56 results on '"Andrea G. Cochran"'
Search Results
2. GNE-064: A Potent, Selective, and Orally Bioavailable Chemical Probe for the Bromodomains of SMARCA2 and SMARCA4 and the Fifth Bromodomain of PBRM1
- Author
-
Alexander M. Taylor, Chris Bailey, Lisa D. Belmont, Robert Campbell, Nico Cantone, Alexandre Côté, Terry D. Crawford, Richard Cummings, Kevin DeMent, Martin Duplessis, Megan Flynn, Andrew C. Good, Hon-Ren Huang, Shivangi Joshi, Yves Leblanc, Jeremy Murray, Christopher G. Nasveschuk, Adrianne Neiss, Florence Poy, F. Anthony Romero, Peter Sandy, Yong Tang, Vickie Tsui, Laura Zawadzke, Robert J. Sims, James E. Audia, Steven F. Bellon, Steven R. Magnuson, Brian K. Albrecht, and Andrea G. Cochran
- Subjects
DNA-Binding Proteins ,Protein Domains ,Drug Discovery ,DNA Helicases ,Molecular Medicine ,Humans ,Nuclear Proteins ,Transcription Factors - Abstract
Bromodomains are acetyllysine recognition domains present in a variety of human proteins. Bromodomains also bind small molecules that compete with acetyllysine, and therefore bromodomains have been targets for drug discovery efforts. Highly potent and selective ligands with good cellular permeability have been proposed as chemical probes for use in exploring the functions of many of the bromodomain proteins. We report here the discovery of a class of such inhibitors targeting the family VIII bromodomains of SMARCA2 (BRM) and SMARCA4 (BRG1), and PBRM1 (polybromo-1) bromodomain 5. We propose one example from this series, GNE-064, as a chemical probe for the bromodomains SMARCA2, SMARCA4, and PBRM1(5) with the potential for
- Published
- 2022
3. Bromodomains: a new target class for drug development
- Author
-
Robert J. Sims, Andrea G. Cochran, and Andrew R. Conery
- Subjects
0301 basic medicine ,Pharmacology ,Clinical Trials as Topic ,Class (computer programming) ,Anti-Inflammatory Agents ,Antineoplastic Agents ,Chemical probe ,General Medicine ,Computational biology ,Biological potential ,Biology ,Epigenesis, Genetic ,Bromodomain ,BET inhibitor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Drug Development ,Drug development ,030220 oncology & carcinogenesis ,Drug Discovery ,Humans ,Molecular Targeted Therapy ,Transcription Factors - Abstract
Less than a decade ago, it was shown that bromodomains, acetyl lysine 'reader' modules found in proteins with varied functions, were highly tractable small-molecule targets. This is an unusual property for protein-protein or protein-peptide interaction domains, and it prompted a wave of chemical probe discovery to understand the biological potential of new agents that targeted bromodomains. The original examples, inhibitors of the bromodomain and extra-terminal (BET) class of bromodomains, showed enticing anti-inflammatory and anticancer activities, and several compounds have since advanced to human clinical trials. Here, we review the current state of BET inhibitor biology in relation to clinical development, and we discuss the next wave of bromodomain inhibitors with clinical potential in oncology and non-oncology indications. The lessons learned from BET inhibitor programmes should affect efforts to develop drugs that target non-BET bromodomains and other epigenetic readers.
- Published
- 2019
- Full Text
- View/download PDF
4. Computational learning reveals coiled coil-like motifs in histidine kinase linker domains.
- Author
-
Mona Singh 0001, Bonnie Berger, Peter S. Kim, James M. Berger, and Andrea G. Cochran
- Published
- 1998
- Full Text
- View/download PDF
5. Expansion of the ISWI chromatin remodeler family with new active complexes
- Author
-
Yutian Gan, Peter Liu, Tianyi Bai, Claudio Ciferri, Mariano Oppikofer, Benjamin Haley, Andrea G. Cochran, and Wendy Sandoval
- Subjects
0301 basic medicine ,Genetics ,Protein subunit ,Biology ,Biochemistry ,Chromatin ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,chemistry ,Nucleosome ,Molecular Biology ,DNA ,Function (biology) - Abstract
ISWI chromatin remodelers mobilize nucleosomes to control DNA accessibility. Complexes isolated to date pair one of six regulatory subunits with one of two highly similar ATPases. However, we find that each endogenously expressed ATPase co-purifies with every regulatory subunit, substantially increasing the diversity of ISWI complexes, and we additionally identify BAZ2B as a novel, seventh regulatory subunit. Through reconstitution of catalytically active human ISWI complexes, we demonstrate that the new interactions described here are stable and direct. Finally, we profile the nucleosome remodeling functions of the now expanded family of ISWI chromatin remodelers. By revealing the combinatorial nature of ISWI complexes, we provide a basis for better understanding ISWI function in normal settings and disease.
- Published
- 2017
- Full Text
- View/download PDF
6. Inhibition of bromodomain-containing protein 9 for the prevention of epigenetically-defined drug resistance
- Author
-
Shumei Wang, Peter Sandy, Christopher G. Nasveschuk, Terry Crawford, Brian K. Albrecht, Andrea G. Cochran, Steffan Vartanian, Hon-Ren Huang, Vickie Tsui, David Stokoe, Steve Bellon, Yong Tang, E. Megan Flynn, Jian Wang, James R. Kiefer, Jeremy Murray, F. Anthony Romero, Laura Zawadzke, Michael C. Hewitt, Martin Duplessis, Alexander M. Taylor, Eneida Pardo, Alexandre Côté, and Steven Magnuson
- Subjects
0301 basic medicine ,Pyridones ,Clinical Biochemistry ,Population ,Drug Resistance ,Pharmaceutical Science ,Drug resistance ,Pharmacology ,Biochemistry ,Aldehyde Dehydrogenase 1 Family ,Epigenesis, Genetic ,Small Molecule Libraries ,03 medical and health sciences ,Drug tolerance ,Cell Line, Tumor ,Drug Discovery ,Gene expression ,Humans ,Epigenetics ,education ,Molecular Biology ,education.field_of_study ,biology ,Chemistry ,Organic Chemistry ,Retinal Dehydrogenase ,Aldehyde Dehydrogenase ,Chromatin ,Bromodomain ,Molecular Docking Simulation ,030104 developmental biology ,Histone ,Drug Resistance, Neoplasm ,Drug Design ,biology.protein ,Cancer research ,Molecular Medicine ,Transcription Factors - Abstract
Bromodomain-containing protein 9 (BRD9), an epigenetic "reader" of acetylated lysines on post-translationally modified histone proteins, is upregulated in multiple cancer cell lines. To assess the functional role of BRD9 in cancer cell lines, we identified a small-molecule inhibitor of the BRD9 bromodomain. Starting from a pyrrolopyridone lead, we used structure-based drug design to identify a potent and highly selective in vitro tool compound 11, (GNE-375). While this compound showed minimal effects in cell viability or gene expression assays, it showed remarkable potency in preventing the emergence of a drug tolerant population in EGFR mutant PC9 cells treated with EGFR inhibitors. Such tolerance has been linked to an altered epigenetic state, and 11 decreased BRD9 binding to chromatin, and this was associated with decreased expression of ALDH1A1, a gene previously shown to be important in drug tolerance. BRD9 inhibitors may therefore show utility in preventing epigenetically-defined drug resistance.
- Published
- 2017
- Full Text
- View/download PDF
7. GNE-886: A Potent and Selective Inhibitor of the Cat Eye Syndrome Chromosome Region Candidate 2 Bromodomain (CECR2)
- Author
-
Arianne Neiss, Martin Duplessis, Hariharan Jayaram, Shumei Wang, F. Anthony Romero, Christopher G. Nasveschuk, Daniel J. Burdick, James E. Audia, Terry Crawford, Steve Bellon, Hon-Ren Huang, Zhaowu Xu, Yongyun Wang, Ying Jiang, Michael C. Hewitt, Jeremy Murray, Andrea G. Cochran, Peter Sandy, Yong Tang, Brian K. Albrecht, E. Megan Flynn, James R. Kiefer, Archana Bommi-Reddy, Jian Wang, Laura Zawadzke, Shivangi Joshi, Vickie Tsui, Xiaoqin Zhu, Alexander M. Taylor, Robert J. Sims, Eneida Pardo, Alexandre Côté, Steven Magnuson, and Richard T. Cummings
- Subjects
0301 basic medicine ,Genetics ,Organic Chemistry ,Lysine ,Transcription inhibitor ,Biology ,Biochemistry ,In vitro ,Bromodomain ,03 medical and health sciences ,030104 developmental biology ,Acetylation ,Drug Discovery ,Cancer research ,Epigenetics ,Cat eye syndrome chromosome region - Abstract
The biological function of bromodomains, epigenetic readers of acetylated lysine residues, remains largely unknown. Herein we report our efforts to discover a potent and selective inhibitor of the bromodomain of cat eye syndrome chromosome region candidate 2 (CECR2). Screening of our internal medicinal chemistry collection led to the identification of a pyrrolopyridone chemical lead, and subsequent structure-based drug design led to a potent and selective CECR2 bromodomain inhibitor (GNE-886) suitable for use as an in vitro tool compound.
- Published
- 2017
- Full Text
- View/download PDF
8. GNE-371, a Potent and Selective Chemical Probe for the Second Bromodomains of Human Transcription-Initiation-Factor TFIID Subunit 1 and Transcription-Initiation-Factor TFIID Subunit 1-like
- Author
-
Shumei Wang, Vickie Tsui, Terry D. Crawford, James E. Audia, Daniel J. Burdick, Maureen H. Beresini, Alexandre Côté, Richard Cummings, Martin Duplessis, E. Megan Flynn, Michael C. Hewitt, Hon-Ren Huang, Hariharan Jayaram, Ying Jiang, Shivangi Joshi, Jeremy Murray, Christopher G. Nasveschuk, Eneida Pardo, Florence Poy, F. Anthony Romero, Yong Tang, Alexander M. Taylor, Jian Wang, Zhaowu Xu, Laura E. Zawadzke, Xiaoyu Zhu, Brian K. Albrecht, Steven R. Magnuson, Steve Bellon, and Andrea G. Cochran
- Subjects
0301 basic medicine ,Models, Molecular ,010405 organic chemistry ,Protein Conformation ,01 natural sciences ,0104 chemical sciences ,03 medical and health sciences ,030104 developmental biology ,Protein Domains ,Drug Design ,Molecular Probes ,Drug Discovery ,Molecular Medicine ,Humans ,Benzimidazoles ,Transcription Factor TFIID - Abstract
The biological functions of the dual bromodomains of human transcription-initiation-factor TFIID subunit 1 (TAF1(1,2)) remain unknown, although TAF1 has been identified as a potential target for oncology research. Here, we describe the discovery of a potent and selective in vitro tool compound for TAF1(2), starting from a previously reported lead. A cocrystal structure of lead compound 2 bound to TAF1(2) enabled structure-based design and structure-activity-relationship studies that ultimately led to our in vitro tool compound, 27 (GNE-371). Compound 27 binds TAF1(2) with an IC
- Published
- 2018
9. Deubiquitinase DUBA is a post-translational brake on interleukin-17 production in T cells
- Author
-
Kim Newton, Jennie R. Lill, Mark Vasser, Zora Modrusan, Jason DeVoss, Rajkumar Noubade, Oscar W. Huang, Donald S. Kirkpatrick, Vishva M. Dixit, Qui T. Phung, Joshua D. Webster, Andrea G. Cochran, Benjamin Fauber, Justin Lesch, Rongze Lu, Lauri Diehl, Xiaoting Wang, Sascha Rutz, Nobuhiko Kayagaki, Celine Eidenschenk, and Wenjun Ouyang
- Subjects
Proteasome Endopeptidase Complex ,Ubiquitin-Protein Ligases ,Inflammation ,Lymphocyte Activation ,Substrate Specificity ,Mice ,Ubiquitin ,Transforming Growth Factor beta ,RAR-related orphan receptor gamma ,Enzyme Stability ,Intestine, Small ,medicine ,Animals ,Transcription factor ,Multidisciplinary ,biology ,Interleukin-17 ,Ubiquitination ,Transforming growth factor beta ,Nuclear Receptor Subfamily 1, Group F, Member 3 ,Acquired immune system ,Research Highlight ,Ubiquitin ligase ,Cell biology ,Mice, Inbred C57BL ,Protein Biosynthesis ,Immunology ,biology.protein ,Th17 Cells ,Female ,Ubiquitin-Specific Proteases ,Interleukin 17 ,medicine.symptom ,Protein Binding ,Signal Transduction - Abstract
T-helper type 17 (TH17) cells that produce the cytokines interleukin-17A (IL-17A) and IL-17F are implicated in the pathogenesis of several autoimmune diseases. The differentiation of TH17 cells is regulated by transcription factors such as RORγt, but post-translational mechanisms preventing the rampant production of pro-inflammatory IL-17A have received less attention. Here we show that the deubiquitylating enzyme DUBA is a negative regulator of IL-17A production in T cells. Mice with DUBA-deficient T cells developed exacerbated inflammation in the small intestine after challenge with anti-CD3 antibodies. DUBA interacted with the ubiquitin ligase UBR5, which suppressed DUBA abundance in naive T cells. DUBA accumulated in activated T cells and stabilized UBR5, which then ubiquitylated RORγt in response to TGF-β signalling. Our data identify DUBA as a cell-intrinsic suppressor of IL-17 production.
- Published
- 2014
- Full Text
- View/download PDF
10. Regulation of deubiquitinase proteolytic activity
- Author
-
Oscar W. Huang and Andrea G. Cochran
- Subjects
Ubiquitin-Specific Proteases ,medicine.diagnostic_test ,biology ,Chemistry ,Proteolysis ,Proteolytic enzymes ,Plasma protein binding ,Deubiquitinating enzyme ,Ubiquitins ,Allosteric Regulation ,Biochemistry ,Ubiquitin ,Structural Biology ,Biocatalysis ,medicine ,biology.protein ,Animals ,Humans ,Reactive Oxygen Species ,Protein Processing, Post-Translational ,Molecular Biology ,Function (biology) - Abstract
Deubiquitinases (DUBs) are proteolytic enzymes whose function is to oppose the process of the conjugation of ubiquitin to a specific substrate. This task is accomplished through an enzymatic cascade involving E1, E2, and E3 enzymes, which collectively produce a product that is either monoubiquitinated, or polyubiquitinated with multiple single ubiquitins or with ubiquitin chains. The resulting modifications may impact protein function or may lead to the degradation of the ubiquitinated species, so the removal of such modifications must be tightly regulated. On the basis of recent work featuring crystal structures and detailed biochemical or biophysical studies of DUBs, we will discuss here how posttranslational modifications, protein binding partners, and reactive oxygen species regulate their catalytic activity.
- Published
- 2013
- Full Text
- View/download PDF
11. Discovery of a Potent and Selective in Vivo Probe (GNE-272) for the Bromodomains of CBP/EP300
- Author
-
Jonathan Maher, F. Anthony Romero, Samir Kharbanda, Mark Merchant, Ivana Yen, Yingjie Li, Edna F. Choo, Jiangpeng Liao, Xiaocang Wei, Kwong Wah Lai, Karen E. Gascoigne, Richard D. Cummings, Hariharan Jayaram, Thomas Hunsaker, Steven F. Bellon, Jian Wang, Cameron L. Noland, Brian K. Albrecht, Ying Jiang, Jeremy Murray, Susan Kaufman, Kerry L. Spillane, Florence Poy, Zhaowu Xu, Fei Wang, James R. Kiefer, Laura Zawadzke, Zhongguo Chen, Alexander M. Taylor, Andrew R. Conery, Xiaoyu Zhu, Wenfeng Liu, Eneida Pardo, Alexandre Côté, Andrea G. Cochran, Vickie Tsui, Steven Magnuson, Terry Crawford, Maureen Beresini, Emily Chan, Kevin X. Chen, Gladys de Leon Boenig, Justin Ly, Tracy Kleinheinz, and Zhongya Xu
- Subjects
0301 basic medicine ,Models, Molecular ,BRD4 ,Pyridones ,Mice, Nude ,Antineoplastic Agents ,P300-CBP Transcription Factors ,01 natural sciences ,Madin Darby Canine Kidney Cells ,03 medical and health sciences ,Mice ,Structure-Activity Relationship ,Dogs ,In vivo ,Cell Line, Tumor ,Drug Discovery ,Structure–activity relationship ,Animals ,Humans ,p300-CBP Transcription Factors ,Cell Proliferation ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Drug discovery ,Chemistry ,Cell growth ,0104 chemical sciences ,Cell biology ,Bromodomain ,030104 developmental biology ,Biochemistry ,Acetylation ,Molecular Medicine ,Pyrazoles ,Female ,Drug Screening Assays, Antitumor - Abstract
The single bromodomain of the closely related transcriptional regulators CBP/EP300 is a target of much recent interest in cancer and immune system regulation. A co-crystal structure of a ligand-efficient screening hit and the CBP bromodomain guided initial design targeting the LPF shelf, ZA loop, and acetylated lysine binding regions. Structure–activity relationship studies allowed us to identify a more potent analogue. Optimization of permeability and microsomal stability and subsequent improvement of mouse hepatocyte stability afforded 59 (GNE-272, TR-FRET IC50 = 0.02 μM, BRET IC50 = 0.41 μM, BRD4(1) IC50 = 13 μM) that retained the best balance of cell potency, selectivity, and in vivo PK. Compound 59 showed a marked antiproliferative effect in hematologic cancer cell lines and modulates MYC expression in vivo that corresponds with antitumor activity in an AML tumor model.
- Published
- 2016
12. Diving into the Water: Inducible Binding Conformations for BRD4, TAF1(2), BRD9, and CECR2 Bromodomains
- Author
-
Brian K. Albrecht, Laura Zawadzke, F. Anthony Romero, Steve Bellon, Yong Tang, Shumei Wang, Christopher G. Nasveschuk, Jian Wang, James E. Audia, Michael C. Hewitt, Alexander M. Taylor, Eneida Pardo, Alexandre Côté, Daniel J. Burdick, Steven Magnuson, Xiaoyu Zhu, Maureen Beresini, Jeremy Murray, Richard D. Cummings, Ying Jiang, Andrew C. Good, Vickie Tsui, Hariharan Jayaram, Florence Poy, Zhaowu Xu, Terry Crawford, Les A. Dakin, E. Megan Flynn, James R. Kiefer, Martin Duplessis, Hon-Ren Huang, and Andrea G. Cochran
- Subjects
0301 basic medicine ,Models, Molecular ,BRD4 ,Stereochemistry ,Pyridones ,Molecular Conformation ,RNA-binding protein ,Cell Cycle Proteins ,Ligands ,03 medical and health sciences ,Structure-Activity Relationship ,Transcription (biology) ,Drug Discovery ,Fluorescence Resonance Energy Transfer ,Structure–activity relationship ,Humans ,Fluorometry ,Pyrroles ,Histone Acetyltransferases ,TATA-Binding Protein Associated Factors ,Binding Sites ,Dose-Response Relationship, Drug ,Chemistry ,Nuclear Proteins ,Water ,Highly selective ,Bromodomain ,TAF1 ,030104 developmental biology ,Förster resonance energy transfer ,Biochemistry ,Molecular Medicine ,Transcription Factor TFIID ,Transcription Factors - Abstract
The biological role played by non-BET bromodomains remains poorly understood, and it is therefore imperative to identify potent and highly selective inhibitors to effectively explore the biology of individual bromodomain proteins. A ligand-efficient nonselective bromodomain inhibitor was identified from a 6-methyl pyrrolopyridone fragment. Small hydrophobic substituents replacing the N-methyl group were designed directing toward the conserved bromodomain water pocket, and two distinct binding conformations were then observed. The substituents either directly displaced and rearranged the conserved solvent network, as in BRD4(1) and TAF1(2), or induced a narrow hydrophobic channel adjacent to the lipophilic shelf, as in BRD9 and CECR2. The preference of distinct substituents for individual bromodomains provided selectivity handles useful for future lead optimization efforts for selective BRD9, CECR2, and TAF1(2) inhibitors.
- Published
- 2016
13. Regulatory T Cell Modulation by CBP/EP300 Bromodomain Inhibition*
- Author
-
Pranal J. Dakle, F. Anthony Romero, Alexander B. Taylor, Charlie Hatton, Venita G. Watson, Hari Jayaram, Srimoyee Ghosh, Andres Salmeron, Peter Sandy, Steve Bellon, Deanna A. Mele, Jennifer A. Mertz, Thornik Reimer, Hon-Ren Huang, Jose M. Lora, Georgia Hatzivassiliou, Laura Zawadzke, Richard T. Cummings, Robert J. Sims, Jeremy W. Setser, Eneida Pardo, Alexandre Côté, Steven Magnuson, Barbara M. Bryant, James E. Audia, Eugene Chiang, Andrew R. Conery, Florence Poy, Andrea G. Cochran, Melissa Chin, Jean-Christophe Harmange, Vickie Tsui, Brian K. Albrecht, Terry Crawford, Denise DeAlmeida-Nagata, and Jane L. Grogan
- Subjects
0301 basic medicine ,Cellular differentiation ,Immunology ,Chemical biology ,Biochemistry ,T-Lymphocytes, Regulatory ,Cell Line ,Histones ,Small Molecule Libraries ,03 medical and health sciences ,Humans ,Epigenetics ,CREB-binding protein ,Molecular Biology ,Cells, Cultured ,biology ,Acetylation ,Cell Differentiation ,Forkhead Transcription Factors ,Cell Biology ,CREB-Binding Protein ,Bromodomain ,Chromatin ,Cell biology ,Protein Structure, Tertiary ,Molecular Docking Simulation ,030104 developmental biology ,Histone ,biology.protein ,Cancer research ,Transcriptome ,E1A-Associated p300 Protein - Abstract
Covalent modification of histones is a fundamental mechanism of regulated gene expression in eukaryotes, and interpretation of histone modifications is an essential feature of epigenetic control. Bromodomains are specialized binding modules that interact with acetylated histones, linking chromatin recognition to gene transcription. Because of their ability to function in a domain-specific fashion, selective disruption of bromodomain:acetylated histone interactions with chemical probes serves as a powerful means for understanding biological processes regulated by these chromatin adaptors. Here we describe the discovery and characterization of potent and selective small molecule inhibitors for the bromodomains of CREBBP/EP300 that engage their target in cellular assays. We use these tools to demonstrate a critical role for CREBBP/EP300 bromodomains in regulatory T cell biology. Because regulatory T cell recruitment to tumors is a major mechanism of immune evasion by cancer cells, our data highlight the importance of CREBBP/EP300 bromodomain inhibition as a novel, small molecule-based approach for cancer immunotherapy.
- Published
- 2016
14. Phosphorylation-dependent activity of the deubiquitinase DUBA
- Author
-
Sarah G. Hymowitz, Vishva M. Dixit, Melissa A. Starovasnik, Till Maurer, Jianping Yin, Ivan Bosanac, Qui Phung, David Arnott, Oscar W. Huang, Jeremy Flinders, Andrea G. Cochran, Xiaolei Ma, and Nobuhiko Kayagaki
- Subjects
chemistry.chemical_classification ,Cell signaling ,Protease ,biology ,medicine.medical_treatment ,Deubiquitinating enzyme ,Cell biology ,Protein structure ,Enzyme ,chemistry ,Biochemistry ,Ubiquitin ,Structural Biology ,Hydrolase ,biology.protein ,medicine ,Phosphorylation ,Molecular Biology - Abstract
Addition and removal of ubiquitin or ubiquitin chains to and from proteins is a tightly regulated process that contributes to cellular signaling and protein stability. Here we show that phosphorylation of the human deubiquitinase DUBA (OTUD5) at a single residue, Ser177, is both necessary and sufficient to activate the enzyme. The crystal structure of the ubiquitin aldehyde adduct of active DUBA reveals a marked cooperation between phosphorylation and substrate binding. An intricate web of interactions involving the phosphate and the C-terminal tail of ubiquitin cause DUBA to fold around its substrate, revealing why phosphorylation is essential for deubiquitinase activity. Phosphoactivation of DUBA represents an unprecedented mode of protease regulation and a clear link between two major cellular signal transduction systems: phosphorylation and ubiquitin modification.
- Published
- 2012
- Full Text
- View/download PDF
15. Recognition of UbcH5c and the nucleosome by the Bmi1/Ring1b ubiquitin ligase complex
- Author
-
Jacob E. Corn, Ken C. Dong, Qui Phung, Andrea G. Cochran, Tommy K. Cheung, and Matthew L. Bentley
- Subjects
chemistry.chemical_classification ,DNA ligase ,General Immunology and Microbiology ,biology ,General Neuroscience ,macromolecular substances ,General Biochemistry, Genetics and Molecular Biology ,Ubiquitin ligase ,Histone H4 ,chemistry ,Ubiquitin ,Biochemistry ,Ubiquitin ligase complex ,Histone H2A ,biology.protein ,Biophysics ,Monoubiquitination ,Nucleosome ,Molecular Biology - Abstract
The Polycomb repressive complex 1 (PRC1) mediates gene silencing, in part by monoubiquitination of histone H2A on lysine 119 (uH2A). Bmi1 and Ring1b are critical components of PRC1 that heterodimerize via their N-terminal RING domains to form an active E3 ubiquitin ligase. We have determined the crystal structure of a complex between the Bmi1/Ring1b RING–RING heterodimer and the E2 enzyme UbcH5c and find that UbcH5c interacts with Ring1b only, in a manner fairly typical of E2–E3 interactions. However, we further show that the Bmi1/Ring1b RING domains bind directly to duplex DNA through a basic surface patch unique to the Bmi1/Ring1b RING–RING dimer. Mutation of residues on this interaction surface leads to a loss of H2A ubiquitination activity. Computational modelling of the interface between Bmi1/Ring1b–UbcH5c and the nucleosome suggests that Bmi1/Ring1b interacts with both nucleosomal DNA and an acidic patch on histone H4 to achieve specific monoubiquitination of H2A. Our results point to a novel mechanism of substrate recognition, and control of product formation, by Bmi1/Ring1b.
- Published
- 2011
- Full Text
- View/download PDF
16. Phosphorylation of a Borealin Dimerization Domain Is Required for Proper Chromosome Segregation
- Author
-
Andreas Lingel, Wayne J. Fairbrother, Qui Phung, Eric Bourhis, and Andrea G. Cochran
- Subjects
Protein Conformation ,Molecular Sequence Data ,Aurora B kinase ,Cell Cycle Proteins ,macromolecular substances ,Biology ,Biochemistry ,Chromosome segregation ,Tandem Mass Spectrometry ,Chromosomes, Human ,Humans ,Amino Acid Sequence ,Phosphorylation ,Nuclear Magnetic Resonance, Biomolecular ,Mitosis ,Alanine ,Sequence Homology, Amino Acid ,INCENP ,Cell cycle ,Cell biology ,Microscopy, Fluorescence ,Mutagenesis, Site-Directed ,Dimerization ,Cytokinesis - Abstract
The chromosomal passenger complex (CPC) has been identified as a master regulator of mitosis. In particular, proper chromosome segregation and cytokinesis depend on the correct localization and function of the CPC. Within the complex, the kinase Aurora B associates with Incenp, Survivin, and Borealin. The stoichiometry of the complex as well as a complete understanding of how these four components interact with each other remains to be elucidated. Here, we identify a new domain of Borealin. We determined its structure using NMR spectroscopy and discovered a novel dimerization motif. Interestingly, we found that substitutions at Borealin T230, recently identified as an Mps1 phosphorylation site, can modulate the dimerization state of Borealin. Mutation of this single residue to alanine or valine impairs Aurora B activity during mitosis and causes chromosome segregation defects. This study reveals that Mps1 regulates the CPC through a novel Borealin domain.
- Published
- 2009
- Full Text
- View/download PDF
17. A Class of 2,4-Bisanilinopyrimidine Aurora A Inhibitors with Unusually High Selectivity against Aurora B
- Author
-
Bing-Yan Zhu, Andrea G. Cochran, Carter Fields, Oscar W. Huang, Elaine Krueger, Tracy Kleinheinz, Thomas Hunsaker, Jun Liang, Thomas E. Rawson, Jennafer Dotson, Rebecca Pulk, Leslie Walker, John Moffat, Mark Ultsch, Dan Burdick, Birong Zhang, Christian Wiesmann, Ignacio Aliagas-Martin, Gail Lewis Phillips, Laura Corson, and Jason Drummond
- Subjects
Protein Conformation ,Aurora B kinase ,Aurora inhibitor ,Antineoplastic Agents ,macromolecular substances ,Protein Serine-Threonine Kinases ,Crystallography, X-Ray ,medicine.disease_cause ,Structure-Activity Relationship ,Aurora Kinases ,Cell Line, Tumor ,Drug Discovery ,medicine ,Aurora Kinase B ,Humans ,Transferase ,Enzyme Inhibitors ,Protein Kinase Inhibitors ,Mitosis ,chemistry.chemical_classification ,Mutation ,Molecular Structure ,biology ,Chemistry ,Kinase ,enzymes and coenzymes (carbohydrates) ,Pyrimidines ,Enzyme ,Biochemistry ,Enzyme inhibitor ,embryonic structures ,biology.protein ,Molecular Medicine ,Drug Screening Assays, Antitumor ,biological phenomena, cell phenomena, and immunity - Abstract
The two major Aurora kinases carry out critical functions at distinct mitotic stages. Selective inhibitors of these kinases, as well as pan-Aurora inhibitors, show antitumor efficacy and are now under clinical investigation. However, the ATP-binding sites of Aurora A and Aurora B are virtually identical, and the structural basis for selective inhibition has therefore not been clear. We report here a class of bisanilinopyrimidine Aurora A inhibitors with excellent selectivity for Aurora A over Aurora B, both in enzymatic assays and in cellular phenotypic assays. Crystal structures of two of the inhibitors in complex with Aurora A implicate a single amino acid difference in Aurora B as responsible for poor inhibitory activity against this enzyme. Mutation of this residue in Aurora B (E161T) or Aurora A (T217E) is sufficient to swap the inhibition profile, suggesting that this difference might be exploited more generally to achieve high selectivity for Aurora A.
- Published
- 2009
- Full Text
- View/download PDF
18. A Pentacyclic Aurora Kinase Inhibitor (AKI-001) with High in Vivo Potency and Oral Bioavailability
- Author
-
Laura Corson, Thomas Hunsaker, Yingqing Ran, Andrea G. Cochran, Elizabeth Blackwood, Hans-Willi Krell, Leslie Walker, Jun Li, Birong Zhang, Aihe Zhou, Jason Halladay, Thomas E. Rawson, Bing-Yan Zhu, Carter Fields, Brian Safina, Mark Ultsch, Matthias Rüth, Tracy Kleinheinz, Angela McNutt, Dan Burdick, Jun Liang, Jason Drummond, Petra Rüger, Christian Wiesmann, Bernhard Goller, Guy Georges, Gail Lewis Phillips, Anja Limberg, John Moffat, and Jenna Dotson
- Subjects
Models, Molecular ,Lactams ,Aurora B kinase ,Aurora inhibitor ,Administration, Oral ,Biological Availability ,Protein Serine-Threonine Kinases ,Pharmacology ,Crystallography, X-Ray ,Heterocyclic Compounds, 4 or More Rings ,Mice ,Dogs ,Aurora kinase ,Aurora Kinases ,In vivo ,Cell Line, Tumor ,Drug Discovery ,Animals ,Aurora Kinase B ,Humans ,Potency ,Protein Kinase Inhibitors ,IC50 ,Aurora Kinase A ,Molecular Structure ,Chemistry ,Rats ,Biochemistry ,embryonic structures ,Molecular Medicine ,Benzimidazoles ,biological phenomena, cell phenomena, and immunity - Abstract
Aurora kinase inhibitors have attracted a great deal of interest as a new class of antimitotic agents. We report a novel class of Aurora inhibitors based on a pentacyclic scaffold. A prototype pentacyclic inhibitor 32 (AKI-001) derived from two early lead structures improves upon the best properties of each parent and compares favorably to a previously reported Aurora inhibitor, 39 (VX-680). The inhibitor exhibits low nanomolar potency against both Aurora A and Aurora B enzymes, excellent cellular potency (IC50 < 100 nM), and good oral bioavailability. Phenotypic cellular assays show that both Aurora A and Aurora B are inhibited at inhibitor concentrations sufficient to block proliferation. Importantly, the cellular activity translates to potent inhibition of tumor growth in vivo. An oral dose of 5 mg/kg QD is well tolerated and results in near stasis (92% TGI) in an HCT116 mouse xenograft model.
- Published
- 2008
- Full Text
- View/download PDF
19. Fragment-Based Discovery of a Selective and Cell-Active Benzodiazepinone CBP/EP300 Bromodomain Inhibitor (CPI-637)
- Author
-
Yongyun Wang, Yingjie Li, James E. Audia, F. Anthony Romero, Zhongguo Chen, Alexander M. Taylor, Robert J. Sims, Nico Cantone, Wenfeng Liu, Eneida Pardo, Alexandre Côté, Richard Pastor, Michael C. Hewitt, Steven Magnuson, Jeremy Murray, Steve Bellon, Richard T. Cummings, Jian Wang, Brian K. Albrecht, Hariharan Jayaram, Maureen Beresini, Leslie A. Dakin, Susan Kaufman, Henry Lu, Terry Crawford, Laura Zawadzke, Vickie Tsui, Xiaoqin Zhu, Gladys de Leon Boenig, Xiaoyu Zhu, Christopher G. Nasveschuk, Fen Yan, Yves Leblanc, Jiangpeng Liao, Andrew R. Conery, Zhaowu Xu, E. Megan Flynn, James R. Kiefer, Andrea G. Cochran, Dong Yu, Oscar W. Huang, Jeremy W. Setser, Tommy Lai, and Patricia J. Keller
- Subjects
0301 basic medicine ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Cell ,Cellular pathways ,Target engagement ,Computational biology ,01 natural sciences ,Biochemistry ,In vitro ,0104 chemical sciences ,Bromodomain ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Transcription (biology) ,Drug Discovery ,medicine ,Cancer research ,EP300 - Abstract
CBP and EP300 are highly homologous, bromodomain-containing transcription coactivators involved in numerous cellular pathways relevant to oncology. As part of our effort to explore the potential therapeutic implications of selectively targeting bromodomains, we set out to identify a CBP/EP300 bromodomain inhibitor that was potent both in vitro and in cellular target engagement assays and was selective over the other members of the bromodomain family. Reported here is a series of cell-potent and selective probes of the CBP/EP300 bromodomains, derived from the fragment screening hit 4-methyl-1,3,4,5-tetrahydro-2H-benzo[b][1,4]diazepin-2-one.
- Published
- 2016
20. An inhibitor of KDM5 demethylases reduces survival of drug-tolerant cancer cells
- Author
-
Erica L. Jackson, Shobu Odate, Robert M. Pitti, Gulfem D. Guler, David Arnott, Brian K. Albrecht, Jean-Christophe Harmange, Ganapati V. Hegde, Victor S. Gehling, Matthew Wongchenko, Shilpi Arora, Pranoti Gangurde, Wanda Manieri, Yibin Yang, Catherine Wilson, Patrick Trojer, Hyojin Kim, Jennifer Busby, Amy Gustafson, E. Megan Flynn, Christopher M. Bailey, Marie Classon, Yichin Liu, Tommy K. Cheung, Andrea G. Cochran, Tobias M. Maile, James R. Kiefer, Fei Lan, Charles Tindell, Kaylyn E. Williamson, Ted Lau, Steve Bellon, Maia Vinogradova, Richard T. Cummings, and Michael R. Costa
- Subjects
0301 basic medicine ,Models, Molecular ,Cell Survival ,Antineoplastic Agents ,03 medical and health sciences ,Histone H3 ,Structure-Activity Relationship ,Neoplasms ,Humans ,Enzyme Inhibitors ,Molecular Biology ,Demethylation ,biology ,Dose-Response Relationship, Drug ,Molecular Structure ,Cell Biology ,Oxidoreductase inhibitor ,Molecular biology ,030104 developmental biology ,Cell culture ,Drug Resistance, Neoplasm ,KDM5A ,Cancer cell ,Cancer research ,biology.protein ,H3K4me3 ,Histone Demethylases ,Drug Screening Assays, Antitumor ,Retinoblastoma-Binding Protein 2 - Abstract
The KDM5 family of histone demethylases catalyzes the demethylation of histone H3 on lysine 4 (H3K4) and is required for the survival of drug-tolerant persister cancer cells (DTPs). Here we report the discovery and characterization of the specific KDM5 inhibitor CPI-455. The crystal structure of KDM5A revealed the mechanism of inhibition of CPI-455 as well as the topological arrangements of protein domains that influence substrate binding. CPI-455 mediated KDM5 inhibition, elevated global levels of H3K4 trimethylation (H3K4me3) and decreased the number of DTPs in multiple cancer cell line models treated with standard chemotherapy or targeted agents. These findings show that pretreatment of cancer cells with a KDM5-specific inhibitor results in the ablation of a subpopulation of cancer cells that can serve as the founders for therapeutic relapse.
- Published
- 2015
21. A Subset of Human Bromodomains Recognizes Butyryllysine and Crotonyllysine Histone Peptide Modifications
- Author
-
E. Megan Flynn, Mariano Oppikofer, Steve Bellon, Florence Poy, Yong Tang, Oscar W. Huang, and Andrea G. Cochran
- Subjects
Models, Molecular ,Acylation ,Protein Array Analysis ,Gene Expression ,Plasma protein binding ,Computational biology ,Crystallography, X-Ray ,Protein Structure, Secondary ,Epigenesis, Genetic ,Histones ,chemistry.chemical_compound ,Structural Biology ,Escherichia coli ,Humans ,Epigenetics ,Binding site ,Molecular Biology ,Histone Acetyltransferases ,Genetics ,TATA-Binding Protein Associated Factors ,Binding Sites ,biology ,Lysine ,Water ,Recombinant Proteins ,Bromodomain ,Protein Structure, Tertiary ,Butyrates ,Kinetics ,Histone ,chemistry ,Crotonates ,Acetyllysine ,Transcription Factor TFIID ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Protein Processing, Post-Translational ,Protein Binding ,Transcription Factors - Abstract
SummaryBromodomains are epigenetic readers that are recruited to acetyllysine residues in histone tails. Recent studies have identified non-acetyl acyllysine modifications, raising the possibility that these might be read by bromodomains. Profiling the nearly complete human bromodomain family revealed that while most human bromodomains bind only the shorter acetyl and propionyl marks, the bromodomains of BRD9, CECR2, and the second bromodomain of TAF1 also recognize the longer butyryl mark. In addition, the TAF1 second bromodomain is capable of binding crotonyl marks. None of the human bromodomains tested binds succinyl marks. We characterized structurally and biochemically the binding to different acyl groups, identifying bromodomain residues and structural attributes that contribute to specificity. These studies demonstrate a surprising degree of plasticity in some human bromodomains but no single factor controlling specificity across the family. The identification of candidate butyryl- and crotonyllysine readers supports the idea that these marks could have specific physiological functions.
- Published
- 2015
22. Activation of the Proapoptotic Death Receptor DR5 by Oligomeric Peptide and Antibody Agonists
- Author
-
Stephen J. Russell, Avi Ashkenazi, Andrea G. Cochran, Klara Totpal, Scot A. Marsters, Sachdev S. Sidhu, Sarah G. Hymowitz, Bing Li, and Deanne M. Compaan
- Subjects
Models, Molecular ,Programmed cell death ,Phage display ,Protein Conformation ,medicine.drug_class ,Molecular Sequence Data ,Apoptosis ,Peptide ,Complementarity determining region ,Crystallography, X-Ray ,Monoclonal antibody ,Antibodies ,Receptors, Tumor Necrosis Factor ,Immunoglobulin Fab Fragments ,Peptide Library ,Structural Biology ,Cell Line, Tumor ,medicine ,Humans ,Amino Acid Sequence ,Receptor ,Peptide library ,Molecular Biology ,chemistry.chemical_classification ,Binding Sites ,biology ,Molecular biology ,Cell biology ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,chemistry ,biology.protein ,Antibody ,Oligopeptides - Abstract
The cell-extrinsic apoptotic pathway triggers programmed cell death in response to certain ligands that bind to cell-surface death receptors. Apoptosis is essential for normal development and homeostasis in metazoans, and furthermore, selective activation of the cell-extrinsic pathway in tumor cells holds considerable promise for cancer therapy. We used phage display to identify peptides and synthetic antibodies that specifically bind to the human proapoptotic death receptor DR5. Despite great differences in overall size and structure, the DR5-binding peptides and antibodies shared a tripeptide motif, which was conserved within a disulfide-constrained loop of the peptides and the third complementarity determining region of the antibody heavy chains. The X-ray crystal structure of an antibody in complex with DR5 revealed that the tripeptide motif is buried at the core of the interface, confirming its central role in antigen recognition. We found that certain peptides and antibodies exhibited potent proapoptotic activity against DR5-expressing SK-MES-1 lung carcinoma cells. These phage-derived ligands may be useful for elucidating DR5 activation at the molecular level and for creating synthetic agonists of proapoptotic death receptors.
- Published
- 2006
- Full Text
- View/download PDF
23. Phage-display as a tool for quantifying protein stability determinants
- Author
-
Joanne D. Kotz, Andrea G. Cochran, and Christopher J. Bond
- Subjects
Protein Folding ,Phage display ,Heavy-chain antibody ,biology ,In silico ,Stability (learning theory) ,Robustness (evolution) ,Nanotechnology ,Computational biology ,Alanine scanning ,Complementarity Determining Regions ,Biochemistry ,Protein Structure, Secondary ,Protein structure ,Bacterial Proteins ,Peptide Library ,biology.protein ,Protein G ,Immunoglobulin Heavy Chains - Abstract
To address questions of protein stability, researchers have increasingly turned to combinatorial approaches that permit the rapid analysis of libraries of protein variants. Phage-display has proved to be a powerful tool for analyzing protein stability due to the large library size and the robustness of the phage particle to a variety of denaturing conditions. With the B1 domain of protein G (GB1) and a camelid heavy chain antibody as model systems, we are using phage-display libraries to experimentally address questions that have generally been addressed in silico, either through computational studies or statistical analysis of known protein structures. One effort has focused on identifying novel solutions to repacking the hydrophobic core of GB1, while maintaining stability comparable to the wild type protein. In a second study, a small set of substitutions in complimentarity-determining region 3 was found to stabilize the framework of the camelid antibody. Another major focus has been to obtain quantitative data on beta-sheet stability determinants. We have successfully adapted a phage-display method for quantitating affinities of protein variants (shotgun alanine scanning) to analysis of GB1 stability. Using this method, we have analyzed the energetic contributions of cross-strand side chain-side chain interactions. Finally, we discuss parameters to consider in using phage-display to discriminate subtle stability differences among fully folded variants. Overall, this method provides a fast approach for quantitatively addressing biophysical questions.
- Published
- 2004
- Full Text
- View/download PDF
24. BAFF/BLyS Receptor 3 Comprises a Minimal TNF Receptor-like Module That Encodes a Highly Focused Ligand-Binding Site
- Author
-
Minhong Yan, Nathaniel C. Gordon, Sarah G. Hymowitz, Wayne J. Fairbrother, Andrea G. Cochran, Melissa A. Starovasnik, Vishva M. Dixit, Borlan Pan, Robert F. Kelley, and Jianping Yin
- Subjects
Models, Molecular ,Molecular Sequence Data ,In Vitro Techniques ,Ligands ,Biochemistry ,Protein Structure, Secondary ,Receptors, Tumor Necrosis Factor ,B-Cell Activating Factor ,Extracellular ,medicine ,Humans ,Amino Acid Sequence ,Cysteine ,B-cell activating factor ,Receptor ,Nuclear Magnetic Resonance, Biomolecular ,B cell ,Binding Sites ,Tumor Necrosis Factor-alpha ,Chemistry ,Mutagenesis ,Membrane Proteins ,Recombinant Proteins ,Protein Structure, Tertiary ,Cell biology ,Solutions ,medicine.anatomical_structure ,Immunology ,Tumor necrosis factor alpha ,Function (biology) ,B-Cell Activation Factor Receptor - Abstract
BAFF/BLyS, a member of the tumor necrosis family (TNF) superfamily of ligands, is a crucial survival factor for B cells. BAFF binds three receptors, TACI, BCMA, and BR3, with signaling through BR3 being essential for promoting B cell function. Typical TNF receptor (TNFR) family members bind their cognate ligands through interactions with two cysteine-rich domains (CRDs). However, the extracellular domain (ECD) of BR3 consists of only a partial CRD, with cysteine spacing distinct from other modules described previously. Herein, we report the solution structure of the BR3 ECD. A core region of only 19 residues adopts a stable structure in solution. The BR3 fold is analogous to the first half of a canonical TNFR CRD but is stabilized by an additional noncanonical disulfide bond. BAFF-binding determinants were identified by shotgun alanine-scanning mutagenesis of the BR3 ECD expressed on phage. Several of the key BAFF-binding residues are presented from a beta-turn that we have shown previously to be sufficient for ligand binding when transferred to a structured beta-hairpin scaffold [Kayagaki, N., Yan, M., Seshasayee, D., Wang, H., Lee, W., French, D. M., Grewal, I. S., Cochran, A. G., Gordon, N. C., Yin, J., Starovasnik, M. A, and Dixit, V. M. (2002) Immunity 10, 515-524]. Outside of the turn, mutagenesis identifies additional hydrophobic contacts that enhance the BAFF-BR3 interaction. The crystal structure of the minimal hairpin peptide, bhpBR3, in complex with BAFF reveals intimate packing of the six-residue BR3 turn into a cavity on the ligand surface. Thus, BR3 binds BAFF through a highly focused interaction site, unprecedented in the TNFR family.
- Published
- 2003
- Full Text
- View/download PDF
25. Turn stability in β-hairpin peptides: Investigation of peptides containing 3:5 type I G1 bulge turns
- Author
-
Nicholas J. Skelton, Andrea G. Cochran, and Tamas Blandl
- Subjects
Models, Molecular ,Protein Folding ,Circular dichroism ,Magnetic Resonance Spectroscopy ,Stereochemistry ,Amino Acid Motifs ,Molecular Sequence Data ,Protein design ,Population ,Peptide ,Antiparallel (biochemistry) ,Biochemistry ,Article ,Protein Structure, Secondary ,Structure-Activity Relationship ,Amino Acid Sequence ,Disulfides ,education ,Molecular Biology ,Peptide sequence ,chemistry.chemical_classification ,education.field_of_study ,Chemistry ,Circular Dichroism ,Amino acid ,Thermodynamics ,Protein folding ,Peptides - Abstract
The turn-forming ability of a series of three-residue sequences was investigated by substituting them into a well-characterized beta-hairpin peptide. The starting scaffold, bhpW, is a disulfide-cyclized 10-residue peptide that folds into a stable beta-hairpin with two antiparallel strands connected by a two-residue reverse turn. Substitution of the central two residues with the three-residue test sequences leads to less stable hairpins, as judged by thiol-disulfide equilibrium measurements. However, analysis of NMR parameters indicated that each molecule retains a significant folded population, and that the type of turn adopted by the three-residue sequence is the same in all cases. The solution structure of a selected peptide with a PDG turn contained an antiparallel beta-hairpin with a 3:5 type I + G1 bulge turn. Analysis of the energetic contributions of individual turn residues in the series of peptides indicates that substitution effects have significant context dependence, limiting the predictive power of individual amino acid propensities for turn formation. The most stable and least stable sequences were also substituted into a more stable disulfide-cyclized scaffold and a linear beta-hairpin scaffold. The relative stabilities remained the same, suggesting that experimental measurements in the bhpW context are a useful way to evaluate turn stability for use in protein design projects. Moreover, these scaffolds are capable of displaying a diverse set of turns, which can be exploited for the mimicry of protein loops or for generating libraries of reverse turns.
- Published
- 2003
- Full Text
- View/download PDF
26. β‒hairpin polypeptides by design and selection
- Author
-
Melissa A. Starovasnik, Nicholas J. Skelton, Stephen J. Russell, Andrea G. Cochran, and Tamas Blandl
- Subjects
chemistry.chemical_classification ,1h nmr spectroscopy ,chemistry ,Biochemistry ,Stereochemistry ,Disulfide bond ,Tryptophan ,Peptide ,Antiparallel (biochemistry) ,Spectroscopy - Abstract
We have developed polypeptide scaffolds that readily adopt aβ‒hairpin conformation (a pair of antiparallel strands connected by a turn) in solution. The study of such peptides allows us to understand the factors that govern stability and folding of these motifs in proteins, and permits mimicry of functionally important regions of proteins. Spectroscopic and biophysical methods have been used to characterize the conformational preferences and stability of these peptides, with a strong emphasis on using restraints generated from1H NMR spectroscopy to determine their three‒dimensional structure. By optimization of inter‒strand interactions, we have developed highly stable disulfide‒cyclized and linearβ‒hairpin peptides. In particular, tryptophan residues at non‒hydrogen bonded strand sites (NHB) are highly stabilizing. A variety of turn types have been presented from these scaffolds, suggesting that they might generally be useful in turn presentation. Interestingly,β‒hairpin peptides (containing a disulfide and a NHB tryptophan) have recently been discovered as antagonists of protein–protein interactions from naïve peptide libraries displayed on phage. Comparison of one suchβ‒hairpin peptide with anα‒helical peptide of very similar sequence provides further insight into the role that residue type and context play in determining polypeptide conformation.
- Published
- 2003
- Full Text
- View/download PDF
27. BAFF/BLyS Receptor 3 Binds the B Cell Survival Factor BAFF Ligand through a Discrete Surface Loop and Promotes Processing of NF-κB2
- Author
-
Melissa A. Starovasnik, Dhaya Seshasayee, Vishva M. Dixit, Andrea G. Cochran, Iqbal S. Grewal, Minhong Yan, Nobuhiko Kayagaki, Hua Wang, Nathaniel C. Gordon, Wyne P. Lee, Jianping Yin, and Dorothy French
- Subjects
Models, Molecular ,Mice, Inbred A ,Molecular Sequence Data ,Immunology ,Apoptosis ,Plasma protein binding ,Biology ,Ligands ,Receptors, Tumor Necrosis Factor ,Cell Line ,Mice ,NF-kappa B p52 Subunit ,B-Cell Activating Factor ,medicine ,Animals ,Lupus Erythematosus, Systemic ,Immunology and Allergy ,Amino Acid Sequence ,BAFF receptor ,B-cell activating factor ,Receptor ,Transcription factor ,B cell ,Mice, Knockout ,B-Lymphocytes ,Binding Sites ,Mice, Inbred NZB ,Tumor Necrosis Factor-alpha ,NF-kappa B ,Membrane Proteins ,Ligand (biochemistry) ,Molecular biology ,Mice, Mutant Strains ,Protein Structure, Tertiary ,Mice, Inbred C57BL ,Infectious Diseases ,medicine.anatomical_structure ,Female ,Signal transduction ,Protein Processing, Post-Translational ,B-Cell Activation Factor Receptor ,Signal Transduction - Abstract
The TNF-like ligand BAFF/BLyS is a potent survival factor for B cells. It binds three receptors: TACI, BCMA, and BR3. We show that BR3 signaling promotes processing of the transcription factor NF-kappaB2/p100 to p52. NF-kappaB2/p100 cleavage was abrogated in B cells from A/WySnJ mice possessing a mutant BR3 gene, but not in TACI or BCMA null B cells. Furthermore, wild-type mice injected with BAFF-neutralizing BR3-Fc protein showed reduced basal NF-kappaB2 activation. BR3-Fc treatment of NZB/WF1 mice, which develop a fatal lupus-like syndrome, inhibited NF-kappaB2 processing and attenuated the disease process. Since inhibiting the BR3-BAFF interaction has therapeutic ramifications, the ligand binding interface of BR3 was investigated and found to reside within a 26 residue core domain. When stabilized within a structured beta-hairpin peptide, six of these residues were sufficient to confer binding to BAFF.
- Published
- 2002
- Full Text
- View/download PDF
28. Amino acid determinants of β-hairpin conformation in erythropoeitin receptor agonist peptides derived from a phage display library
- Author
-
Nicholas J. Skelton, Andrea G. Cochran, Frederic J. de Sauvage, and Stephen J. Russell
- Subjects
Models, Molecular ,Magnetic Resonance Spectroscopy ,Phage display ,Stereochemistry ,Beta hairpin ,Molecular Sequence Data ,Peptide ,Ligands ,Protein Structure, Secondary ,Cell Line ,Turn (biochemistry) ,Viral Proteins ,Residue (chemistry) ,Peptide Library ,Structural Biology ,Receptors, Erythropoietin ,Humans ,Peptide bond ,Amino Acid Sequence ,Databases, Protein ,Molecular Biology ,Peptide sequence ,Sequence Deletion ,chemistry.chemical_classification ,Circular Dichroism ,Computational Biology ,Hydrogen Bonding ,Protein Structure, Tertiary ,Amino acid ,chemistry ,Biochemistry ,Thermodynamics ,Peptides ,Oxidation-Reduction ,Sequence Alignment ,Cell Division - Abstract
Display of peptide libraries on filamentous phage has led to the identification of peptides of the form X 2-5 CX 2 GPXTWXCX 2-5 (where X is a variable residue) that bind to the extra-cellular portion of the erythropoietin receptor (EPO-R). These peptides adopt β-hairpin conformations when co-crystallized with EPO-R. Solution NMR studies reveal that the peptide is conformationally heterogeneous in the absence of receptor due to cis - trans isomerization about the Gly-Pro peptide bond. Replacement of the conserved threonine residue with glycine at the turn i + 3 position produces a stable β-hairpin conformation in solution, although this peptide no longer has activity in an EPO-R-dependent cell proliferation assay. A truncated form of the EPO-R-binding peptide (containing the i + 3 glycine residue) also forms a highly populated, monomeric β-hairpin. In contrast, phage-derived peptide antagonists of insulin-like growth factor binding protein 1 (IGFBP-1) have a high level of sequence identity with the truncated EPO-R peptide (eight of 12 residues) yet adopt a turn-α-helix conformation in solution. Peptides containing all possible pairwise amino acid substitutions between the EPO-R and IGFBP-1 peptides have been analyzed to assess the degree to which the non-conserved residues stabilize the hairpin or helix conformation. All four residues present in the original sequence are required for maximum population of either the β-hairpin or α-helix conformation, although some substitutions have a more dominant effect. The results demonstrate that, within a given sequence, the observed conformation can be dictated by a small subset of the residues (in this case four out of 12).
- Published
- 2002
- Full Text
- View/download PDF
29. BMI1-RING1B is an autoinhibited RING E3 ubiquitin ligase
- Author
-
Oscar W. Huang, Asad M. Taherbhoy, and Andrea G. Cochran
- Subjects
Polycomb Repressive Complex 1 ,Multidisciplinary ,Protein subunit ,Ubiquitin-Protein Ligases ,General Physics and Astronomy ,macromolecular substances ,General Chemistry ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Gene Expression Regulation, Enzymologic ,Cell biology ,Ubiquitin ligase ,Biochemistry ,Ubiquitin ,BMI1 ,Histone H2A ,Ubiquitin-Conjugating Enzymes ,biology.protein ,Escherichia coli ,Monoubiquitination ,Nucleosome ,Humans ,Salt bridge ,Mitogen-Activated Protein Kinase 7 - Abstract
Polycomb repressive complex 1 (PRC1) is required for ubiquitination of histone H2A lysine 119, an epigenetic mark associated with repression of genes important in developmental regulation. The E3 ligase activity of PRC1 resides in the RING1A/B subunit when paired with one of six PCGF partners. The best known of these is the oncogene BMI1/PCGF4. We find that canonical PRC1 E3 ligases such as PCGF4-RING1B have intrinsically very low enzymatic activity compared with non-canonical PRC1 RING dimers. The structure of a high-activity variant in complex with E2 (PCGF5-RING1B-UbcH5c) reveals only subtle differences from an earlier PCGF4 complex structure. However, two charged residues present in the modelled interface with E2-conjugated ubiquitin prove critical: in BMI1/PCGF4, these residues form a salt bridge that may limit efficient ubiquitin transfer. The intrinsically low activity of the PCGF4-RING1B heterodimer is offset by a relatively favourable interaction with nucleosome substrates, resulting in an efficient site-specific monoubiquitination.
- Published
- 2014
30. Structure−Function Analysis of a Phage Display-Derived Peptide That Binds to Insulin-like Growth Factor Binding Protein 1
- Author
-
David Y. Jackson, Kurt Deshayes, Henry B. Lowman, Nicholas J. Skelton, Yvonne Chen, M.T. Pisabarro, N. Dubree, Manuel Baca, Kerry Zobel, Andrea G. Cochran, and Cynthia P. Quan
- Subjects
Phage display ,Surface Properties ,medicine.medical_treatment ,Molecular Sequence Data ,Peptide ,CHO Cells ,Crystallography, X-Ray ,Binding, Competitive ,Biochemistry ,Protein Structure, Secondary ,Insulin-like growth factor-binding protein ,Structure-Activity Relationship ,Peptide Library ,Cricetinae ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Insulin-Like Growth Factor I ,Conserved Sequence ,chemistry.chemical_classification ,Alanine ,biology ,Chemistry ,Binding protein ,GRB10 ,Growth factor ,Molecular Mimicry ,Structure function ,Antagonist ,Surface Plasmon Resonance ,Molecular biology ,Insulin-Like Growth Factor Binding Protein 1 ,Kinetics ,Amino Acid Substitution ,Mutagenesis, Site-Directed ,biology.protein ,Peptides ,Bacteriophage M13 ,Protein Binding - Abstract
Highly structured, peptide antagonists of the interaction between insulin-like growth factor 1 (IGF-I) and IGF binding protein 1 (IGFBP-1) have recently been discovered by phage display of naïve peptide libraries [Lowman, H. B., et al. (1998) Biochemistry 37, 8870--8878]. We now report a detailed analysis of the features of this turn-helix peptide motif that are necessary for IGFBP-1 binding and structural integrity. Further rounds of phage randomization indicate the importance of residues contributing to a hydrophobic patch on one face of the helix. Alanine-scanning substitutions confirm that the hydrophobic residues are necessary for binding. However, structural analysis by NMR spectroscopy indicates that some of these analogues are less well folded. Structured, high-affinity analogues that lack the disulfide bond were prepared by introducing a covalent constraint between side chains at positions i and i + 7 or i + 8 within the helix. Analogues based on this scaffold demonstrate that a helical conformation is present in the bound state, and that hydrophobic side chains in this helix, and residues immediately preceding it, interact with IGFBP-1. By comparison of alanine scanning data for IGF-I and the turn-helix peptide, we propose a model for common surface features of these molecules that recognize IGFBP-1.
- Published
- 2001
- Full Text
- View/download PDF
31. Crystal Structure of the Complex between VEGF and a Receptor-Blocking Peptide
- Author
-
Brian C. Cunningham, C.J. Keenan, Wayne J. Fairbrother, Andrea G. Cochran, Christian Wiesmann, H.W. Christinger, A.M. de Vos, and Gloria Meng
- Subjects
Models, Molecular ,Vascular Endothelial Growth Factor A ,Magnetic Resonance Spectroscopy ,Phage display ,Macromolecular Substances ,Stereochemistry ,Molecular Sequence Data ,Receptor Protein-Tyrosine Kinases ,Peptide ,Endothelial Growth Factors ,Plasma protein binding ,Crystallography, X-Ray ,Biochemistry ,chemistry.chemical_compound ,Humans ,Computer Simulation ,Receptors, Growth Factor ,Amino Acid Sequence ,Binding site ,Receptor ,Peptide sequence ,chemistry.chemical_classification ,Lymphokines ,Vascular Endothelial Growth Factors ,Peptide Fragments ,Solutions ,Vascular endothelial growth factor ,Receptors, Vascular Endothelial Growth Factor ,chemistry ,Crystallization ,Peptides ,Protein Binding - Abstract
Vascular endothelial growth factor (VEGF) is a specific and potent angiogenic factor and, therefore, a prime therapeutic target for the development of antagonists for the treatment of cancer. As a first step toward this goal, phage display was used to generate peptides that bind to the receptor-binding domain (residues 8-109) of VEGF and compete with receptor [Fairbrother, W. J., Christinger, H. W., Cochran, A. G., Fuh, G., Keenan, C. J., Quan, C., Shriver, S. K., Tom, J. Y. K., Wells, J. A., and Cunningham, B. C. (1999) Biochemistry 38, 17754-17764]. The crystal structure of VEGF in complex with one of these peptides was solved and refined to a resolution of 1.9 A. The 20-mer peptide is unstructured in solution and adopts a largely extended conformation when bound to VEGF. Residues 3-8 form a beta-strand which pairs with strand beta6 of VEGF via six hydrogen bonds. The C-terminal four residues of the peptide point away from the growth factor, consistent with NMR data indicating that these residues are flexible in the complex in solution. In contrast, shortening the N-terminus of the peptide leads to decreased binding affinities. Truncation studies show that the peptide can be reduced to 14 residues with only moderate effect on binding affinity. However, because of the extended conformation and the scarcity of specific side-chain interactions with VEGF, the peptide is not a promising lead for small-molecule development. The interface between the peptide and VEGF contains a subset of the residues recognized by a neutralizing Fab fragment and overlaps partially with the binding site for the Flt-1 receptor. The location of the peptide-binding site and the hydrophilic character of the interactions with VEGF resemble more the binding mode of the Fab fragment than that of the receptor.
- Published
- 1998
- Full Text
- View/download PDF
32. Novel Peptides Selected to Bind Vascular Endothelial Growth Factor Target the Receptor-Binding Site
- Author
-
Germaine Fuh, Jeffrey Tom, Clifford Quan, Stephanie Shriver, Wayne J. Fairbrother, Christopher J. Keenan, Andrea G. Cochran, Brian C. Cunningham, James A. Wells, and H.W. Christinger
- Subjects
Models, Molecular ,Vascular Endothelial Growth Factor A ,Phage display ,Molecular Sequence Data ,Peptide ,Endothelial Growth Factors ,Biology ,Biochemistry ,Random Allocation ,chemistry.chemical_compound ,Peptide Library ,Humans ,Receptors, Growth Factor ,Amino Acid Sequence ,Receptor ,Peptide library ,Nuclear Magnetic Resonance, Biomolecular ,Peptide sequence ,Cells, Cultured ,chemistry.chemical_classification ,Lymphokines ,Binding Sites ,Base Sequence ,Vascular Endothelial Growth Factors ,Receptor Protein-Tyrosine Kinases ,Kinase insert domain receptor ,Molecular biology ,Growth Inhibitors ,Cell biology ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,Receptors, Vascular Endothelial Growth Factor ,chemistry ,Endothelium, Vascular ,Peptides ,Cell Division - Abstract
Peptides that inhibit binding of vascular endothelial growth factor (VEGF) to its receptors, KDR and Flt-1, have been produced using phage display. Libraries of short disulfide-constrained peptides yielded three distinct classes of peptides that bind to the receptor-binding domain of VEGF with micromolar affinities. The highest affinity peptide was also shown to antagonize VEGF-induced proliferation of primary human umbilical vascular endothelial cells. The peptides bind to a region of VEGF known to contain the contact surface for Flt-1 and the functional determinants for KDR binding. This suggests that the receptor-binding region of VEGF is a binding "hot spot" that is readily targeted by selected peptides and supports earlier assertions that phage-derived peptides frequently target protein-protein interaction sites. Such peptides may lead to the development of pharmacologically useful VEGF antagonists.
- Published
- 1998
- Full Text
- View/download PDF
33. Mechanistic Studies of Antibody-Catalyzed Pyrimidine Dimer Photocleavage
- Author
-
John R. Jacobsen, Andrea G. Cochran, James C. Stephans, David S. King, and Peter G. Schultz
- Subjects
Colloid and Surface Chemistry ,biology ,Chemistry ,Stereochemistry ,biology.protein ,Pyrimidine dimer ,General Chemistry ,Antibody ,Biochemistry ,Catalysis - Published
- 1995
- Full Text
- View/download PDF
34. Recognition of UbcH5c and the nucleosome by the Bmi1/Ring1b ubiquitin ligase complex
- Author
-
Matthew L, Bentley, Jacob E, Corn, Ken C, Dong, Qui, Phung, Tommy K, Cheung, and Andrea G, Cochran
- Subjects
Models, Molecular ,Polycomb Repressive Complex 1 ,Binding Sites ,Protein Conformation ,Recombinant Fusion Proteins ,Ubiquitin-Protein Ligases ,Ubiquitination ,Nuclear Proteins ,DNA ,Sodium Chloride ,Crystallography, X-Ray ,Article ,Nucleosomes ,DNA-Binding Proteins ,Histones ,Repressor Proteins ,Proto-Oncogene Proteins ,Ubiquitin-Conjugating Enzymes ,Mutagenesis, Site-Directed ,Humans ,Protein Binding - Abstract
The Polycomb repressive complex 1 (PRC1) mediates gene silencing, in part by monoubiquitination of histone H2A on lysine 119 (uH2A). Bmi1 and Ring1b are critical components of PRC1 that heterodimerize via their N-terminal RING domains to form an active E3 ubiquitin ligase. We have determined the crystal structure of a complex between the Bmi1/Ring1b RING-RING heterodimer and the E2 enzyme UbcH5c and find that UbcH5c interacts with Ring1b only, in a manner fairly typical of E2-E3 interactions. However, we further show that the Bmi1/Ring1b RING domains bind directly to duplex DNA through a basic surface patch unique to the Bmi1/Ring1b RING-RING dimer. Mutation of residues on this interaction surface leads to a loss of H2A ubiquitination activity. Computational modelling of the interface between Bmi1/Ring1b-UbcH5c and the nucleosome suggests that Bmi1/Ring1b interacts with both nucleosomal DNA and an acidic patch on histone H4 to achieve specific monoubiquitination of H2A. Our results point to a novel mechanism of substrate recognition, and control of product formation, by Bmi1/Ring1b.
- Published
- 2011
35. Reconstitution of a Frizzled8·Wnt3a·LRP6 Signaling Complex Reveals Multiple Wnt and Dkk1 Binding Sites on LRP6
- Author
-
Christine Tam, Mike Costa, Jiyoung Hwang, Andrea G. Cochran, J. Fernando Bazan, James A. Ernst, Rami N. Hannoush, Yvonne Franke, and Eric Bourhis
- Subjects
Frizzled ,Beta-catenin ,Insecta ,Receptors, Cell Surface ,Plasma protein binding ,Biochemistry ,Receptors, G-Protein-Coupled ,Wnt3 Protein ,Inhibitory Concentration 50 ,Mice ,Wnt3A Protein ,Animals ,Humans ,Molecular Biology ,LDL-Receptor Related Proteins ,beta Catenin ,Binding Sites ,biology ,Dose-Response Relationship, Drug ,Wnt signaling pathway ,LRP6 ,LRP5 ,Cell Biology ,Cell biology ,Protein Structure, Tertiary ,Wnt Proteins ,Kinetics ,DKK1 ,Low Density Lipoprotein Receptor-Related Protein-6 ,biology.protein ,Intercellular Signaling Peptides and Proteins ,Signal Transduction ,Protein Binding - Abstract
Wnt/beta-catenin signaling is initiated at the cell surface by association of secreted Wnt with its receptors Frizzled (Fz) and low density lipoprotein receptor-related protein 5/6 (LRP5/6). The study of these molecular interactions has been a significant technical challenge because the proteins have been inaccessible in sufficient purity and quantity. In this report we describe insect cell expression and purification of soluble mouse Fz8 cysteine-rich domain and human LRP6 extracellular domain and show that they inhibit Wnt/beta-catenin signaling in cellular assays. We determine the binding affinities of Wnts and Dickkopf 1 (Dkk1) to the relevant co-receptors and reconstitute in vitro the Fz8 CRD.Wnt3a.LRP6 signaling complex. Using purified fragments of LRP6, we further show that Wnt3a binds to a region including only the third and fourth beta-propeller domains of LRP6 (E3E4). Surprisingly, we find that Wnt9b binds to a different part of the LRP6 extracellular domain, E1E2, and we demonstrate that Wnt3a and Wnt9b can bind to LRP6 simultaneously. Dkk1 binds to both E1E2 and E3E4 fragments and competes with both Wnt3a and Wnt9b for binding to LRP6. The existence of multiple, independent Wnt binding sites on the LRP6 co-receptor suggests new possibilities for the architecture of Wnt signaling complexes and a model for broad-spectrum inhibition of Wnt/beta-catenin signaling by Dkk1.
- Published
- 2010
36. The mitotic regulator Survivin binds as a monomer to its functional interactor Borealin
- Author
-
Sarah G. Hymowitz, Eric Bourhis, and Andrea G. Cochran
- Subjects
Protein family ,Survivin ,Mutant ,Molecular Sequence Data ,Regulator ,Molecular Conformation ,Mitosis ,Cell Cycle Proteins ,Biology ,Inhibitor of apoptosis ,Crystallography, X-Ray ,Ligands ,Biochemistry ,Protein Structure, Secondary ,Inhibitor of Apoptosis Proteins ,Mice ,Animals ,Humans ,Amino Acid Sequence ,neoplasms ,Molecular Biology ,Alanine ,Sequence Homology, Amino Acid ,Mutagenesis ,Cell Biology ,Cell cycle ,Molecular biology ,Cell biology ,Neoplasm Proteins ,Mutation ,Dimerization ,Microtubule-Associated Proteins ,HeLa Cells ,Protein Binding - Abstract
Survivin is a member of the IAP (inhibitor of apoptosis) protein family, defined in part by the presence of a zinc-binding baculoviral inhibitory repeat (BIR) domain. Most BIR domains bind short sequences beginning with alanine, and in this manner, they recognize and block the action of key targets in apoptotic pathways. However, Survivin binds only very weakly to typical IAP ligands. Unique features of Survivin are the long C-terminal helix following the BIR domain and a short segment (linking the helix and BIR domains) that mediates Survivin homodimerization. Despite this detailed knowledge of the structure of Survivin itself, there is a current lack of understanding about how Survivin recognizes cellular binding partners, and consequently, many questions about Survivin function remain unanswered. We determined two co-crystal structures of Survivin and a minimal binding fragment from the chromosomal passenger protein Borealin, a well validated functional interactor. The interaction between Survivin and Borealin involves extensive packing between the long C-terminal helix of Survivin and a long Borealin helix. Surprisingly, an additional important interaction occurs between the Survivin homodimerization interface and a short segment of Borealin. This segment both structurally mimics and displaces one Survivin monomer. The relevance of this unexpected interaction was tested by mutagenesis of two key Borealin residues. Mutant Borealin introduced into HeLa cells failed to localize properly during mitosis and also caused mislocalization of other chromosomal passenger proteins. This suggests that the mutant is dominant-negative and confirms the functional importance of the interaction surface identified in the crystal structures.
- Published
- 2007
37. Stability of cyclic beta-hairpins: asymmetric contributions from side chains of a hydrogen-bonded cross-strand residue pair
- Author
-
Andrea G. Cochran, Nicholas J. Skelton, Stephen J. Russell, and Tamas Blandl
- Subjects
Models, Molecular ,Protein Folding ,Stereochemistry ,Protein design ,Molecular Sequence Data ,Peptide ,Biochemistry ,Catalysis ,Protein Structure, Secondary ,Colloid and Surface Chemistry ,Side chain ,Molecule ,Amino Acid Sequence ,Cysteine ,Disulfides ,Isoleucine ,chemistry.chemical_classification ,Hydrogen bond ,Tryptophan ,Hydrogen Bonding ,Valine ,General Chemistry ,Protein structure prediction ,Amino acid ,chemistry ,Thermodynamics ,Peptides - Abstract
Amino acid structural propensities measured in "host-guest" model studies are often used in protein structure prediction or to choose appropriate residues in de novo protein design. While this concept has proven useful for helical structures, it is more difficult to apply successfully to beta-sheets. We have developed a cyclic beta-hairpin scaffold as a host for measurement of individual residue contributions to hairpin structural stability. Previously, we have characterized substitutions in non-backbone-hydrogen-bonded strand sites; relative stability differences measured in the cyclic host are highly predictive of changes in folding free energy for linear beta-hairpin peptides. Here, we examine the hydrogen-bonded strand positions of our host. Surprisingly, we find a large favorable contribution to stability from a valine (or isoleucine) substitution immediately preceding the C-terminal cysteine of the host peptide, but not at the cross-strand position of the host or in either strand of a folded linear beta-hairpin (trpzip peptide). Further substitutions in the peptides and NMR structural analysis indicate that the stabilizing effect of valine is general for CX(8)C cyclic hairpins and cannot be explained by particular side-chain-side-chain interactions. Instead, a localized decrease in twist of the peptide backbone on the N-terminal side of the cysteine allows the valine side chain to adopt a unique conformation that decreases the solvent accessibility of the peptide backbone. The conformation differs from the highly twisted (coiled) conformation of the trpzip hairpins and is more typical of conformations present in multistranded beta-sheets. This unexpected structural fine-tuning may explain why cyclic hairpins selected from phage-displayed libraries often have valine in the same position, preceding the C-terminal cysteine. It also emphasizes the diversity of structures accessible to beta-strands and the importance of considering not only "beta-propensity", but also hydrogen-bonding pattern and strand twist, when designing beta structures. Finally, we observe correlated, cooperative stabilization from side-chain substitutions on opposite faces of the hairpin. This suggests that cooperative folding in beta-hairpins and other small beta-structures is driven by cooperative strand-strand association.
- Published
- 2003
38. Short constrained peptides that inhibit HIV-1 entry
- Author
-
Peter A. Carr, Vladimir N. Malashkevich, Samuel K. Sia, Andrea G. Cochran, and Peter S. Kim
- Subjects
chemistry.chemical_classification ,Coiled coil ,Circular dichroism ,Multidisciplinary ,Chemistry ,Stereochemistry ,Anti-HIV Agents ,Circular Dichroism ,Entropy ,Peptide ,Hydrogen Bonding ,Calorimetry ,Biological Sciences ,Gp41 ,Crystallography, X-Ray ,Amino acid ,Heptad repeat ,HIV-1 ,Potency ,Humans ,Thermodynamics ,Amino Acid Sequence ,Glycoprotein ,Oligopeptides ,HIV Long Terminal Repeat - Abstract
Peptides corresponding to the C-terminal heptad repeat of HIV-1 gp41 (C-peptides) are potent inhibitors of HIV-1 entry into cells. Their mechanism of inhibition involves binding in a helical conformation to the central coiled coil of HIV-1 gp41 in a dominant–negative manner. Short C-peptides, however, have low binding affinity for gp41 and poor inhibitory activity, which creates an obstacle to the development of small drug-like C-peptides. To improve the inhibitory potency of short C-peptides that target the hydrophobic pocket region of gp41, we use two strategies to stabilize the C-peptide helix: chemical crosslinking and substitution with unnatural helix-favoring amino acids. In this study, the short linear peptide shows no significant inhibitory activity, but a constrained peptide (C14linkmid) inhibits cell–cell fusion at micromolar potency. Structural studies confirm that the constrained peptides bind to the gp41 hydrophobic pocket. Calorimetry reveals that, of the peptides analyzed, the most potent are those that best balance the changes in binding enthalpy and entropy, and surprisingly not those with the highest helical propensity as measured by circular dichroism spectroscopy. Our study reveals the thermodynamic basis of inhibition of an HIV C-peptide, demonstrates the utility of constraining methods for a short antiviral peptide inhibitor, and has implications for the future design of constrained peptides.
- Published
- 2002
39. Quantifying beta-sheet stability by phage display
- Author
-
Andrea G. Cochran, Alan Zhong, and Mark D. Distefano
- Subjects
Models, Molecular ,Protein Folding ,Phage display ,Sequence analysis ,Protein design ,Mutant ,Beta sheet ,Computational biology ,Biology ,Protein Structure, Secondary ,Protein stability ,Structural Biology ,Peptide Library ,Thermal stability ,Bacteriophages ,Cloning, Molecular ,Molecular Biology ,Genetics ,Binding Sites ,Temperature ,Reproducibility of Results ,Pairing ,Immunoglobulin G ,Mutation ,Thermodynamics ,Carrier Proteins - Abstract
The small immunoglobulin G (IgG)-binding protein GB1 is a favored model system for the study of individual residue contributions to the stability of β-sheets. Nevertheless, only a few of the many possible combinations of mutations have been characterized, leaving many questions unanswered. In order to allow the simultaneous evaluation of libraries of mutants, we have adapted a phage-display method, called shotgun scanning. This method combines a binding (i.e. stability) selection with high-throughput sequence analysis. Relative folding free energies determined from GB1-phage sequence data agree well with published GB1 thermal stability studies, validating the use of phage display to conduct quantitative stability studies on GB1, and further suggesting that this method is generally applicable to mutational analysis of protein stability. Examination of residue pairing in our large collection of GB1 mutants indicates that specific side-chain–side-chain interactions are much less important to β-sheet stability than individual residue contributions. The discrepancy between this observation and published studies can be traced to anomalous stability of the alanine-substituted GB1 variants typically used as reference states in double mutant-cycle analyses. Finally, the combination of large library sizes and a quantitative stability selection should allow phage-based “computation” to be applied to protein design problems.
- Published
- 2002
40. Solution structure of a phage-derived peptide antagonist in complex with vascular endothelial growth factor
- Author
-
Jeffrey Tom, Borlan Pan, Andrea G. Cochran, Bing Li, Wayne J. Fairbrother, and Stephen J. Russell
- Subjects
Models, Molecular ,Vascular Endothelial Growth Factor A ,Phage display ,Stereochemistry ,Angiogenesis ,Molecular Sequence Data ,Peptide ,Endothelial Growth Factors ,Crystallography, X-Ray ,Protein Structure, Secondary ,chemistry.chemical_compound ,Protein structure ,Structural Biology ,Proto-Oncogene Proteins ,Humans ,Bacteriophages ,Receptors, Growth Factor ,Amino Acid Sequence ,Disulfides ,Binding site ,Protein Structure, Quaternary ,Molecular Biology ,Peptide sequence ,Nuclear Magnetic Resonance, Biomolecular ,chemistry.chemical_classification ,Lymphokines ,Alanine ,Binding Sites ,Vascular Endothelial Growth Factor Receptor-1 ,Vascular Endothelial Growth Factors ,Receptor Protein-Tyrosine Kinases ,Protein Structure, Tertiary ,Vascular endothelial growth factor ,Solutions ,Vascular endothelial growth factor A ,Receptors, Vascular Endothelial Growth Factor ,chemistry ,Mutation ,Peptides ,Dimerization ,Hydrophobic and Hydrophilic Interactions - Abstract
Vascular endothelial growth factor (VEGF) is a potent endothelial cell-specific mediator of angiogenesis and vasculogenesis. VEGF is involved pathologically in cancer, proliferative retinopathy and rheumatoid arthritis, and as such represents an important therapeutic target. Three classes of disulfide-constrained peptides that antagonize binding of the VEGF dimer to its receptors, KDR and Flt-1, were identified previously using phage display methods. NMR studies of a representative peptide from the most potent class of these peptide antagonists, v107 (GGNECDAIRMWEWECFERL), were undertaken to characterize its interactions with VEGF. v107 has no defined structure free in solution, but binding to VEGF induces folding of the peptide. The solution structure of the VEGF receptor-binding domain-v107 complex was determined using 3940 (1970 per VEGF monomer) internuclear distance and 476 (238 per VEGF monomer) dihedral angle restraints derived from NMR data obtained using samples containing either (13)C/(15)N-labeled protein plus excess unlabeled peptide or (13)C/(15)N-labeled peptide plus excess unlabeled protein. Residual dipolar coupling restraints supplemented the structure determination of the complex and were found to increase significantly both the global precision of VEGF in the complex and the agreement with available crystal structures of VEGF. The calculated ensemble of structures is of high precision and is in excellent agreement with the experimental restraints. v107 has a turn-helix conformation with hydrophobic residues partitioned to one face of the peptide and polar or charged residues at the other face. Contacts between two v107 peptides and the VEGF dimer are mediated by primarily hydrophobic side-chain interactions. The v107-binding site on VEGF overlaps partially with the binding site of KDR and is similar to that for domain 2 of Flt-1. The structure of the VEGF-v107 complex provides new insight into how binding to VEGF can be achieved that may be useful for the design of small molecule antagonists.
- Published
- 2002
41. Protein–protein interfaces: mimics and inhibitors
- Author
-
Andrea G. Cochran
- Subjects
Models, Molecular ,Peptidomimetic ,Protein Conformation ,Chemical biology ,Biology ,Biochemistry ,Protein–protein interaction ,Analytical Chemistry ,Molecular recognition ,Proto-Oncogene Proteins ,medicine ,Protein protein ,Molecular Mimicry ,Cancer ,Membrane Proteins ,Nuclear Proteins ,Proteins ,Proto-Oncogene Proteins c-mdm2 ,medicine.disease ,Small molecule ,bcl-2 Homologous Antagonist-Killer Protein ,Proto-Oncogene Proteins c-bcl-2 ,Tumor Suppressor Protein p53 ,Protein ligand ,Protein Binding - Abstract
Many biological processes are mediated by specific molecular recognition between proteins. However, the thermodynamic and structural ‘rules’ for such recognition are incompletely understood, as is the potential for inhibition by small molecules. Recent progress has included the discovery of small-molecule inhibitors for several targets important in cancer.
- Published
- 2001
- Full Text
- View/download PDF
42. A minimal peptide scaffold for beta-turn display: optimizing a strand position in disulfide-cyclized beta-hairpins
- Author
-
Robert S. McDowell, and J. E. Theaker, Andrea G. Cochran, Melissa A. Starovasnik, Ricky T. Tong, Eleanor J. Park, and Nicholas J. Skelton
- Subjects
chemistry.chemical_classification ,Models, Molecular ,Scaffold ,Protein Folding ,Phage display ,Disulfide bond ,Peptide ,General Chemistry ,Biochemistry ,Combinatorial chemistry ,Catalysis ,Protein Structure, Secondary ,Structure-Activity Relationship ,Colloid and Surface Chemistry ,chemistry ,Peptide Library ,Thermodynamics ,Disulfides ,Protein target ,Nuclear Magnetic Resonance, Biomolecular - Abstract
Phage display of peptide libraries has become a powerful tool for the evolution of novel ligands that bind virtually any protein target. However, the rules governing conformational preferences in natural peptides are poorly understood, and consequently, structure-activity relationships in these molecules can be difficult to define. In an effort to simplify this process, we have investigated the structural stability of 10-residue, disulfide-constrained beta-hairpins and assessed their suitability as scaffolds for beta-turn display. Using disulfide formation as a probe, relative free energies of folding were measured for 19 peptides that differ at a one strand position. A tryptophan substitution promotes folding to a remarkable degree. NMR analysis confirms that the measured energies correlate well with the degree of beta-hairpin structure in the disulfide-cyclized peptides. Reexamination of a subset of the strand substitutions in peptides with different turn sequences reveals linear free energy relationships, indicating that turns and strand-strand interactions make independent, additive contributions to hairpin stability. Significantly, the tryptophan strand substitution is highly stabilizing with all turns tested, and peptides that display model turns or the less stable C'-C' ' turn of CD4 on this tryptophan "stem" are highly structured beta-hairpins in water. Thus, we have developed a small, structured beta-turn scaffold, containing only natural L-amino acids, that may be used to display peptide libraries of limited conformational diversity on phage.
- Published
- 2001
43. Drug Bioavailability. Edited by Han van de Waterbeemd and Bernard Testa
- Author
-
Andrea G. Cochran
- Subjects
Pharmacology ,Philosophy ,Organic Chemistry ,Drug Discovery ,Molecular Medicine ,General Pharmacology, Toxicology and Pharmaceutics ,Biochemistry ,Bioavailability - Published
- 2009
- Full Text
- View/download PDF
44. Antibody-catalyzed bimolecular imine formation
- Author
-
Tony Pham, Andrea G. Cochran, Peter G. Schultz, and Renee Sugasawara
- Subjects
chemistry.chemical_compound ,Colloid and Surface Chemistry ,Reaction rate constant ,chemistry ,Imine ,Kinetics ,Organic chemistry ,Stereoselectivity ,General Chemistry ,Biochemistry ,Medicinal chemistry ,Catalysis - Published
- 1991
- Full Text
- View/download PDF
45. Antibody-Catalyzed Porphyrin Metallation
- Author
-
Andrea G. Cochran and Peter G. Schultz
- Subjects
inorganic chemicals ,Porphyrins ,Stereochemistry ,Medicinal chemistry ,Redox ,Antibodies ,Catalysis ,chemistry.chemical_compound ,Chelation ,Enzyme kinetics ,Antigens ,Manganese ,Multidisciplinary ,Protoporphyrin IX ,biology ,Cobalt ,Ferrochelatase ,Porphyrin ,Turnover number ,Kinetics ,Zinc ,Mesoporphyrins ,chemistry ,Metals ,biology.protein ,Hapten ,Copper - Abstract
An antibody elicited to a distorted N-methyl porphyrin catalyzed metal ion chelation by the planar porphyrin. At fixed Zn2+ and Cu2+ concentrations, the antibody-catalyzed reaction showed saturation kinetics with respect to the substrate mesoporphyrin IX (2) and was inhibited by the hapten, N-methylmesoporphyrin IX (1). The turnover number of 80 hour-1 for antibody-catalyzed metallation of 2 with Zn2+ compares with an estimated value of 800 hour-1 for ferrochelatase. The antibody also catalyzed the insertion of Co2+ and Mn2+ into 2, but it did not catalyze the metallation of protoporphyrin IX (3) or deuteroporphyrin IX (4). The antibody has high affinity for several metalloporphyrins, suggesting an approach to developing antibody-heme catalysts for redox or electron transfer reactions.
- Published
- 1990
- Full Text
- View/download PDF
46. Imitation of Escherichia coli aspartate receptor signaling in engineered dimers of the cytoplasmic domain
- Author
-
Peter S. Kim and Andrea G. Cochran
- Subjects
Conformational change ,Cytoplasm ,Histidine Kinase ,Protein Conformation ,Recombinant Fusion Proteins ,Allosteric regulation ,Molecular Sequence Data ,Methyl-Accepting Chemotaxis Proteins ,Receptors, Cell Surface ,Biology ,Ligands ,Methylation ,Protein Structure, Secondary ,Bacterial Proteins ,Escherichia coli ,Receptors, Amino Acid ,5-HT5A receptor ,Amino Acid Sequence ,Phosphorylation ,Receptor ,Leucine Zippers ,Multidisciplinary ,Chemotaxis ,Escherichia coli Proteins ,Membrane Proteins ,Ligand (biochemistry) ,Chemoreceptor Cells ,Enzyme Activation ,Transmembrane domain ,Biochemistry ,Biophysics ,Signal transduction ,Protein Kinases ,Signal Transduction - Abstract
Transmembrane signaling by bacterial chemotaxis receptors appears to require a conformational change within a receptor dimer. Dimers were engineered of the cytoplasmic domain of the Escherichia coli aspartate receptor that stimulated the kinase CheA in vitro. The folding free energy of the leucine-zipper dimerization domain was harnessed to twist the dimer interface of the receptor, which markedly affected the extent of CheA activation. Response to this twist was attenuated by modification of receptor regulatory sites, in the same manner as adaptation resets sensitivity to ligand in vivo. These results suggest that the normal allosteric activation of the chemotaxis receptor has been mimicked in a system that lacks both ligand-binding and transmembrane domains. The most stimulatory receptor dimer formed a species of tetrameric size.
- Published
- 1996
47. Abstract A160: Recognition of UbcH5c and the nucleosome by the Bmi1/Ring1b ubiquitin ligase complex
- Author
-
Qui Phung, Tommy K. Cheung, Matthew L. Bentley, Jacob E. Corn, Andrea G. Cochran, and Ken C. Dong
- Subjects
Cancer Research ,biology ,macromolecular substances ,Ubiquitin ligase ,Histone H4 ,Ubiquitins ,Oncology ,Biochemistry ,Ubiquitin ,Ubiquitin ligase complex ,Histone H2A ,biology.protein ,Biophysics ,Nucleosome ,Monoubiquitination - Abstract
Background and Objectives: The Polycomb repressive complex 1 (PRC1) mediates gene silencing, in part by monoubiquitination of histone H2A on lysine 119 (uH2A). Bmi1 and Ring1b are critical components of PRC1 that heterodimerize via their N-terminal RING domains to form an active E3 ubiquitin ligase. A longstanding question in this field is how a single lysine residue in the nucleosome is singled out for ubiquitin modification and why only one ubiquitin is added rather than a chain of ubiquitins. To better understand how Bmi1/Ring1b controls monoubiquitination of H2A, we structurally characterized the E2 and nucleosome interaction sites on Bmi1/Ring1b. Methods: X-ray crystallography was used to determine the structure of a complex between Bmi1/Ring1b and UbcH5c, its cognate E2 enzyme. Fluorescence-polarization assays were used to measure the binding of Bmi1/Ring1b to a synthetic DNA duplex. Site-directed mutagenesis was used to assess the importance of residues at the potential Bmi1/Ring1b-DNA interface, and in vitro ubiquitin ligase assays were used to measure the catalytic activity of mutant complexes. Molecular modeling studies were performed using HADDOCK v2.0. Results: We have determined the crystal structure of a complex between the Bmi1/Ring1b RING-RING heterodimer and the E2 enzyme UbcH5c and find that UbcH5c interacts with Ring1b only, in a manner fairly typical of E2-E3 interactions. However, we further show that the Bmi1/Ring1b RING domains bind directly to duplex DNA through a basic surface patch unique to the Bmi1/Ring1b RING-RING dimer. Mutation of residues on this interaction surface leads to a loss of H2A ubiquitination activity. Through site-directed mutagenesis we have been able to uncouple the E2 binding and the DNA binding activities of Bmi1/Ring1b, indicating that Bmi1/Ring1b uses distinct binding surfaces to recognize E2 and the nucleosomal substrate. Computational modeling of the interface between Bmi1/Ring1b-UbcH5c and the nucleosome suggests that Bmi1/Ring1b interacts with both nucleosomal DNA and an acidic patch on histone H4 to achieve specific mono-ubiquitination of H2A. Conclusions: Our data show that the direct interaction of the RING domains with nucleosomal DNA is crucial for the ubiquitin ligase activity of Bmi1/Ring1b. To our knowledge, this is the first example of a RING-domain E3 ligase binding directly to its substrate via the RING domain. Our results point to a novel mechanism of substrate recognition, and control of product formation, by Bmi1/Ring1b. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr A160.
- Published
- 2011
- Full Text
- View/download PDF
48. Designing Stable β-Hairpins: Energetic Contributions from Cross-Strand Residues
- Author
-
Andrea G. Cochran and Stephen J. Russell and
- Subjects
Colloid and Surface Chemistry ,Chemistry ,Stereochemistry ,General Chemistry ,Biochemistry ,Catalysis - Published
- 2000
- Full Text
- View/download PDF
49. Peroxidase activity of an antibody-heme complex
- Author
-
Andrea G. Cochran and Peter G. Schultz
- Subjects
chemistry.chemical_classification ,biology ,General Chemistry ,Biochemistry ,Catalysis ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,Enzyme ,chemistry ,biology.protein ,Antibody ,Heme ,Peroxidase - Published
- 1990
- Full Text
- View/download PDF
50. Substrate Capacity Considerations in Developing Kinase Assays.
- Author
-
Laura E. DeForge, Andrea G. Cochran, Sherry H. Yeh, Brian S. Robinson, Karen L. Billeci, and Wai Lee T. Wong
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.