kinases in vitro. Using interaction proteomics, we validate the significance of our in vitro studies and also discover that the phosphorylation-dependent recruitment of PP6 phosphatase and Rho guanine exchange factor protein complexes differ in key respects from that elucidated for MST1 and MST2. Together our studies confirm and extend previous work to delineate the intricate regulatory steps in key signaling pathways.
- Published
- 2017
28. Structures of CRISPR Cas3 offer mechanistic insights into Cascade-activated DNA unwinding and degradation
- Author
-
Farchione, YW Huo, Yibei Xiao, RG Zhang, Fran Ding, Ki Hyun Nam, S Zhou, Lijie Wu, Ailong Ke, Kanagalaghatta R. Rajashankar, Igor Kurinov, and HJ Lee
- Subjects
Models, Molecular ,Protein Conformation ,CRISPR-Associated Proteins ,Molecular Sequence Data ,DNA, Single-Stranded ,Crystallography, X-Ray ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Adenosine Triphosphate ,Protein structure ,Structural Biology ,Cas3 ,Actinomycetales ,CRISPR ,nuclease ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Nuclease ,Base Sequence ,biology ,030302 biochemistry & molecular biology ,DNA Helicases ,Cas ,Helicase ,RNA ,helicase ,Biochemistry ,chemistry ,biology.protein ,Biophysics ,Nucleic acid ,Adenosine triphosphate ,DNA - Abstract
CRISPR drives prokaryotic adaptation to invasive nucleic acids such as phages and plasmids, using an RNA-mediated interference mechanism. Interference in type I CRISPR-Cas systems requires a targeting Cascade complex and a degradation machine, Cas3, which contains both nuclease and helicase activities. Here we report the crystal structures of Thermobifida fusca Cas3 bound to single-stranded (ss) DNA substrate and show that it is an obligate 3'-to-5' ssDNase that preferentially accepts substrate directly from the helicase moiety. Conserved residues in the HD-type nuclease coordinate two irons for ssDNA cleavage. We demonstrate ATP coordination and conformational flexibility of the SF2-type helicase domain. Cas3 is specifically guided toward Cascade-bound target DNA by a PAM sequence, through physical interactions with both the nontarget substrate strand and the CasA protein. The sequence of recognition events ensures well-controlled DNA targeting and degradation of foreign DNA by Cascade and Cas3.
- Published
- 2014
- Full Text
- View/download PDF
29. Reconstitution and characterization of eukaryotic N6-threonylcarbamoylation of tRNA using a minimal enzyme system
- Author
-
David Chiovitti, Dante Neculai, Amy A. Caudy, Jonathan Strecker, Daniel Durocher, Frank Sicheri, Rachel K. Szilard, Leo C. K. Wan, Fang Yuan, Elena Lissina, Igor Kurinov, Gennadiy Poda, Corey Nislow, Daniel Y.L. Mao, and Neroshan Thevakumaran
- Subjects
Models, Molecular ,Adenosine ,Saccharomyces cerevisiae Proteins ,Protein family ,Protein subunit ,Saccharomyces cerevisiae ,DNA-binding protein ,Ribosome ,Mitochondrial Proteins ,03 medical and health sciences ,Telomere Homeostasis ,RNA, Transfer ,Structural Biology ,Transcription (biology) ,Genetics ,030304 developmental biology ,0303 health sciences ,biology ,030302 biochemistry & molecular biology ,Metalloendopeptidases ,biology.organism_classification ,DNA-Binding Proteins ,Protein Subunits ,Transfer RNA ,Dimerization - Abstract
The universally conserved Kae1/Qri7/YgjD and Sua5/YrdC protein families have been implicated in growth, telomere homeostasis, transcription and the N6-threonylcarbamoylation (t(6)A) of tRNA, an essential modification required for translational fidelity by the ribosome. In bacteria, YgjD orthologues operate in concert with the bacterial-specific proteins YeaZ and YjeE, whereas in archaeal and eukaryotic systems, Kae1 operates as part of a larger macromolecular assembly called KEOPS with Bud32, Cgi121, Gon7 and Pcc1 subunits. Qri7 orthologues function in the mitochondria and may represent the most primitive member of the Kae1/Qri7/YgjD protein family. In accordance with previous findings, we confirm that Qri7 complements Kae1 function and uncover that Qri7 complements the function of all KEOPS subunits in growth, t(6)A biosynthesis and, to a partial degree, telomere maintenance. These observations suggest that Kae1 provides a core essential function that other subunits within KEOPS have evolved to support. Consistent with this inference, Qri7 alone is sufficient for t(6)A biosynthesis with Sua5 in vitro. In addition, the 2.9 Å crystal structure of Qri7 reveals a simple homodimer arrangement that is supplanted by the heterodimerization of YgjD with YeaZ in bacteria and heterodimerization of Kae1 with Pcc1 in KEOPS. The partial complementation of telomere maintenance by Qri7 hints that KEOPS has evolved novel functions in higher organisms.
- Published
- 2013
- Full Text
- View/download PDF
30. MOB1 Mediated Phospho-recognition in the Core Mammalian Hippo Pathway
- Author
-
Amber L. Couzens, Sebastian Guettler, Panagis Filippakopoulos, Daniel Y. Mao, James D.R. Knight, Frank Sicheri, Shawn Xiong, Sarah Picaud, Anne-Claude Gingras, and Igor Kurinov
- Subjects
0301 basic medicine ,Phosphopeptides ,Plasma protein binding ,WWTR1 ,Biology ,Protein Serine-Threonine Kinases ,Biochemistry ,Serine-Threonine Kinase 3 ,Analytical Chemistry ,03 medical and health sciences ,Humans ,Binding site ,Phosphorylation ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,YAP1 ,Hippo signaling pathway ,Phosphopeptide ,Research ,Autophosphorylation ,Intracellular Signaling Peptides and Proteins ,Recombinant Proteins ,030104 developmental biology ,HeLa Cells ,Protein Binding ,Signal Transduction - Abstract
The Hippo tumor suppressor pathway regulates organ size and tissue homoeostasis in response to diverse signaling inputs. The core of the pathway consists of a short kinase cascade: MST1 and MST2 phosphorylate and activate LATS1 and LATS2, which in turn phosphorylate and inactivate key transcriptional co-activators, YAP1 and TAZ (gene WWTR1). The MOB1 adapter protein regulates both phosphorylation reactions firstly by concurrently binding to the upstream MST and downstream LATS kinases to enable the trans phosphorylation reaction, and secondly by allosterically activating the catalytic function of LATS1 and LATS2 to directly stimulate phosphorylation of YAP and TAZ. Studies of yeast Mob1 and human MOB1 revealed that the ability to recognize phosphopeptide sequences in their interactors, Nud1 and MST2 respectively, was critical to their roles in regulating the Mitotic Exit Network in yeast and the Hippo pathway in metazoans. However, the underlying rules of phosphopeptide recognition by human MOB1, the implications of binding specificity for Hippo pathway signaling, and the generality of phosphopeptide binding function to other human MOB family members remained elusive. Employing proteomics, peptide arrays and biochemical analyses, we systematically examine the phosphopeptide binding specificity of MOB1 and find it to be highly complementary to the substrate phosphorylation specificity of MST1 and MST2. We demonstrate that autophosphorylation of MST1 and MST2 on several threonine residues provides multiple MOB1 binding sites with varying binding affinities which in turn contribute to a redundancy of MST1-MOB1 protein interactions in cells. The crystal structures of MOB1A in complex with two favored phosphopeptide sites in MST1 allow for a full description of the MOB1A phosphopeptide-binding consensus. Lastly, we show that the phosphopeptide binding properties of MOB1A are conserved in all but one of the seven MOB family members in humans, thus providing a starting point for uncovering their elusive cellular functions.
- Published
- 2016
31. Author response: Structural mechanism of ligand activation in human calcium-sensing receptor
- Author
-
Lidia Mosyak, Baohua Cao, Martin Bush, Hee-Chang Mun, Donald D. Chang, Yan Chen, Trang Nguyen, Prakash Subramanyam, Emmanuel Sturchler, Alice P Brown, Tat Cheung Cheng, Yong Geng, Patricia McDonald, Sarah C. Brennan, Hao Zuo, Igor Kurinov, Matthias Quick, Henry M. Colecraft, Qing R. Fan, and Arthur D. Conigrave
- Subjects
Chemistry ,Biophysics ,Calcium-sensing receptor ,Ligand (biochemistry) ,Mechanism (sociology) - Published
- 2016
- Full Text
- View/download PDF
32. Norathyriol Suppresses Skin Cancers Induced by Solar Ultraviolet Radiation by Targeting ERK Kinases
- Author
-
Zigang Dong, Keyuan Zhou, Andria Carper, Margarita Malakhova, Ann M. Bode, Kanamata Reddy, Carlos P. Sosa, Myoung Ok Kim, Alyssa Langfald, Jixia Li, Naomi Oi, Feng Zhu, Igor Kurinov, and Madhusoodanan Mottamal
- Subjects
MAPK/ERK pathway ,Cancer Research ,integumentary system ,Cell growth ,Kinase ,Biology ,medicine.disease ,Molecular biology ,chemistry.chemical_compound ,Oncology ,chemistry ,medicine ,Cancer research ,Phosphorylation ,Skin cancer ,Mangiferin ,Protein kinase A ,Transcription factor - Abstract
Ultraviolet (UV) irradiation is the leading factor in the development of skin cancer, prompting great interest in chemopreventive agents for this disease. In this study, we report the discovery of norathyriol, a plant-derived chemopreventive compound identified through an in silico virtual screening of the Chinese Medicine Library. Norathyriol is a metabolite of mangiferin found in mango, Hypericum elegans, and Tripterospermum lanceolatum and is known to have anticancer activity. Mechanistic investigations determined that norathyriol acted as an inhibitor of extracellular signal–regulated kinase (ERK)1/2 activity to attenuate UVB-induced phosphorylation in mitogen-activated protein kinases signaling cascades. We confirmed the direct and specific binding of norathyriol with ERK2 through a cocrystal structural analysis. The xanthone moiety in norathyriol acted as an adenine mimetic to anchor the compound by hydrogen bonds to the hinge region of the protein ATP-binding site on ERK2. Norathyriol inhibited in vitro cell growth in mouse skin epidermal JB6 P+ cells at the level of G2–M phase arrest. In mouse skin tumorigenesis assays, norathyriol significantly suppressed solar UV–induced skin carcinogenesis. Further analysis indicated that norathyriol mediates its chemopreventive activity by inhibiting the ERK-dependent activity of transcriptional factors AP-1 and NF-κB during UV-induced skin carcinogenesis. Taken together, our results identify norathyriol as a safe new chemopreventive agent that is highly effective against development of UV-induced skin cancer. Cancer Res; 72(1); 260–70. ©2011 AACR.
- Published
- 2012
- Full Text
- View/download PDF
33. High Resolution X-Ray Structure and Potent Anti-HIV Activity of Recombinant Dianthin Antiviral Protein
- Author
-
Fatih M. Uckun, Francis Rajamohan, and Igor Kurinov
- Subjects
Models, Molecular ,Molecular model ,Anti-HIV Agents ,Immunoblotting ,Antiviral protein ,Virus Replication ,Peripheral blood mononuclear cell ,law.invention ,Active center ,X-Ray Diffraction ,Dianthus ,law ,Escherichia ,Drug Discovery ,Escherichia coli ,Humans ,Plant Proteins ,chemistry.chemical_classification ,Aniline Compounds ,Binding Sites ,Cell-Free System ,biology ,RNA ,biology.organism_classification ,Recombinant Proteins ,Amino acid ,Plant Leaves ,chemistry ,Biochemistry ,Protein Biosynthesis ,HIV-1 ,Ribosome Inactivating Proteins, Type 1 ,Recombinant DNA ,Crystallization - Abstract
Dianthin antiviral protein (DAP) is a naturally occurring antiviral protein from the leaves of carnation (Dianthus caryophyllus) capable of depurinating HIV-1 RNA and inhibiting HIV-1 replication in human peripheral blood mononuclear cells. Escherichia coli-derived recombinant DAP (rDAP, amino acids 1-254) was purified to homogeneity for structural and functional studies. In the following paper the X-ray crystal structure of rDAP as well as its complexes with cyclic AMP and adenyl-guanosine (ApG) as substrate analogs at 1.7 A resolution are reported. Molecular modeling studies of the interactions of DAP and the structurally similar pokeweed antiviral protein (PAP) with a single-stranded RNA heptamer predicted a more potent anti-HIV activity for rDAP due to its unique surface topology and more favorable charge distribution in its 20 A-long RNA binding active center cleft. In accordance with the predictions of the modeling studies, rDAP was more potent than rPAP in depurinating HIV-1 RNA. To the knowledge of the authors, this is the first structural and functional characterization of recombinant DAP.
- Published
- 2011
- Full Text
- View/download PDF
34. Atg8 Transfer from Atg7 to Atg3: A Distinctive E1-E2 Architecture and Mechanism in the Autophagy Pathway
- Author
-
Amanda Nourse, Michal Hammel, Alan Deng, Stephen E. Kaiser, Igor Kurinov, Douglas R. Green, Allison H. Williams, Stephen W.G. Tait, Asad M. Taherbhoy, Charles O. Rock, and Brenda A. Schulman
- Subjects
Models, Molecular ,Protein Conformation ,Recombinant Fusion Proteins ,ATG8 ,Molecular Sequence Data ,Autophagy-Related Proteins ,Protomer ,Biology ,Ubiquitin-conjugating enzyme ,Crystallography, X-Ray ,Transfection ,Autophagy-Related Protein 7 ,Article ,Cell Line ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,0302 clinical medicine ,Protein structure ,Multienzyme Complexes ,Protein Interaction Mapping ,Autophagy ,Animals ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Binding site ,Molecular Biology ,030304 developmental biology ,Mice, Knockout ,chemistry.chemical_classification ,0303 health sciences ,DNA ligase ,Binding Sites ,Intracellular Signaling Peptides and Proteins ,Autophagy-Related Protein 8 Family ,Cell Biology ,Fibroblasts ,chemistry ,Biochemistry ,Mutation ,Ubiquitin-Conjugating Enzymes ,Biophysics ,Trans-acting ,Protein Multimerization ,Carrier Proteins ,Hydrophobic and Hydrophilic Interactions ,Microtubule-Associated Proteins ,030217 neurology & neurosurgery - Abstract
Summary Atg7 is a noncanonical, homodimeric E1 enzyme that interacts with the noncanonical E2 enzyme, Atg3, to mediate conjugation of the ubiquitin-like protein (UBL) Atg8 during autophagy. Here we report that the unique N-terminal domain of Atg7 (Atg7 NTD ) recruits a unique "flexible region" from Atg3 (Atg3 FR ). The structure of an Atg7 NTD -Atg3 FR complex reveals hydrophobic residues from Atg3 engaging a conserved groove in Atg7, important for Atg8 conjugation. We also report the structure of the homodimeric Atg7 C-terminal domain, which is homologous to canonical E1s and bacterial antecedents. The structures, SAXS, and crosslinking data allow modeling of a full-length, dimeric (Atg7∼Atg8-Atg3) 2 complex. The model and biochemical data provide a rationale for Atg7 dimerization: Atg8 is transferred in trans from the catalytic cysteine of one Atg7 protomer to Atg3 bound to the N-terminal domain of the opposite Atg7 protomer within the homodimer. The studies reveal a distinctive E1∼UBL-E2 architecture for enzymes mediating autophagy.
- Published
- 2011
- Full Text
- View/download PDF
35. Crystal Structure of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-associated Csn2 Protein Revealed Ca2+-dependent Double-stranded DNA Binding Activity
- Author
-
Igor Kurinov, Ki Hyun Nam, and Ailong Ke
- Subjects
Models, Molecular ,Molecular Sequence Data ,Molecular Conformation ,Protomer ,Biology ,Crystallography, X-Ray ,Biochemistry ,Double-stranded DNA binding ,chemistry.chemical_compound ,Protein structure ,Bacterial Proteins ,Enterococcus faecalis ,CRISPR ,Protein–DNA interaction ,Amino Acid Sequence ,Cloning, Molecular ,Protein Structure, Quaternary ,Molecular Biology ,Genetics ,Models, Genetic ,Sequence Homology, Amino Acid ,DNA ,Cell Biology ,Protein Structure, Tertiary ,DNA-Binding Proteins ,chemistry ,Multigene Family ,Protein Structure and Folding ,CRISPR Loci ,RNA Interference ,Protein quaternary structure ,Protein Binding - Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) and their associated protein genes (cas genes) are widespread in bacteria and archaea. They form a line of RNA-based immunity to eradicate invading bacteriophages and malicious plasmids. A key molecular event during this process is the acquisition of new spacers into the CRISPR loci to guide the selective degradation of the matching foreign genetic elements. Csn2 is a Nmeni subtype-specific cas gene required for new spacer acquisition. Here we characterize the Enterococcus faecalis Csn2 protein as a double-stranded (ds-) DNA-binding protein and report its 2.7 Å tetrameric ring structure. The inner circle of the Csn2 tetrameric ring is ∼26 Å wide and populated with conserved lysine residues poised for nonspecific interactions with ds-DNA. Each Csn2 protomer contains an α/β domain and an α-helical domain; significant hinge motion was observed between these two domains. Ca(2+) was located at strategic positions in the oligomerization interface. We further showed that removal of Ca(2+) ions altered the oligomerization state of Csn2, which in turn severely decreased its affinity for ds-DNA. In summary, our results provided the first insight into the function of the Csn2 protein in CRISPR adaptation by revealing that it is a ds-DNA-binding protein functioning at the quaternary structure level and regulated by Ca(2+) ions.
- Published
- 2011
- Full Text
- View/download PDF
36. A Structure-Based Strategy for Engineering Selective Ubiquitin Variant Inhibitors of Skp1-Cul1-F-Box Ubiquitin Ligases
- Author
-
Frank Sicheri, Sachdev S. Sidhu, Alevtina Pavlenco, Maryna Gorelik, Noah Manczyk, and Igor Kurinov
- Subjects
Models, Molecular ,0301 basic medicine ,Jumonji Domain-Containing Histone Demethylases ,Phage display ,KDM2B ,Computational biology ,Crystallography, X-Ray ,Article ,Small Molecule Libraries ,Structure-Activity Relationship ,03 medical and health sciences ,Ubiquitin ,Structural Biology ,Skp1 ,Humans ,Ligase activity ,Enzyme Inhibitors ,Ubiquitins ,Molecular Biology ,Binding Sites ,SKP Cullin F-Box Protein Ligases ,biology ,Chemistry ,F-Box Proteins ,Cullin Proteins ,3. Good health ,Ubiquitin ligase ,HEK293 Cells ,030104 developmental biology ,Structural biology ,biology.protein ,Female ,CUL1 ,Cell Surface Display Techniques ,Protein Binding - Abstract
Skp1-Cul1-F-box (SCF) E3 ligases constitute the largest and best-characterized family of the multisubunit E3 ligases with important cellular functions and numerous disease links. The specificity of an SCF E3 ligase is established by one of the 69 human F-box proteins that are recruited to Cul1 through the Skp1 adaptor. We previously reported generation of ubiquitin variants (UbVs) targeting Fbw7 and Fbw11, which inhibit ligase activity by binding at the F-box-Skp1 interface to competitively displace Cul1. In the present study, we employed an optimized engineering strategy to generate specific binding UbVs against 17 additional Skp1-F-box complexes. We validated our design strategy and uncovered the structural basis of binding specificity by crystallographic analyses of representative UbVs bound to Skp1-Fbl10 and Skp1-Fbl11. Our study highlights the power of combining phage display with structure-based design to develop UbVs targeting specific protein surfaces.
- Published
- 2018
- Full Text
- View/download PDF
37. Atomic Structure of the KEOPS Complex: An Ancient Protein Kinase-Containing Molecular Machine
- Author
-
Daniel Durocher, Michael Downey, Sigrun Rumpel, Jenny S.L. Ho, Cheryl H. Arrowsmith, Leo C. K. Wan, Cynthia S.W. Ho, Daniel Y.L. Mao, Rachel K. Szilard, Dante Neculai, Stephen Orlicky, Wei Zhang, Yosr Z. Haffani, Frank Sicheri, Derek F. Ceccarelli, Christophe Farès, and Igor Kurinov
- Subjects
Models, Molecular ,Methanococcus ,Transcription, Genetic ,Thermoplasma ,Archaeal Proteins ,Protein subunit ,Biology ,Crystallography, X-Ray ,Article ,Structural genomics ,Transcription (biology) ,Escherichia coli ,Humans ,Protein kinase A ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,Gene ,Genetics ,Sequence Homology, Amino Acid ,Kinase ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,Telomere ,biology.organism_classification ,Protein Structure, Tertiary ,Cell biology ,Protein Subunits ,Essential gene ,Multiprotein Complexes ,Carrier Proteins ,Protein Kinases - Abstract
Kae1 is a universally conserved ATPase and part of the essential gene set in bacteria. In archaea and eukaryotes, Kae1 is embedded within the protein kinase-containing KEOPS complex. Mutation of KEOPS subunits in yeast leads to striking telomere and transcription defects, but the exact biochemical function of KEOPS is not known. As a first step to elucidating its function, we solved the atomic structure of archaea-derived KEOPS complexes involving Kae1, Bud32, Pcc1, and Cgi121 subunits. Our studies suggest that Kae1 is regulated at two levels by the primordial protein kinase Bud32, which is itself regulated by Cgi121. Moreover, Pcc1 appears to function as a dimerization module, perhaps suggesting that KEOPS may be a processive molecular machine. Lastly, as Bud32 lacks the conventional substrate-recognition infrastructure of eukaryotic protein kinases including an activation segment, Bud32 may provide a glimpse of the evolutionary history of the protein kinase family.
- Published
- 2008
- Full Text
- View/download PDF
38. CNK and HYP form a discrete dimer by their SAM domains to mediate RAF kinase signaling
- Author
-
Igor Kurinov, Thanashan Rajakulendran, Malha Sahmi, Marc Therrien, Mike Tyers, and Frank Sicheri
- Subjects
Models, Molecular ,MAPK/ERK pathway ,law.invention ,Protein structure ,law ,Escherichia coli ,Drosophila Proteins ,Humans ,c-Raf ,Protein kinase A ,Enhancer ,Adaptor Proteins, Signal Transducing ,Multidisciplinary ,Chemistry ,Kinase ,Signal transducing adaptor protein ,Biological Sciences ,Protein Structure, Tertiary ,Cell biology ,Biochemistry ,Suppressor ,raf Kinases ,Crystallization ,Dimerization ,Protein Kinases ,Signal Transduction - Abstract
RAF kinase functions in the mitogen-activated protein kinase (MAPK) pathway to transmit growth signals to the downstream kinases MEK and ERK. Activation of RAF catalytic activity is facilitated by a regulatory complex comprising the proteins CNK ( C onnector e n hancer of K SR), HYP (Hyphen), and KSR ( K inase S uppressor of R as). The sterile α-motif (SAM) domain found in both CNK and HYP plays an essential role in complex formation. Here, we have determined the x-ray crystal structure of the SAM domain of CNK in complex with the SAM domain of HYP. The structure reveals a single-junction SAM domain dimer of 1:1 stoichiometry in which the binding mode is a variation of polymeric SAM domain interactions. Through in vitro and in vivo mutational analyses, we show that the specific mode of dimerization revealed by the crystal structure is essential for RAF signaling and facilitates the recruitment of KSR to form the CNK/HYP/KSR regulatory complex. We present two docking-site models to account for how SAM domain dimerization might influence the formation of a higher-order CNK/HYP/KSR complex.
- Published
- 2008
- Full Text
- View/download PDF
39. Implementation of a k/k0 Method to Identify Long-Range Structure in Transition States during Conformational Folding/Unfolding of Proteins
- Author
-
Lovy Pradeep, Harold A. Scheraga, Igor Kurinov, and Steven E. Ealick
- Subjects
Protein Denaturation ,Protein Folding ,RNase P ,Protein Conformation ,Phi value analysis ,Bovine pancreatic ribonuclease ,Arginine ,Crystallography, X-Ray ,Article ,Structure-Activity Relationship ,Protein structure ,Structural Biology ,Animals ,Molecular Biology ,Guanidine ,Alanine ,biology ,Chemistry ,Valine ,Ribonuclease, Pancreatic ,Hydrogen-Ion Concentration ,Contact order ,Folding (chemistry) ,Crystallography ,Kinetics ,Covalent bond ,biology.protein ,Thermodynamics ,Protein folding ,Cattle - Abstract
A previously-introduced kinetic-rate constant (k/k0) method, where k and k0 are the folding (unfolding) rate constants in the mutant and the wild-type forms, respectively, of a protein, has been applied to obtain qualitative information about structure in the transition state (TS) ensemble of bovine pancreatic ribonuclease A (RNase A) which contains four native disulfide bonds. The method compares the folding (unfolding) kinetics of two versions of RNase A, with and without a covalent crosslink (in the form of a fifth disulfide bond); the method tests whether the crosslinked residues are associated in the folding (unfolding) transition state of the non-crosslinked version. To confirm that the fifth disulfide bond has not introduced a significant structural perturbation, we solved the crystal structure of the V43C-R85C mutant to 1.6 Å resolution. Our findings suggest that residues Val 43 and Arg 85 are not associated in the folding (unfolding) TS ensemble of RNase A, and also that Ala 4 and Val 118 may form non-native contacts in the folding (unfolding) TS ensemble.
- Published
- 2007
- Full Text
- View/download PDF
40. A Localized Specific Interaction Alters the Unfolding Pathways of Structural Homologues
- Author
-
Mahesh Narayan, Harold A. Scheraga, Igor Kurinov, Guoqiang Xu, Ervin Welker, Mey Khalili, Daniel R. Ripoll, and Steven E. Ealick
- Subjects
Models, Molecular ,Protein Folding ,Protein Conformation ,Stereochemistry ,Bovine pancreatic ribonuclease ,Biochemistry ,Article ,Catalysis ,Ribonucleases ,Colloid and Surface Chemistry ,Protein structure ,Native state ,Animals ,Ribonuclease ,biology ,Chemistry ,Protein dynamics ,Ribonuclease, Pancreatic ,General Chemistry ,Recombinant Proteins ,Folding (chemistry) ,Mutagenesis, Site-Directed ,biology.protein ,Thermodynamics ,Cattle ,Pancreatic ribonuclease ,Protein folding ,Oxidation-Reduction - Abstract
Reductive unfolding studies of proteins are designed to provide information about intramolecular interactions that govern the formation (and stabilization) of the native state and about folding/unfolding pathways. By mutating Tyr92 to G, A, or L in the model protein, bovine pancreatic ribonuclease A, and through analysis of temperature factors and molecular dynamics simulations of the crystal structures of these mutants, it is demonstrated that the markedly different reductive unfolding rates and pathways of ribonuclease A and its structural homologue onconase can be attributed to a single, localized, ring-stacking interaction between Tyr92 and Pro93 in the bovine variant. The fortuitous location of this specific stabilizing interaction in a disulfide-bond-containing loop region of ribonuclease A results in the localized modulation of protein dynamics which, in turn, enhances the susceptibility of the disulfide bond to reduction leading to an alteration in the reductive unfolding behavior of the homologues. These results have important implications for folding studies involving topological determinants to obtain folding/unfolding rates and pathways, for protein structure-function prediction through fold recognition, and for predicting proteolytic cleavage sites.
- Published
- 2006
- Full Text
- View/download PDF
41. Structural basis for the recruitment of glycogen synthase by glycogenin
- Author
-
Frank Sicheri, Roger W. Hunter, Xiaojing Tang, Mike Tyers, Andrew Judd, Mar García-Rocha, Joan J. Guinovart, Maria Deak, Elton Zeqiraj, Alexander von Wilamowitz-Moellendorff, Kei Sakamoto, and Igor Kurinov
- Subjects
Glycogenin ,Glycosylation ,Crystallography, X-Ray ,Glycogen debranching enzyme ,chemistry.chemical_compound ,Glycogen phosphorylase ,Mice ,Structure-Activity Relationship ,Glycogen branching enzyme ,Glucose homeostasis ,Animals ,Glycogen synthase ,Caenorhabditis elegans ,Caenorhabditis elegans Proteins ,Protein Structure, Quaternary ,Cells, Cultured ,Glycoproteins ,Mice, Knockout ,Multidisciplinary ,Glycogen ,biology ,Cell-Free System ,Glycogen Synthase ,chemistry ,Biochemistry ,PNAS Plus ,Glycogenesis ,Glucosyltransferases ,biology.protein ,Protein Multimerization ,Protein Binding - Abstract
Glycogen is a primary form of energy storage in eukaryotes that is essential for glucose homeostasis. The glycogen polymer is synthesized from glucose through the cooperative action of glycogen synthase (GS), glycogenin (GN), and glycogen branching enzyme and forms particles that range in size from 10 to 290 nm. GS is regulated by allosteric activation upon glucose-6-phosphate binding and inactivation by phosphorylation on its N- and C-terminal regulatory tails. GS alone is incapable of starting synthesis of a glycogen particle de novo, but instead it extends preexisting chains initiated by glycogenin. The molecular determinants by which GS recognizes self-glucosylated GN, the first step in glycogenesis, are unknown. We describe the crystal structure of Caenorhabditis elegans GS in complex with a minimal GS targeting sequence in GN and show that a 34-residue region of GN binds to a conserved surface on GS that is distinct from previously characterized allosteric and binding surfaces on the enzyme. The interaction identified in the GS-GN costructure is required for GS–GN interaction and for glycogen synthesis in a cell-free system and in intact cells. The interaction of full-length GS-GN proteins is enhanced by an avidity effect imparted by a dimeric state of GN and a tetrameric state of GS. Finally, the structure of the N- and C-terminal regulatory tails of GS provide a basis for understanding phosphoregulation of glycogen synthesis. These results uncover a central molecular mechanism that governs glycogen metabolism.
- Published
- 2014
42. Structure and mechanism of action of the hydroxy-aryl-aldehyde class of IRE1 endoribonuclease inhibitors
- Author
-
Michael Prakesch, Rima Al-awar, Kenneth Lee, Lynn Lehmann, Gennadiy Poda, Danka Vuga, David Chiovitti, Colleen Schweitzer, Marella D. Canny, Julie L. Lucas, John B. Patterson, Nero Thevakumaran, Andras Toro, Nicole M. Duffy, Qingping Zeng, Igor Kurinov, Daniel Durocher, David Uehling, Victor Tam, Brian J. Wilson, Manisha Talukdar, Mario Sanches, and Frank Sicheri
- Subjects
RNase P ,Protein Conformation ,Morpholines ,Endoribonuclease ,DNA Mutational Analysis ,General Physics and Astronomy ,CD59 Antigens ,Regulatory Factor X Transcription Factors ,Biology ,Protein Serine-Threonine Kinases ,Crystallography, X-Ray ,General Biochemistry, Genetics and Molecular Biology ,Article ,Small Molecule Libraries ,03 medical and health sciences ,Structure-Activity Relationship ,0302 clinical medicine ,Protein structure ,Ribonucleases ,Coumarins ,Catalytic Domain ,Cell Line, Tumor ,Structure–activity relationship ,Humans ,Binding site ,Enzyme Inhibitors ,030304 developmental biology ,0303 health sciences ,Aldehydes ,Multidisciplinary ,Binding Sites ,Molecular Structure ,Endoplasmic reticulum ,Membrane Proteins ,General Chemistry ,Small molecule ,DNA-Binding Proteins ,Biochemistry ,030220 oncology & carcinogenesis ,Benzaldehydes ,Unfolded protein response ,Plasmacytoma ,Transcription Factors - Abstract
Endoplasmic reticulum (ER) stress activates the unfolded protein response and its dysfunction is linked to multiple diseases. The stress transducer IRE1α is a transmembrane kinase endoribonuclease (RNase) that cleaves mRNA substrates to re-establish ER homeostasis. Aromatic ring systems containing hydroxy-aldehyde moieties, termed hydroxy-aryl-aldehydes (HAA), selectively inhibit IRE1α RNase and thus represent a novel chemical series for therapeutic development. We solved crystal structures of murine IRE1α in complex with three HAA inhibitors. HAA inhibitors engage a shallow pocket at the RNase-active site through pi-stacking interactions with His910 and Phe889, an essential Schiff base with Lys907 and a hydrogen bond with Tyr892. Structure-activity studies and mutational analysis of contact residues define the optimal chemical space of inhibitors and validate the inhibitor-binding site. These studies lay the foundation for understanding both the biochemical and cellular functions of IRE1α using small molecule inhibitors and suggest new avenues for inhibitor design.
- Published
- 2014
43. Author response: Mechanism of ubiquitin ligation and lysine prioritization by a HECT E3
- Author
-
Chris MacDonald, Michal Hammel, Brenda A. Schulman, Judith Souphron, Robert C. Piper, Igor Kurinov, Brian Kuhlman, Steven M. Lewis, Marcelo Actis, Hari B. Kamadurai, Joseph S. Harrison, Patrick Rodrigues, Alan Deng, Darcie J. Miller, Naoaki Fujii, and Yu Qiu
- Subjects
Prioritization ,Ubiquitin ,biology ,Mechanism (biology) ,Chemistry ,Lysine ,biology.protein ,Ligation ,Cell biology - Published
- 2013
- Full Text
- View/download PDF
44. Mechanism of ubiquitin ligation and lysine prioritization by a HECT E3
- Author
-
Brenda A. Schulman, Joseph S. Harrison, Hari B. Kamadurai, Brian Kuhlman, Patrick Rodrigues, Judith Souphron, Steven M. Lewis, Naoaki Fujii, Igor Kurinov, Marcelo Actis, Alan Deng, Robert C. Piper, Chris MacDonald, Darcie J. Miller, Michal Hammel, and Yu Qiu
- Subjects
HECT domain ,Ubiquitylation ,QH301-705.5 ,Protein Conformation ,HECT ,Science ,Ubiquitin-Protein Ligases ,Molecular Sequence Data ,S. cerevisiae ,NEDD4 ,macromolecular substances ,Ubiquitin-conjugating enzyme ,Biochemistry ,General Biochemistry, Genetics and Molecular Biology ,Deubiquitinating enzyme ,03 medical and health sciences ,Protein structure ,Ubiquitin ,Catalytic Domain ,Rsp5 ,Amino Acid Sequence ,Biology (General) ,E2 conjugating enzyme ,E3 ligase ,030304 developmental biology ,0303 health sciences ,General Immunology and Microbiology ,biology ,Sequence Homology, Amino Acid ,General Neuroscience ,Lysine ,030302 biochemistry & molecular biology ,General Medicine ,Biophysics and Structural Biology ,3. Good health ,Ubiquitin ligase ,Cell biology ,Mutagenesis ,biology.protein ,Medicine ,sense organs ,Insight - Abstract
Ubiquitination by HECT E3 enzymes regulates myriad processes, including tumor suppression, transcription, protein trafficking, and degradation. HECT E3s use a two-step mechanism to ligate ubiquitin to target proteins. The first step is guided by interactions between the catalytic HECT domain and the E2∼ubiquitin intermediate, which promote formation of a transient, thioester-bonded HECT∼ubiquitin intermediate. Here we report that the second step of ligation is mediated by a distinct catalytic architecture established by both the HECT E3 and its covalently linked ubiquitin. The structure of a chemically trapped proxy for an E3∼ubiquitin-substrate intermediate reveals three-way interactions between ubiquitin and the bilobal HECT domain orienting the E3∼ubiquitin thioester bond for ligation, and restricting the location of the substrate-binding domain to prioritize target lysines for ubiquitination. The data allow visualization of an E2-to-E3-to-substrate ubiquitin transfer cascade, and show how HECT-specific ubiquitin interactions driving multiple reactions are repurposed by a major E3 conformational change to promote ligation. DOI:http://dx.doi.org/10.7554/eLife.00828.001.
- Published
- 2013
45. Abstract 821: Discovery of catechol moiety-containing natural compounds as direct ERK2 inhibitors by in vitro kinase assay and co-crystallography
- Author
-
Seung-Ho Shin, Margarita Malakhova, Do Young Lim, Ann M. Bode, Zigang Dong, and Igor Kurinov
- Subjects
Cancer Research ,Chemistry ,Kinase ,Cell growth ,Molecular biology ,chemistry.chemical_compound ,Oncology ,Biochemistry ,Caffeic acid ,Viability assay ,Kinase activity ,Quercetin ,Luteolin ,Fisetin - Abstract
Catechol (pyrocathechol, 1,2-dihydroxybenzene) is a chemical that is used by industry in the manufacture of certain products. Many natural compounds contain catechol as a functional group found in various fruits and vegetables such as apple, apricot, grape, bananas, soybean, peanut, pear, plum, mango, avocado, and mushroom. Extracellular signal-regulated kinase 2 (ERK2), a protein kinase that belongs to MAPK family, modulates many important functions such as cell growth, apoptosis and transcriptional regulation in cancer. The expression levels of ERK2 mRNA and protein are reportedly very high in many human cancers, including leukemia, colon, breast, lung and skin cancer. Previously, we reported that norathyriol and caffeic acid, which possess the catechol moiety, and catechol itself directly bind and inhibit ERK2 kinase activity. Those findings led us to test catechol moiety-containing natural compounds, luteolin, quercetin, fisetin, 7,3’,4’-trihydroxyisoflavone, and cyanidin, against ERK2 kinase activity. We found that all of these compounds directly bind to ERK2 and inhibit its activity. We further confirmed the results by resolving a co-crystal structure of ERK2 bound with luteolin in the ATP binding site. To apply this finding to cancer, the effect of the compounds was tested on the K562 human myelogenous leukemia cell line where ERK2 is highly expressed and knock-down of the protein reduced anchorage-independent growth. Five newly-found ERK2 inhibitors (luteolin, quercetin, fisetin, 7,3’,4’-trihydroxyisoflavone and cyanidin) and three known ERK2 inhibitors (norathyriol, caffeic acid and catechol) decreased cell viability of K562 at 40 ìM. When the eight compounds were used at the same dose in combination (5 ìM each and 40 ìM total), the mixture inhibited ERK2 and reduced cell viability of K562 cells. In summary, using in vitro kinase assays and co-crystallography, we identified new ERK2 inhibitors that contain the catechol functional group, and showed anti-cancer effects of the compounds by cell viability assay and soft agar assay. This work impacts the cancer prevention community by showing that many natural compounds can work together targeting ERK2, an important target in cancer. Citation Format: Seung Ho Shin, Margarita Malakhova, Igor Kurinov, Do Young Lim, Ann M. Bode, Zigang Dong. Discovery of catechol moiety-containing natural compounds as direct ERK2 inhibitors by in vitro kinase assay and co-crystallography. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 821.
- Published
- 2016
- Full Text
- View/download PDF
46. Noncanonical E2 recruitment by the autophagy E1 revealed by Atg7-Atg3 and Atg7-Atg10 structures
- Author
-
Brenda A. Schulman, Jennifer L. Olszewski, Alan Deng, Shanshan Yu, Stephen E. Kaiser, Asad M. Taherbhoy, David M. Duda, Timothy D. Fenn, Igor Kurinov, Kai Mao, and Daniel J. Klionsky
- Subjects
Models, Molecular ,0303 health sciences ,Protein Conformation ,ATG8 ,Ubiquitin-activating enzyme ,Autophagy ,Ubiquitin-Activating Enzymes ,Biology ,Crystallography, X-Ray ,Article ,Transport protein ,Cell biology ,ATG12 ,03 medical and health sciences ,0302 clinical medicine ,Protein structure ,Biochemistry ,Structural Biology ,Electrophoresis, Polyacrylamide Gel ,Molecular Biology ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Core functions of autophagy are mediated by ubiquitin-like protein (UBL) cascades, in which a homodimeric E1 enzyme, Atg7, directs the UBLs Atg8 and Atg12 to their respective E2 enzymes, Atg3 and Atg10. Crystallographic and mutational analyses of yeast (Atg7 – Atg3)2 and (Atg7 –Atg10)2 complexes reveal noncanonical, multisite E1 –E2 recognition in autophagy. Atg7’s unique N-terminal domain recruits distinctive elements from the Atg3 and Atg10 ‘backsides’. This, along with E1 and E2 conformational variability, allows presentation of ‘frontside’ Atg3 and Atg10 active sites to the catalytic cysteine in the C-terminal domain from the opposite Atg7 protomer in the homodimer. Despite different modes of binding, the data suggest that common principles underlie conjugation in both noncanonical and canonical UBL cascades, whereby flexibly tethered E1 domains recruit E2s through surfaces remote from their active sites to juxtapose the E1 and E2 catalytic cysteines.
- Published
- 2012
47. Structural Diversity of the Active N-Terminal Kinase Domain of p90 Ribosomal S6 Kinase 2
- Author
-
Kangdong Liu, Zigang Dong, Margarita Malakhova, Ann M. Bode, Jung-Hyun Shim, Duo Zheng, Valerie Steinman, Igor D'Angelo, and Igor Kurinov
- Subjects
Multidisciplinary ,Terminal (telecommunication) ,business.industry ,Computer science ,Science ,lcsh:R ,lcsh:Medicine ,Correction ,Structural diversity ,Advanced Photon Source ,Bioinformatics ,Protein kinase domain ,P90 Ribosomal S6 Kinase ,Medicine ,lcsh:Q ,lcsh:Science ,Telecommunications ,business - Abstract
The Acknowledgments section was omitted. It should read: A part of the work is based upon research conducted at the Northeastern Collaborative Access Team beamline 24ID of the Advanced Photon Source, supported by award RR-15301 from the National Center for Research Resources at the U.S. National Institutes of Health. Use of the Advanced Photon Source is supported by the U.S. Department of Energy, Office of Basic Energy Sciences, under contract No. W-31-109-ENG-38.
- Published
- 2009
- Full Text
- View/download PDF
48. Modeling and alanine scanning mutagenesis studies of recombinant pokeweed antiviral protein
- Author
-
Francis Rajamohan, Fatih M. Uckun, Matthew J. Pugmire, and Igor Kurinov
- Subjects
Protein Conformation ,Molecular Sequence Data ,Biochemistry ,Ribosome ,Antiviral Agents ,Active center ,Protein Isoforms ,Amino Acid Sequence ,Molecular Biology ,N-Glycosyl Hydrolases ,Plant Proteins ,Alanine ,biology ,RNA ,Active site ,Cell Biology ,Ribosomal RNA ,Alanine scanning ,Stem-loop ,Recombinant Proteins ,biology.protein ,Mutagenesis, Site-Directed ,Ribosome Inactivating Proteins, Type 1 ,Nucleic Acid Conformation - Abstract
The Phytolacca americana-derived naturally occurring ribosome inhibitory protein pokeweed antiviral protein (PAP) is an N-glycosidase that catalytically removes a specific adenine residue from the stem loop of ribosomal RNA. We have employed molecular modeling studies using a novel model of PAP-RNA complexes and site-directed mutagenesis combined with bioassays to evaluate the importance of the residues at the catalytic site and a putative RNA binding active center cleft between the catalytic site and C-terminal domain for the enzymatic deadenylation of ribosomal RNA by PAP. As anticipated, alanine substitutions by site-directed mutagenesis of the PAP active site residues Tyr(72), Tyr(123), Glu(176), and Arg(179) that directly participate in the catalytic deadenylation of RNA resulted in greater than 3 logs of loss in depurinating and ribosome inhibitory activity. Similarly, alanine substitution of the conserved active site residue Trp(208), which results in the loss of stabilizing hydrophobic interactions with the ribose as well as a hydrogen bond to the phosphate backbone of the RNA substrate, caused greater than 3 logs of loss in enzymatic activity. By comparison, alanine substitutions of residues (28)KD(29), (80)FE(81), (111)SR(112), (166)FL(167) that are distant from the active site did not significantly reduce the enzymatic activity of PAP. Our modeling studies predicted that the residues of the active center cleft could via electrostatic interactions contribute to both the correct orientation and stable binding of the substrate RNA molecule in the active site pocket. Notably, alanine substitutions of the highly conserved, charged, and polar residues of the active site cleft including (48)KY(49), (67)RR(68), (69)NN(70), and (90)FND(92) substantially reduced the depurinating and ribosome inhibitory activity of PAP. These results provide unprecedented evidence that besides the active site residues of PAP, the conserved, charged, and polar side chains located at its active center cleft also play a critical role in the PAP-mediated depurination of ribosomal RNA.
- Published
- 2000
49. Drug-resistant HIV-1 proteases identify enzyme residues important for substrate selection and catalytic rate
- Author
-
Jonathan Leis, Todd W. Ridky, Alexandra Kikonyogo, Alexander Wlodawer, John W. Erickson, S V Gulnik, Igor Kurinov, Terry D. Copeland, Robert W. Harrison, and Irene T. Weber
- Subjects
Proteases ,Stereochemistry ,Proteolysis ,medicine.medical_treatment ,Mutant ,Biology ,Cleavage (embryo) ,Biochemistry ,Catalysis ,Substrate Specificity ,Capsid ,HIV Protease ,medicine ,Aspartic Acid Endopeptidases ,chemistry.chemical_classification ,Protease ,Binding Sites ,medicine.diagnostic_test ,Hydrolysis ,Parallel study ,Drug Resistance, Microbial ,Valine ,Nucleocapsid Proteins ,Peptide Fragments ,Amino acid ,Enzyme Activation ,Enzyme ,chemistry ,Amino Acid Substitution ,Avian Sarcoma Viruses ,HIV-1 ,Mutagenesis, Site-Directed ,Crystallization - Abstract
A series of mutations, first identified in protease inhibitor-resistant HIV-1 viral isolates, were introduced into HIV-1 PR as individual substitutions. Mutants containing R8K, V32I, V82T, I84V, G48V/L90M, or V82T/I84V substitutions were analyzed for differences in substrate preference and catalytic efficiency using a set of single amino acid substituted HIV-1 CA-NCa cleavage site peptides. All mutants exhibited wild-type preference for large hydrophobic residues, especially Phe, in the P1' substrate position. Only the R8K and V32I mutants showed significant differences in subsite selection compared to wild-type enzyme. In a parallel study, the individual mutations R10K, L12V, I44V, A60M, I71V, and I108V were introduced into RSV PR. These amino acid positions are structurally equivalent to Arg8, Leu10, Val32, Met46, Ile54, and Ile84 in HIV-1 PR, respectively, which mutate in drug-resistance. The RSV R10K substitution significantly altered substrate specificity and catalytic rate, compared to wild-type, in a manner similar to that of the HIV-1 R8K mutant. Crystal structures of the RSV PR R10K, I44V, I71V, and Il08V mutant enzymes presented here indicate that each of these substitutions has little effect on the overall structure of the respective enzymes. Taken together, these data provide an explanation for the reported in vivo predilection for selection of large hydrophobic residues in the P1' substrate position of second locus mutations in the Gag polyprotein PR cleavage sites. The data also suggest that the selection of resistant enzymes is not simply limited to loss of binding to inhibitor but affects other steps in proteolysis.
- Published
- 1998
50. Abstract 2234: Catechol suppresses EGF-induced cell transformation by inhibiting ERK2 activity as confirmed by a crystallographic study
- Author
-
Seung-Ho Shin, Mee-Hyun Lee, Ann M. Bode, Margarita Malakhova, Do Young Lim, Igor Kurinov, and Zigang Dong
- Subjects
MAPK/ERK pathway ,Cancer Research ,Catechol ,Cell growth ,Kinase ,Growth factor ,medicine.medical_treatment ,chemistry.chemical_compound ,Crystallography ,Oncology ,Biochemistry ,chemistry ,ELK1 ,Epidermal growth factor ,medicine ,Kinase activity - Abstract
Catechol (pyrocathechol, 1,2-dihydroxybenzene) is a chemical that is used by industry in the manufacture of certain products. It also occurs naturally in many fruits such as apple, apricot, grape, bananas, pear, plum, mango, avocado, potato, and mushrooms. Browning of fruit occurs because of a chemical reaction between catechol and oxygen. Even though, catechol has toxicity at a high dose as shown by an in vivo study, the Environmental Protection Agency has not classified catechol with respect to potential carcinogenicity. Whether a lower concentration (< 40 μM) of catechol treatment has effects on carcinogenesis is not known. We found that catechol binds to ERK2 and inhibits its activity. The extracellular signal-regulated kinases (ERKs) belong to the mitogen-activated protein (MAP) kinase family, and regulate important biological processes such as cell growth, proliferation, differentiation, and transcription regulation. ERK2 is activated in response of UV, cytokines and growth factors, such as epidermal growth factor (EGF) and insulin-like growth factor. Constitutive activation of ERK2 is observed in many human cancers such as colon, breast, lung and skin cancers. In this study we present the co-crystal structure of ERK2 bound with catechol in the ATP-active site. The conserved amino acid residues within the hinge region that are involved in the interaction of ERK and its inhibitors make the contacts with catechol. Contacts include side chain of Q105, and the main chains of the D106 and M108 residues. We found that catechol inhibits ERK2 in vitro kinase activity more than two-fold at a 20 μM concentration. This was confirmed by the observed decrease of the phosphorylation of Elk1, a specific downstream target of ERK2. In addition, 20 μM catechol inhibited EGF-induced JB6 cell transformation. Interestingly, JB6 cell proliferation was not affected at 20 μM concentration. Taken together, the results showed that catechol inhibits EGF-induced cell transformation without any toxicity through the inhibition of ERK2 activity. Citation Format: Do Young Lim, Margarita Malakhova, Igor Kurinov, Seung Ho Shin, Mee-Hyun Lee, Ann M. Bode, Zigang Dong. Catechol suppresses EGF-induced cell transformation by inhibiting ERK2 activity as confirmed by a crystallographic study. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2234. doi:10.1158/1538-7445.AM2013-2234
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.