18 results on '"Coronavirus 3C Proteases"'
Search Results
2. Hepatitis C virus NS3/4A inhibitors and other drug-like compounds as covalent binders of SARS-CoV-2 main protease.
- Author
-
Andi, Babak, Andi, Babak, Kumaran, Desigan, Kreitler, Dale F, Soares, Alexei S, Keereetaweep, Jantana, Jakoncic, Jean, Lazo, Edwin O, Shi, Wuxian, Fuchs, Martin R, Sweet, Robert M, Shanklin, John, Adams, Paul D, Schmidt, Jurgen G, Head, Martha S, McSweeney, Sean, Andi, Babak, Andi, Babak, Kumaran, Desigan, Kreitler, Dale F, Soares, Alexei S, Keereetaweep, Jantana, Jakoncic, Jean, Lazo, Edwin O, Shi, Wuxian, Fuchs, Martin R, Sweet, Robert M, Shanklin, John, Adams, Paul D, Schmidt, Jurgen G, Head, Martha S, and McSweeney, Sean
- Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), threatens global public health. The world needs rapid development of new antivirals and vaccines to control the current pandemic and to control the spread of the variants. Among the proteins synthesized by the SARS-CoV-2 genome, main protease (Mpro also known as 3CLpro) is a primary drug target, due to its essential role in maturation of the viral polyproteins. In this study, we provide crystallographic evidence, along with some binding assay data, that three clinically approved anti hepatitis C virus drugs and two other drug-like compounds covalently bind to the Mpro Cys145 catalytic residue in the active site. Also, molecular docking studies can provide additional insight for the design of new antiviral inhibitors for SARS-CoV-2 using these drugs as lead compounds. One might consider derivatives of these lead compounds with higher affinity to the Mpro as potential COVID-19 therapeutics for further testing and possibly clinical trials.
- Published
- 2022
3. Benchmarking the ability of novel compounds to inhibit SARS-CoV-2 main protease using steered molecular dynamics simulations
- Author
-
Singh, R., Bhardwaj, V. K., Das, P., Bhattacherjee, D., Zyryanov, G. V., Purohit, R., Singh, R., Bhardwaj, V. K., Das, P., Bhattacherjee, D., Zyryanov, G. V., and Purohit, R.
- Abstract
Background: The SARS-CoV-2 main protease (Mpro) is an attractive target in the COVID-19 drug development process. It catalyzes the polyprotein's translation from viral RNA and specifies a particular cleavage site. Due to the absence of identical cleavage specificity in human cell proteases, targeting Mpro with chemical compounds can obstruct the replication of the virus. Methods: To explore the potential binding mechanisms of 1,2,3-triazole scaffolds in comparison to co-crystallized inhibitors 11a and 11b towards Mpro, we herein utilized molecular dynamics and enhanced sampling simulation studies. Results and conclusion: All the 1,2,3-triazole scaffolds interacted with catalytic residues (Cys145 and His41) and binding pocket residues of Mpro involving Met165, Glu166, Ser144, Gln189, His163, and Met49. Furthermore, the adequate binding free energy and potential mean force of the topmost compound 3h was comparable to the experimental inhibitors 11a and 11b of Mpro. Overall, the current analysis could be beneficial in developing the SARS-CoV-2 Mpro potential inhibitors. © 2022 Elsevier Ltd
- Published
- 2022
4. Hepatitis C virus NS3/4A inhibitors and other drug-like compounds as covalent binders of SARS-CoV-2 main protease.
- Author
-
Andi, Babak, Andi, Babak, Kumaran, Desigan, Kreitler, Dale F, Soares, Alexei S, Keereetaweep, Jantana, Jakoncic, Jean, Lazo, Edwin O, Shi, Wuxian, Fuchs, Martin R, Sweet, Robert M, Shanklin, John, Adams, Paul D, Schmidt, Jurgen G, Head, Martha S, McSweeney, Sean, Andi, Babak, Andi, Babak, Kumaran, Desigan, Kreitler, Dale F, Soares, Alexei S, Keereetaweep, Jantana, Jakoncic, Jean, Lazo, Edwin O, Shi, Wuxian, Fuchs, Martin R, Sweet, Robert M, Shanklin, John, Adams, Paul D, Schmidt, Jurgen G, Head, Martha S, and McSweeney, Sean
- Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), threatens global public health. The world needs rapid development of new antivirals and vaccines to control the current pandemic and to control the spread of the variants. Among the proteins synthesized by the SARS-CoV-2 genome, main protease (Mpro also known as 3CLpro) is a primary drug target, due to its essential role in maturation of the viral polyproteins. In this study, we provide crystallographic evidence, along with some binding assay data, that three clinically approved anti hepatitis C virus drugs and two other drug-like compounds covalently bind to the Mpro Cys145 catalytic residue in the active site. Also, molecular docking studies can provide additional insight for the design of new antiviral inhibitors for SARS-CoV-2 using these drugs as lead compounds. One might consider derivatives of these lead compounds with higher affinity to the Mpro as potential COVID-19 therapeutics for further testing and possibly clinical trials.
- Published
- 2022
5. Discovery and Mechanism of SARS-CoV-2 Main Protease Inhibitors.
- Author
-
Huff, Sarah, Huff, Sarah, Kummetha, Indrasena Reddy, Tiwari, Shashi Kant, Huante, Matthew B, Clark, Alex E, Wang, Shaobo, Bray, William, Smith, Davey, Carlin, Aaron F, Endsley, Mark, Rana, Tariq M, Huff, Sarah, Huff, Sarah, Kummetha, Indrasena Reddy, Tiwari, Shashi Kant, Huante, Matthew B, Clark, Alex E, Wang, Shaobo, Bray, William, Smith, Davey, Carlin, Aaron F, Endsley, Mark, and Rana, Tariq M
- Abstract
The emergence of a new coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presents an urgent public health crisis. Without available targeted therapies, treatment options remain limited for COVID-19 patients. Using medicinal chemistry and rational drug design strategies, we identify a 2-phenyl-1,2-benzoselenazol-3-one class of compounds targeting the SARS-CoV-2 main protease (Mpro). FRET-based screening against recombinant SARS-CoV-2 Mpro identified six compounds that inhibit proteolysis with nanomolar IC50 values. Preincubation dilution experiments and molecular docking determined that the inhibition of SARS-CoV-2 Mpro can occur by either covalent or noncovalent mechanisms, and lead E04 was determined to inhibit Mpro competitively. Lead E24 inhibited viral replication with a nanomolar EC50 value (844 nM) in SARS-CoV-2-infected Vero E6 cells and was further confirmed to impair SARS-CoV-2 replication in human lung epithelial cells and human-induced pluripotent stem cell-derived 3D lung organoids. Altogether, these studies provide a structural framework and mechanism of Mpro inhibition that should facilitate the design of future COVID-19 treatments.
- Published
- 2022
6. A Self-Immolative Fluorescent Probe for Selective Detection of SARS-CoV-2 Main Protease.
- Author
-
Xu, Ming, Xu, Ming, Zhou, Jiajing, Cheng, Yong, Jin, Zhicheng, Clark, Alex E, He, Tengyu, Yim, Wonjun, Li, Yi, Chang, Yu-Ci, Wu, Zhuohong, Fajtová, Pavla, O'Donoghue, Anthony J, Carlin, Aaron F, Todd, Michael D, Jokerst, Jesse V, Xu, Ming, Xu, Ming, Zhou, Jiajing, Cheng, Yong, Jin, Zhicheng, Clark, Alex E, He, Tengyu, Yim, Wonjun, Li, Yi, Chang, Yu-Ci, Wu, Zhuohong, Fajtová, Pavla, O'Donoghue, Anthony J, Carlin, Aaron F, Todd, Michael D, and Jokerst, Jesse V
- Abstract
Existing tools to detect and visualize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) suffer from low selectivity, poor cell permeability, and high cytotoxicity. Here we report a novel self-immolative fluorescent probe (MP590) for the highly selective and sensitive detection of the SARS-CoV-2 main protease (Mpro). This fluorescent probe was prepared by connecting a Mpro-cleavable peptide (N-acetyl-Abu-Tle-Leu-Gln) with a fluorophore (i.e., resorufin) via a self-immolative aromatic linker. Fluorescent titration results show that MP590 can detect Mpro with a limit of detection (LoD) of 35 nM and is selective over interferents such as hemoglobin, bovine serum albumin (BSA), thrombin, amylase, SARS-CoV-2 papain-like protease (PLpro), and trypsin. The cell imaging data indicate that this probe can report Mpro in HEK 293T cells transfected with a Mpro expression plasmid as well as in TMPRSS2-VeroE6 cells infected with SARS-CoV-2. Our results suggest that MP590 can both measure and monitor Mpro activity and quantitatively evaluate Mpro inhibition in infected cells, making it an important tool for diagnostic and therapeutic research on SARS-CoV-2.
- Published
- 2022
7. Identification of SARS-CoV-2 inhibitors targeting Mpro and PLpro using in-cell-protease assay.
- Author
-
Narayanan, Anoop, Narayanan, Anoop, Narwal, Manju, Majowicz, Sydney A, Varricchio, Carmine, Toner, Shay A, Ballatore, Carlo, Brancale, Andrea, Murakami, Katsuhiko S, Jose, Joyce, Narayanan, Anoop, Narayanan, Anoop, Narwal, Manju, Majowicz, Sydney A, Varricchio, Carmine, Toner, Shay A, Ballatore, Carlo, Brancale, Andrea, Murakami, Katsuhiko S, and Jose, Joyce
- Abstract
SARS-CoV-2 proteases Mpro and PLpro are promising targets for antiviral drug development. In this study, we present an antiviral screening strategy involving a novel in-cell protease assay, antiviral and biochemical activity assessments, as well as structural determinations for rapid identification of protease inhibitors with low cytotoxicity. We identified eight compounds with anti-SARS-CoV-2 activity from a library of 64 repurposed drugs and modeled at protease active sites by in silico docking. We demonstrate that Sitagliptin and Daclatasvir inhibit PLpro, and MG-101, Lycorine HCl, and Nelfinavir mesylate inhibit Mpro of SARS-CoV-2. The X-ray crystal structure of Mpro in complex with MG-101 shows a covalent bond formation between the inhibitor and the active site Cys145 residue indicating its mechanism of inhibition is by blocking the substrate binding at the active site. Thus, we provide methods for rapid and effective screening and development of inhibitors for blocking virus polyprotein processing as SARS-CoV-2 antivirals. Additionally, we show that the combined inhibition of Mpro and PLpro is more effective in inhibiting SARS-CoV-2 and the delta variant.
- Published
- 2022
8. Protease-Responsive Peptide-Conjugated Mitochondrial-Targeting AIEgens for Selective Imaging and Inhibition of SARS-CoV-2-Infected Cells.
- Author
-
Cheng, Yong, Cheng, Yong, Clark, Alex, Zhou, Jiajing, He, Tengyu, Li, Yi, Borum, Raina, Creyer, Matthew, Xu, Ming, Jin, Zhicheng, Zhou, Jingcheng, Yim, Wonjun, Wu, Zhuohong, Fajtová, Pavla, ODonoghue, Anthony, Carlin, Aaron, Jokerst, Jesse, Cheng, Yong, Cheng, Yong, Clark, Alex, Zhou, Jiajing, He, Tengyu, Li, Yi, Borum, Raina, Creyer, Matthew, Xu, Ming, Jin, Zhicheng, Zhou, Jingcheng, Yim, Wonjun, Wu, Zhuohong, Fajtová, Pavla, ODonoghue, Anthony, Carlin, Aaron, and Jokerst, Jesse
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a serious threat to human health and lacks an effective treatment. There is an urgent need for both real-time tracking and precise treatment of the SARS-CoV-2-infected cells to mitigate and ultimately prevent viral transmission. However, selective triggering and tracking of the therapeutic process in the infected cells remains challenging. Here, we report a main protease (Mpro)-responsive, mitochondrial-targeting, and modular-peptide-conjugated probe (PSGMR) for selective imaging and inhibition of SARS-CoV-2-infected cells via enzyme-instructed self-assembly and aggregation-induced emission (AIE) effect. The amphiphilic PSGMR was constructed with tunable structure and responsive efficiency and validated with recombinant proteins, cells transfected with Mpro plasmid or infected by SARS-CoV-2, and a Mpro inhibitor. By rational construction of AIE luminogen (AIEgen) with modular peptides and Mpro, we verified that the cleavage of PSGMR yielded gradual aggregation with bright fluorescence and enhanced cytotoxicity to induce mitochondrial interference of the infected cells. This strategy may have value for selective detection and treatment of SARS-CoV-2-infected cells.
- Published
- 2022
9. Structure-based drug design of an inhibitor of the SARS-CoV-2 (COVID-19) main protease using free software: A tutorial for students and scientists.
- Author
-
Zhang, Sheng, Zhang, Sheng, Krumberger, Maj, Morris, Michael A, Parrocha, Chelsea Marie T, Kreutzer, Adam G, Nowick, James S, Zhang, Sheng, Zhang, Sheng, Krumberger, Maj, Morris, Michael A, Parrocha, Chelsea Marie T, Kreutzer, Adam G, and Nowick, James S
- Abstract
This paper describes the structure-based design of a preliminary drug candidate against COVID-19 using free software and publicly available X-ray crystallographic structures. The goal of this tutorial is to disseminate skills in structure-based drug design and to allow others to unleash their own creativity to design new drugs to fight the current pandemic. The tutorial begins with the X-ray crystallographic structure of the main protease (Mpro) of the SARS coronavirus (SARS-CoV) bound to a peptide substrate and then uses the UCSF Chimera software to modify the substrate to create a cyclic peptide inhibitor within the Mpro active site. Finally, the tutorial uses the molecular docking software AutoDock Vina to show the interaction of the cyclic peptide inhibitor with both SARS-CoV Mpro and the highly homologous SARS-CoV-2 Mpro. The supporting information provides an illustrated step-by-step protocol, as well as a video showing the inhibitor design process, to help readers design their own drug candidates for COVID-19 and the coronaviruses that will cause future pandemics. An accompanying preprint in bioRxiv [https://doi.org/10.1101/2020.08.03.234872] describes the synthesis of the cyclic peptide and the experimental validation as an inhibitor of SARS-CoV-2 Mpro.
- Published
- 2021
10. A cyclic peptide inhibitor of the SARS-CoV-2 main protease.
- Author
-
Kreutzer, Adam G, Kreutzer, Adam G, Krumberger, Maj, Diessner, Elizabeth M, Parrocha, Chelsea Marie T, Morris, Michael A, Guaglianone, Gretchen, Butts, Carter T, Nowick, James S, Kreutzer, Adam G, Kreutzer, Adam G, Krumberger, Maj, Diessner, Elizabeth M, Parrocha, Chelsea Marie T, Morris, Michael A, Guaglianone, Gretchen, Butts, Carter T, and Nowick, James S
- Abstract
This paper presents the design and study of a first-in-class cyclic peptide inhibitor against the SARS-CoV-2 main protease (Mpro). The cyclic peptide inhibitor is designed to mimic the conformation of a substrate at a C-terminal autolytic cleavage site of Mpro. The cyclic peptide contains a [4-(2-aminoethyl)phenyl]-acetic acid (AEPA) linker that is designed to enforce a conformation that mimics a peptide substrate of Mpro. In vitro evaluation of the cyclic peptide inhibitor reveals that the inhibitor exhibits modest activity against Mpro and does not appear to be cleaved by the enzyme. Conformational searching predicts that the cyclic peptide inhibitor is fairly rigid, adopting a favorable conformation for binding to the active site of Mpro. Computational docking to the SARS-CoV-2 Mpro suggests that the cyclic peptide inhibitor can bind the active site of Mpro in the predicted manner. Molecular dynamics simulations provide further insights into how the cyclic peptide inhibitor may bind the active site of Mpro. Although the activity of the cyclic peptide inhibitor is modest, its design and study lays the groundwork for the development of additional cyclic peptide inhibitors against Mpro with improved activities.
- Published
- 2021
11. Protease cleavage of RNF20 facilitates coronavirus replication via stabilization of SREBP1.
- Author
-
Zhang, Shilei, Zhang, Shilei, Wang, Jingfeng, Cheng, Genhong, Zhang, Shilei, Zhang, Shilei, Wang, Jingfeng, and Cheng, Genhong
- Abstract
COVID-19, caused by severe acute respiratory coronavirus 2 (SARS-CoV-2), has presented a serious risk to global public health. The viral main protease Mpro (also called 3Clpro) encoded by NSP5 is an enzyme essential for viral replication. However, very few host proteins have been experimentally validated as targets of 3Clpro. Here, through bioinformatics analysis of 300 interferon stimulatory genes (ISGs) based on the prediction method NetCorona, we identify RNF20 (Ring Finger Protein 20) as a novel target of 3Clpro. We have also provided evidence that 3Clpro, but not the mutant 3ClproC145A without catalytic activity, cleaves RNF20 at a conserved Gln521 across species, which subsequently prevents SREBP1 from RNF20-mediated degradation and promotes SARS-CoV-2 replication. We show that RNA interference (RNAi)-mediated depletion of either RNF20 or RNF40 significantly enhances viral replication, indicating the antiviral role of RNF20/RNF40 complex against SARS-CoV-2. The involvement of SREBP1 in SARS-CoV-2 infection is evidenced by a decrease of viral replication in the cells with SREBP1 knockdown and inhibitor AM580. Taken together, our findings reveal RNF20 as a novel host target for SARS-CoV-2 main protease and indicate that 3Clpro inhibitors may treat COVID-19 through not only blocking viral polyprotein cleavage but also enhancing host antiviral response.
- Published
- 2021
12. DNA-encoded chemistry technology yields expedient access to SARS-CoV-2 Mpro inhibitors.
- Author
-
Chamakuri, Srinivas, Chamakuri, Srinivas, Lu, Shuo, Ucisik, Melek Nihan, Bohren, Kurt M, Chen, Ying-Chu, Du, Huang-Chi, Faver, John C, Jimmidi, Ravikumar, Li, Feng, Li, Jian-Yuan, Nyshadham, Pranavanand, Palmer, Stephen S, Pollet, Jeroen, Qin, Xuan, Ronca, Shannon E, Sankaran, Banumathi, Sharma, Kiran L, Tan, Zhi, Versteeg, Leroy, Yu, Zhifeng, Matzuk, Martin M, Palzkill, Timothy, Young, Damian W, Chamakuri, Srinivas, Chamakuri, Srinivas, Lu, Shuo, Ucisik, Melek Nihan, Bohren, Kurt M, Chen, Ying-Chu, Du, Huang-Chi, Faver, John C, Jimmidi, Ravikumar, Li, Feng, Li, Jian-Yuan, Nyshadham, Pranavanand, Palmer, Stephen S, Pollet, Jeroen, Qin, Xuan, Ronca, Shannon E, Sankaran, Banumathi, Sharma, Kiran L, Tan, Zhi, Versteeg, Leroy, Yu, Zhifeng, Matzuk, Martin M, Palzkill, Timothy, and Young, Damian W
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has killed more than 4 million humans globally, but there is no bona fide Food and Drug Administration-approved drug-like molecule to impede the COVID-19 pandemic. The sluggish pace of traditional therapeutic discovery is poorly suited to producing targeted treatments against rapidly evolving viruses. Here, we used an affinity-based screen of 4 billion DNA-encoded molecules en masse to identify a potent class of virus-specific inhibitors of the SARS-CoV-2 main protease (Mpro) without extensive and time-consuming medicinal chemistry. CDD-1714, the initial three-building-block screening hit (molecular weight [MW] = 542.5 g/mol), was a potent inhibitor (inhibition constant [K i] = 20 nM). CDD-1713, a smaller two-building-block analog (MW = 353.3 g/mol) of CDD-1714, is a reversible covalent inhibitor of Mpro (K i = 45 nM) that binds in the protease pocket, has specificity over human proteases, and shows in vitro efficacy in a SARS-CoV-2 infectivity model. Subsequently, key regions of CDD-1713 that were necessary for inhibitory activity were identified and a potent (K i = 37 nM), smaller (MW = 323.4 g/mol), and metabolically more stable analog (CDD-1976) was generated. Thus, screening of DNA-encoded chemical libraries can accelerate the discovery of efficacious drug-like inhibitors of emerging viral disease targets.
- Published
- 2021
13. Protease cleavage of RNF20 facilitates coronavirus replication via stabilization of SREBP1.
- Author
-
Zhang, Shilei, Zhang, Shilei, Wang, Jingfeng, Cheng, Genhong, Zhang, Shilei, Zhang, Shilei, Wang, Jingfeng, and Cheng, Genhong
- Abstract
COVID-19, caused by severe acute respiratory coronavirus 2 (SARS-CoV-2), has presented a serious risk to global public health. The viral main protease Mpro (also called 3Clpro) encoded by NSP5 is an enzyme essential for viral replication. However, very few host proteins have been experimentally validated as targets of 3Clpro. Here, through bioinformatics analysis of 300 interferon stimulatory genes (ISGs) based on the prediction method NetCorona, we identify RNF20 (Ring Finger Protein 20) as a novel target of 3Clpro. We have also provided evidence that 3Clpro, but not the mutant 3ClproC145A without catalytic activity, cleaves RNF20 at a conserved Gln521 across species, which subsequently prevents SREBP1 from RNF20-mediated degradation and promotes SARS-CoV-2 replication. We show that RNA interference (RNAi)-mediated depletion of either RNF20 or RNF40 significantly enhances viral replication, indicating the antiviral role of RNF20/RNF40 complex against SARS-CoV-2. The involvement of SREBP1 in SARS-CoV-2 infection is evidenced by a decrease of viral replication in the cells with SREBP1 knockdown and inhibitor AM580. Taken together, our findings reveal RNF20 as a novel host target for SARS-CoV-2 main protease and indicate that 3Clpro inhibitors may treat COVID-19 through not only blocking viral polyprotein cleavage but also enhancing host antiviral response.
- Published
- 2021
14. DNA-encoded chemistry technology yields expedient access to SARS-CoV-2 Mpro inhibitors.
- Author
-
Chamakuri, Srinivas, Chamakuri, Srinivas, Lu, Shuo, Ucisik, Melek Nihan, Bohren, Kurt M, Chen, Ying-Chu, Du, Huang-Chi, Faver, John C, Jimmidi, Ravikumar, Li, Feng, Li, Jian-Yuan, Nyshadham, Pranavanand, Palmer, Stephen S, Pollet, Jeroen, Qin, Xuan, Ronca, Shannon E, Sankaran, Banumathi, Sharma, Kiran L, Tan, Zhi, Versteeg, Leroy, Yu, Zhifeng, Matzuk, Martin M, Palzkill, Timothy, Young, Damian W, Chamakuri, Srinivas, Chamakuri, Srinivas, Lu, Shuo, Ucisik, Melek Nihan, Bohren, Kurt M, Chen, Ying-Chu, Du, Huang-Chi, Faver, John C, Jimmidi, Ravikumar, Li, Feng, Li, Jian-Yuan, Nyshadham, Pranavanand, Palmer, Stephen S, Pollet, Jeroen, Qin, Xuan, Ronca, Shannon E, Sankaran, Banumathi, Sharma, Kiran L, Tan, Zhi, Versteeg, Leroy, Yu, Zhifeng, Matzuk, Martin M, Palzkill, Timothy, and Young, Damian W
- Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has killed more than 4 million humans globally, but there is no bona fide Food and Drug Administration-approved drug-like molecule to impede the COVID-19 pandemic. The sluggish pace of traditional therapeutic discovery is poorly suited to producing targeted treatments against rapidly evolving viruses. Here, we used an affinity-based screen of 4 billion DNA-encoded molecules en masse to identify a potent class of virus-specific inhibitors of the SARS-CoV-2 main protease (Mpro) without extensive and time-consuming medicinal chemistry. CDD-1714, the initial three-building-block screening hit (molecular weight [MW] = 542.5 g/mol), was a potent inhibitor (inhibition constant [Ki] = 20 nM). CDD-1713, a smaller two-building-block analog (MW = 353.3 g/mol) of CDD-1714, is a reversible covalent inhibitor of Mpro (Ki = 45 nM) that binds in the protease pocket, has specificity over human proteases, and shows in vitro efficacy in a SARS-CoV-2 infectivity model. Subsequently, key regions of CDD-1713 that were necessary for inhibitory activity were identified and a potent (Ki = 37 nM), smaller (MW = 323.4 g/mol), and metabolically more stable analog (CDD-1976) was generated. Thus, screening of DNA-encoded chemical libraries can accelerate the discovery of efficacious drug-like inhibitors of emerging viral disease targets.
- Published
- 2021
15. ReI Tricarbonyl Complexes as Coordinate Covalent Inhibitors for the SARS-CoV-2 Main Cysteine Protease.
- Author
-
Karges, Johannes, Karges, Johannes, Kalaj, Mark, Gembicky, Milan, Cohen, Seth M, Karges, Johannes, Karges, Johannes, Kalaj, Mark, Gembicky, Milan, and Cohen, Seth M
- Abstract
Since its outbreak, the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) has impacted the quality of life and cost hundreds-of-thousands of lives worldwide. Based on its global spread and mortality, there is an urgent need for novel treatments which can combat this disease. To date, the 3-chymotrypsin-like protease (3CLpro ), which is also known as the main protease, is considered among the most important pharmacological targets. The vast majority of investigated 3CLpro inhibitors are organic, non-covalent binders. Herein, the use of inorganic, coordinate covalent binders is proposed that can attenuate the activity of the protease. ReI tricarbonyl complexes were identified that demonstrate coordinate covalent enzymatic inhibition of 3CLpro . Preliminary studies indicate the selective inhibition of 3CLpro over several human proteases. This study presents the first example of metal complexes as inhibitors for the 3CLpro cysteine protease.
- Published
- 2021
16. Structure-based drug design of an inhibitor of the SARS-CoV-2 (COVID-19) main protease using free software: A tutorial for students and scientists.
- Author
-
Zhang, Sheng, Zhang, Sheng, Krumberger, Maj, Morris, Michael A, Parrocha, Chelsea Marie T, Kreutzer, Adam G, Nowick, James S, Zhang, Sheng, Zhang, Sheng, Krumberger, Maj, Morris, Michael A, Parrocha, Chelsea Marie T, Kreutzer, Adam G, and Nowick, James S
- Abstract
This paper describes the structure-based design of a preliminary drug candidate against COVID-19 using free software and publicly available X-ray crystallographic structures. The goal of this tutorial is to disseminate skills in structure-based drug design and to allow others to unleash their own creativity to design new drugs to fight the current pandemic. The tutorial begins with the X-ray crystallographic structure of the main protease (Mpro) of the SARS coronavirus (SARS-CoV) bound to a peptide substrate and then uses the UCSF Chimera software to modify the substrate to create a cyclic peptide inhibitor within the Mpro active site. Finally, the tutorial uses the molecular docking software AutoDock Vina to show the interaction of the cyclic peptide inhibitor with both SARS-CoV Mpro and the highly homologous SARS-CoV-2 Mpro. The supporting information provides an illustrated step-by-step protocol, as well as a video showing the inhibitor design process, to help readers design their own drug candidates for COVID-19 and the coronaviruses that will cause future pandemics. An accompanying preprint in bioRxiv [https://doi.org/10.1101/2020.08.03.234872] describes the synthesis of the cyclic peptide and the experimental validation as an inhibitor of SARS-CoV-2 Mpro.
- Published
- 2021
17. A cyclic peptide inhibitor of the SARS-CoV-2 main protease.
- Author
-
Kreutzer, Adam G, Kreutzer, Adam G, Krumberger, Maj, Diessner, Elizabeth M, Parrocha, Chelsea Marie T, Morris, Michael A, Guaglianone, Gretchen, Butts, Carter T, Nowick, James S, Kreutzer, Adam G, Kreutzer, Adam G, Krumberger, Maj, Diessner, Elizabeth M, Parrocha, Chelsea Marie T, Morris, Michael A, Guaglianone, Gretchen, Butts, Carter T, and Nowick, James S
- Abstract
This paper presents the design and study of a first-in-class cyclic peptide inhibitor against the SARS-CoV-2 main protease (Mpro). The cyclic peptide inhibitor is designed to mimic the conformation of a substrate at a C-terminal autolytic cleavage site of Mpro. The cyclic peptide contains a [4-(2-aminoethyl)phenyl]-acetic acid (AEPA) linker that is designed to enforce a conformation that mimics a peptide substrate of Mpro. In vitro evaluation of the cyclic peptide inhibitor reveals that the inhibitor exhibits modest activity against Mpro and does not appear to be cleaved by the enzyme. Conformational searching predicts that the cyclic peptide inhibitor is fairly rigid, adopting a favorable conformation for binding to the active site of Mpro. Computational docking to the SARS-CoV-2 Mpro suggests that the cyclic peptide inhibitor can bind the active site of Mpro in the predicted manner. Molecular dynamics simulations provide further insights into how the cyclic peptide inhibitor may bind the active site of Mpro. Although the activity of the cyclic peptide inhibitor is modest, its design and study lays the groundwork for the development of additional cyclic peptide inhibitors against Mpro with improved activities.
- Published
- 2021
18. Protease cleavage of RNF20 facilitates coronavirus replication via stabilization of SREBP1.
- Author
-
Zhang, Shilei, Zhang, Shilei, Wang, Jingfeng, Cheng, Genhong, Zhang, Shilei, Zhang, Shilei, Wang, Jingfeng, and Cheng, Genhong
- Abstract
COVID-19, caused by severe acute respiratory coronavirus 2 (SARS-CoV-2), has presented a serious risk to global public health. The viral main protease Mpro (also called 3Clpro) encoded by NSP5 is an enzyme essential for viral replication. However, very few host proteins have been experimentally validated as targets of 3Clpro. Here, through bioinformatics analysis of 300 interferon stimulatory genes (ISGs) based on the prediction method NetCorona, we identify RNF20 (Ring Finger Protein 20) as a novel target of 3Clpro. We have also provided evidence that 3Clpro, but not the mutant 3ClproC145A without catalytic activity, cleaves RNF20 at a conserved Gln521 across species, which subsequently prevents SREBP1 from RNF20-mediated degradation and promotes SARS-CoV-2 replication. We show that RNA interference (RNAi)-mediated depletion of either RNF20 or RNF40 significantly enhances viral replication, indicating the antiviral role of RNF20/RNF40 complex against SARS-CoV-2. The involvement of SREBP1 in SARS-CoV-2 infection is evidenced by a decrease of viral replication in the cells with SREBP1 knockdown and inhibitor AM580. Taken together, our findings reveal RNF20 as a novel host target for SARS-CoV-2 main protease and indicate that 3Clpro inhibitors may treat COVID-19 through not only blocking viral polyprotein cleavage but also enhancing host antiviral response.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.