203 results on '"Andrew D. Mesecar"'
Search Results
2. SHIP1 therapeutic target enablement: Identification and evaluation of inhibitors for the treatment of late‐onset Alzheimer's disease
- Author
-
Cynthia D. Jesudason, Emily R. Mason, Shaoyou Chu, Adrian L. Oblak, June Javens‐Wolfe, Mustapha Moussaif, Greg Durst, Philip Hipskind, Daniel E. Beck, Jiajun Dong, Ovini Amarasinghe, Zhong‐Yin Zhang, Adam K. Hamdani, Kratika Singhal, Andrew D. Mesecar, Sarah Souza, Marlene Jacobson, Jerry Di Salvo, Disha M. Soni, Murugesh Kandasamy, Andrea R. Masters, Sara K Quinney, Suzanne Doolen, Hasi Huhe, Stacey J. Sukoff Rizzo, Bruce T. Lamb, Alan D. Palkowitz, and Timothy I. Richardson
- Subjects
cellular thermal shift assay (CETSA) ,INPP5D ,pharmacokinetics ,phenotypic high‐content imaging assay ,SHIP1 ,SHIP1 inhibitors ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract INTRODUCTION The risk of developing Alzheimer's disease is associated with genes involved in microglial function. Inositol polyphosphate‐5‐phosphatase (INPP5D), which encodes Src homology 2 (SH2) domain–containing inositol polyphosphate 5‐phosphatase 1 (SHIP1), is a risk gene expressed in microglia. Because SHIP1 binds receptor immunoreceptor tyrosine‐based inhibitory motifs (ITIMs), competes with kinases, and converts PI(3,4,5)P3 to PI(3,4)P2, it is a negative regulator of microglia function. Validated inhibitors are needed to evaluate SHIP1 as a potential therapeutic target. METHODS We identified inhibitors and screened the enzymatic domain of SHIP1. A protein construct containing two domains was used to evaluate enzyme inhibitor potency and selectivity versus SHIP2. Inhibitors were tested against a construct containing all ordered domains of the human and mouse proteins. A cellular thermal shift assay (CETSA) provided evidence of target engagement in cells. Phospho‐AKT levels provided further evidence of on‐target pharmacology. A high‐content imaging assay was used to study the pharmacology of SHIP1 inhibition while monitoring cell health. Physicochemical and absorption, distribution, metabolism, and excretion (ADME) properties were evaluated to select a compound suitable for in vivo studies. RESULTS SHIP1 inhibitors displayed a remarkable array of activities and cellular pharmacology. Inhibitory potency was dependent on the protein construct used to assess enzymatic activity. Some inhibitors failed to engage the target in cells. Inhibitors that were active in the CETSA consistently destabilized the protein and reduced pAKT levels. Many SHIP1 inhibitors were cytotoxic either at high concentration due to cell stress or they potently induced cell death depending on the compound and cell type. One compound activated microglia, inducing phagocytosis at concentrations that did not result in significant cell death. A pharmacokinetic study demonstrated brain exposures in mice upon oral administration. DISCUSSION 3‐((2,4‐Dichlorobenzyl)oxy)‐5‐(1‐(piperidin‐4‐yl)‐1H‐pyrazol‐4‐yl) pyridine activated primary mouse microglia and demonstrated exposures in mouse brain upon oral dosing. Although this compound is our recommended chemical probe for investigating the pharmacology of SHIP1 inhibition at this time, further optimization is required for clinical studies. Highlights Cellular thermal shift assay (CETSA) and signaling (pAKT) assays were developed to provide evidence of src homology 2 (SH2) domain‐contaning inositol phosphatase 1 (SHIP1) target engagement and on‐target activity in cellular assays. A phenotypic high‐content imaging assay with simultaneous measures of phagocytosis, cell number, and nuclear intensity was developed to explore cellular pharmacology and monitor cell health. SHIP1 inhibitors demonstrate a wide range of activity and cellular pharmacology, and many reported inhibitors are cytotoxic. The chemical probe 3‐((2,4‐dichlorobenzyl)oxy)−5‐(1‐(piperidin‐4‐yl)−1H‐pyrazol‐4‐yl) pyridine is recommended to explore SHIP1 pharmacology.
- Published
- 2023
- Full Text
- View/download PDF
3. Preclinical characterization of an intravenous coronavirus 3CL protease inhibitor for the potential treatment of COVID19
- Author
-
Britton Boras, Rhys M. Jones, Brandon J. Anson, Dan Arenson, Lisa Aschenbrenner, Malina A. Bakowski, Nathan Beutler, Joseph Binder, Emily Chen, Heather Eng, Holly Hammond, Jennifer Hammond, Robert E. Haupt, Robert Hoffman, Eugene P. Kadar, Rob Kania, Emi Kimoto, Melanie G. Kirkpatrick, Lorraine Lanyon, Emma K. Lendy, Jonathan R. Lillis, James Logue, Suman A. Luthra, Chunlong Ma, Stephen W. Mason, Marisa E. McGrath, Stephen Noell, R. Scott Obach, Matthew N. O’ Brien, Rebecca O’Connor, Kevin Ogilvie, Dafydd Owen, Martin Pettersson, Matthew R. Reese, Thomas F. Rogers, Romel Rosales, Michelle I. Rossulek, Jean G. Sathish, Norimitsu Shirai, Claire Steppan, Martyn Ticehurst, Lawrence W. Updyke, Stuart Weston, Yuao Zhu, Kris M. White, Adolfo García-Sastre, Jun Wang, Arnab K. Chatterjee, Andrew D. Mesecar, Matthew B. Frieman, Annaliesa S. Anderson, and Charlotte Allerton
- Subjects
Science - Abstract
The 3CL protease of SARS-CoV-2 is inhibited by PF-00835231 in vitro. Here, the authors show that the prodrug PF-07304814 has broad spectrum activity, inhibiting SARS-CoV and SARS-CoV-2 in mice and its ADME and safety profile support clinical development.
- Published
- 2021
- Full Text
- View/download PDF
4. A small molecule compound with an indole moiety inhibits the main protease of SARS-CoV-2 and blocks virus replication
- Author
-
Shin-ichiro Hattori, Nobuyo Higashi-Kuwata, Hironori Hayashi, Srinivasa Rao Allu, Jakka Raghavaiah, Haydar Bulut, Debananda Das, Brandon J. Anson, Emma K. Lendy, Yuki Takamatsu, Nobutoki Takamune, Naoki Kishimoto, Kazutaka Murayama, Kazuya Hasegawa, Mi Li, David A. Davis, Eiichi N. Kodama, Robert Yarchoan, Alexander Wlodawer, Shogo Misumi, Andrew D. Mesecar, Arun K. Ghosh, and Hiroaki Mitsuya
- Subjects
Science - Abstract
Here, using in vitro assays and structural analysis, the authors characterize the anti-SARS-CoV-2 properties of two small molcules, showing these to bind and target the virus main protease (Mpro), and to exhibit a synergistic antiviral effect when combined with remdesivir in vitro.
- Published
- 2021
- Full Text
- View/download PDF
5. Chloropyridinyl Esters of Nonsteroidal Anti-Inflammatory Agents and Related Derivatives as Potent SARS-CoV-2 3CL Protease Inhibitors
- Author
-
Arun K. Ghosh, Dana Shahabi, Monika Yadav, Satish Kovela, Brandon J. Anson, Emma K. Lendy, Connie Bonham, Devika Sirohi, Carlos A. Brito-Sierra, Shin-ichiro Hattori, Richard Kuhn, Hiroaki Mitsuya, and Andrew D. Mesecar
- Subjects
indomethacin derivative ,antiviral activity ,COVID-19 ,3CLpro inhibitors ,covalent inhibitors ,ibuprofen derivative ,Organic chemistry ,QD241-441 - Abstract
We report the design and synthesis of a series of new 5-chloropyridinyl esters of salicylic acid, ibuprofen, indomethacin, and related aromatic carboxylic acids for evaluation against SARS-CoV-2 3CL protease enzyme. These ester derivatives were synthesized using EDC in the presence of DMAP to provide various esters in good to excellent yields. Compounds are stable and purified by silica gel chromatography and characterized using 1H-NMR, 13C-NMR, and mass spectral analysis. These synthetic derivatives were evaluated in our in vitro SARS-CoV-2 3CLpro inhibition assay using authentic SARS-CoV-2 3CLpro enzyme. Compounds were also evaluated in our in vitro antiviral assay using quantitative VeroE6 cell-based assay with RNAqPCR. A number of compounds exhibited potent SARS-CoV-2 3CLpro inhibitory activity and antiviral activity. Compound 9a was the most potent inhibitor, with an enzyme IC50 value of 160 nM. Compound 13b exhibited an enzyme IC50 value of 4.9 µM. However, it exhibited a potent antiviral EC50 value of 24 µM in VeroE6 cells. Remdesivir, an RdRp inhibitor, exhibited an antiviral EC50 value of 2.4 µM in the same assay. We assessed the mode of inhibition using mass spectral analysis which suggested the formation of a covalent bond with the enzyme. To obtain molecular insight, we have created a model of compound 9a bound to SARS-CoV-2 3CLpro in the active site.
- Published
- 2021
- Full Text
- View/download PDF
6. Development and validation of a yeast high-throughput screen for inhibitors of Aβ42 oligomerization
- Author
-
Sei-Kyoung Park, Scott D. Pegan, Andrew D. Mesecar, Lisa M. Jungbauer, Mary Jo LaDu, and Susan W. Liebman
- Subjects
Medicine ,Pathology ,RB1-214 - Abstract
SUMMARY Recent reports point to small soluble oligomers, rather than insoluble fibrils, of amyloid β (Aβ), as the primary toxic species in Alzheimer’s disease. Previously, we developed a low-throughput assay in yeast that is capable of detecting small Aβ42 oligomer formation. Specifically, Aβ42 fused to the functional release factor domain of yeast translational termination factor, Sup35p, formed sodium dodecyl sulfate (SDS)-stable low-n oligomers in living yeast, which impaired release factor activity. As a result, the assay for oligomer formation uses yeast growth to indicate restored release factor activity and presumably reduced oligomer formation. We now describe our translation of this assay into a high-throughput screen (HTS) for anti-oligomeric compounds. By doing so, we also identified two presumptive anti-oligomeric compounds from a sub-library of 12,800 drug-like small molecules. Subsequent biochemical analysis confirmed their anti-oligomeric activity, suggesting that this form of HTS is an efficient, sensitive and cost-effective approach to identify new inhibitors of Aβ42 oligomerization.
- Published
- 2011
- Full Text
- View/download PDF
7. Supplemental Figure 3 from Cholesterol Sulfonation Enzyme, SULT2B1b, Modulates AR and Cell Growth Properties in Prostate Cancer
- Author
-
Timothy L. Ratliff, Liang Cheng, Andrew D. Mesecar, Chang-Deng Hu, Kimberly K. Buhman, Grant N. Burcham, R. Graham Cooks, Livia Eberlin, Kevin Kerian, Scott A. Crist, and Renee E. Vickman
- Abstract
SULT2B1b KD reduces RWPE-1 cell viability.
- Published
- 2023
8. Supplemental Table 1 from Cholesterol Sulfonation Enzyme, SULT2B1b, Modulates AR and Cell Growth Properties in Prostate Cancer
- Author
-
Timothy L. Ratliff, Liang Cheng, Andrew D. Mesecar, Chang-Deng Hu, Kimberly K. Buhman, Grant N. Burcham, R. Graham Cooks, Livia Eberlin, Kevin Kerian, Scott A. Crist, and Renee E. Vickman
- Abstract
Cell line authentication.
- Published
- 2023
9. Supplemental Figure 1 from Cholesterol Sulfonation Enzyme, SULT2B1b, Modulates AR and Cell Growth Properties in Prostate Cancer
- Author
-
Timothy L. Ratliff, Liang Cheng, Andrew D. Mesecar, Chang-Deng Hu, Kimberly K. Buhman, Grant N. Burcham, R. Graham Cooks, Livia Eberlin, Kevin Kerian, Scott A. Crist, and Renee E. Vickman
- Abstract
SULT2B1b expression and CS accumulation in human prostate specimens.
- Published
- 2023
10. Supplemental Materials and Methods from Cholesterol Sulfotransferase SULT2B1b Modulates Sensitivity to Death Receptor Ligand TNFα in Castration-Resistant Prostate Cancer
- Author
-
Timothy L. Ratliff, Chang-Deng Hu, Andrew D. Mesecar, Scott A. Crist, R. W. Doerge, Chi Zhang, Faye Zheng, Gregory M. Cresswell, Nadia A. Lanman, Jiang Yang, and Renee E. Vickman
- Abstract
The Supplemental Methods give additional details for methods in the main text.
- Published
- 2023
11. Supplemental Figure 2 from Cholesterol Sulfonation Enzyme, SULT2B1b, Modulates AR and Cell Growth Properties in Prostate Cancer
- Author
-
Timothy L. Ratliff, Liang Cheng, Andrew D. Mesecar, Chang-Deng Hu, Kimberly K. Buhman, Grant N. Burcham, R. Graham Cooks, Livia Eberlin, Kevin Kerian, Scott A. Crist, and Renee E. Vickman
- Abstract
Modulation of SULT2B1b alters relative abundance of CS in LNCaP cells.
- Published
- 2023
12. Supplemental Materials and Methods from Cholesterol Sulfonation Enzyme, SULT2B1b, Modulates AR and Cell Growth Properties in Prostate Cancer
- Author
-
Timothy L. Ratliff, Liang Cheng, Andrew D. Mesecar, Chang-Deng Hu, Kimberly K. Buhman, Grant N. Burcham, R. Graham Cooks, Livia Eberlin, Kevin Kerian, Scott A. Crist, and Renee E. Vickman
- Abstract
Additional information for Materials and Methods section.
- Published
- 2023
13. Supplemental Figure 5 from Cholesterol Sulfonation Enzyme, SULT2B1b, Modulates AR and Cell Growth Properties in Prostate Cancer
- Author
-
Timothy L. Ratliff, Liang Cheng, Andrew D. Mesecar, Chang-Deng Hu, Kimberly K. Buhman, Grant N. Burcham, R. Graham Cooks, Livia Eberlin, Kevin Kerian, Scott A. Crist, and Renee E. Vickman
- Abstract
SULT2B1b KD in C4-2 cells induces cell death.
- Published
- 2023
14. Supplemental Figures 1-7 and Supplemental Table 1 from Cholesterol Sulfotransferase SULT2B1b Modulates Sensitivity to Death Receptor Ligand TNFα in Castration-Resistant Prostate Cancer
- Author
-
Timothy L. Ratliff, Chang-Deng Hu, Andrew D. Mesecar, Scott A. Crist, R. W. Doerge, Chi Zhang, Faye Zheng, Gregory M. Cresswell, Nadia A. Lanman, Jiang Yang, and Renee E. Vickman
- Abstract
Figure S1 provides quality control from scRNA-seq analysis. Figure S2 indicates altered pathways in SULT2B1b KD cells. Figure S3 demonstrates TNF and SULT2B1b KD interact to further reduce AR activity in LNCaP cells. Figure S4 demonstrates that TNFR expression is not altered by SULT2B1b KD. Figure S5 demonstrates that SULT2B1b KD induces TRADD expression in LNCaP cells. Figure S6 indicates SULT2B1 is expressed at low levels. Figure S7 supports that TRAIL, TL1, and DAXX are not involved in apoptosis in SULT2B1b KD cells. Table S1 gives correlations of SULT2B1 with TNF-related genes.
- Published
- 2023
15. Supplemental Figure 6 from Cholesterol Sulfonation Enzyme, SULT2B1b, Modulates AR and Cell Growth Properties in Prostate Cancer
- Author
-
Timothy L. Ratliff, Liang Cheng, Andrew D. Mesecar, Chang-Deng Hu, Kimberly K. Buhman, Grant N. Burcham, R. Graham Cooks, Livia Eberlin, Kevin Kerian, Scott A. Crist, and Renee E. Vickman
- Abstract
Transcriptional activity of LXRα vs. LXRβ in LNCaP cells.
- Published
- 2023
16. Supplemental Figure 4 from Cholesterol Sulfonation Enzyme, SULT2B1b, Modulates AR and Cell Growth Properties in Prostate Cancer
- Author
-
Timothy L. Ratliff, Liang Cheng, Andrew D. Mesecar, Chang-Deng Hu, Kimberly K. Buhman, Grant N. Burcham, R. Graham Cooks, Livia Eberlin, Kevin Kerian, Scott A. Crist, and Renee E. Vickman
- Abstract
shRNA KD of SULT2B1b expression yields similar results to siRNA KD.
- Published
- 2023
17. Genetic Variants of Phospholipase C-γ2 Confer Altered Microglial Phenotypes and Differential Risk for Alzheimer’s Disease
- Author
-
Andy P. Tsai, Chuanpeng Dong, Peter Bor-Chian Lin, Adrian L. Oblak, Gonzalo Viana Di Prisco, Nian Wang, Nicole Hajicek, Adam J. Carr, Emma K. Lendy, Oliver Hahn, Micaiah Atkins, Aulden G. Foltz, Jheel Patel, Guixiang Xu, Miguel Moutinho, John Sondek, Qisheng Zhang, Andrew D. Mesecar, Yunlong Liu, Brady K. Atwood, Tony Wyss-Coray, Kwangsik Nho, Stephanie J. Bissel, Bruce T. Lamb, and Gary E. Landreth
- Abstract
SummaryGenetic association studies have demonstrated the critical involvement of the microglial immune response in Alzheimer’s disease (AD) pathogenesis. Phospholipase C-gamma-2 (PLCG2) is selectively expressed by microglia and acts in many immune receptor signaling pathways. In AD, PLCG2 is induced uniquely in plaque-associated microglia. A genetic variant ofPLCG2, PLCG2P522R, is a mild hypermorph that attenuates AD risk. We report the identification of aPLCG2variant,PLCG2M28L, associated with loss-of-function and confers increased AD risk. PLCG2P522Rattenuates disease in an amyloidogenic murine AD model, whereasPLCG2M28Lexacerbates the plaque burden associated with altered phagocytosis and Aβ clearance. The variants bidirectionally modulate disease pathology by inducing distinct transcriptional programs that identify microglial subpopulations associated with protective or detrimental phenotypes. In summary, these findings identify PLCG2M28Las a new AD risk variant and demonstrate that PLCG2 variants can differentially orchestrate microglial responses in AD pathogenesis that can be therapeutically targeted.Graphical abstractHighlightsA genetic variant of PLCG2, M28L, is associated with an increased risk for Alzheimer’s disease (AD)In an amyloidogenic AD mouse model, PLCG2M28L exacerbates disease pathogenesisConversely, PLCG2P522R, a protective PLCG2 variant, attenuates AD pathogenesisThe PLCG2 variants uniquely alter the microglial transcriptome and phenotypes
- Published
- 2022
18. A Target Enablement Package for the Inhibition of SHIP1 as a Therapeutic Strategy for the Treatment of Alzheimer’s Disease
- Author
-
Timothy I Richardson, Cynthia D Jesudason, Karen L Lobb, Gregory L Durst, Brent Clayton, Steven M Massey, Daniel E Beck, Zhong‐Yin Zhang, Jiajun Dong, Jianping Lin, Jinmin Miao, Karson S Putt, Andrew D Mesecar, Adam K Hamdani, Emma K Lendy, Sarah Souza, Marlene Jacobson, Jerry Di Salvo, Shaoyou Chu, Emily R Mason, Adrian L Oblak, Disha Soni, Sara K Quinney, Larissa L Silva, Murugesh Kandasamy, Andrea R Masters, Stacey J Sukoff Rizzo, Suzanne Doolen, Kun Huang, Jie Zhang, Bruce T Lamb, and Alan D Palkowitz
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
19. A Structure-Based Discovery Platform for BACE2 and the Development of Selective BACE Inhibitors
- Author
-
Andrew D. Mesecar, Yu-Chen Yen, Jagannadharao Tirlangi, Annalissa M Kammeyer, and Arun K. Ghosh
- Subjects
Drug ,Physiology ,Cognitive Neuroscience ,media_common.quotation_subject ,Biochemistry ,Article ,law.invention ,Type ii diabetes ,03 medical and health sciences ,0302 clinical medicine ,Alzheimer Disease ,law ,Escherichia coli ,Aspartic Acid Endopeptidases ,Humans ,Crystallization ,030304 developmental biology ,media_common ,0303 health sciences ,Chemistry ,Cell Biology ,General Medicine ,Combinatorial chemistry ,Diabetes Mellitus, Type 2 ,Structure based ,Amyloid Precursor Protein Secretases ,Selectivity ,030217 neurology & neurosurgery - Abstract
The ability to perform routine structure-guided drug design for selective BACE inhibitors has been limited because of the lack of robust platform for BACE2 expression, purification, and crystallization. To overcome this limitation, we developed a platform that produces 2–3 mg of pure BACE2 protein per liter of E. coli culture, and we used this protein to design macrocyclic compounds that potently and selectively inhibit BACE1 over BACE2. Compound 2 was found to potently inhibit BACE 1 (K(i) = 5 nM) with a selectivity of 214-fold over BACE2. The X-ray crystal structures of unbound BACE2 (2.2 Å) and BACE2 bound to compound 3 (3.0 Å and K(i) = 7 nM) were determined and compared to the X-ray structures of BACE1 revealing the S1–S3 subsite as a selectivity determinant. This platform should enable a more rapid development of new and selective BACE inhibitors for the treatment of Alzheimer’s disease or type II diabetes.
- Published
- 2021
20. Discovery of SARS-CoV-2 antiviral drugs through large-scale compound repurposing
- Author
-
Laura Riva, Shuofeng Yuan, Xin Yin, Laura Martin-Sancho, Naoko Matsunaga, Lars Pache, Sebastian Burgstaller-Muehlbacher, Paul D. De Jesus, Peter Teriete, Mitchell V. Hull, Max W. Chang, Jasper Fuk-Woo Chan, Jianli Cao, Vincent Kwok-Man Poon, Kristina M. Herbert, Kuoyuan Cheng, Tu-Trinh H. Nguyen, Andrey Rubanov, Yuan Pu, Courtney Nguyen, Angela Choi, Raveen Rathnasinghe, Michael Schotsaert, Lisa Miorin, Marion Dejosez, Thomas P. Zwaka, Ko-Yung Sit, Luis Martinez-Sobrido, Wen-Chun Liu, Kris M. White, Mackenzie E. Chapman, Emma K. Lendy, Richard J. Glynne, Randy Albrecht, Eytan Ruppin, Andrew D. Mesecar, Jeffrey R. Johnson, Christopher Benner, Ren Sun, Peter G. Schultz, Andrew I. Su, Adolfo García-Sastre, Arnab K. Chatterjee, Kwok-Yung Yuen, and Sumit K. Chanda
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Coronavirus disease 2019 (COVID-19) ,Morpholines ,Induced Pluripotent Stem Cells ,Pneumonia, Viral ,Drug Evaluation, Preclinical ,Cysteine Proteinase Inhibitors ,Virus Replication ,Antiviral Agents ,Models, Biological ,Article ,Cell Line ,Unit (housing) ,Small Molecule Libraries ,Betacoronavirus ,03 medical and health sciences ,0302 clinical medicine ,Pandemic ,medicine ,Humans ,Pandemics ,Alanine ,Multidisciplinary ,Dose-Response Relationship, Drug ,SARS-CoV-2 ,Triazines ,business.industry ,General surgery ,Drug Repositioning ,Hydrazones ,COVID-19 ,Reproducibility of Results ,Drug Synergism ,Virus Internalization ,Adenosine Monophosphate ,COVID-19 Drug Treatment ,Pyrimidines ,030104 developmental biology ,Gene Expression Regulation ,Cardiothoracic surgery ,Alveolar Epithelial Cells ,030220 oncology & carcinogenesis ,Quality standard ,Coronavirus Infections ,business - Abstract
Summary The emergence of the novel SARS coronavirus 2 (SARS-CoV-2) in 2019 has triggered an ongoing global pandemic of severe pneumonia-like disease designated as coronavirus disease 2019 (COVID-19)1. The development of a vaccine is likely to require at least 12-18 months, and the typical timeline for approval of a novel antiviral therapeutic can exceed 10 years. Thus, repurposing of known drugs could significantly accelerate the deployment of novel therapies for COVID-19. Towards this end, we profiled a library of known drugs encompassing approximately 12,000 clinical-stage or FDA-approved small molecules. We report the identification of 100 molecules that inhibit viral replication, including 21 known drugs that exhibit dose response relationships. Of these, thirteen were found to harbor effective concentrations likely commensurate with achievable therapeutic doses in patients, including the PIKfyve kinase inhibitor apilimod2–4, and the cysteine protease inhibitors MDL-28170, Z LVG CHN2, VBY-825, and ONO 5334. Notably, MDL-28170, ONO 5334, and apilimod were found to antagonize viral replication in human iPSC-derived pneumocyte-like cells, and the PIKfyve inhibitor also demonstrated antiviral efficacy in a primary human lung explant model. Since most of the molecules identified in this study have already advanced into the clinic, the known pharmacological and human safety profiles of these compounds will enable accelerated preclinical and clinical evaluation of these drugs for the treatment of COVID-19.
- Published
- 2020
21. Indole Chloropyridinyl Ester-Derived SARS-CoV-2 3CLpro Inhibitors: Enzyme Inhibition, Antiviral Efficacy, Structure-Activity Relationship, and X-ray Structural Studies
- Author
-
Brandon J. Anson, Dana Shahabi, Emma K. Lendy, Andrew D. Mesecar, Monika Yadav, Hiroaki Mitsuya, Jakka Raghavaiah, Nobuyo Higashi-Kuwata, Arun K. Ghosh, and Shin-ichiro Hattori
- Subjects
Indoles ,Stereochemistry ,Pyridines ,medicine.medical_treatment ,Molecular Dynamics Simulation ,Crystallography, X-Ray ,Article ,chemistry.chemical_compound ,Structure-Activity Relationship ,Drug Discovery ,Chlorocebus aethiops ,medicine ,Structure–activity relationship ,Animals ,Humans ,Protease Inhibitors ,Carboxylate ,Binding site ,IC50 ,Vero Cells ,Coronavirus 3C Proteases ,Indole test ,Protease ,Alanine ,Binding Sites ,Chemistry ,SARS-CoV-2 ,COVID-19 ,Adenosine Monophosphate ,RNA Polymerase Inhibitor ,Vero cell ,Molecular Medicine - Abstract
Here, we report the synthesis, structure-activity relationship studies, enzyme inhibition, antiviral activity, and X-ray crystallographic studies of 5-chloropyridinyl indole carboxylate derivatives as a potent class of SARS-CoV-2 chymotrypsin-like protease inhibitors. Compound 1 exhibited a SARS-CoV-2 3CLpro inhibitory IC50 value of 250 nM and an antiviral EC50 value of 2.8 µM in VeroE6 cells. Remdesivir, an RNA-dependent RNA polymerase inhibitor, showed an antiviral EC50 value of 1.2 µM in the same assay. Compound 1 showed comparable antiviral activity with remdesivir in immunocytochemistry assays. Compound 7d with an N-allyl derivative showed the most potent enzyme inhibitory IC50 value of 73 nM. To obtain molecular insight into the binding properties of these molecules, X-ray crystal structures of compounds 2, 7b, and 9d-bound to SARS-CoV 3CLpro were determined, and their binding properties were compared.
- Published
- 2021
22. Author response: Structural insight into the dual function of LbpB in mediating Neisserial pathogenesis
- Author
-
Andrew D. Mesecar, Courtney M. Daczkowski, Srinivas Chakravarthy, Ravi Yadav, Srinivas Govindan, and Nicholas Noinaj
- Subjects
Pathogenesis ,Biology ,Neuroscience ,Dual function - Published
- 2021
23. Chloropyridinyl Esters of Nonsteroidal Anti-Inflammatory Agents and Related Derivatives as Potent SARS-CoV-2 3CL Protease Inhibitors
- Author
-
Carlos A Brito-Sierra, Andrew D. Mesecar, Devika Sirohi, Satish Kovela, Monika Yadav, Hiroaki Mitsuya, Dana Shahabi, Emma K. Lendy, Arun K. Ghosh, Brandon J. Anson, Shinichiro Hattori, Richard J. Kuhn, and Connie C. Bonham
- Subjects
Halogenation ,Pyridines ,medicine.medical_treatment ,Indomethacin ,Pharmaceutical Science ,Organic chemistry ,Ibuprofen ,indomethacin derivative ,Analytical Chemistry ,chemistry.chemical_compound ,QD241-441 ,Drug Discovery ,Chlorocebus aethiops ,Coronavirus 3C Proteases ,chemistry.chemical_classification ,biology ,Anti-Inflammatory Agents, Non-Steroidal ,Esters ,Molecular Docking Simulation ,Chemistry (miscellaneous) ,Covalent bond ,antiviral activity ,Molecular Medicine ,covalent inhibitors ,Salicylic Acid ,Stereochemistry ,medicine.drug_class ,Antiviral Agents ,Anti-inflammatory ,Article ,3CLpro inhibitors ,medicine ,Animals ,Humans ,Protease Inhibitors ,Physical and Theoretical Chemistry ,IC50 ,Vero Cells ,Protease ,SARS-CoV-2 ,ibuprofen derivative ,Active site ,COVID-19 ,In vitro ,COVID-19 Drug Treatment ,salicylic acid derivative ,Enzyme ,chemistry ,biology.protein ,Salicylic acid - Abstract
We report the design and synthesis of a series of new 5-chloropyridinyl esters of salicylic acid, ibuprofen, indomethacin, and related aromatic carboxylic acids for evaluation against SARS-CoV-2 3CL protease enzyme. These ester derivatives were synthesized using EDC in the presence of DMAP to provide various esters in good to excellent yields. Compounds are stable and purified by silica gel chromatography and characterized using 1H-NMR, 13C-NMR, and mass spectral analysis. These synthetic derivatives were evaluated in our in vitro SARS-CoV-2 3CLpro inhibition assay using authentic SARS-CoV-2 3CLpro enzyme. Compounds were also evaluated in our in vitro antiviral assay using quantitative VeroE6 cell-based assay with RNAqPCR. A number of compounds exhibited potent SARS-CoV-2 3CLpro inhibitory activity and antiviral activity. Compound 9a was the most potent inhibitor, with an enzyme IC50 value of 160 nM. Compound 13b exhibited an enzyme IC50 value of 4.9 µM. However, it exhibited a potent antiviral EC50 value of 24 µM in VeroE6 cells. Remdesivir, an RdRp inhibitor, exhibited an antiviral EC50 value of 2.4 µM in the same assay. We assessed the mode of inhibition using mass spectral analysis which suggested the formation of a covalent bond with the enzyme. To obtain molecular insight, we have created a model of compound 9a bound to SARS-CoV-2 3CLpro in the active site.
- Published
- 2021
24. Mn 2+ coordinates Cap-0-RNA to align substrates for efficient 2′- O -methyl transfer by SARS-CoV-2 nsp16
- Author
-
Nicole L. Inniss, George Minasov, Courtney M. Daczkowski, Andrew D. Mesecar, Karla J. F. Satchell, Paul Hoover, Joseph S. Brunzelle, Monica Rosas-Lemus, and Ludmilla Shuvalova
- Subjects
Models, Molecular ,S-Adenosylmethionine ,Ribonucleotide ,Methyltransferase ,viruses ,RNA Stability ,Viral Nonstructural Proteins ,Crystallography, X-Ray ,medicine.disease_cause ,Biochemistry ,Insert (molecular biology) ,Substrate Specificity ,0302 clinical medicine ,Structural Biology ,Catalytic Domain ,skin and connective tissue diseases ,Research Articles ,Coronavirus ,0303 health sciences ,biology ,Chemistry ,Recombinant Proteins ,Infectious Diseases ,RNA, Viral ,Research Article ,Signal Transduction ,RNA Caps ,Stereochemistry ,Methylation ,03 medical and health sciences ,Virology ,medicine ,Humans ,Amino Acid Sequence ,RNA, Messenger ,Molecular Biology ,030304 developmental biology ,Manganese ,Messenger RNA ,SARS-CoV-2 ,fungi ,COVID-19 ,Active site ,RNA ,STKE Research Articles ,Methyltransferases ,Cell Biology ,respiratory tract diseases ,body regions ,biology.protein ,Nucleic Acid Conformation ,030217 neurology & neurosurgery - Abstract
Efficient RNA capping by the SARS-CoV-2 methyltransferase requires metal ions and a unique four-residue insert., Uniquely coronavirus Virally encoded 2′-O-methyltransferases catalyze the last step in the capping of viral RNAs, which protects the RNAs from degradation and prevents them from triggering host defenses. Minasov et al. report structures of the SARS-CoV-2 methyltransferase, a heterodimeric complex of the enzyme nsp16 and its coactivator nsp10, in complex with a short, capped RNA (instead of the RNA cap analogs used to generate previous structures), the methyl donor SAM, and divalent metal cations. The metal ions and a four-residue insert of nsp16 were important for precisely aligning the RNA substrate in the active site for efficient catalysis. This insert is present in coronavirus but not in mammalian methyltransferases, suggesting this site as a potential target for the design of coronavirus-specific methyltransferase inhibitors., Capping of viral messenger RNAs is essential for efficient translation, for virus replication, and for preventing detection by the host cell innate response system. The SARS-CoV-2 genome encodes the 2′-O-methyltransferase nsp16, which, when bound to the coactivator nsp10, uses S-adenosylmethionine (SAM) as a donor to transfer a methyl group to the first ribonucleotide of the mRNA in the final step of viral mRNA capping. Here, we provide biochemical and structural evidence that this reaction requires divalent cations, preferably Mn2+, and a coronavirus-specific four-residue insert. We determined the x-ray structures of the SARS-CoV-2 2′-O-methyltransferase (the nsp16-nsp10 heterodimer) in complex with its reaction substrates, products, and divalent metal cations. These structural snapshots revealed that metal ions and the insert stabilize interactions between the capped RNA and nsp16, resulting in the precise alignment of the ribonucleotides in the active site. Comparison of available structures of 2′-O-methyltransferases with capped RNAs from different organisms revealed that the four-residue insert unique to coronavirus nsp16 alters the backbone conformation of the capped RNA in the binding groove, thereby promoting catalysis. This insert is highly conserved across coronaviruses, and its absence in mammalian methyltransferases makes this region a promising site for structure-guided drug design of selective coronavirus inhibitors.
- Published
- 2021
25. Structural insight into the dual function of LbpB in mediating Neisserial pathogenesis
- Author
-
Srinivas Chakravarthy, Srinivas Govindan, Andrew D. Mesecar, Ravi Yadav, Nicholas Noinaj, and Courtney M. Daczkowski
- Subjects
QH301-705.5 ,Science ,Structural Biology and Molecular Biophysics ,Antimicrobial peptides ,Mutagenesis (molecular biology technique) ,lactoferrin binding protein b ,Peptide ,Neisseria meningitidis ,General Biochemistry, Genetics and Molecular Biology ,chemistry.chemical_compound ,antimicrobial peptides ,Immune system ,Bacterial Proteins ,iron scavenging ,Lactoferricin ,multidrug resistance ,Biology (General) ,chemistry.chemical_classification ,Microbiology and Infectious Disease ,General Immunology and Microbiology ,biology ,Chemistry ,Lactoferrin ,General Neuroscience ,Binding protein ,Mutagenesis ,food and beverages ,Transporter ,General Medicine ,biology.organism_classification ,Neisseria gonorrhoeae ,Cell biology ,lactoferrin ,Structural biology ,biology.protein ,Medicine ,Neisseria ,Other ,Carrier Proteins ,Protein Binding ,Research Article - Abstract
Lactoferrin-binding protein B (LbpB) is a lipoprotein present on the surface of Neisseria that has been postulated to serve dual functions during pathogenesis in both iron acquisition from lactoferrin (Lf), and in providing protection against the cationic antimicrobial peptide lactoferricin (Lfcn). While previous studies support a dual role for LbpB, exactly how these ligands interact with LbpB has remained unknown. Here, we present the structures of LbpB from N. meningitidis and N. gonorrhoeae in complex with human holo-Lf, forming a 1:1 complex and confirmed by size-exclusion chromatography small-angle X-ray scattering. LbpB consists of N- and C-lobes with the N-lobe interacting extensively with the C-lobe of Lf. Our structures provide insight into LbpB’s preference towards holo-Lf, and our mutagenesis and binding studies show that Lf and Lfcn bind independently. Our studies provide the molecular details for how LbpB serves to capture and preserve Lf in an iron-bound state for delivery to the membrane transporter LbpA for iron piracy, and as an antimicrobial peptide sink to evade host immune defenses.
- Published
- 2021
26. Catalytic function and substrate specificity of alpha and beta coronavirus papain-like proteases: targeting human NL63, SARS-CoV and SARS-CoV-2 (Covid-19) with designed inhibitors
- Author
-
Mackenzie E. Chapman, Courtney M. Daczkowski, Arun K. Ghosh, and Andrew D. Mesecar
- Subjects
Biophysics - Published
- 2022
27. Development of an Efficient Enzyme Production and Structure-Based Discovery Platform for BACE1 Inhibitors
- Author
-
Jagannadharao Tirlangi, Katherine C. Jensen, Andrew D. Mesecar, Annalissa M Kammeyer, Yu-Chen Yen, and Arun K. Ghosh
- Subjects
Macrocyclic Compounds ,Kinetics ,Crystallography, X-Ray ,Biochemistry ,Protein Refolding ,Article ,law.invention ,03 medical and health sciences ,law ,Catalytic Domain ,Cell Line, Tumor ,Drug Discovery ,mental disorders ,Amyloid precursor protein ,Aspartic Acid Endopeptidases ,Humans ,Protease Inhibitors ,Crystallization ,Enzyme Assays ,chemistry.chemical_classification ,0303 health sciences ,biology ,Chemistry ,Drug discovery ,030302 biochemistry & molecular biology ,Isothermal titration calorimetry ,Combinatorial chemistry ,Enzyme assay ,Dissociation constant ,Enzyme ,biology.protein ,Amyloid Precursor Protein Secretases ,Protein Binding - Abstract
BACE1 (Beta-site Amyloid Precursor Protein (APP) Cleaving Enzyme 1) is a promising therapeutic target for Alzheimer’s Disease (AD). However, efficient expression, purification, and crystallization systems are not well described or detailed in the literature nor are approaches for treatment of enzyme kinetic data for potent inhibitors well described. We therefore developed a platform for expression and purification of BACE1, including protein refolding from E.coli inclusion bodies, in addition to optimizing a reproducible crystallization procedure of BACE1 bound with inhibitors. We also report a detailed approach to the proper analysis of enzyme kinetic data for compounds that exhibit either rapid-equilibrium or tight-binding mechanisms. Our methods allow for the purification of ~15 mg of BACE1 enzyme from 1 L of culture which is higher than reported yields in the current literature. To evaluate the data analysis approach developed here, a well-known potent inhibitor and two of its derivatives were tested, analyzed, and compared. The inhibitory constants (K(i)) obtained from the kinetic studies are in agreement with dissociation constants (K(d)) that were also determined using isothermal titration calorimetry (ITC) experiments. The X-ray structures of these three compounds in complex with BACE1 were readily obtained and provide important insight into the structure and thermodynamics of the BACE1-inhibitor interactions.
- Published
- 2019
28. Mn2+ coordinates Cap-0-RNA to align substrates for efficient 2′-O-methyl transfer by SARS-CoV-2 nsp16
- Author
-
George Minasov, Monica Rosas-Lemus, Karla J. F. Satchell, Courtney M. Daczkowski, Paul Hoover, Ludmilla Shuvalova, Joseph S. Brunzelle, Andrew D. Mesecar, and Nicole L. Inniss
- Subjects
chemistry.chemical_classification ,Residue (chemistry) ,Methyltransferase ,Ribonucleotide ,RNA capping ,Chemistry ,Activator (genetics) ,Stereochemistry ,RNA ,Translation (biology) ,Divalent - Abstract
Capping viral messenger RNAs is essential for efficient translation and prevents their detection by host innate immune responses. For SARS-CoV-2, RNA capping includes 2′-O-methylation of the first ribonucleotide by methyltransferase nsp16 in complex with activator nsp10. The reaction requires substrates, a short RNA and SAM, and is catalyzed by divalent cations, with preference for Mn2+. Crystal structures of nsp16-nsp10 with capped RNAs revealed a critical role of metal ions in stabilizing interactions between ribonucleotides and nsp16, resulting in precise alignment of the substrates for methyl transfer. An aspartate residue that is highly conserved among coronaviruses alters the backbone conformation of the capped RNA in the binding groove. This aspartate is absent in mammalian methyltransferases and is a promising site for designing coronavirus-specific inhibitors.
- Published
- 2021
29. Preclinical characterization of an intravenous coronavirus 3CL protease inhibitor for the potential treatment of COVID19
- Author
-
Heather Eng, Annaliesa S. Anderson, Norimitsu Shirai, Brandon J. Anson, James Logue, Stuart Weston, Marisa McGrath, Martyn D. Ticehurst, Rebecca E. O’Connor, Michelle Rossulek, Martin Pettersson, Matthew N O' Brien, Jean G. Sathish, Matthew B. Frieman, Emi Kimoto, Jun Wang, R. Scott Obach, Emily I. Chen, Robert Haupt, Yuao Zhu, Thomas F. Rogers, Andrew D. Mesecar, Suman Luthra, Adolfo García-Sastre, Dafydd R. Owen, Rhys M. Jones, Eugene P. Kadar, Chunlong Ma, Rob Kania, Lisa Aschenbrenner, Arnab K. Chatterjee, Charlotte Moira Norfor Allerton, Joseph John Binder, Kevin Ogilvie, Holly L. Hammond, Nathan Beutler, Claire M. Steppan, Jennifer Hammond, Stephen Noell, Romel Rosales, Robert M. Hoffman, Lillis Jonathan Richard, Matthew R. Reese, Stephen W. Mason, Dan Arenson, Malina A. Bakowski, Lawrence W. Updyke, Lorraine F. Lanyon, Kris M. White, Emma K. Lendy, Melanie G. Kirkpatrick, and Britton Boras
- Subjects
Indoles ,Science ,medicine.medical_treatment ,viruses ,General Physics and Astronomy ,Pharmacology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Article ,Mice ,In vivo ,Coronavirus 229E, Human ,Leucine ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Protease inhibitor (pharmacology) ,skin and connective tissue diseases ,Infusions, Intravenous ,Vero Cells ,Coronavirus 3C Proteases ,Coronavirus ,ADME ,Multidisciplinary ,Protease ,Alanine ,Chemistry ,SARS-CoV-2 ,fungi ,COVID-19 ,Drug Synergism ,General Chemistry ,Prodrug ,In vitro ,Adenosine Monophosphate ,Pyrrolidinones ,respiratory tract diseases ,COVID-19 Drug Treatment ,body regions ,Disease Models, Animal ,Coronavirus Protease Inhibitors ,Severe acute respiratory syndrome-related coronavirus ,Drug Design ,Drug Therapy, Combination ,HeLa Cells - Abstract
COVID-19 caused by the SARS-CoV-2 virus has become a global pandemic. 3CL protease is a virally encoded protein that is essential across a broad spectrum of coronaviruses with no close human analogs. PF-00835231, a 3CL protease inhibitor, has exhibited potent in vitro antiviral activity against SARS-CoV-2 as a single agent. Here we report, the design and characterization of a phosphate prodrug PF-07304814 to enable the delivery and projected sustained systemic exposure in human of PF-00835231 to inhibit coronavirus family 3CL protease activity with selectivity over human host protease targets. Furthermore, we show that PF-00835231 has additive/synergistic activity in combination with remdesivir. We present the ADME, safety, in vitro, and in vivo antiviral activity data that supports the clinical evaluation of PF-07304814 as a potential COVID-19 treatment.
- Published
- 2021
30. Report of the National Institutes of Health SARS-CoV-2 Antiviral Therapeutics Summit
- Author
-
Kumar Singh Saikatendu, Michael J. Sofia, Matthew D. Disney, David Baker, Jennifer O. Nwankwo, Marla Weetall, Annaliesa S. Anderson, Christopher P. Austin, Mindy I. Davis, Matthias Götte, Emmie de Wit, Andrew D. Mesecar, Matthew D. Hall, Richard J. Whitley, Stephanie Moore, James M. Anderson, Kara Carter, George R. Painter, Anthony J. Conley, Charlotte A. Lanteri, Sandra K. Weller, Jay Bradner, Celia A. Schiffer, Tomas Cihlar, Abigail Grossman, Timothy P. Sheahan, Kizzmekia S. Corbett, Stephanie L. Ford-Scheimer, Kyle R. Brimacombe, Lillian Chiang, Elizabeth A. Campbell, Daria J. Hazuda, Mark R. Denison, Frederick G. Hayden, Sara Cherry, Pei Yong Shi, Courtney V. Fletcher, Hilary D. Marston, Jules O'Rear, Hugh D. C. Smyth, Francis S. Collins, and Anthony S. Fauci
- Subjects
0301 basic medicine ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,medicine.disease_cause ,NIH Virtual SARS-CoV-2 Supplement ,03 medical and health sciences ,0302 clinical medicine ,Research community ,preclinical ,Immunology and Allergy ,Medicine ,State of the science ,Coronavirus ,Medical education ,geography ,Summit ,geography.geographical_feature_category ,SARS-CoV-2 ,business.industry ,Research needs ,antiviral therapeutics ,viral replication machinery ,emerging modalities ,AcademicSubjects/MED00290 ,030104 developmental biology ,Infectious Diseases ,Drug development ,proteases ,business ,030217 neurology & neurosurgery - Abstract
The NIH Virtual SARS-CoV-2 Antiviral Summit, held on 6 November 2020, was organized to provide an overview on the status and challenges in developing antiviral therapeutics for coronavirus disease 2019 (COVID-19), including combinations of antivirals. Scientific experts from the public and private sectors convened virtually during a live videocast to discuss severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) targets for drug discovery as well as the preclinical tools needed to develop and evaluate effective small-molecule antivirals. The goals of the Summit were to review the current state of the science, identify unmet research needs, share insights and lessons learned from treating other infectious diseases, identify opportunities for public-private partnerships, and assist the research community in designing and developing antiviral therapeutics. This report includes an overview of therapeutic approaches, individual panel summaries, and a summary of the discussions and perspectives on the challenges ahead for antiviral development.
- Published
- 2021
- Full Text
- View/download PDF
31. Discovery of a Novel Inhibitor of Coronavirus 3CL Protease for the Potential Treatment of COVID-19
- Author
-
Emma K. Lendy, Martyn D. Ticehurst, Robert Steven Kania, Lisa Aschenbrenner, Chuang Ma, Michelle Rossulek, Emily I. Chen, Charlotte Moira Norfor Allerton, Rhys M. Jones, Stephen W. Mason, Kevin Ogilvie, Heather Eng, Dan Arenson, Lorraine F. Lanyon, Abhishek Chatterjee, Rob Haupt, Martin Pettersson, Britton Boras, Eugene P. Kadar, Malina A. Bakowski, Yuao Zhu, Obach Rs, Suman Luthra, Stuart Weston, Joseph John Binder, Lillis, Stephen Noell, Thomas F. Rogers, Dafydd R. Owen, O’Brien Mn, Claire M. Steppan, Lawrence W. Updyke, Jennifer Hammond, Jun Wang, Norimitsu Shirai, Brandon J. Anson, Nathan Beutler, Jean G. Sathish, Melanie G. Kirkpatrick, Annaliesa S. Anderson, James Logue, Matthew B. Frieman, Andrew D. Mesecar, Holly L. Hammond, Robert M. Hoffman, Reese Mr, Marisa McGrath, Rebecca E. O’Connor, and Emi Kimoto
- Subjects
Protease ,Coronavirus disease 2019 (COVID-19) ,SARS-CoV-2 ,Chemistry ,medicine.medical_treatment ,Prodrug ,medicine.disease_cause ,Virology ,Article ,In vitro ,Virus ,Pharmacodynamics ,Preclinical research ,In vivo ,medicine ,Pharmacokinetics ,ADME ,Coronavirus - Abstract
COVID-19 caused by the SARS-CoV-2 virus has become a global pandemic. 3CL protease is a virally encoded protein that is essential across a broad spectrum of coronaviruses with no close human analogs. PF-00835231, a 3CL protease inhibitor, has exhibited potent in vitro antiviral activity against SARS-CoV-2 as a single agent. Here we report, the design and characterization of a phosphate prodrug PF-07304814 to enable the delivery and projected sustained systemic exposure in human of PF-00835231 to inhibit coronavirus family 3CL protease activity with selectivity over human host protease targets. Furthermore, we show that PF-00835231 has additive/synergistic activity in combination with remdesivir. We present the ADME, safety, in vitro, and in vivo antiviral activity data that supports the clinical evaluation of PF-07304814 as a potential COVID-19 treatment., The 3CL protease of SARS-CoV-2 is inhibited by PF-00835231 in vitro. Here, the authors show that the prodrug PF-07304814 has broad spectrum activity, inhibiting SARS-CoV and SARS-CoV-2 in mice and its ADME and safety profile support clinical development.
- Published
- 2020
32. pH dependent inhibition from ammonium ions in the Pseudomonas mevalonii HMG-CoA Reductase crystallization environment
- Author
-
Olaf Wiest, Andrew D. Mesecar, Tim Schmidt, C. D. Critchelow, Cynthia V. Stauffacher, Paul Helquist, Vatsal Purohit, Anthony R. Rosales, and Calvin Steussy
- Subjects
chemistry.chemical_classification ,biology ,Stereochemistry ,Coenzyme A ,Reaction intermediate ,Reductase ,Cofactor ,law.invention ,chemistry.chemical_compound ,Enzyme ,chemistry ,law ,biology.protein ,Ammonium ,NAD+ kinase ,Crystallization - Abstract
HMG-CoA reductase (Pseudomonas mevalonii) utilizes mevalonate, coenzyme A (CoA) and the cofactor NAD in a complex mechanism involving two hydride transfers with cofactor exchange, accompanied by large conformational changes by a 50 residue subdomain, to generate HMG-CoA. Details about this mechanism such as the conformational changes that allow intermediate formation, cofactor exchange and product release remain unknown. The formation of the proposed intermediates has also not been observed in structural studies with natural substrates. Having been shown to be an essential enzyme for the survival of gram-positive antibiotic resistant pathogenic bacteria, studying its mechanism in detail will be beneficial in developing novel antibacterials. The enzyme has been shown to be catalytically active inside the crystal with dithio-HMG-CoA and NADH but curiously is found to be inactive in the reverse direction in the structure bound to mevalonate, CoA and NAD.To understand the factors limiting activity in the HMGR crystal with mevalonate, CoA and NAD, we studied the effect of crystallization components and pH on enzymatic activity. We observed a strong inhibition in the crystallization buffer and an increase in activity with increasing pH. We attribute this inhibitive effect to the presence of ammonium ions present in the crystal since inhibition is also observed with several other ammonium salt buffers. Additionally, the lack of inhibition was observed in the absence of ammonium. The effect of each ligand (mevalonate, CoA and NAD) on the rate of the enzymatic reaction in the crystallization environment was further investigated by measuring their Kmin the crystallization buffer. The Kmmeasurements indicate that the hydride transfer step between NAD and mevalonate is inhibited in the crystallization environment. To test this further, we solved a crystal structure of pmHMGR bound to the post-hydride transfer intermediate (mevaldehyde) and cofactor Coenzyme A. The resulting turnover with the formation of a thiohemiacetal indicated that the crystallization environment inhibited the oxidative acylation of mevalonate and the reaction intermediate mevaldyl-CoA.
- Published
- 2020
33. A small molecule compound with an indole moiety inhibits the main protease of SARS-CoV-2 and blocks virus replication
- Author
-
Srinivasa Rao Allu, Emma K. Lendy, Jakka Raghavaiah, Alexander Wlodawer, Arun K. Ghosh, Debananda Das, David A. Davis, Kazutaka Murayama, Hiroaki Mitsuya, Shogo Misumi, Yuki Takamatsu, Nobuyo Higashi-Kuwata, Mi Li, Brandon J. Anson, Hironori Hayashi, Kazuya Hasegawa, Naoki Kishimoto, Nobutoki Takamune, Shin ichiro Hattori, Eiichi Kodama, Robert Yarchoan, Haydar Bulut, and Andrew D. Mesecar
- Subjects
0301 basic medicine ,Indoles ,Pyridines ,Science ,medicine.medical_treatment ,viruses ,General Physics and Astronomy ,Antiviral Agents ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Chlorocebus aethiops ,medicine ,Moiety ,Animals ,Humans ,Vero Cells ,Indole test ,Multidisciplinary ,Protease ,Alanine ,biology ,Chemistry ,SARS-CoV-2 ,Viral Proteases ,Antimicrobials ,Active site ,virus diseases ,General Chemistry ,Small molecule ,In vitro ,Adenosine Monophosphate ,COVID-19 Drug Treatment ,030104 developmental biology ,Coronavirus Protease Inhibitors ,Viral replication ,Biochemistry ,030220 oncology & carcinogenesis ,Indoline ,biology.protein - Abstract
Except remdesivir, no specific antivirals for SARS-CoV-2 infection are currently available. Here, we characterize two small-molecule-compounds, named GRL-1720 and 5h, containing an indoline and indole moiety, respectively, which target the SARS-CoV-2 main protease (Mpro). We use VeroE6 cell-based assays with RNA-qPCR, cytopathic assays, and immunocytochemistry and show both compounds to block the infectivity of SARS-CoV-2 with EC50 values of 15 ± 4 and 4.2 ± 0.7 μM for GRL-1720 and 5h, respectively. Remdesivir permitted viral breakthrough at high concentrations; however, compound 5h completely blocks SARS-CoV-2 infection in vitro without viral breakthrough or detectable cytotoxicity. Combination of 5h and remdesivir exhibits synergism against SARS-CoV-2. Additional X-ray structural analysis show that 5h forms a covalent bond with Mpro and makes polar interactions with multiple active site amino acid residues. The present data suggest that 5h might serve as a lead Mpro inhibitor for the development of therapeutics for SARS-CoV-2 infection., Here, using in vitro assays and structural analysis, the authors characterize the anti-SARS-CoV-2 properties of two small molcules, showing these to bind and target the virus main protease (Mpro), and to exhibit a synergistic antiviral effect when combined with remdesivir in vitro.
- Published
- 2020
34. Structure-Guided Mutagenesis Alters Deubiquitinating Activity and Attenuates Pathogenesis of a Murine Coronavirus
- Author
-
Matthew Hackbart, Susan C. Baker, Andrew D. Mesecar, Robert C. Mettelman, Amornrat O'Brien, Kristina R. Kesely, Mackenzie E. Chapman, Yafang Chen, Xufang Deng, and Anna M. Mielech
- Subjects
Models, Molecular ,Ubiquitin binding ,Protein Conformation ,viruses ,medicine.medical_treatment ,Mutant ,coronavirus ,Virus Replication ,medicine.disease_cause ,Mice ,Interferon ,Coronavirus ,0303 health sciences ,Virulence ,Chemistry ,030302 biochemistry & molecular biology ,Cell biology ,3. Good health ,Host-Pathogen Interactions ,Interferon Type I ,IFN antagonist ,Coronavirus Infections ,DUB activity ,medicine.drug ,Immunology ,RNA-dependent RNA polymerase ,Mutagenesis (molecular biology technique) ,papain-like protease ,Biology ,Microbiology ,Virus ,PLP2 ,Structure-Activity Relationship ,Viral Proteins ,03 medical and health sciences ,Virology ,medicine ,Animals ,Amino Acid Sequence ,030304 developmental biology ,Murine hepatitis virus ,Protease ,030306 microbiology ,Macrophages ,Ubiquitination ,Viral replication ,Mutagenesis ,Insect Science ,Pathogenesis and Immunity ,PLP2 structure - Abstract
Coronaviruses employ a genetic economy by encoding multifunctional proteins that function in viral replication and also modify the host environment to disarm the innate immune response. The coronavirus papain-like protease 2 (PLP2) domain possesses protease activity, which cleaves the viral replicase polyprotein, and also DUB activity (deconjugating ubiquitin/ubiquitin-like molecules from modified substrates) using identical catalytic residues. To separate the DUB activity from the protease activity, we employed a structure-guided mutagenesis approach and identified residues that are important for ubiquitin binding. We found that mutating the ubiquitin-binding residues results in a PLP2 that has reduced DUB activity but retains protease activity. We engineered a recombinant murine coronavirus to express the DUB mutant and showed that the DUB mutant virus activated an earlier type I interferon response in macrophages and exhibited reduced replication in mice. The results of this study demonstrate that PLP2/DUB is an interferon antagonist and a virulence trait of coronaviruses., Coronaviruses express a multifunctional papain-like protease, termed papain-like protease 2 (PLP2). PLP2 acts as a protease that cleaves the viral replicase polyprotein and as a deubiquitinating (DUB) enzyme which removes ubiquitin (Ub) moieties from ubiquitin-conjugated proteins. Previous in vitro studies implicated PLP2/DUB activity as a negative regulator of the host interferon (IFN) response, but the role of DUB activity during virus infection was unknown. Here, we used X-ray structure-guided mutagenesis and functional studies to identify amino acid substitutions within the ubiquitin-binding surface of PLP2 that reduced DUB activity without affecting polyprotein processing activity. We engineered a DUB mutation (Asp1772 to Ala) into a murine coronavirus and evaluated the replication and pathogenesis of the DUB mutant virus (DUBmut) in cultured macrophages and in mice. We found that the DUBmut virus replicates similarly to the wild-type (WT) virus in cultured cells, but the DUBmut virus activates an IFN response at earlier times compared to the wild-type virus infection in macrophages, consistent with DUB activity negatively regulating the IFN response. We compared the pathogenesis of the DUBmut virus to that of the wild-type virus and found that the DUBmut-infected mice had a statistically significant reduction (P
- Published
- 2020
- Full Text
- View/download PDF
35. Drug Development and Medicinal Chemistry Efforts toward SARS‐Coronavirus and Covid‐19 Therapeutics
- Author
-
Margherita Brindisi, Mackenzie E. Chapman, Arun K. Ghosh, Andrew D. Mesecar, Dana Shahabi, Ghosh, A. K., Brindisi, M., Shahabi, D., Chapman, M. E., and Mesecar, A. D.
- Subjects
Models, Molecular ,Coronavirus disease 2019 (COVID-19) ,Chemistry, Pharmaceutical ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Protease Inhibitor ,Pneumonia, Viral ,coronavirus ,protease inhibitors ,Reviews ,Review ,Computational biology ,Biology ,medicine.disease_cause ,Antiviral Agents ,01 natural sciences ,Approved drug ,Biochemistry ,Drug Development ,Very Important Paper ,Pandemic ,Drug Discovery ,medicine ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,Pandemics ,Coronavirus ,Antiviral Agent ,Pharmacology ,Coronavirus Infection ,010405 organic chemistry ,Drug discovery ,Organic Chemistry ,Proteolytic enzymes ,COVID-19 ,SARS-CoV ,0104 chemical sciences ,coronaviru ,010404 medicinal & biomolecular chemistry ,Drug development ,Molecular Medicine ,Coronavirus Infections ,Human - Abstract
The COVID‐19 pandemic caused by SARS‐CoV‐2 infection is spreading at an alarming rate and has created an unprecedented health emergency around the globe. There is no effective vaccine or approved drug treatment against COVID‐19 and other pathogenic coronaviruses. The development of antiviral agents is an urgent priority. Biochemical events critical to the coronavirus replication cycle provided a number of attractive targets for drug development. These include, spike protein for binding to host cell‐surface receptors, proteolytic enzymes that are essential for processing polyproteins into mature viruses, and RNA‐dependent RNA polymerase for RNA replication. There has been a lot of ground work for drug discovery and development against these targets. Also, high‐throughput screening efforts have led to the identification of diverse lead structures, including natural product‐derived molecules. This review highlights past and present drug discovery and medicinal‐chemistry approaches against SARS‐CoV, MERS‐CoV and COVID‐19 targets. The review hopes to stimulate further research and will be a useful guide to the development of effective therapies against COVID‐19 and other pathogenic coronaviruses., Effective antiviral treatment? The review highlights potential drug design targets against pathogenic coronaviruses, particularly Covid‐19 as well as past and recent progress towards the development of small‐molecule drug‐like compounds including protein‐X‐ray structure‐based design of cysteine protease inhibitors. Hopefully, the present review will stimulate development of much needed effective drugs against Covid‐19.
- Published
- 2020
- Full Text
- View/download PDF
36. Broad-spectrum inhibition of coronavirus main and papain-like proteases by HCV drugs
- Author
-
Richard T. D’Aquila, Sergii Pshenychnyi, Andrew D. Mesecar, Emma K. Lendy, Brandon J. Anson, Mackenzie E. Chapman, and Karla J. F. Satchell
- Subjects
Proteases ,Broad spectrum ,Papain ,chemistry.chemical_compound ,Chemistry ,viruses ,medicine ,virus diseases ,medicine.disease_cause ,Virology ,Coronavirus - Abstract
Coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 has led to over 200,000 deaths thus far. We screened a library of approved antiviral drugs against the two SARS-CoV-2 proteases, 3C-like/main protease (3CLpro/Mpro) and papain-like protease (PLpro), which are essential for viral replication and attractive drug targets. Three HCV protease inhibitors were tested and found to inhibit 3CLpro and PLpro enzymes from Alpha-, Beta- and Gamma-coronaviruses. Anti-HIV drugs had no activity. Boceprevir and telaprevir inhibited 3CLpro, with boceprevir inhibiting eight of nine coronavirus 3CLpro enzymes tested including from SARS-CoV-2, MERS and SARS-CoV. Asunaprevir inhibited PLpro from SARS-CoV-2 and four other coronaviruses. The 1.4 Å X-ray structure of boceprevir bound to 3CLpro was determined to explain its broad-spectrum activity and guide structure-based design of inhibitors of multiple coronaviruses.Authors Brandon J. Anson, Mackenzie E. Chapman, and Emma K. Lendy contributed equally to this work.
- Published
- 2020
37. Characterization of human sulfotransferase 1A1 (SULT1A1) as a new target for structure-based design of drugs to treat cancer
- Author
-
Samadhi C. Kulathunga, Qing Zhou, Nicolás M. Morato, R. Graham Cooks, and Andrew D. Mesecar
- Subjects
Biophysics - Published
- 2022
38. Exploring the structural determinants for inhibitor selectivity towards the BACE protein family using structure-based molecular design
- Author
-
Emma K. Lendy, Yu-Chen Yen, Emilio L. Cardenas, Margherita Brindisi, Arun K. Ghosh, and Andrew D. Mesecar
- Subjects
Biophysics - Published
- 2022
39. A structure-guided taxonomicbased approach to the design of broad-spectrum coronavirus protease inhibitors
- Author
-
Andrew D. Mesecar, Sarah E. St John, Emma K. Lendy, Brandon J. Anson, Mackenzie E. Chapman, and Arun K. Ghosh
- Subjects
Inorganic Chemistry ,Structural Biology ,General Materials Science ,Physical and Theoretical Chemistry ,Condensed Matter Physics ,Biochemistry - Published
- 2021
40. Structural insight into dual function of Neisserial lactoferrin binding protein B
- Author
-
Srinivas Chakravarthy, Srinivas Govindan, Andrew D. Mesecar, Courtney M. Daczkowski, Nicholas Noinaj, and Ravi Yadav
- Subjects
Inorganic Chemistry ,biology ,Structural Biology ,Chemistry ,Lactoferrin ,Binding protein ,biology.protein ,General Materials Science ,Physical and Theoretical Chemistry ,Condensed Matter Physics ,Biochemistry ,Dual function ,Cell biology - Published
- 2021
41. Mg2+ and Mn2+ coordinate Cap-0-RNA to position substrates for efficient 2′-O-methyl transfer by SARS-CoV-2 nsp16
- Author
-
Paul Hoover, George Minasov, Andrew D. Mesecar, Monica R. Lemus, Karla J. F. Satchell, Ludmilla Shuvalova, Nicole L. Inniss, Courtney M. Daczkowski, and Joseph S. Brunzelle
- Subjects
Inorganic Chemistry ,Structural Biology ,Chemistry ,Position (vector) ,Stereochemistry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,RNA ,General Materials Science ,Physical and Theoretical Chemistry ,Condensed Matter Physics ,Biochemistry - Published
- 2021
42. Structural Insights into the Interaction of Coronavirus Papain-Like Proteases and Interferon-Stimulated Gene Product 15 from Different Species
- Author
-
Scott D. Pegan, Andrew D. Mesecar, John V. Dzimianski, Courtney M. Daczkowski, Octavia Y. Goodwin, and Jozlyn R. Clasman
- Subjects
0301 basic medicine ,Proteases ,Middle East respiratory syndrome coronavirus ,Protein Conformation ,viruses ,medicine.disease_cause ,Crystallography, X-Ray ,Article ,Gene product ,03 medical and health sciences ,Mice ,Viral Proteins ,Mouse hepatitis virus ,Structural Biology ,medicine ,Animals ,Humans ,Molecular Biology ,Ubiquitins ,Coronavirus ,Genetics ,Murine hepatitis virus ,biology ,Interferon-stimulated gene ,3C Viral Proteases ,biology.organism_classification ,ISG15 ,Virology ,3. Good health ,Cysteine Endopeptidases ,Kinetics ,030104 developmental biology ,Severe acute respiratory syndrome-related coronavirus ,Middle East Respiratory Syndrome Coronavirus ,Binding domain ,Protein Binding - Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV) encode multifunctional papain-like proteases (PLPs) that have the ability to process the viral polyprotein to facilitate RNA replication and antagonize the host innate immune response. The latter function involves reversing the post-translational modification of cellular proteins conjugated with either ubiquitin (Ub) or Ub-like interferon-stimulated gene product 15 (ISG15). Ub is known to be highly conserved among eukaryotes, but surprisingly, ISG15 is highly divergent among animals. The ramifications of this sequence divergence to the recognition of ISG15 by coronavirus PLPs at a structural and biochemical level are poorly understood. Therefore, the activity of PLPs from SARS-CoV, MERS-CoV, and mouse hepatitis virus was evaluated against seven ISG15s originating from an assortment of animal species susceptible, and not, to certain coronavirus infections. Excitingly, our kinetic, thermodynamic, and structural analysis revealed an array of different preferences among PLPs. Included in these studies is the first insight into a coronavirus PLP's interface with ISG15 via SARS-CoV PLpro in complex with the principle binding domain of human ISG15 (hISG15) and mouse ISG15s (mISG15s). The first X-ray structure of the full-length mISG15 protein is also reported and highlights a unique, twisted hinge region of ISG15 that is not conserved in hISG15, suggesting a potential role in differential recognition. Taken together, this new information provides a structural and biochemical understanding of the distinct specificities among coronavirus PLPs observed and addresses a critical gap of how PLPs can interact with ISG15s from a wide variety of species.
- Published
- 2017
- Full Text
- View/download PDF
43. Steady-state kinetic studies reveal that the anti-cancer target Ubiquitin-Specific Protease 17 (USP17) is a highly efficient deubiquitinating enzyme
- Author
-
Nicole M. Hjortland and Andrew D. Mesecar
- Subjects
0301 basic medicine ,Protein Denaturation ,Protein Folding ,Light ,Biophysics ,lac operon ,Sf9 ,Cleavage (embryo) ,Biochemistry ,Article ,Deubiquitinating enzyme ,03 medical and health sciences ,chemistry.chemical_compound ,Ubiquitin ,Cell Line, Tumor ,Neoplasms ,Endopeptidases ,Escherichia coli ,Humans ,Scattering, Radiation ,Molecular Biology ,chemistry.chemical_classification ,Deubiquitinating Enzymes ,biology ,Lysine ,Ubiquitination ,Hydrogen-Ion Concentration ,Up-Regulation ,Kinetics ,030104 developmental biology ,Enzyme ,Gene Expression Regulation ,chemistry ,Mutagenesis, Site-Directed ,biology.protein ,TCEP ,Protein quaternary structure ,Ultracentrifugation ,HeLa Cells - Abstract
USP17 is a deubiquitinating enzyme that is upregulated in numerous cancers and therefore a drug target. We developed a robust expression, purification, and assay system for USP17 enabling its enzymatic and structural characterization. USP17 was expressed in E. coli as inclusion bodies and then solubilized, refolded, and purified using affinity and size-exclusion chromatography. Milligram quantities of pure USP17 can be produced that is catalytically more efficient ( k cat /K m = 1500 (x10 3 ) M −1 sec −1 ) than other human USPs studied to date. Analytical size-exclusion chromatography, analytical ultracentrifugation, and dynamic light scattering studies suggest that the quaternary structure of USP17 is a monomer. Steady-state kinetic studies show that USP17 efficiently hydrolyzes both ubiquitin-AMC ( k cat = 1.5 sec −1 and K m = 1.0 μM) and ubiquitin-rhodamine110 ( k cat = 1.8 sec −1 and K m = 2.0 μM) substrates. Ubiquitin chain cleavage assays reveal that USP17 efficiently cleaves di-ubiquitin chains with Lys 11 , Lys 33 , Lys 48 and Lys 63 linkages and tetra-ubiquitin chains with Lys 11 , Lys 48 and Lys 63 linkages but is inefficient in cleaving di-ubiquitin chains with Lys 6 , Lys 27 , or Lys 29 linkages or linear ubiquitin chains. The substrate specificity of USP17 is most similar to that of USP1, where both USPs display higher specificity than other characterized members of the USP family.
- Published
- 2016
44. Crystallization of SARS Coronavirus 3CL Protease to Identify Inhibitor Targets
- Author
-
Samantha Zinnia Fernandes, Andrew D. Mesecar, and Brandon J. Anson
- Subjects
Protease ,Chemistry ,law ,medicine.medical_treatment ,Genetics ,medicine ,Severe acute respiratory syndrome coronavirus ,Crystallization ,Molecular Biology ,Biochemistry ,Virology ,Biotechnology ,law.invention - Published
- 2019
45. Cholesterol Sulfonation Enzyme, SULT2B1b, Modulates AR and Cell Growth Properties in Prostate Cancer
- Author
-
Scott A. Crist, Kevin S. Kerian, Livia S. Eberlin, Chang-Deng Hu, Timothy L. Ratliff, Kimberly K. Buhman, R. Graham Cooks, Andrew D. Mesecar, Renee E. Vickman, Grant N. Burcham, and Liang Cheng
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Prostate cancer ,0302 clinical medicine ,Prostate ,Cell Line, Tumor ,LNCaP ,medicine ,Humans ,Molecular Biology ,Cell Proliferation ,Cell Death ,Cell growth ,Prostatic Neoplasms ,Cancer ,medicine.disease ,Androgen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,Receptors, Androgen ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Cholesterol Esters ,Sulfotransferases ,Growth inhibition - Abstract
Cholesterol accumulates in prostate lesions and has been linked to prostate cancer incidence and progression. However, how accumulated cholesterol contributes to prostate cancer development and progression is not completely understood. Cholesterol sulfate (CS), the primary sulfonation product of cholesterol sulfotransferase (SULT2B1b), accumulates in human prostate adenocarcinoma and precancerous prostatic intraepithelial neoplasia (PIN) lesions compared with normal regions of the same tissue sample. Given the enhanced accumulation of CS in these lesions, it was hypothesized that SULT2B1b-mediated production of CS provides a growth advantage to these cells. To address this, prostate cancer cells with RNAi-mediated knockdown (KD) of SULT2B1b were used to assess the impact on cell growth and survival. SULT2B1b is expressed and functional in a variety of prostate cells, and the data demonstrate that SULT2B1b KD, in LNCaP and other androgen-responsive (VCaP and C4-2) cells, results in decreased cell growth/viability and induces cell death. SULT2B1b KD also decreases androgen receptor (AR) activity and expression at mRNA and protein levels. While AR overexpression has no impact on SULT2B1b KD-mediated cell death, the addition of exogenous androgen is able to partially rescue the growth inhibition induced by SULT2B1b KD in LNCaP cells. These results suggest that SULT2B1b positively regulates the AR either through alterations in ligand availability or by interaction with critical coregulators that influence AR activity. Implications: These findings provide evidence that SULT2B1b is a novel regulator of AR activity and cell growth in prostate cancer and should be further investigated for therapeutic potential. Mol Cancer Res; 14(9); 776–86. ©2016 AACR.
- Published
- 2016
46. Design of potent and highly selective inhibitors for human β-secretase 2 (memapsin 1), a target for type 2 diabetes
- Author
-
Andrew D. Mesecar, Emilio L. Cárdenas, Deborah Downs, Kanury V. S. Rao, Yu-Chen Yen, Xiangping Huang, Jordan Tang, Arun K. Ghosh, and Bhavanam Sekhara Reddy
- Subjects
0301 basic medicine ,Cathepsin ,Chemistry ,General Chemistry ,Type 2 diabetes ,Pharmacology ,Highly selective ,medicine.disease ,Combinatorial chemistry ,3. Good health ,03 medical and health sciences ,Memapsin 1 ,030104 developmental biology ,β secretase ,medicine - Abstract
Design, synthesis and evaluation of very potent and selective β-secretase 2 (memapsin 1, BACE 2) inhibitors are described. The inhibitors were designed specifically to interact with the S2′-site of β-secretase 2 to provide >170000-fold selectivity over β-secretase (BACE 1) and >15000-fold selectivity over cathepsin D. BACE 2 is implicated in type 2 diabetes. The studies serve as an important guide to selective BACE 2 inhibitors.
- Published
- 2016
47. Kinetic control of MERS‐CoV 3CL pro is mediated by non‐conserved residues distal to the active site
- Author
-
Andrew D. Mesecar, Brandon J. Anson, and Courtney M. Daczkowski
- Subjects
chemistry.chemical_classification ,Proteases ,biology ,Active site ,RNA ,medicine.disease_cause ,Biochemistry ,Enzyme ,chemistry ,Viral replication ,Genetics ,biology.protein ,medicine ,Enzyme kinetics ,Molecular Biology ,Biotechnology ,Coronavirus ,Cysteine - Abstract
Middle-East Respiratory Syndrome (MERS) Coronavirus is a re-emergent zoonotic pathogen with a 30% fatality rate in humans The MERS positive-sense single-stranded RNA genome is translated into two polyproteins that must be processed by two viral cysteine proteases (3CLpro and PLpro) into 16 non-structural proteins (nsp) before viral replication can occur Attempts to drug either of these cysteine proteases have proven difficult MERS 3CLpro functions as dimer and is unique among all of the coronavirus 3CLpro enzymes studied to date because competitive inhibitors activate the wild-type MERS 3CLpro enzyme at low inhibitor concentrations and inhibit at higher concentrations This mechanism of substrate/inhibitor-induced dimerization seems to be mediated by non-conserved residues in structural regions distal to the catalytic site We have identified unique residues that are involved in the inhibitor-induced dimerization mechanism and we show that these ?competitive? inhibitors activate and inhibit in a dose-dependent manner In an attempt to provide a structural basis for this mechanism, we crystallized the apo and inhibitor-bound forms of this enzyme and diffracted them to 1 5 ? 2 2 A We also show that residues in distinct structural regions synergize to modulate the intrinsic kinetic parameters of this family of enzymes These parameters include increased turnover (kcat) and a lower dimer dissociation constant (KD) Additionally, a rapid-equilibrium kinetic mechanism was developed to model the kinetic response of MERS 3CLpro to inhibitors that act via substrate/inhibitor induced activation/inhibition mechanisms Support or Funding Information This project has been funded in whole or in part with Federal funds from the National Institute of Allergy and Infectious Diseases, National Institutes of Health, Department of Health and Human Services, under Contracts No HHSN272200700058C, HHSN272201200026C, and HHSN272201700060C This research used resources of the Advanced Photon Source, a U S Department of Energy (DOE) Office of Science User Facility operated for the DOE Office of Science by Argonne National Laboratory under Contract No DE-AC02-06CH11357 Use of the Lilly Research Laboratories Collaborative Access Team (LRL-CAT) beamline at Sector 31 of the Advanced Photon Source was provided by Eli Lilly Company, which operates the facility
- Published
- 2020
48. Decoupling deISGylating and deubiquitinating activities of the MERS virus papain-like protease
- Author
-
Andrew D. Mesecar, Karthik Srinivasan, Jozlyn R. Clasman, and Renata K. Everett
- Subjects
0301 basic medicine ,ISG15 ,Proteases ,USP, ubiquitin specific protease ,medicine.medical_treatment ,CoV, coronavirus ,030106 microbiology ,Mutant ,deISGylase ,DUB, deubiquitinating ,PA, propargylamine ,Viral Nonstructural Proteins ,medicine.disease_cause ,Article ,Host-Parasite Interactions ,03 medical and health sciences ,Ubiquitin ,Virology ,medicine ,Humans ,Ubiquitins ,Ub, ubiquitin ,Coronavirus 3C Proteases ,Coronavirus ,Pharmacology ,Mutation ,PLpro, papain-like protease ,Protease ,biology ,Chemistry ,Crystal structure ,ISG15, interferon stimulating gene 15 ,Papain-like protease ,3. Good health ,Cell biology ,Cysteine Endopeptidases ,PLpro ,030104 developmental biology ,Viral replication ,Middle East Respiratory Syndrome Coronavirus ,biology.protein ,Cytokines ,Ubl, ubiquitin-like ,Coronavirus Infections ,Protein Processing, Post-Translational ,Protein Binding - Abstract
Coronavirus papain-like proteases (PLPs or PLpro), such as the one encoded in the genome of the infectious Middle East Respiratory Syndrome (MERS) virus, have multiple enzymatic activities that promote viral infection. PLpro acts as a protease and processes the large coronavirus polyprotein for virus replication. PLpro also functions as both a deubiquitinating (DUB) and deISGylating (deISG) enzyme and removes ubiquitin (Ub) and interferon-stimulated gene 15 (ISG15) from cellular proteins. Both DUB and deISG activities are implicated in suppressing innate immune responses; however, the precise role of each activity in this process is still unclear due in part to the difficulties in separating each activity. In this study, we determine the first structure of MERS PLpro in complex with the full-length human ISG15 to a resolution of 2.3 Å. This structure and available structures of MERS PLpro-Ub complexes were used as molecular guides to design PLpro mutants that lack either or both DUB/deISG activities. We tested 13 different PLpro mutants for protease, DUB, and deISG activitites using fluorescence-based assays. Results show that we can selectively modulate DUB activity at amino acid positions 1649 and 1653 while mutation of Val1691 or His1652 of PLpro to a positive charged residue completely impairs both DUB/deISG activities. These mutant enzymes will provide new functional tools for delineating the importance of DUB versus deISG activity in virus-infected cells and may serve as potential candidates for attenuating the MERS virus in vivo for modified vaccine design efforts., Graphical abstract Image 1, Highlights • The X-ray structure of MERS-CoV papain-like protease bound to full-length human ISG15 was determined. • This is the first structure of a viral-encoded Ubiquitin Specific Protease bound to full-length ISG15. • Structure-guided protein design was used to generate MERS PLpro mutants that have attenuated DUB and/or deISG activities. • Side chain properties, charge and/or steric bulk, are critical in designing mutants with desired substrate specificities. • These new MERS PLpro mutants will aid in delineating the importance of DUB versus deISG activity in virus-infected cells.
- Published
- 2020
49. Highly Selective and Potent Human β-Secretase 2 (BACE2) Inhibitors against Type 2 Diabetes: Design, Synthesis, X-ray Structure and Structure-Activity Relationship Studies
- Author
-
Emma K. Lendy, Bhavanam Sekhara Reddy, Satish Kovela, Emilio L. Cárdenas, Andrew D. Mesecar, Yu-Chen Yen, Margherita Brindisi, Xiangping Huang, Jordan Tang, Arun K. Ghosh, Deborah Downs, Kalapala Venketeswara Rao, Ghosh, A. K., Brindisi, M., Yen, Y. -C., Lendy, E. K., Kovela, S., Cardenas, E. L., Reddy, B. S., Rao, K. V., Downs, D., Huang, X., Tang, J., and Mesecar, A. D.
- Subjects
Isostere ,Stereochemistry ,BACE2 inhibitor ,Drug Evaluation, Preclinical ,Crystallography, X-Ray ,Biochemistry ,Article ,chemistry.chemical_compound ,Structure-Activity Relationship ,Tmem27 ,Drug Discovery ,Amyloid precursor protein ,Ethylamines ,Structure–activity relationship ,Aspartic Acid Endopeptidases ,Humans ,Hypoglycemic Agents ,General Pharmacology, Toxicology and Pharmaceutics ,Enzyme Inhibitors ,Pharmacology ,chemistry.chemical_classification ,type 2 diabete ,Trifluoromethyl ,Binding Sites ,biology ,Molecular Structure ,Organic Chemistry ,BACE2 inhibitors ,structure-based design ,type 2 diabetes ,β-secretase 2 ,Molecular Docking Simulation ,Memapsin 1 ,Enzyme ,chemistry ,Diabetes Mellitus, Type 2 ,Docking (molecular) ,biology.protein ,Molecular Medicine ,Thermodynamics ,Amyloid Precursor Protein Secretases ,Selectivity ,Protein Binding - Abstract
Herein we present the design, synthesis, and biological evaluation of potent and highly selective β-secretase 2 (memapsin 1, beta-site amyloid precursor protein cleaving enzyme 2, or BACE 2) inhibitors. BACE2 has been recognized as an exciting new target for type 2 diabetes. The X-ray structure of BACE1 bound to inhibitor 2 a {N3 -[(1S,2R)-1-benzyl-2-hydroxy-3-[[(1S,2S)-2-hydroxy-1-(isobutylcarbamoyl)propyl]amino]propyl]-5-[methyl(methylsulfonyl)amino]-N1 -[(1R)-1-phenylpropyl]benzene-1,3-dicarboxamide} containing a hydroxyethylamine isostere was determined. Based on this structure, a computational docking study was performed which led to inhibitor 2 a-bound BACE2 models. These were used to optimize the potency and selectivity of inhibitors. A systematic structure-activity relationship study led to the identification of determinants of the inhibitors' potency and selectivity toward the BACE2 enzyme. Inhibitors 2 d [N3 -[(1S,2R)-1-benzyl-2-hydroxy-3-[[(1S,2S)-2-hydroxy-1-(isobutylcarbamoyl)pentyl]amino]propyl]-N1 -methyl-N1 -[(1R)-1-phenylpropyl]benzene-1,3-dicarboxamide; Ki =0.031 nm, selectivity over BACE1: ≈174 000-fold] and 3 l [N1 -((2S,3R)-3-hydroxy-1-phenyl-4-((3-(trifluoromethyl)benzyl)amino)butan-2-yl)-N3 ,5-dimethyl-N3 -((R)-1-phenylethyl)isophthalamide; Ki =1.6 nm, selectivity over BACE1: >500-fold] displayed outstanding potency and selectivity. Inhibitor 3 l is nonpeptide in nature and may pave the way to the development of a new class of potent and selective BACE2 inhibitors with clinical potential.
- Published
- 2018
50. Cholesterol Sulfotransferase SULT2B1b Modulates Sensitivity to Death Receptor Ligand TNFα in Castration-Resistant Prostate Cancer
- Author
-
Scott A. Crist, Timothy L. Ratliff, Gregory M. Cresswell, Andrew D. Mesecar, Jiang Yang, Renee E. Vickman, Faye Zheng, Chang-Deng Hu, Rebecca W. Doerge, Chi Zhang, and Nadia A. Lanman
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Programmed cell death ,Fas-Associated Death Domain Protein ,Recombinant Fusion Proteins ,Apoptosis ,Article ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Cell Line, Tumor ,LNCaP ,medicine ,Humans ,FADD ,Molecular Biology ,biology ,Cell Death ,Chemistry ,Tumor Necrosis Factor-alpha ,Prostate ,Cancer ,NF-kappa B p50 Subunit ,medicine.disease ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,MRNA Sequencing ,Oncology ,Receptors, Androgen ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Tumor necrosis factor alpha ,Sulfotransferases ,Signal Transduction - Abstract
Cholesterol sulfotransferase, SULT2B1b, has been demonstrated to modulate both androgen receptor activity and cell growth properties. However, the mechanism(s) by which SULT2B1b alters these properties within prostate cancer cells has not been described. Furthermore, specific advantages of SULT2B1b expression in prostate cancer cells are not understood. In these studies, single-cell mRNA sequencing was conducted to compare the transcriptomes of SULT2B1b knockdown (KD) versus Control KD LNCaP cells. Over 2,000 differentially expressed genes were identified along with alterations in numerous canonical pathways, including the death receptor signaling pathway. The studies herein demonstrate that SULT2B1b KD increases TNFα expression in prostate cancer cells and results in NF-κB activation in a TNF-dependent manner. More importantly, SULT2B1b KD significantly enhances TNF-mediated apoptosis in both TNF-sensitive LNCaP cells and TNF-resistant C4-2 cells. Overexpression of SULT2B1b in LNCaP cells also decreases sensitivity to TNF-mediated cell death, suggesting that SULT2B1b modulates pathways dictating the TNF sensitivity capacity of prostate cancer cells. Probing human prostate cancer patient datasets further supports this work by providing evidence that SULT2B1b expression is inversely correlated with TNF-related genes, including TNF, CD40LG, FADD, and NFKB1. Together, these data provide evidence that SULT2B1b expression in prostate cancer cells enhances resistance to TNF and may provide a growth advantage. In addition, targeting SULT2B1b may induce an enhanced therapeutic response to TNF treatment in advanced prostate cancer. Implications: These data suggest that SULT2B1b expression enhances resistance to TNF and may promote prostate cancer.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.