134 results on '"Lyn H. Jones"'
Search Results
2. Covalent drug discovery using sulfur(VI) fluoride exchange warheads
- Author
-
Huang Huang and Lyn H. Jones
- Subjects
Drug Discovery - Published
- 2023
- Full Text
- View/download PDF
3. Catalytic Degraders Effectively Address Kinase Site Mutations in EML4-ALK Oncogenic Fusions
- Author
-
Yang Gao, Baishan Jiang, Hellen Kim, Matthew J. Berberich, Jianwei Che, Katherine A. Donovan, John M. Hatcher, Fidel Huerta, Nicholas P. Kwiatkowski, Yingpeng Liu, Peter P. Liuni, Rebecca J. Metivier, Vineeth K. Murali, Radosław P. Nowak, Tinghu Zhang, Eric S. Fischer, Nathanael S. Gray, and Lyn H. Jones
- Subjects
Drug Discovery ,Molecular Medicine - Published
- 2023
- Full Text
- View/download PDF
4. HiBiT-SpyTag: A Minimal Tag for Covalent Protein Capture and Degrader Development
- Author
-
Tiffany Tsang, Fidel Huerta, Yingpeng Liu, Jianwei Che, Rebecca J. Metivier, Silas Ferrao, Katherine A. Donovan, Lyn H. Jones, Breanna L. Zerfas, and Radosław P. Nowak
- Subjects
Molecular Medicine ,General Medicine ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
5. Beyond Inhibition
- Author
-
Milka Kostic and Lyn H. Jones
- Published
- 2022
- Full Text
- View/download PDF
6. Introduction to the themed collection on Covalent Drug Discovery
- Author
-
Keriann M. Backus, Zhengying Pan, and Lyn H. Jones
- Subjects
Pharmacology ,Organic Chemistry ,Drug Discovery ,Pharmaceutical Science ,Molecular Medicine ,Biochemistry - Abstract
Guest editors Keriann Backus, Zhengying Pan and Lyn Jones introduce the themed collection on Covalent Drug Discovery.
- Published
- 2023
7. Cereblon target validation using a covalent inhibitor of neosubstrate recruitment
- Author
-
Geoffrey P. Dann, Hu Liu, Radosław P. Nowak, and Lyn H. Jones
- Published
- 2023
- Full Text
- View/download PDF
8. Advancing targeted protein degrader discovery by measuring cereblon engagement in cells
- Author
-
Breanna L. Zerfas, Fidel Huerta, Hu Liu, Guangyan Du, Nathanael S. Gray, Lyn H. Jones, and Radosław P. Nowak
- Published
- 2023
- Full Text
- View/download PDF
9. Physicochemistry of Cereblon Modulating Drugs Determines Pharmacokinetics and Disposition
- Author
-
Hu Liu, Nikki R. Kong, Jianwei Che, and Lyn H. Jones
- Subjects
Zinc finger ,Drug ,Drug discovery ,Chemistry ,Cereblon ,media_common.quotation_subject ,Organic Chemistry ,Protein degradation ,Pharmacology ,Pomalidomide ,Biochemistry ,Thalidomide ,Pharmacokinetics ,Drug Discovery ,medicine ,medicine.drug ,media_common - Abstract
[Image: see text] Immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide engage cereblon and mediate a protein interface with neosubstrates such as zinc finger transcription factors promoting their polyubiquitination and degradation. The IMiDs have garnered considerable excitement in drug discovery, leading to exploration of targeted protein degradation strategies. Although the molecular modes-of-action of the IMiDs and related degraders have been the subject of intense research, their pharmacokinetics and disposition have been relatively understudied. Here, we assess the effects of physicochemistry of the IMiDs, the phthalimide EM-12, and the candidate drug CC-220 (iberdomide) on lipophilicity, solubility, metabolism, permeability, intracellular bioavailability, and cell-based potency. The insights yielded in this study will enable the rational property-based design and development of targeted protein degraders in the future.
- Published
- 2021
- Full Text
- View/download PDF
10. In vitro and in vivo effects of 2,4 diaminoquinazoline inhibitors of the decapping scavenger enzyme DcpS: Context-specific modulation of SMN transcript levels.
- Author
-
Jonathan J Cherry, Christine J DiDonato, Elliot J Androphy, Alessandro Calo, Kyle Potter, Sara K Custer, Sarah Du, Timothy L Foley, Ariamala Gopalsamy, Emily J Reedich, Susana M Gordo, William Gordon, Natalie Hosea, Lyn H Jones, Daniel K Krizay, Gregory LaRosa, Hongxia Li, Sachin Mathur, Carol A Menard, Paraj Patel, Rebeca Ramos-Zayas, Anne Rietz, Haojing Rong, Baohong Zhang, and Michael A Tones
- Subjects
Medicine ,Science - Abstract
C5-substituted 2,4-diaminoquinazoline inhibitors of the decapping scavenger enzyme DcpS (DAQ-DcpSi) have been developed for the treatment of spinal muscular atrophy (SMA), which is caused by genetic deficiency in the Survival Motor Neuron (SMN) protein. These compounds are claimed to act as SMN2 transcriptional activators but data underlying that claim are equivocal. In addition it is unclear whether the claimed effects on SMN2 are a direct consequence of DcpS inhibitor or might be a consequence of lysosomotropism, which is known to be neuroprotective. DAQ-DcpSi effects were characterized in cells in vitro utilizing DcpS knockdown and 7-methyl analogues as probes for DcpS vs non-DcpS-mediated effects. We also performed analysis of Smn transcript levels, RNA-Seq analysis of the transcriptome and SMN protein in order to identify affected pathways underlying the therapeutic effect, and studied lysosomotropic and non-lysosomotropic DAQ-DCpSi effects in 2B/- SMA mice. Treatment of cells caused modest and transient SMN2 mRNA increases with either no change or a decrease in SMNΔ7 and no change in SMN1 transcripts or SMN protein. RNA-Seq analysis of DAQ-DcpSi-treated N2a cells revealed significant changes in expression (both up and down) of approximately 2,000 genes across a broad range of pathways. Treatment of 2B/- SMA mice with both lysomotropic and non-lysosomotropic DAQ-DcpSi compounds had similar effects on disease phenotype indicating that the therapeutic mechanism of action is not a consequence of lysosomotropism. In striking contrast to the findings in vitro, Smn transcripts were robustly changed in tissues but there was no increase in SMN protein levels in spinal cord. We conclude that DAQ-DcpSi have reproducible benefit in SMA mice and a broad spectrum of biological effects in vitro and in vivo, but these are complex, context specific, and not the result of simple SMN2 transcriptional activation.
- Published
- 2017
- Full Text
- View/download PDF
11. Covalent drugs targeting histidine - an unexploited opportunity?
- Author
-
Jianwei Che and Lyn H. Jones
- Subjects
Pharmacology ,Organic Chemistry ,Drug Discovery ,Pharmaceutical Science ,Molecular Medicine ,Biochemistry - Abstract
Covalent drugs and chemical probes often possess pharmacological advantages over reversible binding ligands, such as enhanced potency and pharmacodynamic duration. The highly nucleophilic cysteine thiol is commonly targeted using acrylamide electrophiles, but the amino acid is rarely present in protein binding sites. Sulfonyl exchange chemistry has expanded the covalent drug discovery toolkit by enabling the rational design of irreversible inhibitors targeting tyrosine, lysine, serine and threonine. Probes containing the sulfonyl fluoride warhead have also been shown to serendipitously label histidine residues in proteins. Histidine targeting is an attractive prospect because the residue is frequently proximal to protein small molecule ligands and the imidazole side chain possesses desirable nucleophilicity. We recently reported the design of cereblon molecular glues to site-selectively modify a histidine in the thalidomide binding site using sulfonyl exchange chemistry. We believe that histidine targeting holds great promise for future covalent drug development and this Opinion highlights these opportunities.
- Published
- 2022
12. Medicinal Chemical Biology
- Author
-
Lyn H. Jones
- Subjects
Chemical biology ,Chemoproteomics ,Computational biology ,Biology ,Protein labeling - Published
- 2021
- Full Text
- View/download PDF
13. Target Validation Using PROTACs: Applying the Four Pillars Framework
- Author
-
Radosław P. Nowak and Lyn H. Jones
- Subjects
Proteasome Endopeptidase Complex ,Computer science ,Ubiquitin-Protein Ligases ,Computational biology ,Protein degradation ,01 natural sciences ,Biochemistry ,Analytical Chemistry ,Structure-Activity Relationship ,03 medical and health sciences ,Ubiquitin ,Drug Discovery ,Humans ,Molecular Targeted Therapy ,030304 developmental biology ,0303 health sciences ,biology ,010405 organic chemistry ,Drug discovery ,Ubiquitination ,Pillar ,0104 chemical sciences ,Ubiquitin ligase ,Eukaryotic Cells ,Proteolysis ,biology.protein ,Molecular Medicine ,Biological Assay ,sense organs ,Experimental methods ,Protein Processing, Post-Translational ,Site of action ,Function (biology) ,Biotechnology - Abstract
Proteolysis targeting chimeras (PROTACs) are heterobifunctional compounds that recruit the E3 ubiquitin ligase machinery to proteins of interest, resulting in their ubiquitination and subsequent proteasomal degradation. Targeted protein degradation has generated considerable interest in drug discovery because inhibition of one particular function of a protein often does not deliver the therapeutic efficacy that results from whole-protein depletion. However, the physicochemistry and intrinsically complex pharmacology of PROTACs present challenges, particularly for the development of orally bioavailable drugs. Here we describe the application of a translational pharmacology framework (called the four pillars) to expedite PROTAC development by informing pharmacokinetic-pharmacodynamic (PKPD) understanding and helping elucidate structure-activity relationships. Experimental methods are reviewed that help illuminate exposure of the drug or probe at the site of action (pillar 1) and engagement of its target(s) (pillar 2) that drive functional pharmacological effects (pillar 3) resulting in modulation of a relevant phenotype (pillar 4). We hope the guidance will be useful to those developing targeted protein degraders and help establish PROTAC molecules as robust target validation chemical probes.
- Published
- 2021
- Full Text
- View/download PDF
14. Fragment-based covalent ligand discovery
- Author
-
Tinghu Zhang, Edward T. Chouchani, Nathanael S. Gray, Matthew P. Patricelli, Milka Kostic, Lyn H. Jones, Jianwei Che, and Wenchao Lu
- Subjects
0303 health sciences ,010405 organic chemistry ,Drug discovery ,Chemistry ,Ligand ,Sample processing ,Computational biology ,01 natural sciences ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Biochemistry ,Small molecule ,0104 chemical sciences ,03 medical and health sciences ,Fragment (logic) ,Chemistry (miscellaneous) ,Covalent bond ,Molecular Biology ,030304 developmental biology - Abstract
Targeted covalent inhibitors have regained widespread attention in drug discovery and have emerged as powerful tools for basic biomedical research. Fueled by considerable improvements in mass spectrometry sensitivity and sample processing, chemoproteomic strategies have revealed thousands of proteins that can be covalently modified by reactive small molecules. Fragment-based drug discovery, which has traditionally been used in a target-centric fashion, is now being deployed on a proteome-wide scale thereby expanding its utility to both the discovery of novel covalent ligands and their cognate protein targets. This powerful approach is allowing ‘high-throughput’ serendipitous discovery of cryptic pockets leading to the identification of pharmacological modulators of proteins previously viewed as “undruggable”. The reactive fragment toolkit has been enabled by recent advances in the development of new chemistries that target residues other than cysteine including lysine and tyrosine. Here, we review the emerging area of covalent fragment-based ligand discovery, which integrates the benefits of covalent targeting and fragment-based medicinal chemistry. We discuss how the two strategies synergize to facilitate the efficient discovery of new pharmacological modulators of established and new therapeutic target proteins., Covalent fragment-based ligand discovery greatly facilitates the discovery of useful fragments for drug discovery and helps unveil chemical-tractable biological targets in native biological systems.
- Published
- 2021
15. PF-07059013: A Noncovalent Modulator of Hemoglobin for Treatment of Sickle Cell Disease
- Author
-
Ariamala Gopalsamy, Ann E. Aulabaugh, Amey Barakat, Kevin C. Beaumont, Shawn Cabral, Daniel P. Canterbury, Agustin Casimiro-Garcia, Jeanne S. Chang, Ming Z. Chen, Chulho Choi, Robert L. Dow, Olugbeminiyi O. Fadeyi, Xidong Feng, Scott P. France, Roger M. Howard, Jay M. Janz, Jayasankar Jasti, Reema Jasuja, Lyn H. Jones, Amanda King-Ahmad, Kelly M. Knee, Jeffrey T. Kohrt, Chris Limberakis, Spiros Liras, Carlos A. Martinez, Kim F. McClure, Arjun Narayanan, Jatin Narula, Jonathan J. Novak, Thomas N. O’Connell, Mihir D. Parikh, David W. Piotrowski, Olga Plotnikova, Ralph P. Robinson, Parag V. Sahasrabudhe, Raman Sharma, Benjamin A. Thuma, Dipy Vasa, Liuqing Wei, A. Zane Wenzel, Jane M. Withka, Jun Xiao, and Hatice G. Yayla
- Subjects
Hemolytic anemia ,Erythrocytes ,Chemistry ,Point mutation ,Hemoglobin, Sickle ,Cell ,Genetic disorder ,Phases of clinical research ,Hemoglobin A ,Anemia, Sickle Cell ,Disease ,Pharmacology ,Multiple dose ,medicine.disease ,Oxygen ,Mice ,medicine.anatomical_structure ,Drug Discovery ,Quinolines ,medicine ,Animals ,Molecular Medicine ,Hemoglobin - Abstract
Sickle cell disease (SCD) is a genetic disorder caused by a single point mutation (β6 Glu → Val) on the β-chain of adult hemoglobin (HbA) that results in sickled hemoglobin (HbS). In the deoxygenated state, polymerization of HbS leads to sickling of red blood cells (RBC). Several downstream consequences of polymerization and RBC sickling include vaso-occlusion, hemolytic anemia, and stroke. We report the design of a noncovalent modulator of HbS, clinical candidate PF-07059013 (23). The seminal hit molecule was discovered by virtual screening and confirmed through a series of biochemical and biophysical studies. After a significant optimization effort, we arrived at 23, a compound that specifically binds to Hb with nanomolar affinity and displays strong partitioning into RBCs. In a 2-week multiple dose study using Townes SCD mice, 23 showed a 37.8% (±9.0%) reduction in sickling compared to vehicle treated mice. 23 (PF-07059013) has advanced to phase 1 clinical trials.
- Published
- 2020
- Full Text
- View/download PDF
16. Cereblon covalent modulation through structure-based design of histidine targeting chemical probes
- Author
-
Justin T. Cruite, Geoffrey P. Dann, Jianwei Che, Katherine A. Donovan, Silas Ferrao, Scott B. Ficarro, Eric S. Fischer, Nathanael S. Gray, Fidel Huerta, Nikki R. Kong, Hu Liu, Jarrod A. Marto, Rebecca J. Metivier, Radosław P. Nowak, Breanna L. Zerfas, and Lyn H. Jones
- Subjects
Chemistry (miscellaneous) ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Molecular Biology ,Biochemistry - Abstract
Electrophilic biocompatible warheads, particularly cysteine-reactive acrylamides, have enabled the development of covalent inhibitor drugs and chemical biology probes, but cysteine is rarely present in protein binding sites. Therefore, expansion of the list of targetable amino acid residues is required to augment the synthetic bology toolkit of site-selective protein modifications. This work describes the first rational targeting of a specific histidine residue in a protein binding site using sulfonyl exchange chemistry. Structure-based drug design was used to incorporate sulfonyl fluoride and triazole reactive groups into the isoindolinone thalidomide congener EM12 to yield potent covalent inhibitors of the cereblon E3 ubiquitin ligase complex through engagement of His353. Conversely, the fluorosulfate derivative EM12-FS labels His353, but degrades a novel neosubstrate, the protein N-terminal glutamine amidohydrolase NTAQ1, which is involved in the N-end rule pathway and DNA damage response. Targeted protein degradation using cereblon ligands has become an important new drug discovery modality and the chemical probes and covalent labeling strategy described here will broadly impact this exciting area of therapeutic research.
- Published
- 2022
17. Target Validation—Prosecuting the Target
- Author
-
Lyn H. Jones
- Subjects
Chemistry - Published
- 2022
- Full Text
- View/download PDF
18. Targeted protein degraders: a call for collective action to advance safety assessment
- Author
-
Lyn H, Jones, Constance A, Mitchell, Lise, Loberg, Mira, Pavkovic, Mohan, Rao, Ruth, Roberts, Katie, Stamp, Laurie, Volak, Matthias B, Wittwer, and Syril, Pettit
- Subjects
Pharmacology ,Proteolysis ,Drug Discovery ,Humans ,Proteins ,General Medicine - Published
- 2022
- Full Text
- View/download PDF
19. Labeling Preferences of Diazirines with Protein Biomolecules
- Author
-
Andrew C. Huang, Lyn H. Jones, Giovanni Muncipinto, Christina M. Woo, Hung-Yi Wu, Matthew T Labenski, and Alexander V West
- Subjects
Proteome ,Protein Conformation ,Photoaffinity Labels ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Catalysis ,chemistry.chemical_compound ,Colloid and Surface Chemistry ,Protein structure ,Humans ,Reactivity (chemistry) ,Amino Acids ,Alkyl ,chemistry.chemical_classification ,Binding Sites ,Photoaffinity labeling ,Chemistry ,Biomolecule ,Voltage-Dependent Anion Channel 1 ,Proteins ,General Chemistry ,Diazonium Compounds ,Hydrogen-Ion Concentration ,Combinatorial chemistry ,0104 chemical sciences ,Amino acid ,Diazomethane ,Diazirine ,Diazo - Abstract
Diazirines are widely used in photoaffinity labeling (PAL) to trap noncovalent interactions with biomolecules. However, design and interpretation of PAL experiments is challenging without a molecular understanding of the reactivity of diazirines with protein biomolecules. Herein, we report a systematic evaluation of the labeling preferences of alkyl and aryl diazirines with individual amino acids, single proteins, and in the whole cell proteome. We find that alkyl diazirines exhibit preferential labeling of acidic amino acids in a pH-dependent manner that is characteristic of a reactive alkyl diazo intermediate, while the aryl-fluorodiazirine labeling pattern reflects reaction primarily through a carbene intermediate. From a survey of 32 alkyl diazirine probes, we use this reactivity profile to rationalize why alkyl diazirine probes preferentially enrich highly acidic proteins or those embedded in membranes and why probes with a net positive charge tend to produce higher labeling yields in cells and in vitro. These results indicate that alkyl diazirines are an especially effective chemistry for surveying the membrane proteome and will facilitate design and interpretation of biomolecular labeling experiments with diazirines.
- Published
- 2021
20. Design of next-generation covalent inhibitors: Targeting residues beyond cysteine
- Author
-
Lyn H. Jones
- Subjects
Serine ,chemistry.chemical_classification ,chemistry.chemical_compound ,Methionine ,Biochemistry ,Chemistry ,Chemical biology ,Plasma protein binding ,Small molecule ,Histidine ,Amino acid ,Cysteine - Abstract
Covalent protein inhibitors have found considerable utility in medicinal chemistry and chemical biology. Chemical probes and drugs have been rationally designed and fine-tuned to engage cysteine residues predominantly using electrophilic acrylamide warheads that deliver inhibitors with improved pharmacological efficacy, duration and selectivity over reversible binders. However, cysteine is rarely present in protein binding sites and so there is a need to advance inhibitors that engage alternative amino acid residues. This review describes recent progress in the development of covalent small molecules that target residues beyond cysteine. Numerous electrophiles have been incorporated into inhibitors that selectively label various reactive amino acid side chains including lysine, tyrosine, histidine, serine, threonine, glutamate, aspartate and methionine. Several synthetic strategies have facilitated these advances, particularly in the area of sulfonyl-exchange chemistry, though other emerging approaches are presented. We hope the review will inspire future research into next-generation covalent protein inhibitors that positively effect therapeutic discovery and drive fundamental biological insights through novel chemical probe development.
- Published
- 2021
- Full Text
- View/download PDF
21. Selection of a novel anti-nicotine vaccine: influence of antigen design on antibody function in mice.
- Author
-
David C Pryde, Lyn H Jones, David P Gervais, David R Stead, David C Blakemore, Matthew D Selby, Alan D Brown, Jotham W Coe, Matthew Badland, David M Beal, Rebecca Glen, Yvonne Wharton, Gavin J Miller, Phil White, Ningli Zhang, Michelle Benoit, Karen Robertson, James R Merson, Heather L Davis, and Michael J McCluskie
- Subjects
Medicine ,Science - Abstract
Anti-nicotine vaccines may aid smoking cessation via the induction of anti-nicotine antibodies (Ab) which reduce nicotine entering the brain, and hence the associated reward. Ab function depends on both the quantity (titer) and the quality (affinity) of the Ab. Anti-nicotine vaccines tested previously in clinical studies had poor efficacy despite high Ab titer, and this may be due to inadequate function if Ab of low affinity were induced. In this study, we designed and synthesized a series of novel nicotine-like haptens which were all linked to diphtheria toxoid (DT) as carrier, but which differed in the site of attachment of linker to nicotine, the nature of linker used, and the handle used to attach the hapten to DT. The resulting hapten conjugates were evaluated in a mouse model, using CpG (a TLR9 agonist) and aluminum hydroxide (Al(OH)3) as adjuvants, whereby Ab titers, affinity and function were evaluated using a radiolabeled nicotine challenge model. A series of additional linkers varying in length, rigidity and polarity were used with a single hapten to generate additional DT-conjugates, which were also tested in mice. Conjugates made with different haptens resulted in various titers of anti-nicotine Ab. Several haptens gave similarly high Ab titers, but among these, Ab affinity and hence function varied considerably. Linker also influenced Ab titer, affinity and function. These results demonstrate that immune responses induced in mice by nicotine-conjugate antigens are greatly influenced by hapten design including site of attachment of linker to nicotine, the nature of linker used, and the handle used to attach the hapten to DT. While both Ab titer and affinity contributed to function, affinity was more sensitive to antigen differences.
- Published
- 2013
- Full Text
- View/download PDF
22. LanCLs add glutathione to dehydroamino acids generated at phosphorylated sites in the proteome
- Author
-
Ritu Raj, Wilfred A. van der Donk, Kuan Yu Lai, Neha Garg, Satish K. Nair, Shabaz Mohammed, Jie Chen, Benjamin G. Davis, Sébastien R. G. Galan, Graham J. Hutchings, Min Zeng, Chang He, Jitka Riedl, Yibo Zeng, Tianhui Hina Zhou, K. Phin Chooi, Shi Liu, and Lyn H. Jones
- Subjects
Male ,Proteome ,Antimicrobial peptides ,MAP Kinase Kinase 1 ,Biology ,Sulfides ,General Biochemistry, Genetics and Molecular Biology ,Article ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Protein Domains ,Dehydroalanine ,Animals ,Humans ,Phosphorylation ,030304 developmental biology ,chemistry.chemical_classification ,Mice, Knockout ,Mitogen-Activated Protein Kinase Kinases ,0303 health sciences ,Alanine ,Kinase ,Aminobutyrates ,Membrane Proteins ,Glutathione ,Phosphate-Binding Proteins ,Enzyme ,HEK293 Cells ,chemistry ,Biochemistry ,Nucleic acid ,Female ,030217 neurology & neurosurgery ,Antimicrobial Cationic Peptides - Abstract
Enzyme-mediated damage repair or mitigation, while common for nucleic acids, is rare for proteins. Examples of protein damage are elimination of phosphorylated Ser/Thr to dehydroalanine/dehydrobutyrine (Dha/Dhb) in pathogenesis and aging. Bacterial LanC enzymes use Dha/Dhb to form carbon-sulfur linkages in antimicrobial peptides, but the functions of eukaryotic LanC-like (LanCL) counterparts are unknown. We show that LanCLs catalyze the addition of glutathione to Dha/Dhb in proteins, driving irreversible C-glutathionylation. Chemo-enzymatic methods were developed to site-selectively incorporate Dha/Dhb at phospho-regulated sites in kinases. In human MAPK-MEK1, such "elimination damage" generated aberrantly activated kinases, which were deactivated by LanCL-mediated C-glutathionylation. Surveys of endogenous proteins bearing damage from elimination (the eliminylome) also suggest it is a source of electrophilic reactivity. LanCLs thus remove these reactive electrophiles and their potentially dysregulatory effects from the proteome. As knockout of LanCL in mice can result in premature death, repair of this kind of protein damage appears important physiologically.
- Published
- 2020
23. Reaktive chemische Sonden: Jenseits des Kinase-Cysteinoms
- Author
-
Lyn H. Jones
- Subjects
010405 organic chemistry ,Chemistry ,General Medicine ,010402 general chemistry ,01 natural sciences ,0104 chemical sciences - Published
- 2018
- Full Text
- View/download PDF
24. Reactive Chemical Probes: Beyond the Kinase Cysteinome
- Author
-
Lyn H. Jones
- Subjects
Proteomics ,010405 organic chemistry ,Kinase ,Chemistry ,Lysine ,Molecular Conformation ,Chemical biology ,General Chemistry ,Computational biology ,010402 general chemistry ,01 natural sciences ,Mass Spectrometry ,Catalysis ,0104 chemical sciences ,Humans ,Kinome ,Chemoproteomics ,Cysteine ,Protein Kinase Inhibitors ,Protein Kinases - Abstract
The reaction of small-molecule chemical probes with proteins has been harnessed to develop covalent inhibitor drugs and protein-profiling technologies. This Essay discusses some of the recent enhancements to the chemical biology toolkit that are enabling the study of previously unchartered areas of chemoproteomic space. An analysis of the kinome is used to illustrate the potential for these approaches enable the pursuit of new targets using reactive chemical probes.
- Published
- 2018
- Full Text
- View/download PDF
25. Small-Molecule Kinase Downregulators
- Author
-
Lyn H. Jones
- Subjects
0301 basic medicine ,Clinical Biochemistry ,Down-Regulation ,Mitogen-activated protein kinase kinase ,Ligands ,Biochemistry ,MAP2K7 ,03 medical and health sciences ,0302 clinical medicine ,TANK-binding kinase 1 ,Drug Discovery ,Humans ,ASK1 ,Protein Kinase Inhibitors ,Molecular Biology ,Pharmacology ,MAP kinase kinase kinase ,biology ,Cyclin-dependent kinase 4 ,Phosphotransferases ,Cyclin-dependent kinase 2 ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Cyclin-dependent kinase 9 - Abstract
New opportunities to advance small-molecule kinase ligands that downregulate their cognate target binding proteins are discussed. Rationally designed heterobifunctional kinase degraders are compared with ATP site ligands that were serendipitously found to cause kinase downregulation. These approaches could be particularly useful in the treatment of cancers since many kinases are known to remodel pro-oncogenic protein-protein interactions, which could be destroyed by small-molecule-mediated kinase depletion.
- Published
- 2018
- Full Text
- View/download PDF
26. Correction: Fragment-based covalent ligand discovery
- Author
-
Nathanael S. Gray, Wenchao Lu, Tinghu Zhang, Milka Kostic, Jianwei Che, Edward T. Chouchani, Matthew P. Patricelli, and Lyn H. Jones
- Subjects
biology ,Chemistry (miscellaneous) ,Fragment (computer graphics) ,Covalent bond ,Chemistry ,Stereochemistry ,biology.protein ,Chromatin structure remodeling (RSC) complex ,Ligand (biochemistry) ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Molecular Biology ,Biochemistry - Abstract
Correction for ‘Fragment-based covalent ligand discovery’ by Wenchao Lu et al., RSC Chem. Biol., 2021, DOI: 10.1039/d0cb00222d.
- Published
- 2021
- Full Text
- View/download PDF
27. Microfluidic-Enabled Intracellular Delivery of Membrane Impermeable Inhibitors to Study Target Engagement in Human Primary Cells
- Author
-
Brian Juba, Betsy Pierce, Steve Kortum, Armon Sharei, Jing Li, Erik C. Hett, Jean-Baptiste Telliez, Bu Wang, Mark E. Bunnage, Lee R. Roberts, Strohbach Joseph Walter, Michael Pacheco, Atli Thorarensen, Lyn H. Jones, Michael L. Vazquez, and Jonathan B. Gilbert
- Subjects
0301 basic medicine ,Membrane permeability ,02 engineering and technology ,Biology ,Biochemistry ,Cell membrane ,Structure-Activity Relationship ,03 medical and health sciences ,Lab-On-A-Chip Devices ,medicine ,Humans ,Janus Kinase Inhibitors ,Structure–activity relationship ,Cells, Cultured ,Janus Kinases ,Molecular Structure ,Electroporation ,Cell Membrane ,General Medicine ,Biochemical Activity ,021001 nanoscience & nanotechnology ,Small molecule ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Leukocytes, Mononuclear ,Molecular Medicine ,0210 nano-technology ,Janus kinase ,Intracellular - Abstract
Biochemical screening is a major source of lead generation for novel targets. However, during the process of small molecule lead optimization, compounds with excellent biochemical activity may show poor cellular potency, making structure-activity relationships difficult to decipher. This may be due to low membrane permeability of the molecule, resulting in insufficient intracellular drug concentration. The Cell Squeeze platform increases permeability regardless of compound structure by mechanically disrupting the membrane, which can overcome permeability limitations and bridge the gap between biochemical and cellular studies. In this study, we show that poorly permeable Janus kinase (JAK) inhibitors are delivered into primary cells using Cell Squeeze, inhibiting up to 90% of the JAK pathway, while incubation of JAK inhibitors with or without electroporation had no significant effect. We believe this robust intracellular delivery approach could enable more effective lead optimization and deepen our understanding of target engagement by small molecules and functional probes.
- Published
- 2017
- Full Text
- View/download PDF
28. Highly potent and selective NaV1.7 inhibitors for use as intravenous agents and chemical probes
- Author
-
Jianmin Sun, Thomas Ryckmans, David Fengas, Colin R. Rose, M. Scott Johnson, Matthew Corbett, David James Rawson, Nigel Alan Swain, Joseph S. Warmus, Lyn H. Jones, Brian E. Marron, Aristos J. Alexandrou, David Printzenhoff, C. Elizabeth Payne, Bruce M. Bechle, Rubben Torella, Neil A. Castle, Jonathan W. Theile, Elaine Tseng, David C. Blakemore, Andrew Pike, R. Ian Storer, Neil J. Flanagan, and Alan Daniel Brown
- Subjects
0301 basic medicine ,Voltage-gated ion channel ,Chemistry ,Sodium channel ,Organic Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Pharmacology ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,In vivo ,Drug Discovery ,NAV1 ,Molecular Medicine ,Potency ,Molecular Biology ,030217 neurology & neurosurgery ,Ion channel ,Conjugate ,ADME - Abstract
The discovery and selection of a highly potent and selective NaV1.7 inhibitor PF-06456384, designed specifically for intravenous infusion, is disclosed. Extensive in vitro pharmacology and ADME profiling followed by in vivo preclinical PK and efficacy model data are discussed. A proposed protein-ligand binding mode for this compound is also provided to rationalise the high levels of potency and selectivity over inhibition of related sodium channels. To further support the proposed binding mode, potent conjugates are described which illustrate the potential for development of chemical probes to enable further target evaluation.
- Published
- 2017
- Full Text
- View/download PDF
29. Covalent Enzyme Inhibition through Fluorosulfate Modification of a Noncatalytic Serine Residue
- Author
-
Ariamala Gopalsamy, Lyn H. Jones, Robert E. Kyne, Ralph P. Robinson, Xidong Feng, Lise R. Hoth, Olugbeminiyi O. Fadeyi, Erik C. Hett, and Chulho Choi
- Subjects
Stereochemistry ,DCPS ,Chemical biology ,Crystallography, X-Ray ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Cell Line ,Serine ,Fluorides ,Residue (chemistry) ,Catalytic Domain ,Enzyme Stability ,Animals ,Humans ,Enzyme Inhibitors ,chemistry.chemical_classification ,biology ,010405 organic chemistry ,Chemistry ,Active site ,General Medicine ,Sulfuric Acids ,0104 chemical sciences ,Amino acid ,Electrophile ,biology.protein ,Molecular Medicine ,Cysteine - Abstract
Irreversible enzyme inhibitors and covalent chemical biology probes often utilize the reaction of a protein cysteine residue with an appropriately positioned electrophile (e.g., acrylamide) on the ligand template. However, cysteine residues are not always available for site-specific protein labeling, and therefore new approaches are needed to expand the toolkit of appropriate electrophiles ("warheads") that target alternative amino acids. We previously described the rational targeting of tyrosine residues in the active site of a protein (the mRNA decapping scavenger enzyme, DcpS) using inhibitors armed with a sulfonyl fluoride electrophile. These inhibitors subsequently enabled the development of clickable probe technology to measure drug-target occupancy in live cells. Here we describe a fluorosulfate-containing inhibitor (aryl fluorosulfate probe (FS-p1)) with excellent chemical and metabolic stability that reacts selectively with a noncatalytic serine residue in the same active site of DcpS as confirmed by peptide mapping experiments. Our results suggest that noncatalytic serine targeting using fluorosulfate electrophilic warheads could be a suitable strategy for the development of covalent inhibitor drugs and chemical probes.
- Published
- 2017
- Full Text
- View/download PDF
30. Applications of chemogenomic library screening in drug discovery
- Author
-
Mark E. Bunnage and Lyn H. Jones
- Subjects
0301 basic medicine ,Pharmacology ,Drug discovery ,Computer science ,Phenotypic screening ,Drug Repositioning ,Chemical biology ,Compound management ,General Medicine ,Predictive toxicology ,Computational biology ,Small Molecule Libraries ,03 medical and health sciences ,Drug repositioning ,Phenotype ,030104 developmental biology ,Drug Design ,Drug Discovery ,Animals ,Humans ,Identification (biology) ,Molecular Targeted Therapy - Abstract
The allure of phenotypic screening, combined with the industry preference for target-based approaches, has prompted the development of innovative chemical biology technologies that facilitate the identification of new therapeutic targets for accelerated drug discovery. A chemogenomic library is a collection of selective small-molecule pharmacological agents, and a hit from such a set in a phenotypic screen suggests that the annotated target or targets of that pharmacological agent may be involved in perturbing the observable phenotype. In this Review, we describe opportunities for chemogenomic screening to considerably expedite the conversion of phenotypic screening projects into target-based drug discovery approaches. Other applications are explored, including drug repositioning, predictive toxicology and the discovery of novel pharmacological modalities.
- Published
- 2017
- Full Text
- View/download PDF
31. Broad-Spectrum Kinase Profiling in Live Cells with Lysine-Targeted Sulfonyl Fluoride Probes
- Author
-
Jack Taunton, Q Zhao, John Charles Kath, Alma L. Burlingame, Xiaohu Ouyang, Xiaobo Wan, Ketan S. Gajiwala, and Lyn H. Jones
- Subjects
Cells ,Lysine ,Dasatinib ,Endogeny ,010402 general chemistry ,Models, Biological ,01 natural sciences ,Biochemistry ,Article ,Mass Spectrometry ,Catalysis ,Adenosine Triphosphate ,Drug Delivery Systems ,Colloid and Surface Chemistry ,Models ,medicine ,Kinome ,Binding site ,Cells, Cultured ,chemistry.chemical_classification ,Cultured ,Binding Sites ,Molecular Structure ,010405 organic chemistry ,Kinase ,General Chemistry ,Sulfinic Acids ,Biological ,0104 chemical sciences ,Enzyme ,chemistry ,Molecular Probes ,Chemical Sciences ,Generic health relevance ,Protein Kinases ,Intracellular ,medicine.drug - Abstract
Protein kinases comprise a large family of structurally related enzymes. A major goal in kinase-inhibitor development is to selectively engage the desired kinase while avoiding myriad off-target kinases. However, quantifying inhibitor interactions with multiple endogenous kinases in live cells remains an unmet challenge. Here, we report the design of sulfonyl fluoride probes that covalently label a broad swath of the intracellular kinome with high efficiency. Protein crystallography and mass spectrometry confirmed a chemoselective reaction between the sulfonyl fluoride and a conserved lysine in the ATP binding site. Optimized probe 2 (XO44) covalently modified up to 133 endogenous kinases, efficiently competing with high intracellular concentrations of ATP. We employed probe 2 and label-free mass spectrometry to quantify intracellular kinase engagement by the approved drug, dasatinib. The data revealed saturable dasatinib binding to a small subset of kinase targets at clinically relevant concentrations, highlighting the utility of lysine-targeted sulfonyl fluoride probes in demanding chemoproteomic applications.
- Published
- 2017
- Full Text
- View/download PDF
32. Quantitative measurement of intracellular HDAC1/2 drug occupancy using a trans-cyclooctene largazole thiol probe
- Author
-
Song Chou, Lee R. Roberts, Arjun Narayanan, Hua Xu, Betsy Pierce, and Lyn H. Jones
- Subjects
0301 basic medicine ,Cell ,Pharmaceutical Science ,Pharmacology ,Biology ,010402 general chemistry ,01 natural sciences ,Biochemistry ,03 medical and health sciences ,Drug Discovery ,medicine ,Potency ,Histone deacetylase 2 ,Drug discovery ,Organic Chemistry ,Assay ,HDAC1 ,0104 chemical sciences ,030104 developmental biology ,medicine.anatomical_structure ,Histone ,biology.protein ,Molecular Medicine ,Intracellular - Abstract
Histone deacetylases (HDACs) regulate diverse cellular processes, and are promising targets for a number of diseases. Here we describe the design and utilization of a largazole-based chemical probe to quantitatively measure the intracellular occupancy of HDAC1 and HDAC2 by dacinostat. Surprisingly, the probe was unable to enrich HDAC3 despite its nanomolar potency in a biochemical assay, further proving the necessity of cell-based target occupancy assays to understand compound potency in physiologically-relevant settings. This occupancy assay has the potential to aid the development of novel HDAC1/2 inhibitors in drug discovery.
- Published
- 2017
- Full Text
- View/download PDF
33. Pharmacological Correction of Proteinopathies via Lysosomal Degradation
- Author
-
Lyn H. Jones
- Subjects
Text mining ,Chemistry ,business.industry ,Degradation (geology) ,Computational biology ,business ,Biochemistry - Published
- 2020
- Full Text
- View/download PDF
34. Precision Retargeting: A Selective Covalent Inhibitor Illuminates CDK7 Biology
- Author
-
Lyn H. Jones
- Subjects
Clinical Biochemistry ,Cell ,Mutant ,Chemical biology ,Biology ,01 natural sciences ,Biochemistry ,Article ,Drug Discovery ,medicine ,Molecular Biology ,Protein Kinase Inhibitors ,Pharmacology ,010405 organic chemistry ,Kinase ,Cell Cycle ,Cell cycle ,Cyclin-Dependent Kinases ,0104 chemical sciences ,Cell biology ,medicine.anatomical_structure ,Phenotype ,Pyrimidines ,Covalent bond ,Retargeting ,Molecular Medicine ,Cyclin-dependent kinase 7 - Abstract
Cyclin-dependent kinase 7 (CDK7) regulates both cell cycle and transcription, but its precise role remains elusive. We previously described THZ1, a CDK7 inhibitor, which dramatically inhibits super-enhancer associated gene expression. However, potent CDK12/13 off-target activity obscured CDK7s contribution to this phenotype. Here, we describe the discovery of a highly selective, covalent CDK7 inhibitor. YKL-5-124 causes arrest at the G1/S transition and inhibition of E2F-driven gene expression; these effects are rescued by a CDK7 mutant unable to covalently engage YKL-5-124, demonstrating on-target specificity. Unlike THZ1, treatment with YKL-5-124 resulted in no change to Pol II CTD phosphorylation; however, inhibition could be reconstituted by combining YKL-5-124 and THZ531, a selective CDK12/13 inhibitor, revealing potential redundancies in CDK control of gene transcription. These findings highlight the importance of CDK7/12/13 polypharmacology for anti-cancer activity of THZ1 and posit that selective inhibition of CDK7 may be useful for treatment of cancers marked by E2F misregulation.
- Published
- 2019
35. Quantifying drug-target engagement in live cells using sulfonyl fluoride chemical probes
- Author
-
Lyn H, Jones, Hua, Xu, and Olugbeminiyi O, Fadeyi
- Subjects
Models, Molecular ,Drug Discovery ,Endoribonucleases ,Drug Evaluation, Preclinical ,Humans ,Click Chemistry ,Enzyme Inhibitors ,Sulfinic Acids ,Cells, Cultured - Abstract
Phenotypic screening in disease-relevant models identifies small molecule hits with desirable efficacy but often with unknown modes of action. Target identification and validation are integral to successful biomedical research. Technologies are required to validate the biological target (or targets) through which a pharmacological agent is proposed to exert its effects. This work details the rational structure-based design, synthetic preparation and cell-based application of a clickable sulfonyl fluoride chemical probe to directly report on the mechanism of a series of compounds previously discovered in a reporter gene assay. Quantification of drug-target occupancy in living human primary cells enabled a deeper understanding of the molecular pharmacology of the chemotype. The technology described herein should be of broad interest to those involved in chemical biology research and the drug discovery endeavor.
- Published
- 2019
36. Expanding chemogenomic space using chemoproteomics
- Author
-
Lyn H. Jones
- Subjects
Proteomics ,Clinical Biochemistry ,Chemical biology ,Pharmaceutical Science ,Computational biology ,01 natural sciences ,Biochemistry ,Mass Spectrometry ,Small Molecule Libraries ,chemistry.chemical_compound ,Structure-Activity Relationship ,Drug Discovery ,Chemogenomics ,Humans ,Chemoproteomics ,Molecular Biology ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Proteins ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Proteome ,Molecular Medicine - Abstract
Chemogenomics expedites the discovery of therapeutically-relevant targets from phenotypic screens. However, the vast majority of proteins in the proteome lack selective pharmacological modulators, necessitating the development of new technologies that significantly expand chemogenomic space. Chemoproteomics has emerged as a robust platform to map small molecule-protein interactions in cells using functionalized chemical probes in conjunction with mass spectrometry analysis. Exploration of the ligandable proteome in this manner has led to the development of new pharmacological modulators of diverse proteins. Opportunities to further enhance the impact of chemoproteomics using medicinal chemical biology are described.
- Published
- 2019
37. Quantifying drug-target engagement in live cells using sulfonyl fluoride chemical probes
- Author
-
Olugbeminiyi O. Fadeyi, Hua Xu, and Lyn H. Jones
- Subjects
0303 health sciences ,03 medical and health sciences ,Drug discovery ,Chemistry ,Biological target ,Mechanism (biology) ,Phenotypic screening ,030303 biophysics ,Click chemistry ,Chemical biology ,Molecular Pharmacology ,Computational biology ,Small molecule - Abstract
Phenotypic screening in disease-relevant models identifies small molecule hits with desirable efficacy but often with unknown modes of action. Target identification and validation are integral to successful biomedical research. Technologies are required to validate the biological target (or targets) through which a pharmacological agent is proposed to exert its effects. This work details the rational structure-based design, synthetic preparation and cell-based application of a clickable sulfonyl fluoride chemical probe to directly report on the mechanism of a series of compounds previously discovered in a reporter gene assay. Quantification of drug-target occupancy in living human primary cells enabled a deeper understanding of the molecular pharmacology of the chemotype. The technology described herein should be of broad interest to those involved in chemical biology research and the drug discovery endeavor.
- Published
- 2019
- Full Text
- View/download PDF
38. Human amyotrophic lateral sclerosis excitability phenotype screen: Target discovery and validation
- Author
-
Mary K. Dornon, Joseph R. Klim, Ole Wiskow, Allison P. Berg, Kevin Eggan, Clifford J. Woolf, Devlin Frost, Kuchuan Chen, Liying Zhang, Hongying Yang, Marco T. Siekmann, Karen L. Leach, Anthony J. Coyle, Seungkyu Lee, Jackie Klug-McLeod, Alyssa Grantham, Anne B. Jefferson, Dongyi Zhao, Xuan Huang, Lyn H. Jones, Fabien Vincent, Rie Maeda, Amy Brault, and Kasper C.D. Roet
- Subjects
0301 basic medicine ,Phenotypic screening ,Amyotrophic Lateral Sclerosis ,SOD1 ,Cell Differentiation ,AMPA receptor ,Biology ,Motor neuron ,medicine.disease ,Phenotype ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Drug development ,Live cell imaging ,medicine ,Humans ,Amyotrophic lateral sclerosis ,Neuroscience ,030217 neurology & neurosurgery - Abstract
SUMMARY Drug development is hampered by poor target selection. Phenotypic screens using neurons differentiated from patient stem cells offer the possibility to validate known and discover novel disease targets in an unbiased fashion. To identify targets for managing hyperexcitability, a pathological feature of amyotrophic lateral sclerosis (ALS), we design a multi-step screening funnel using patient-derived motor neurons. High-content live cell imaging is used to evaluate neuronal excitability, and from a screen against a chemogenomic library of 2,899 target-annotated compounds, 67 reduce the hyperexcitability of ALS motor neurons carrying the SOD1(A4V) mutation, without cytotoxicity. Bioinformatic deconvolution identifies 13 targets that modulate motor neuron excitability, including two known ALS excitability modulators, AMPA receptors and Kv7.2/3 ion channels, constituting target validation. We also identify D2 dopamine receptors as modulators of ALS motor neuron excitability. This screen demonstrates the power of human disease cell-based phenotypic screens for identifying clinically relevant targets for neurological disorders., Graphical abstract, In brief Motor neuron hyperexcitability is observed in both ALS patients and their iPSC-derived neurons. Combining a high-content live imaging excitability phenotypic assay, high-throughput screening against a cross-annotated chemogenomic library, and bioinformatic enrichment analysis, Huang et al. identify targets modulating the hyperexcitability of ALS patient-derived motor neurons in an unbiased manner.
- Published
- 2021
- Full Text
- View/download PDF
39. Design and development of histone deacetylase (HDAC) chemical probes for cell-based profiling
- Author
-
R. Ian Storer, James A. Clulow, Joseph S. Warmus, Jianmin Sun, Victoria E. Albrow, Colin R. Rose, Rachel L. Grimley, Lyn H. Jones, and Edward W. Tate
- Subjects
Proteomics ,0301 basic medicine ,Cell Membrane Permeability ,Proteome ,Histone Deacetylases ,Mass Spectrometry ,03 medical and health sciences ,Drug Discovery ,Humans ,Molecular Biology ,Regulation of gene expression ,Molecular Structure ,Staining and Labeling ,biology ,Acetylation ,Recombinant Proteins ,Chromatin ,Enzyme Activation ,Histone Deacetylase Inhibitors ,030104 developmental biology ,Histone ,Tubulin ,Biochemistry ,Molecular Probes ,biology.protein ,Histone deacetylase ,Bioorthogonal chemistry ,Biotechnology - Abstract
Histone deacetylases (HDACs) contribute to regulation of gene expression by mediating higher-order chromatin structures. They assemble into large multiprotein complexes that regulate activity and specificity. We report the development of small molecule probes with class IIa and pan-HDAC activity that contain photoreactive crosslinking groups and either a biotin reporter, or a terminal alkyne handle for subsequent bioorthogonal ligation. The probes retained inhibitory activity against recombinant HDAC proteins and caused an accumulation of acetylated histone and tubulin following cell treatment. The versatility of the probes has been demonstrated by their ability to photoaffinity modify HDAC targets in vitro. An affinity enrichment probe was used in conjunction with mass spectrometry proteomics to isolate HDACs and their interacting proteins in a native proteome. The performance of the probes in recombinant versus cell-based systems highlights issues for the development of chemoproteomic technologies targeting class IIa HDACs in particular.
- Published
- 2016
- Full Text
- View/download PDF
40. Label-free technologies for target identification and validation
- Author
-
Jing Li, Hua Xu, Lyn H. Jones, and Graham M. West
- Subjects
0301 basic medicine ,Pharmacology ,Computer science ,Phenotypic screening ,Organic Chemistry ,Target engagement ,Pharmaceutical Science ,Nanotechnology ,Computational biology ,Biochemistry ,03 medical and health sciences ,030104 developmental biology ,Drug Discovery ,Animal Disease Models ,Molecular Medicine ,Identification (biology) ,Label free - Abstract
Phenotypic screening is a powerful strategy for identifying active molecules with particular biological effects in cellular or animal disease models. Functionalized chemical probes have been instrumental in revealing new targets and confirming target engagement. However, substantial effort and resources are required to design and synthesize these bioactive probes. In contrast, label-free technologies have the advantage of bypassing the need for chemical probes. Here we highlight the recent developments in label-free methods and discuss the strengths and limitations of each approach.
- Published
- 2016
- Full Text
- View/download PDF
41. Emerging Utility of Fluorosulfate Chemical Probes
- Author
-
Lyn H. Jones
- Subjects
010405 organic chemistry ,Drug discovery ,Aryl ,Organic Chemistry ,Lysine ,Chemical biology ,010402 general chemistry ,01 natural sciences ,Biochemistry ,0104 chemical sciences ,Serine ,chemistry.chemical_compound ,chemistry ,Drug Discovery ,Chemoproteomics ,Tyrosine ,Histidine - Abstract
[Image: see text] Aryl fluorosulfates are finding widespread utility in chemical biology and medicinal chemistry. The context-dependent engagement of tyrosine, lysine, serine, and histidine amino acid residues in functional protein sites has enabled chemogenomic and chemoproteomic techniques that demonstrate considerable promise for drug discovery and biomedical research.
- Published
- 2018
42. Molecular hybridization yields triazole bronchodilators for the treatment of COPD
- Author
-
Emilio F. Stuart, Nick Clarke, Paul Alan Glossop, Neil Feeder, Strang Ross Sinclair, J. W. Watson, Jane L. Burrows, Amy S. Kenyon, Matthew D. Selby, Michael A. Trevethick, Sheena Patel, Rhys M. Jones, Kim James, Karen N. Wright, Dannielle Frances Roberts, and Lyn H. Jones
- Subjects
Bronchoconstriction ,Clinical Biochemistry ,Triazole ,Biological Availability ,Pharmaceutical Science ,CHO Cells ,Muscarinic Antagonists ,Pharmacology ,Biochemistry ,Pulmonary Disease, Chronic Obstructive ,chemistry.chemical_compound ,Cricetulus ,Dogs ,Drug Discovery ,medicine ,Animals ,Humans ,Tiotropium Bromide ,Bifunctional ,Adrenergic beta-2 Receptor Agonists ,Salmeterol Xinafoate ,Molecular Biology ,Receptor, Muscarinic M3 ,COPD ,β2 agonists ,Ipratropium ,Organic Chemistry ,Triazoles ,medicine.disease ,Bronchodilator Agents ,Rats ,Molecular hybridization ,Biopharmaceutical ,chemistry ,Molecular Medicine ,Bioisostere - Abstract
A 1,2,4-triazole motif was employed as a bioisostere for the ester commonly used in muscarinic antagonists, and subsequent integrative conjugation to a β2 agonist quinolinone furnished a new class of bifunctional MABAs for the treatment of COPD. Medicinal chemistry optimization using the principles of 'inhalation by design' furnished a clinical candidate with desirable pharmacological, pharmacokinetic and biopharmaceutical properties.
- Published
- 2015
- Full Text
- View/download PDF
43. Click chemistry patents and their impact on drug discovery and chemical biology
- Author
-
Hua Xu and Lyn H. Jones
- Subjects
Aldehydes ,Cycloaddition Reaction ,Drug discovery ,Chemistry ,business.industry ,Chemical biology ,Nanotechnology ,General Medicine ,Ketones ,Data science ,Patents as Topic ,Drug Discovery ,Click chemistry ,Click Chemistry ,Benzothiazoles ,Sulfhydryl Compounds ,business ,Pharmaceutical industry - Abstract
First introduced by K Barry Sharpless in 2001, the term ‘click chemistry’ soon became a widely used description of chemical reactions that proceed rapidly, cleanly and in a manner that is often compatible with aqueous solutions. Click chemistry is frequently employed throughout the process of drug discovery, and greatly helps advance research programs in the pharmaceutical industry. It facilitates library synthesis to support medicinal chemistry optimization, helps identify the targets and off-targets of drug candidates, and can facilitate the determination of drug efficacy in clinical trials. In the last decade, a large number of patent applications covering the various types and utilities of click chemistry have been filed. In this review, we provide the first analysis of click chemistry applications.
- Published
- 2015
- Full Text
- View/download PDF
44. Rational Targeting of Active-Site Tyrosine Residues Using Sulfonyl Fluoride Probes
- Author
-
Shenping Liu, Lee R. Roberts, Erik C. Hett, Mihir D. Parikh, Ariamala Gopalsamy, Kieran F. Geoghegan, Ralph P. Robinson, Michael A. Tones, Lyn H. Jones, Robert E. Kyne, Hua Xu, Carol A. Menard, and Arjun Narayanan
- Subjects
DCPS ,Chemical biology ,Chemistry Techniques, Synthetic ,Crystallography, X-Ray ,Biochemistry ,Structure-Activity Relationship ,Catalytic Domain ,Endoribonucleases ,Humans ,Structure–activity relationship ,Molecular Targeted Therapy ,Enzyme Inhibitors ,Binding site ,Tyrosine ,Cells, Cultured ,chemistry.chemical_classification ,biology ,Chemistry ,Active site ,General Medicine ,Sulfinic Acids ,Enzyme ,Covalent bond ,Molecular Probes ,biology.protein ,Molecular Medicine - Abstract
This work describes the first rational targeting of tyrosine residues in a protein binding site by small-molecule covalent probes. Specific tyrosine residues in the active site of the mRNA-decapping scavenger enzyme DcpS were modified using reactive sulfonyl fluoride covalent inhibitors. Structure-based molecular design was used to create an alkyne-tagged probe bearing the sulfonyl fluoride warhead, thus enabling the efficient capture of the protein from a complex proteome. Use of the probe in competition experiments with a diaminoquinazoline DcpS inhibitor permitted the quantification of intracellular target occupancy. As a result, diaminoquinazoline upregulators of survival motor neuron protein that are used for the treatment of spinal muscular atrophy were confirmed as inhibitors of DcpS in human primary cells. This work illustrates the utility of sulfonyl fluoride probes designed to react with specific tyrosine residues of a protein and augments the chemical biology toolkit by these probes uses in target validation and molecular pharmacology.
- Published
- 2015
- Full Text
- View/download PDF
45. TAK1 selective inhibition: state of the art and future opportunities
- Author
-
Iain Kilty and Lyn H. Jones
- Subjects
Pharmacology ,biology ,MAP kinase kinase kinase ,Cyclin-dependent kinase 4 ,Cyclin-dependent kinase 2 ,NF-kappa B ,Molecular Dynamics Simulation ,Mitogen-activated protein kinase kinase ,MAP Kinase Kinase Kinases ,MAP2K7 ,Cell biology ,TANK-binding kinase 1 ,Drug Discovery ,biology.protein ,Humans ,Molecular Medicine ,RNA Interference ,ASK1 ,Cyclin-dependent kinase 9 ,Mitogen-Activated Protein Kinases ,Oligopeptides ,Protein Kinase Inhibitors ,Protein Binding ,Signal Transduction - Abstract
The mitogen activated protein kinase kinase kinase transforming growth factor-β-activated kinase 1 (TAK1) has emerged as an interesting therapeutic target for inflammatory diseases and cancer. TAK1 is a tightly regulated kinase that represents a key signaling node in cellular responses to inflammatory stimuli, modulating both expression of inflammatory mediators and cell death. The first inhibitors described for TAK1 exploit the active site cysteine residue found in this kinase, but more recently both type I ATP hinge-binding inhibitors and type II DFG-out inhibitors have been described. This article will review the emerging role of TAK1 kinase in inflammation, the current state of the art for small molecule inhibitor development and opportunities for chemical biology approaches.
- Published
- 2015
- Full Text
- View/download PDF
46. Direct photocapture of bromodomains using tropolone chemical probes
- Author
-
Erik C. Hett, Parag Sahasrabudhe, Mathew T. Pletcher, R. Aldrin Denny, Alexandria P. Taylor, Shenping Liu, Arindrajit Basak, Andrew C. Flick, Paul D. Bonin, Eugene Lvovich Piatnitski Chekler, Ingrid A. Stock, Lyn H. Jones, Shores C. Salter, and Kieran F. Geoghegan
- Subjects
Pharmacology ,BRD4 ,biology ,Stereochemistry ,Organic Chemistry ,Lysine ,Pharmaceutical Science ,Biochemistry ,Combinatorial chemistry ,Tropolone ,Bromodomain ,BET inhibitor ,chemistry.chemical_compound ,Tubulin ,chemistry ,Drug Discovery ,Proteome ,biology.protein ,Molecular Medicine ,Tropone - Abstract
Medicinal chemistry techniques, including structure-based molecular design, fragment replacement and synthetic library enablement, were used to create potent inhibitors of bromodomain and extraterminal domain (BET) and CREB bindingprotein bromodomains. One inhibitor featured the homoaromatic tropolone methyl ether motif as a mimic of acetyl lysine, as confirmed by X-ray crystallography. The intrinsic photoreactivity of the tropone fragment was harnessed successfully to directly photolabel recombinant bromodomains which inspired further development of a clickable probe to assess BRD4 target engagement by (+)-JQ1 as a representative BET inhibitor in a complex proteome. The antimitoticnatural productcolchicine and related natural products that contain the tropolone ring system were also found to be bromodomain inhibitors, and surprisingly, our probe was shown to photolabel tubulin. These results highlight the caution that should be exercised when considering the selectivity of pharmacological agents, and photoreactive chemical probes should be assessed for their intrinsic ability to directly label their biological targets with a view to creating useful chemoproteomic tools.
- Published
- 2015
- Full Text
- View/download PDF
47. An industry perspective on drug target validation
- Author
-
Lyn H. Jones
- Subjects
0301 basic medicine ,Drug ,Drug Industry ,business.industry ,Drug discovery ,media_common.quotation_subject ,Phenotypic screening ,Perspective (graphical) ,Drug target ,Validation Studies as Topic ,medicine.disease ,Data science ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,Risk analysis (engineering) ,Drug Design ,Drug Discovery ,medicine ,Humans ,Attrition ,Molecular Targeted Therapy ,business ,media_common - Abstract
Drug attrition in clinical trials due to inadequate preclinical target validation is a huge problem facing drug discovery. The issue is compounded by concerns over the reproducibility of published ...
- Published
- 2016
- Full Text
- View/download PDF
48. Understanding the chemically-reactive proteome
- Author
-
Lyn H. Jones
- Subjects
Proteomics ,0301 basic medicine ,Proteome ,Molecular pathology ,fungi ,Chemical biology ,food and beverages ,Biology ,03 medical and health sciences ,030104 developmental biology ,Biochemistry ,Drug Discovery ,Humans ,Reactivity (chemistry) ,Identification (biology) ,Amino Acids ,Amino acid residue ,Protein Processing, Post-Translational ,Molecular Biology ,Biotechnology - Abstract
The reactivity of amino acid residues in proteins is context-dependent and difficult to predict. Chemical biology can be used to understand the chemical modifications of proteins to help elucidate the nature of the reactive proteome. The resulting insights can be applied to pharmacoproteomics, target identification and molecular pathology.
- Published
- 2016
- Full Text
- View/download PDF
49. Protein labelling
- Author
-
Lyn H, Jones and Eranthie, Weerapana
- Subjects
Eukaryotic Cells ,Prokaryotic Cells ,Proteome ,Molecular Probes ,Animals ,Humans ,Proteins ,Protein Processing, Post-Translational ,Molecular Biology ,Biotechnology - Abstract
Professor Eranthie Weerapana and Professor Lyn Jones introduce this Protein Labelling themed issue.
- Published
- 2016
- Full Text
- View/download PDF
50. Discovery of PF-06928215 as a high affinity inhibitor of cGAS enabled by a novel fluorescence polarization assay
- Author
-
Lih-Ling Lin, Guoxing Wang, Joseph P. Gardner, Hong Wang, Suman Shanker, Eicke Latz, Jeffrey S. Culp, Bruce Allen Lefker, Frank Lovering, Daniel C. Schmitt, Dana Castro, Ann Aulabaugh, Ryan Nistler, Timothy A. Subashi, Jimson Wong, Ken Dower, Steven J. Hawrylik, Olga Plotnikova, Karen L. Leach, Wen Lin, Fabien Vincent, James F. Smith, Amy Brault, Darren Dumlao, David Hepworth, Justin Hall, Mark Horn, Shawn Weng, Jing Li, Nootaree Niljanskul, William B. Snyder, Edyta Tyminski, Douglas T. Golenbock, David W. Lin, Leslie Anthony Dakin, Dikran Aivazian, Lyn H. Jones, John I. Trujillo, Ming Chen, Betsy Pierce, and Peter Jones
- Subjects
0301 basic medicine ,Models, Molecular ,GTP' ,Cell Lines ,Druggability ,lcsh:Medicine ,Plasma protein binding ,Biochemistry ,Mass Spectrometry ,0302 clinical medicine ,Spectrum Analysis Techniques ,Drug Discovery ,Enzyme-Linked Immunoassays ,Enzyme Inhibitors ,lcsh:Science ,Genetic Interference ,Multidisciplinary ,ATP synthase ,biology ,Molecular Structure ,Chemistry ,Drug discovery ,Anti-Inflammatory Agents, Non-Steroidal ,Nucleotidyltransferases ,Enzymes ,Bioassays and Physiological Analysis ,Spectrophotometry ,030220 oncology & carcinogenesis ,Second messenger system ,Biological Cultures ,Nucleotides, Cyclic ,Research Article ,Protein Binding ,High-throughput screening ,Fluorescence Polarization ,Enzyme-Linked Immunosorbent Assay ,Research and Analysis Methods ,Antibodies ,03 medical and health sciences ,Genetics ,Humans ,Immunoassays ,Molecular Biology Techniques ,Molecular Biology ,Enzyme Assays ,Molecular Biology Assays and Analysis Techniques ,Hybridomas ,Fluorimetry ,lcsh:R ,Biology and Life Sciences ,Proteins ,Molecular biology ,High Throughput Screening ,030104 developmental biology ,Pyrimidines ,Phosphodiester bond ,biology.protein ,Immunologic Techniques ,Enzymology ,Pyrazoles ,lcsh:Q ,Biochemical Analysis - Abstract
Cyclic GMP-AMP synthase (cGAS) initiates the innate immune system in response to cytosolic dsDNA. After binding and activation from dsDNA, cGAS uses ATP and GTP to synthesize 2', 3' -cGAMP (cGAMP), a cyclic dinucleotide second messenger with mixed 2'-5' and 3'-5' phosphodiester bonds. Inappropriate stimulation of cGAS has been implicated in autoimmune disease such as systemic lupus erythematosus, thus inhibition of cGAS may be of therapeutic benefit in some diseases; however, the size and polarity of the cGAS active site makes it a challenging target for the development of conventional substrate-competitive inhibitors. We report here the development of a high affinity (KD = 200 nM) inhibitor from a low affinity fragment hit with supporting biochemical and structural data showing these molecules bind to the cGAS active site. We also report a new high throughput cGAS fluorescence polarization (FP)-based assay to enable the rapid identification and optimization of cGAS inhibitors. This FP assay uses Cy5-labelled cGAMP in combination with a novel high affinity monoclonal antibody that specifically recognizes cGAMP with no cross reactivity to cAMP, cGMP, ATP, or GTP. Given its role in the innate immune response, cGAS is a promising therapeutic target for autoinflammatory disease. Our results demonstrate its druggability, provide a high affinity tool compound, and establish a high throughput assay for the identification of next generation cGAS inhibitors.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.