1
|
Wu J, Yu L, Li K, Zhang D, Ye L, Xu X, Zhou J, Li Z, Xu H. Electrochemical Synthesis of Quinazolines: N-H/C(sp 3)-H Coupling of o-Carbonyl Anilines with Amino Acids and Amines. J Org Chem 2024; 89:17031-17041. [PMID: 39528414 DOI: 10.1021/acs.joc.4c01173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
A mild and efficient electrochemical protocol for the synthesis of quinazolines through N-H/C(sp3)-H coupling of o-carbonyl anilines with natural amino acids/amines has been developed. The products quinazolines can easily be isolated in moderate to excellent yields under external chemical oxidant-free conditions. Moreover, this reaction can be safely conducted on gram scale.
Collapse
Affiliation(s)
- Jiwei Wu
- College of Chemistry and Materials Engineering, Anhui Science and Technology University, Fengyang 233100, P. R. China
| | - Lingxiang Yu
- College of Chemistry and Materials Engineering, Anhui Science and Technology University, Fengyang 233100, P. R. China
| | - Kaixuan Li
- College of Chemistry and Materials Engineering, Anhui Science and Technology University, Fengyang 233100, P. R. China
| | - Di Zhang
- College of Chemistry and Materials Engineering, Anhui Science and Technology University, Fengyang 233100, P. R. China
| | - Longqiang Ye
- College of Chemistry and Materials Engineering, Anhui Science and Technology University, Fengyang 233100, P. R. China
| | - Xiaolan Xu
- School of Medical Science, Anhui Medical University, Hefei 230009, P. R. China
| | - Jie Zhou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Zirong Li
- College of Chemistry and Materials Engineering, Anhui Science and Technology University, Fengyang 233100, P. R. China
| | - Huajian Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| |
Collapse
|
2
|
Du S, Liu X, Hu X, Zhan P. Viral Protein Dimerization Quality Control: A Design Strategy for a Potential Viral Inhibitor. J Med Chem 2024; 67:16951-16966. [PMID: 39303015 DOI: 10.1021/acs.jmedchem.4c01540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
The global pharmaceutical market has been profoundly impacted by the coronavirus pandemic, leading to an increased demand for specific drugs. Consequently, drug resistance has prompted continuous innovation in drug design strategies to effectively combat resistant pathogens or disease variants. Protein dimers play crucial roles in vivo, including catalytic reactions, signal transduction, and structural stability. The site of action for protein dimerization modulators typically does not reside within the active site of the protein, thereby potentially impeding resistance development. Therefore, harnessing viral protein dimerization modulators could represent a promising avenue for combating viral infections. In this Perspective, we provide a detailed introduction to the design principles and applications of dimerization modulators in antiviral research. Furthermore, we analyze various representative examples to elucidate their modes of action while presenting our perspective on dimerization modulators along with the opportunities and challenges associated with this groundbreaking area of investigation.
Collapse
Affiliation(s)
- Shaoqing Du
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, Shandong 250012, P. R. China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, Shandong 250012, P. R. China
| | - Xueping Hu
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao 266237, P. R. China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan, Shandong 250012, P. R. China
| |
Collapse
|
3
|
Yu SC, Li X, Cheng L, Liu L. Constrution of Quinazoline-Linked Covalent Organic Frameworks via a Multicomponent Reaction for Photocatalysis. Chemistry 2024; 30:e202400668. [PMID: 38822692 DOI: 10.1002/chem.202400668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Quinazoline (Qz)-linked covalent organic frameworks (COFs) have been constructed via a three-component reaction of ortho-acylanilines, benzaldehydes and NH4OAc. The structure of Qz-COFs has been confirmed by solid-state nuclear magnetic resonance spectroscopy, Fourier transform infrared and powder X-ray diffraction patterns. The Qz-COFs possess high chemical stability, showing good endurance to strong acid, strong base, oxidant, reductant and other conditions. Particularly, Qz-COF-3 can catalyze the aerobic photooxidation of toluene and other compounds containing C(sp3)-H bonds.
Collapse
Affiliation(s)
- Song-Chen Yu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Xiaohu Li
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Liang Cheng
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Li Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Laboratory of Molecular Recognition and Function, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| |
Collapse
|
4
|
Zhang DW, Xu XS, Zhou R, Fu Z. Modulation of HIV-1 capsid multimerization by sennoside A and sennoside B via interaction with the NTD/CTD interface in capsid hexamer. Front Microbiol 2023; 14:1270258. [PMID: 37817748 PMCID: PMC10561090 DOI: 10.3389/fmicb.2023.1270258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Small molecules that bind to the pocket targeted by a peptide, termed capsid assembly inhibitor (CAI), have shown antiviral effects with unique mechanisms of action. We report the discovery of two natural compounds, sennoside A (SA) and sennoside B (SB), derived from medicinal plants that bind to this pocket in the C-terminal domain of capsid (CA CTD). Both SA and SB were identified via a drug-screening campaign that utilized a time-resolved fluorescence resonance energy transfer assay. They inhibited the HIV-1 CA CTD/CAI interaction at sub-micromolar concentrations of 0.18 μM and 0.08 μM, respectively. Mutation of key residues (including Tyr 169, Leu 211, Asn 183, and Glu 187) in the CA CTD decreased their binding affinity to the CA monomer, from 1.35-fold to 4.17-fold. Furthermore, both compounds induced CA assembly in vitro and bound directly to the CA hexamer, suggesting that they interact with CA beyond the CA CTD. Molecular docking showed that both compounds were bound to the N-terminal domain (NTD)/CTD interface between adjacent protomers within the CA hexamer. SA established a hydrogen-bonding network with residues N57, V59, Q63, K70, and N74 of CA1-NTD and Q179 of CA2-CTD. SB formed hydrogen bonds with the N53, N70, and N74 residues of CA1-NTD, and the A177and Q179 residues of CA2-CTD. Both compounds, acting as glue, can bring αH4 in the NTD and αH9 in the CTD of the NTD/CTD interface close to each other. Collectively, our research indicates that SA and SB, which enhance CA assembly, could serve as novel chemical tools to identify agents that modulate HIV-1 CA assembly. These natural compounds may potentially lead to the development of new antiviral therapies with unique mechanisms of action.
Collapse
Affiliation(s)
- Da-Wei Zhang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Xiao-Shuang Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Rui Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiguo Fu
- Department of Orthopedics, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China
| |
Collapse
|
5
|
Tamatam R, Shin D. Recent Advances in the Transition-Metal-Free Synthesis of Quinazolines. Molecules 2023; 28:molecules28073227. [PMID: 37049989 PMCID: PMC10147101 DOI: 10.3390/molecules28073227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023] Open
Abstract
Quinazolines are a privileged class of nitrogen-containing heterocycles, widely present in a variety of natural products and synthetic chemicals with a broad spectrum of biological and medicinal activities. Owing to their pharmaceutical applications and promising biological value, a variety of synthetic methodologies have been reported for these scaffolds. From the perspective of green and sustainable chemistry, transition-metal-free synthesis provides an alternative method for accessing several biologically active heterocycles. In this review, we summarize the recent progress achieved in the transition-metal-free synthesis of quinazolines and we cover the literature from 2015 to 2022. This aspect is present alongside the advantages, limitations, mechanistic rationalization, and future perspectives associated with the synthetic methodologies.
Collapse
Affiliation(s)
- Rekha Tamatam
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea
- Gachon Pharmaceutical Research Institute, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Dongyun Shin
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea
- Gachon Pharmaceutical Research Institute, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea
| |
Collapse
|
6
|
Zhao S, Zhang X, da Silva-Júnior EF, Zhan P, Liu X. Computer-aided drug design in seeking viral capsid modulators. Drug Discov Today 2023; 28:103581. [PMID: 37030533 DOI: 10.1016/j.drudis.2023.103581] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/16/2023] [Accepted: 03/30/2023] [Indexed: 04/09/2023]
Abstract
Approved or licensed antiviral drugs have limited applications because of their drug resistance and severe adverse effects. By contrast, by stabilizing or destroying the viral capsid, compounds known as capsid modulators prevent viral replication by acting on new targets and, therefore, overcoming the problem of clinical drug resistance. For example. computer-aided drug design (CADD) methods, using strategies based on structures of biological targets (structure-based drug design; SBDD), such as docking, molecular dynamics (MD) simulations, and virtual screening (VS), have provided opportunities for fast and effective development of viral capsid modulators. In this review, we summarize the application of CADD in the discovery, optimization, and mechanism prediction of capsid-targeting small molecules, providing new insights into antiviral drug discovery modalities. Teaser: Computer-aided drug design will accelerate the development of viral capsid regulators, which brings new hope for the treatment of refractory viral diseases.
Collapse
Affiliation(s)
- Shujie Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Xujie Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China
| | - Edeildo Ferreira da Silva-Júnior
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Lourival Melo Mota Avenue, 57072-970 Maceió, Alagoas, Brazil.
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China.
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, PR China.
| |
Collapse
|
7
|
Tamatam R, Kim SH, Shin D. Transition-metal-catalyzed synthesis of quinazolines: A review. Front Chem 2023; 11:1140562. [PMID: 37007059 PMCID: PMC10060649 DOI: 10.3389/fchem.2023.1140562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
Quinazolines are a class of nitrogen-containing heterocyclic compounds with broad-spectrum of pharmacological activities. Transition-metal-catalyzed reactions have emerged as reliable and indispensable tools for the synthesis of pharmaceuticals. These reactions provide new entries into pharmaceutical ingredients of continuously increasing complexity, and catalysis with these metals has streamlined the synthesis of several marketed drugs. The last few decades have witnessed a tremendous outburst of transition-metal-catalyzed reactions for the construction of quinazoline scaffolds. In this review, the progress achieved in the synthesis of quinazolines under transition metal-catalyzed conditions are summarized and reports from 2010 to date are covered. This is presented along with the mechanistic insights of each representative methodology. The advantages, limitations, and future perspectives of synthesis of quinazolines through such reactions are also discussed.
Collapse
Affiliation(s)
- Rekha Tamatam
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
- Gachon Pharmaceutical Research Institute, Gachon University, Incheon, Republic of Korea
| | - Seok-Ho Kim
- College of Pharmacy, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
- *Correspondence: Seok-Ho Kim, ; Dongyun Shin,
| | - Dongyun Shin
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
- Gachon Pharmaceutical Research Institute, Gachon University, Incheon, Republic of Korea
- *Correspondence: Seok-Ho Kim, ; Dongyun Shin,
| |
Collapse
|
8
|
Zhang DW, Xie L, Xu XS, Li Y, Xu X. A Broad-Spectrum Antiviral Molecule, Protoporphyrin IX, Acts as a Moderator of HIV-1 Capsid Assembly by Targeting the Capsid Hexamer. Microbiol Spectr 2023; 11:e0266322. [PMID: 36475726 PMCID: PMC9927277 DOI: 10.1128/spectrum.02663-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The capsid protein (CA), an essential component of human immunodeficiency virus type 1 (HIV-1), represents an appealing target for antivirals. Small molecules targeting the CAI-binding cavity in the C-terminal domain of HIV-1 CA (CA CTD) confer potent antiviral activities. In this study, we report that a small molecule, protoporphyrin IX (PPIX), targets the HIV-1 CA by binding to this pocket. PPIX was identified via in vitro drug screening, using a homogeneous and time-resolved fluorescence-based assay. CA multimerization and a biolayer interferometry (BLI) assay showed that PPIX promoted CA multimerization and bound directly to CA. The binding model of PPIX to CA CTD revealed that PPIX forms hydrogen bonds with the L211and E212 residues in the CA CTD. Moreover, the BLI assay demonstrated that this compound preferentially binds to the CA hexamer versus the monomer. The superposition of the CAI CTD-PPIX complex and the hexameric CA structure suggests that PPIX binds to the interface formed by the NTD and the CTD between adjacent protomers in the CA hexamer via the T72 and E212 residues, serving as a glue to enhance the multimerization of CA. Taken together, our studies demonstrate that PPIX, a hexamer-targeted CA assembly enhancer, should be a new chemical probe for the discovery of modulators of the HIV-1 capsid assembly. IMPORTANCE CA and its assembled viral core play essential roles in distinct steps during HIV-1 replication, including reverse transcription, integration, nuclear entry, virus assembly, and maturation through CA-CA or CA-host factor interactions. These functions of CA are fundamental for HIV-1 pathogenesis, making it an appealing target for antiviral therapy. In the present study, we identified protoporphyrin IX (PPIX) as a candidate CA modulator that can promote CA assembly and prefers binding the CA hexamer versus the monomer. PPIX, like a glue, bound at the interfaces between CA subunits to accelerate CA multimerization. Therefore, PPIX could be used as a new lead for a CA modulator, and it holds potential research applications.
Collapse
Affiliation(s)
- Da-Wei Zhang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Liangxu Xie
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Xiao-Shuang Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Yimin Li
- College of Pharmacy and Key Laboratory for Research and Development of “Qin Medicine” of Shaanxi Administration of Chinese Medicine, Shaanxi University of Chinese Medicine, Xixian New District, China
| | - Xiaojun Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| |
Collapse
|
9
|
Sharma S, Utreja D. Synthesis and antiviral activity of diverse heterocyclic scaffolds. Chem Biol Drug Des 2022; 100:870-920. [PMID: 34551197 DOI: 10.1111/cbdd.13953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/07/2021] [Accepted: 09/11/2021] [Indexed: 01/25/2023]
Abstract
Heterocyclic moieties form a major part of organic chemistry as they are widely distributed in nature and have wide scale practical applications ranging from extensive clinical use to diverse fields such as medicine, agriculture, photochemistry, biocidal formulations, and polymer science. By virtue of their therapeutic properties, they could be employed in combating many infectious diseases. Among the common infectious diseases, viral infections are of great public health importance worldwide. Thus, there is an urgent need for the discovery and development of antiviral drugs and clinical methods to prevent various viral infections so as to increase the life expectancy. This review presents the comprehensive overview of the synthesis and antiviral activity of different heterocyclic compounds 2015 onwards, which aids in present knowledge and helps the researchers and other stakeholders to explore their field.
Collapse
Affiliation(s)
- Shivali Sharma
- Department of Chemistry, College of Basic Sciences and Humanities, Punjab Agricultural University, Ludhiana, India
| | - Divya Utreja
- Department of Chemistry, College of Basic Sciences and Humanities, Punjab Agricultural University, Ludhiana, India
| |
Collapse
|
10
|
Sadeghi Meresht A, Ezzatzadeh E, Dehbandi B, Salimifard M, Rostamian R. Fe 3O 4/CuO Nanocomposite Promoted Green Synthesis of Functionalized Quinazolines Using Water Extract of Lettuce Leaves as Green Media: Study of Antioxidant Activity. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2021.1913426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Abdollah Sadeghi Meresht
- Active Pharmaceutical Ingeredients Research Center (APIRC), Tehran Medicinal Science Branch, Islamic Azad University, Tehran, Iran
| | - Elham Ezzatzadeh
- Department of Chemistry, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| | - Behnam Dehbandi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoomeh Salimifard
- Department of Chemistry, Qaemshahr Branch, Islamic Azad University, Qaemshahr, Iran
| | - Rezvaneh Rostamian
- Department of Chemistry, Qaemshahr Branch, Islamic Azad University, Qaemshahr, Iran
| |
Collapse
|
11
|
Sheikholeslami-Farahani F, Sadeghi Marasht A, Mirabi A, Ghazvini M, Hosseinnasab Rostam M. Ionic Liquid as Green and Recyclable Solvent for the Synthesis of Pyrazinoquinazolines: Study of Antioxidant Activity. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2020.1871039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
| | - Abdollah Sadeghi Marasht
- Active Pharmaceutical Ingeredients Research Center (APIRC), Tehran Medicinal Science Branch, Islamic Azad University, Tehran, Iran
| | - Ali Mirabi
- Department of Chemistry, Qaemshahr Branch, Islamic Azad University, Qaemshahr, Iran
| | - Maryam Ghazvini
- Department of Chemistry, Payame Noor University, Tehran, Iran
| | | |
Collapse
|
12
|
Sarkar R, Gajurel S, Gupta A, Kumar Pal A. Synergistic Catalysis by Copper Oxide/Graphene Oxide Nanocomposites: A Facile Approach to Prepare Quinazolines and Quinazoline Containing Triazole/Tetrazole Moieties under Mild Reaction Conditions. ChemistrySelect 2022. [DOI: 10.1002/slct.202200297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Rajib Sarkar
- Department of Chemistry, Centre for Advanced Studies North-Eastern Hill University Shillong 793022 India
| | - Sushmita Gajurel
- Department of Chemistry, Centre for Advanced Studies North-Eastern Hill University Shillong 793022 India
| | - Ajay Gupta
- Department of Chemistry, Centre for Advanced Studies North-Eastern Hill University Shillong 793022 India
| | - Amarta Kumar Pal
- Department of Chemistry, Centre for Advanced Studies North-Eastern Hill University Shillong 793022 India
| |
Collapse
|
13
|
McFadden WM, Snyder AA, Kirby KA, Tedbury PR, Raj M, Wang Z, Sarafianos SG. Rotten to the core: antivirals targeting the HIV-1 capsid core. Retrovirology 2021; 18:41. [PMID: 34937567 PMCID: PMC8693499 DOI: 10.1186/s12977-021-00583-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/09/2021] [Indexed: 12/20/2022] Open
Abstract
The capsid core of HIV-1 is a large macromolecular assembly that surrounds the viral genome and is an essential component of the infectious virus. In addition to its multiple roles throughout the viral life cycle, the capsid interacts with multiple host factors. Owing to its indispensable nature, the HIV-1 capsid has been the target of numerous antiretrovirals, though most capsid-targeting molecules have not had clinical success until recently. Lenacapavir, a long-acting drug that targets the HIV-1 capsid, is currently undergoing phase 2/3 clinical trials, making it the most successful capsid inhibitor to-date. In this review, we detail the role of the HIV-1 capsid protein in the virus life cycle, categorize antiviral compounds based on their targeting of five sites within the HIV-1 capsid, and discuss their molecular interactions and mechanisms of action. The diverse range of inhibition mechanisms provides insight into possible new strategies for designing novel HIV-1 drugs and furthers our understanding of HIV-1 biology. ![]()
Collapse
Affiliation(s)
- William M McFadden
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Alexa A Snyder
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Karen A Kirby
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Philip R Tedbury
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Monika Raj
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Zhengqiang Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Stefan G Sarafianos
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA.
| |
Collapse
|
14
|
Identification of an Antiretroviral Small Molecule That Appears To Be a Host-Targeting Inhibitor of HIV-1 Assembly. J Virol 2021; 95:JVI.00883-20. [PMID: 33148797 PMCID: PMC7925099 DOI: 10.1128/jvi.00883-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/25/2020] [Indexed: 12/16/2022] Open
Abstract
Given the projected increase in multidrug-resistant HIV-1, there is an urgent need for development of antiretrovirals that act on virus life cycle stages not targeted by drugs currently in use. Host-targeting compounds are of particular interest because they can offer a high barrier to resistance. Here, we report identification of two related small molecules that inhibit HIV-1 late events, a part of the HIV-1 life cycle for which potent and specific inhibitors are lacking. This chemotype was discovered using cell-free protein synthesis and assembly systems that recapitulate intracellular host-catalyzed viral capsid assembly pathways. These compounds inhibit replication of HIV-1 in human T cell lines and peripheral blood mononuclear cells, and are effective against a primary isolate. They reduce virus production, likely by inhibiting a posttranslational step in HIV-1 Gag assembly. Notably, the compound colocalizes with HIV-1 Gag in situ; however, unexpectedly, selection experiments failed to identify compound-specific resistance mutations in gag or pol, even though known resistance mutations developed upon parallel nelfinavir selection. Thus, we hypothesized that instead of binding to Gag directly, these compounds localize to assembly intermediates, the intracellular multiprotein complexes containing Gag and host factors that form during immature HIV-1 capsid assembly. Indeed, imaging of infected cells shows compound colocalized with two host enzymes found in assembly intermediates, ABCE1 and DDX6, but not two host proteins found in other complexes. While the exact target and mechanism of action of this chemotype remain to be determined, our findings suggest that these compounds represent first-in-class, host-targeting inhibitors of intracellular events in HIV-1 assembly.IMPORTANCE The success of antiretroviral treatment for HIV-1 is at risk of being undermined by the growing problem of drug resistance. Thus, there is a need to identify antiretrovirals that act on viral life cycle stages not targeted by drugs in use, such as the events of HIV-1 Gag assembly. To address this gap, we developed a compound screen that recapitulates the intracellular events of HIV-1 assembly, including virus-host interactions that promote assembly. This effort led to the identification of a new chemotype that inhibits HIV-1 replication at nanomolar concentrations, likely by acting on assembly. This compound colocalized with Gag and two host enzymes that facilitate capsid assembly. However, resistance selection did not result in compound-specific mutations in gag, suggesting that the chemotype does not directly target Gag. We hypothesize that this chemotype represents a first-in-class inhibitor of virus production that acts by targeting a virus-host complex important for HIV-1 Gag assembly.
Collapse
|
15
|
Zhou XR, Liu Y, Huang Z, Yao Q, He F, Gao Y. Gag Protein Oriented Supramolecular Nets as Potential HIV Traps. Bioconjug Chem 2021; 32:106-110. [PMID: 33405891 DOI: 10.1021/acs.bioconjchem.0c00706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
For HIV/AIDS treatment, the cocktail therapy which uses a combination of anti-retroviral drugs remains the most widely accepted practice. However, the potential drug toxicity, patient tolerability, and emerging drug resistance have limited its long-term efficiency. Here, we design a HIV Gag protein-targeting redox supramolecular assembly (ROSA) system for potential HIV inhibition. An assembling precursor was constructed through conjugation of an oxidation-activatable fluorogenic compound BQA with a selected tetrapeptide GGFF. Since BQA shares a similar structure with the known Gag inhibitor, the precursor could bind to HIV Gag protein with moderate affinity. Moreover, after oxidation, the corresponding nanofibers could bind to Gag protein and trap HIV to realize virus control, thus providing a potential anti-HIV strategy.
Collapse
Affiliation(s)
- Xi-Rui Zhou
- Division of Chemical Metrology and Analytical Science, National Institute of Metrology, Beijing 100029, China
| | - Ye Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming 650031, China
| | - Zhentao Huang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Qingxin Yao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Fangfei He
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yuan Gao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory of Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
| |
Collapse
|
16
|
Fernández GA, Castro EF, Rosas RA, Fidalgo DM, Adler NS, Battini L, España de Marco MJ, Fabiani M, Bruno AM, Bollini M, Cavallaro LV. Design and Optimization of Quinazoline Derivatives: New Non-nucleoside Inhibitors of Bovine Viral Diarrhea Virus. Front Chem 2020; 8:590235. [PMID: 33425849 PMCID: PMC7793975 DOI: 10.3389/fchem.2020.590235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/09/2020] [Indexed: 01/30/2023] Open
Abstract
Bovine viral diarrhea virus (BVDV) belongs to the Pestivirus genus (Flaviviridae). In spite of the availability of vaccines, the virus is still causing substantial financial losses to the livestock industry. In this context, the use of antiviral agents could be an alternative strategy to control and reduce viral infections. The viral RNA-dependent RNA polymerase (RdRp) is essential for the replication of the viral genome and constitutes an attractive target for the identification of antiviral compounds. In a previous work, we have identified potential molecules that dock into an allosteric binding pocket of BVDV RdRp via a structure-based virtual screening approach. One of them, N-(2-morpholinoethyl)-2-phenylquinazolin-4-amine [1, 50% effective concentration (EC50) = 9.7 ± 0.5 μM], was selected to perform different chemical modifications. Among 24 derivatives synthesized, eight of them showed considerable antiviral activity. Molecular modeling of the most active compounds showed that they bind to a pocket located in the fingers and thumb domains in BVDV RdRp, which is different from that identified for other non-nucleoside inhibitors (NNIs) such as thiosemicarbazone (TSC). We selected compound 2-[4-(2-phenylquinazolin-4-yl)piperazin-1-yl]ethanol (1.9; EC50 = 1.7 ± 0.4 μM) for further analysis. Compound 1.9 was found to inhibit the in vitro replication of TSC-resistant BVDV variants, which carry the N264D mutation in the RdRp. In addition, 1.9 presented adequate solubility in different media and a high-stability profile in murine and bovine plasma.
Collapse
Affiliation(s)
- Gabriela A Fernández
- Laboratorio de Química Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Eliana F Castro
- Centro de Investigaciones en Ciencias Veterinarias y Agronómicas, Instituto de Virología e Innovaciones Tecnológicas, Instituto Nacional de Tecnología Agropecuaria, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra Virología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rocío A Rosas
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra Virología, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Universidad de Buenos Aires, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Daniela M Fidalgo
- Laboratorio de Química Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Natalia S Adler
- Laboratorio de Química Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Leandro Battini
- Laboratorio de Química Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Maria J España de Marco
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra Virología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Matias Fabiani
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra Virología, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Universidad de Buenos Aires, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ana M Bruno
- Departamento de Química Orgánica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariela Bollini
- Laboratorio de Química Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Lucia V Cavallaro
- Departamento de Microbiología, Inmunología, Biotecnología y Genética, Cátedra Virología, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
17
|
structural characterization of the interaction between the C-terminal domain of the influenza polymerase PA subunit and an optimized small peptide inhibitor. Antiviral Res 2020; 185:104971. [PMID: 33166574 DOI: 10.1016/j.antiviral.2020.104971] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023]
Abstract
Influenza viruses can cause severe respiratory infections in humans, leading to nearly half a million deaths worldwide each year. Improved antiviral drugs are needed to address the threat of development of novel pandemic strains. Current therapeutic interventions target three key proteins in the viral life cycle: neuraminidase, the M2 channel and RNA-dependent-RNA polymerase. Protein-protein interactions between influenza polymerase subunits are potential new targets for drug development. Using a newly developed assay based on AlphaScreen technology, we screened a peptide panel for protein-protein interaction inhibitors to identify a minimal PB1 subunit-derived peptide that retains high inhibition potential and can be further modified. Here, we present an X-ray structure of the resulting decapeptide bound to the C-terminal domain of PA polymerase subunit from pandemic isolate A/California/07/2009 H1N1 at 1.6 Å resolution and discuss its implications for the design of specific, potent influenza polymerase inhibitors.
Collapse
|
18
|
Datir R, Kemp S, El Bouzidi K, Mlchocova P, Goldstein R, Breuer J, Towers GJ, Jolly C, Quiñones-Mateu ME, Dakum PS, Ndembi N, Gupta RK. In Vivo Emergence of a Novel Protease Inhibitor Resistance Signature in HIV-1 Matrix. mBio 2020; 11:e02036-20. [PMID: 33144375 PMCID: PMC7642677 DOI: 10.1128/mbio.02036-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/21/2020] [Indexed: 12/16/2022] Open
Abstract
Protease inhibitors (PIs) are the second- and last-line therapy for the majority of HIV-infected patients worldwide. Only around 20% of individuals who fail PI regimens develop major resistance mutations in protease. We sought to explore the role of mutations in gag-pro genotypic and phenotypic changes in viruses from six Nigerian patients who failed PI-based regimens without known drug resistance-associated protease mutations in order to identify novel determinants of PI resistance. Target enrichment and next-generation sequencing (NGS) with the Illumina MiSeq system were followed by haplotype reconstruction. Full-length Gag-protease gene regions were amplified from baseline (pre-PI) and virologic failure (VF) samples, sequenced, and used to construct gag-pro-pseudotyped viruses. Phylogenetic analysis was performed using maximum-likelihood methods. Susceptibility to lopinavir (LPV) and darunavir (DRV) was measured using a single-cycle replication assay. Western blotting was used to analyze Gag cleavage. In one of six participants (subtype CRF02_AG), we found 4-fold-lower LPV susceptibility in viral clones during failure of second-line treatment. A combination of four mutations (S126del, H127del, T122A, and G123E) in the p17 matrix of baseline virus generated a similar 4-fold decrease in susceptibility to LPV but not darunavir. These four amino acid changes were also able to confer LPV resistance to a subtype B Gag-protease backbone. Western blotting demonstrated significant Gag cleavage differences between sensitive and resistant isolates in the presence of drug. Resistant viruses had around 2-fold-lower infectivity than sensitive clones in the absence of drug. NGS combined with haplotype reconstruction revealed that resistant, less fit clones emerged from a minority population at baseline and thereafter persisted alongside sensitive fitter viruses. We used a multipronged genotypic and phenotypic approach to document emergence and temporal dynamics of a novel protease inhibitor resistance signature in HIV-1 matrix, revealing the interplay between Gag-associated resistance and fitness.
Collapse
Affiliation(s)
| | - Steven Kemp
- University College London, London, United Kingdom
| | | | - Petra Mlchocova
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | - Judy Breuer
- University College London, London, United Kingdom
| | | | - Clare Jolly
- University College London, London, United Kingdom
| | | | - Patrick S Dakum
- Institute for Human Virology, Abuja, Nigeria
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nicaise Ndembi
- Institute for Human Virology, Abuja, Nigeria
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ravindra K Gupta
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Africa Health Research Institute, Durban, South Africa
| |
Collapse
|
19
|
Wang L, Casey MC, Vernekar SKV, Sahani RL, Kankanala J, Kirby KA, Du H, Hachiya A, Zhang H, Tedbury PR, Xie J, Sarafianos SG, Wang Z. Novel HIV-1 capsid-targeting small molecules of the PF74 binding site. Eur J Med Chem 2020; 204:112626. [PMID: 32814250 DOI: 10.1016/j.ejmech.2020.112626] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 10/23/2022]
Abstract
The PF74 binding site in HIV-1 capsid protein (CA) is a compelling antiviral drug target. Although PF74 confers mechanistically distinct antiviral phenotypes by competing against host factors for CA binding, it suffers from prohibitively low metabolic stability. Therefore, there has been increasing interest in designing novel sub-chemotypes of PF74 with similar binding mode and improved metabolic stability. We report herein our efforts to explore the inter-domain interacting indole moiety for designing novel CA-targeting small molecules. Our design includes simple substitution on the indole ring, and more importantly, novel sub-chemotypes with the indole moiety replaced with a few less electron-rich rings. All 56 novel analogs were synthesized and evaluated for antiviral activity, cytotoxicity, and impact on CA hexamer stability. Selected analogs were tested for metabolic stability in liver microsomes. Molecular modeling was performed to verify compound binding to the PF74 site. In the end, 5-hydroxyindole analogs (8,9 and 12) showed improved potency (up to 20-fold) over PF74. Of the novel sub-chemotypes, α- and β-naphthyl analogs (33 and 27) exhibited sub micromolar antiviral potencies comparable to that of PF74. Interestingly, although only moderately inhibiting HIV-1 (single-digit micromolar EC50s), analogs of the 2-indolone sub-chemotype consistently lowered the melting point (Tm) of CA hexamers, some with improved metabolic stability over PF74.
Collapse
Affiliation(s)
- Lei Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Mary C Casey
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center, Columbia, MO, 65211, USA
| | - Sanjeev Kumar V Vernekar
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Rajkumar Lalji Sahani
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Jayakanth Kankanala
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Karen A Kirby
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Haijuan Du
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Atsuko Hachiya
- Clinical Research Center, Nagoya Medical Center, National Hospital Organization, Nagoya, Aichi, 460-0001, Japan
| | - Huanchun Zhang
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Philip R Tedbury
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jiashu Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Stefan G Sarafianos
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zhengqiang Wang
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
20
|
Zhang DW, Luo RH, Xu L, Yang LM, Xu XS, Zheng YT, Luo H. Natural-product-library-based screening for discovery of capsid C-terminal domain targeted HIV-1 inhibitors. Int J Antimicrob Agents 2020; 55:105926. [PMID: 32092396 DOI: 10.1016/j.ijantimicag.2020.105926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/10/2020] [Accepted: 02/14/2020] [Indexed: 01/31/2023]
Abstract
Antiretroviral therapy (ART) can effectively suppress replication of human immunodeficiency virus type 1 (HIV-1) and limit disease progression. However, ART is unable to eradicate the virus, and the requirement for lifelong treatment may have side effects and may lead to the development of resistance. New approaches to prevent and treat HIV-1 infection should therefore be developed. HIV-1 capsid (CA) protein is an unexploited but attractive target for antiviral drug development. The hydrophobic cavity of the C-terminal domain of CA (CA CTD) has been validated as a potential target for antiviral drugs. Binding of compounds to this conserved non-polar groove in CA CTD allosterically disrupts the CA assembly. This study screened 2080 natural products to identify potential antiviral agents for further development to combat HIV-1 infection. From the primary screen at a fixed concentration of 50 µM, 16 compounds were found to be effective against this target. Six compounds observed in the primary screen were confirmed in dose-response experiments, and were tested against HIV-1-induced cytopathic effects. Two compounds were found to inhibit HIV-1 replication, and the most active compound - rubranol - inhibited viral replication at a moderate micromolar concentration (EC50 = 15.85 μM). The binding modes of rubranol and hirsutanonol to CA CTD were analysed by molecular docking, providing insight for the design of drugs targeting HIV-1 CA. This study reports, for the first time, identification of natural products that showed potential as anti-HIV-1 agents by targeting the conserved hydrophobic cavity of HIV-1 CA CTD.
Collapse
Affiliation(s)
- Da-Wei Zhang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Rong-Hua Luo
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, the National Kunming High Level Biosafety Research Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Liu-Meng Yang
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, the National Kunming High Level Biosafety Research Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiao-Shuang Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Yong-Tang Zheng
- Key Laboratory of Bioactive Peptides of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, the National Kunming High Level Biosafety Research Center for Nonhuman Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
21
|
Understanding Flavivirus Capsid Protein Functions: The Tip of the Iceberg. Pathogens 2020; 9:pathogens9010042. [PMID: 31948047 PMCID: PMC7168633 DOI: 10.3390/pathogens9010042] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/01/2020] [Accepted: 01/03/2020] [Indexed: 12/18/2022] Open
Abstract
Flaviviruses are enveloped positive-sense single-stranded RNA arboviruses, infectious to humans and many other animals and are transmitted primarily via tick or mosquito vectors. Capsid is the primary structural protein to interact with viral genome within virus particles and is therefore necessary for efficient packaging. However, in cells, capsid interacts with many proteins and nucleic acids and we are only beginning to understand the broad range of functions of flaviviral capsids. It is known that capsid dimers interact with the membrane of lipid droplets, aiding in both viral packaging and storage of capsid prior to packaging. However, capsid dimers can bind a range of nucleic acid templates in vitro, and likely interact with a range of targets during the flavivirus lifecycle. Capsid may interact with host RNAs, resulting in altered RNA splicing and RNA transcription. Capsid may also bind short interfering-RNAs and has been proposed to sequester these species to protect flaviviruses from the invertebrate siRNA pathways. Capsid can also be found in the nucleolus, where it wreaks havoc on ribosome biogenesis. Here we review flavivirus capsid structure, nucleic acid interactions and how these give rise to multiple functions. We also discuss how these features might be exploited either in the design of effective antivirals or novel vaccine strategies.
Collapse
|
22
|
Xing H, Chen J, Shi Y, Huang T, Hai L, Wu Y. Synthesis of 4-ethenyl quinazolines via rhodium(iii)-catalyzed [5 + 1] annulation reaction of N-arylamidines with cyclopropenones. Org Chem Front 2020. [DOI: 10.1039/c9qo01372e] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An unprecedented synthesis of 4-ethenyl quinazolines/2-benzoyl quinazolines via Rh(iii)-catalyzed C–H annulation reaction was reported.
Collapse
Affiliation(s)
- Huimin Xing
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry
- Department of Medicinal Chemistry
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Jian Chen
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry
- Department of Medicinal Chemistry
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Yuesen Shi
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry
- Department of Medicinal Chemistry
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Tianle Huang
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry
- Department of Medicinal Chemistry
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Li Hai
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry
- Department of Medicinal Chemistry
- West China School of Pharmacy
- Sichuan University
- Chengdu
| | - Yong Wu
- Key Laboratory of Drug Targeting and Drug Delivery System of Education Ministry
- Department of Medicinal Chemistry
- West China School of Pharmacy
- Sichuan University
- Chengdu
| |
Collapse
|
23
|
Chan CK, Lai CY, Wang CC. TMSOTf-catalyzed synthesis of substituted quinazolines using hexamethyldisilazane as a nitrogen source under neat and microwave irradiation conditions. Org Biomol Chem 2020; 18:7201-7212. [DOI: 10.1039/d0ob01507e] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
An efficient synthetic route for the synthesis of substituted quinazolines under neat, metal-free and microwave irradiation conditions has been developed by using TMSOTf as an acid catalyst and HMDS as a nitrogen source.
Collapse
Affiliation(s)
| | - Chien-Yu Lai
- Institute of Chemistry
- Academia Sinica
- Taipei 115
- Taiwan
| | | |
Collapse
|
24
|
Yuan S, Yu B, Liu H. “On‐Water” Palladium‐Catalyzed Tandem Cyclization Reaction for the Synthesis of Biologically Relevant 4‐Arylquinazolines. Chemistry 2019; 25:13109-13113. [DOI: 10.1002/chem.201903464] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/20/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Shuo Yuan
- School of Pharmaceutical sciencesZhengzhou University Zhengzhou 450001 P. R. China
| | - Bin Yu
- School of Pharmaceutical sciencesZhengzhou University Zhengzhou 450001 P. R. China
| | - Hong‐Min Liu
- School of Pharmaceutical sciencesZhengzhou University Zhengzhou 450001 P. R. China
| |
Collapse
|
25
|
Hachiya I, Nagoshi S, Shimizu M. Titanium Tetraiodide/Trimethylsilyl Iodide Synergistically Induced Cyclization of N-(2-Cyanophenyl)benzamides into 2-Aryl-4-iodoquinazolines. ACS OMEGA 2019; 4:10463-10473. [PMID: 31460142 PMCID: PMC6648542 DOI: 10.1021/acsomega.9b01199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/05/2019] [Indexed: 06/10/2023]
Abstract
Synthesis of 2-aryl-4-iodoquinazolines is developed using titanium tetraiodide/trimethylsilyl iodide synergistically induced cyclization of N-(2-cyanophenyl)benzamides. The cyclization reactions proceeded to give the 2-aryl-4-iodoquinazolines in moderate to high yields. Remarkable synergetic effect of titanium tetraiodide and trimethylsilyl iodide was observed to promote the cyclization. The method was applied to the formal synthesis of a potent analgesic agent.
Collapse
Affiliation(s)
- Iwao Hachiya
- Department
of Chemistry for Materials, Graduate School of Engineering, Mie University, Tsu, Mie 514-8507, Japan
| | - Shintaro Nagoshi
- Department
of Chemistry for Materials, Graduate School of Engineering, Mie University, Tsu, Mie 514-8507, Japan
| | - Makoto Shimizu
- Department
of Chemistry for Materials, Graduate School of Engineering, Mie University, Tsu, Mie 514-8507, Japan
- School
of Energy Science and Engineering, Nanjing
Tech University, Nanjing 211816, Jiangsu Province, China
| |
Collapse
|
26
|
Zhang DW, Luo RH, Xu L, Yang LM, Xu XS, Bedwell GJ, Engelman AN, Zheng YT, Chang S. A HTRF based competitive binding assay for screening specific inhibitors of HIV-1 capsid assembly targeting the C-Terminal domain of capsid. Antiviral Res 2019; 169:104544. [PMID: 31254557 DOI: 10.1016/j.antiviral.2019.104544] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 06/13/2019] [Accepted: 06/20/2019] [Indexed: 02/02/2023]
Abstract
Due to its multifaceted essential roles in virus replication and extreme genetic fragility, the human immunodeficiency virus type 1 (HIV-1) capsid (CA) protein is a valued therapeutic target. However, CA is as yet unexploited clinically, as there are no antiviral agents that target it currently on the market. To facilitate the identification of potential HIV-1 CA inhibitors, we established a homogeneous time-resolved fluorescence (HTRF) assay to screen for small molecules that target a biologically active and specific binding pocket in the C-terminal domain of HIV-1 CA (CA CTD). The assay, which is based on competition of small molecules for the binding of a known CA inhibitor (CAI) to the CA CTD, exhibited a signal-to-background ratio (S/B) > 10 and a Z' value > 0.9. In a pilot screen of three kinase inhibitor libraries containing 464 compounds, we identified one compound, TX-1918, as a low micromolecular inhibitor of the HIV-1 CA CTD-CAI interaction (IC50 = 3.81 μM) that also inhibited viral replication at moderate micromolar concentration (EC50 = 15.16 μM) and inhibited CA assembly in vitro. Based on the structure of TX-1918, an additional compound with an antiviral EC50 of 6.57 μM and cellular cytotoxicity CC50 of 102.55 μM was obtained from a compound similarity search. Thus, the HTRF-based assay has properties that are suitable for screening large compound libraries to identify novel anti-HIV-1 inhibitors targeting the CA CTD.
Collapse
Affiliation(s)
- Da-Wei Zhang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, 213001, China; Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Rong-Hua Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, The National Kunming High Level Biosafety Research Center for Nonhuman Primate, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, 213001, China
| | - Liu-Meng Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, The National Kunming High Level Biosafety Research Center for Nonhuman Primate, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Xiao-Shuang Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, 213001, China
| | - Gregory J Bedwell
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Alan N Engelman
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, The National Kunming High Level Biosafety Research Center for Nonhuman Primate, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.
| | - Shan Chang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, 213001, China.
| |
Collapse
|
27
|
Novikova M, Zhang Y, Freed EO, Peng K. Multiple Roles of HIV-1 Capsid during the Virus Replication Cycle. Virol Sin 2019; 34:119-134. [PMID: 31028522 PMCID: PMC6513821 DOI: 10.1007/s12250-019-00095-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/16/2019] [Indexed: 11/29/2022] Open
Abstract
Human immunodeficiency virus-1 capsid (HIV-1 CA) is involved in different stages of the viral replication cycle. During virion assembly, CA drives the formation of the hexameric lattice in immature viral particles, while in mature virions CA monomers assemble in cone-shaped cores surrounding the viral RNA genome and associated proteins. In addition to its functions in late stages of the viral replication cycle, CA plays key roles in a number of processes during early phases of HIV-1 infection including trafficking, uncoating, recognition by host cellular proteins and nuclear import of the viral pre-integration complex. As a result of efficient cooperation of CA with other viral and cellular proteins, integration of the viral genetic material into the host genome, which is an essential step for productive viral infection, successfully occurs. In this review, we will summarize available data on CA functions in HIV-1 replication, describing in detail its roles in late and early phases of the viral replication cycle.
Collapse
Affiliation(s)
- Mariia Novikova
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Yulan Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Eric O Freed
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA.
| | - Ke Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
28
|
Iodine/potassium iodide catalyst for the synthesis of trifluoromethylated quinazolines via intramolecular cyclization of 2,2,2-trifluoro-N-benzyl-N'-arylacetimidamides. Mol Divers 2019; 24:131-139. [PMID: 30868331 DOI: 10.1007/s11030-019-09933-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 02/24/2019] [Indexed: 01/26/2023]
Abstract
An efficient and simple protocol for the synthesis of trifluoromethylated quinazolines has been described by I2-/KI-promoted oxidative C(sp3)-C(sp2) bond under the optimal oxidative cyclization reaction conditions. The required 2,2,2-trifluoro-N-benzyl-N'-arylacetimidamides are readily prepared from the corresponding acetimidoyl chlorides and benzylamines under a nucleophilic substitution reaction in the form of in situ. The merits of this protocol are the use of inexpensive molecular iodine, metal-free oxidative coupling and good to excellent yields.
Collapse
|
29
|
Tran CB, To TA, Vo YH, Do TTL, Nguyen QTP, Nguyen AT, Nguyen TT, Phan NTS. A Green Synthesis of N
,N
- and N
,O
-Heterocycles Using a Recyclable Catalyst in a Bio-Based Solvent. ChemistrySelect 2019. [DOI: 10.1002/slct.201804027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Chau B. Tran
- Faculty of Chemical Engineering; HCMC University of Technology, VNU-HCM; 268 Ly Thuong Kiet, District 10 Ho Chi Minh City Vietnam
| | - Tuong A. To
- Faculty of Chemical Engineering; HCMC University of Technology, VNU-HCM; 268 Ly Thuong Kiet, District 10 Ho Chi Minh City Vietnam
| | - Yen H. Vo
- Faculty of Chemical Engineering; HCMC University of Technology, VNU-HCM; 268 Ly Thuong Kiet, District 10 Ho Chi Minh City Vietnam
| | - Tuong T. L. Do
- Faculty of Chemical Engineering; HCMC University of Technology, VNU-HCM; 268 Ly Thuong Kiet, District 10 Ho Chi Minh City Vietnam
| | - Qui T. P. Nguyen
- Faculty of Chemical Engineering; HCMC University of Technology, VNU-HCM; 268 Ly Thuong Kiet, District 10 Ho Chi Minh City Vietnam
| | - Anh T. Nguyen
- Faculty of Chemical Engineering; HCMC University of Technology, VNU-HCM; 268 Ly Thuong Kiet, District 10 Ho Chi Minh City Vietnam
| | - Tung T. Nguyen
- Faculty of Chemical Engineering; HCMC University of Technology, VNU-HCM; 268 Ly Thuong Kiet, District 10 Ho Chi Minh City Vietnam
| | - Nam T. S. Phan
- Faculty of Chemical Engineering; HCMC University of Technology, VNU-HCM; 268 Ly Thuong Kiet, District 10 Ho Chi Minh City Vietnam
| |
Collapse
|
30
|
Satish G, Polu A, Kota L, Ilangovan A. Copper-catalyzed oxidative amination of methanol to access quinazolines. Org Biomol Chem 2019; 17:4774-4782. [DOI: 10.1039/c9ob00392d] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A novel method for the copper-catalyzed oxidative amination of 2′-aminoarylketones with methanol as a C1 carbon source and ammonium acetate as an amine source to construct quinazolines was established in a one-pot manner.
Collapse
Affiliation(s)
| | - Ashok Polu
- School of Chemistry
- Bharathidasan University
- Tiruchirappalli
- India
| | - Laxman Kota
- School of Chemistry
- Bharathidasan University
- Tiruchirappalli
- India
| | | |
Collapse
|
31
|
Prakash R, Bora BR, Boruah RC, Gogoi S. Ru(II)-Catalyzed C–H Activation and Annulation Reaction via Carbon–Carbon Triple Bond Cleavage. Org Lett 2018; 20:2297-2300. [DOI: 10.1021/acs.orglett.8b00643] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Rashmi Prakash
- Applied Organic Chemistry, Chemical Sciences & Technology Division, CSIR-North East Institute of Science and Technology, AcSIR, Jorhat 785006, India
| | - Bidisha R. Bora
- Applied Organic Chemistry, Chemical Sciences & Technology Division, CSIR-North East Institute of Science and Technology, AcSIR, Jorhat 785006, India
| | - Romesh C. Boruah
- Applied Organic Chemistry, Chemical Sciences & Technology Division, CSIR-North East Institute of Science and Technology, AcSIR, Jorhat 785006, India
| | - Sanjib Gogoi
- Applied Organic Chemistry, Chemical Sciences & Technology Division, CSIR-North East Institute of Science and Technology, AcSIR, Jorhat 785006, India
| |
Collapse
|
32
|
Zhu J, Shao Y, Hu K, Qi L, Cheng T, Chen J. Pd-Catalyzed tandem reaction of N-(2-cyanoaryl)benzamides with arylboronic acids: synthesis of quinazolines. Org Biomol Chem 2018; 16:8596-8603. [DOI: 10.1039/c8ob02421a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A Pd-catalyzed tandem reaction of N-(2-cyanoaryl)benzamides with arylboronic acids for the synthesis of quinazolines has been reported.
Collapse
Affiliation(s)
- Jianghe Zhu
- College of Chemistry & Materials Engineering
- Wenzhou University
- Wenzhou 325035
- P. R. China
| | - Yinlin Shao
- College of Chemistry & Materials Engineering
- Wenzhou University
- Wenzhou 325035
- P. R. China
| | - Kun Hu
- College of Chemistry & Materials Engineering
- Wenzhou University
- Wenzhou 325035
- P. R. China
| | - Linjun Qi
- College of Chemistry & Materials Engineering
- Wenzhou University
- Wenzhou 325035
- P. R. China
| | - Tianxing Cheng
- College of Chemistry & Materials Engineering
- Wenzhou University
- Wenzhou 325035
- P. R. China
| | - Jiuxi Chen
- College of Chemistry & Materials Engineering
- Wenzhou University
- Wenzhou 325035
- P. R. China
| |
Collapse
|
33
|
Ramanathan M, Liu ST. Preparation of Quinazolines via a 2+2+2 Annulation from Aryldiazonium Salts and Nitriles. J Org Chem 2017; 82:8290-8295. [DOI: 10.1021/acs.joc.7b01325] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Mani Ramanathan
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Shiuh-Tzung Liu
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
34
|
Ma J, Wan Y, Hong C, Li M, Hu X, Mo W, Hu B, Sun N, Jin L, Shen Z. ABNO-Catalyzed Aerobic Oxidative Synthesis of 2-Substituted 4H
-3,1-Benzoxazines and Quinazolines. European J Org Chem 2017. [DOI: 10.1002/ejoc.201700384] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jiaqi Ma
- College of Chemical Engineering; Zhejiang University of Technology; 310014 Hangzhou People's Republic of China
| | - Yan Wan
- College of Chemical Engineering; Zhejiang University of Technology; 310014 Hangzhou People's Republic of China
| | - Chao Hong
- College of Chemical Engineering; Zhejiang University of Technology; 310014 Hangzhou People's Republic of China
| | - Meichao Li
- College of Chemical Engineering; Zhejiang University of Technology; 310014 Hangzhou People's Republic of China
| | - Xinquan Hu
- College of Chemical Engineering; Zhejiang University of Technology; 310014 Hangzhou People's Republic of China
| | - Weimin Mo
- College of Chemical Engineering; Zhejiang University of Technology; 310014 Hangzhou People's Republic of China
| | - Baoxiang Hu
- College of Chemical Engineering; Zhejiang University of Technology; 310014 Hangzhou People's Republic of China
| | - Nan Sun
- College of Chemical Engineering; Zhejiang University of Technology; 310014 Hangzhou People's Republic of China
| | - Liqun Jin
- College of Chemical Engineering; Zhejiang University of Technology; 310014 Hangzhou People's Republic of China
| | - Zhenlu Shen
- College of Chemical Engineering; Zhejiang University of Technology; 310014 Hangzhou People's Republic of China
| |
Collapse
|
35
|
Lv Z, Wang B, Hu Z, Zhou Y, Yu W, Chang J. Synthesis of Quinazolines from N,N′-Disubstituted Amidines via I2/KI-Mediated Oxidative C–C Bond Formation. J Org Chem 2016; 81:9924-9930. [DOI: 10.1021/acs.joc.6b02100] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Zhigang Lv
- College
of Chemistry and
Molecular Engineering, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Bingnan Wang
- College
of Chemistry and
Molecular Engineering, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Zhiyuan Hu
- College
of Chemistry and
Molecular Engineering, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Yiming Zhou
- College
of Chemistry and
Molecular Engineering, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Wenquan Yu
- College
of Chemistry and
Molecular Engineering, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Junbiao Chang
- College
of Chemistry and
Molecular Engineering, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| |
Collapse
|
36
|
Doležal M, Hadravová R, Kožíšek M, Bednárová L, Langerová H, Ruml T, Rumlová M. Functional and Structural Characterization of Novel Type of Linker Connecting Capsid and Nucleocapsid Protein Domains in Murine Leukemia Virus. J Biol Chem 2016; 291:20630-42. [PMID: 27514744 DOI: 10.1074/jbc.m116.746461] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Indexed: 12/24/2022] Open
Abstract
The assembly of immature retroviral particles is initiated in the cytoplasm by the binding of the structural polyprotein precursor Gag with viral genomic RNA. The protein interactions necessary for assembly are mediated predominantly by the capsid (CA) and nucleocapsid (NC) domains, which have conserved structures. In contrast, the structural arrangement of the CA-NC connecting region differs between retroviral species. In HIV-1 and Rous sarcoma virus, this region forms a rod-like structure that separates the CA and NC domains, whereas in Mason-Pfizer monkey virus, this region is densely packed, thus holding the CA and NC domains in close proximity. Interestingly, the sequence connecting the CA and NC domains in gammaretroviruses, such as murine leukemia virus (MLV), is unique. The sequence is called a charged assembly helix (CAH) due to a high number of positively and negatively charged residues. Although both computational and deletion analyses suggested that the MLV CAH forms a helical conformation, no structural or biochemical data supporting this hypothesis have been published. Using an in vitro assembly assay, alanine scanning mutagenesis, and biophysical techniques (circular dichroism, NMR, microcalorimetry, and electrophoretic mobility shift assay), we have characterized the structure and function of the MLV CAH. We provide experimental evidence that the MLV CAH belongs to a group of charged, E(R/K)-rich, single α-helices. This is the first single α-helix motif identified in viral proteins.
Collapse
Affiliation(s)
- Michal Doležal
- From the Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 16610 Prague 6
| | - Romana Hadravová
- From the Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 16610 Prague 6
| | - Milan Kožíšek
- From the Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 16610 Prague 6
| | - Lucie Bednárová
- From the Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 16610 Prague 6
| | - Hana Langerová
- the Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague 6, Technická 3, 166 28 Prague, and
| | - Tomáš Ruml
- the Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague 6, Technická 3, 166 28 Prague, and
| | - Michaela Rumlová
- From the Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 2, 16610 Prague 6, the Department of Biotechnology, University of Chemistry and Technology, Technická 5, 166 28 Prague, Czech Republic
| |
Collapse
|
37
|
Kožíšek M, Štěpánek O, Parkan K, Berenguer Albiñana C, Pávová M, Weber J, Krӓusslich HG, Konvalinka J, Machara A. Synthesis and evaluation of 2-pyridinylpyrimidines as inhibitors of HIV-1 structural protein assembly. Bioorg Med Chem Lett 2016; 26:3487-90. [PMID: 27353536 DOI: 10.1016/j.bmcl.2016.06.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 11/28/2022]
Abstract
In an effort to identify an HIV-1 capsid assembly inhibitor with improved solubility and potency, we synthesized two series of pyrimidine analogues based on our earlier lead compound N-(4-(ethoxycarbonyl)phenyl)-2-(pyridine-4-yl)quinazoline-4-amine. In vitro binding experiments showed that our series of 2-pyridine-4-ylpyrimidines had IC50 values higher than 28μM. Our series of 2-pyridine-3-ylpyrimidines exhibited IC50 values ranging from 3 to 60μM. The congeners with a fluoro substituent introduced at the 4-N-phenyl moiety, along with a methyl at C-6, represent potent HIV capsid assembly inhibitors binding to the C-terminal domain of the capsid protein.
Collapse
Affiliation(s)
- Milan Kožíšek
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Gilead Sciences and IOCB Research Center, Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | - Ondřej Štěpánek
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Gilead Sciences and IOCB Research Center, Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | - Kamil Parkan
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Gilead Sciences and IOCB Research Center, Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | - Carlos Berenguer Albiñana
- Department of Organic Chemistry, Faculty of Science, Charles University, Hlavova 8, 128 43 Prague 2, Czech Republic
| | - Marcela Pávová
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Gilead Sciences and IOCB Research Center, Flemingovo n. 2, 166 10 Prague 6, Czech Republic; Department of Infectious Diseases, Virology, University Hospital Heidelberg, Im Neuenheimer Feld 324, 691 20 Heidelberg, Germany
| | - Jan Weber
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Gilead Sciences and IOCB Research Center, Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | - Hans-Georg Krӓusslich
- Department of Infectious Diseases, Virology, University Hospital Heidelberg, Im Neuenheimer Feld 324, 691 20 Heidelberg, Germany
| | - Jan Konvalinka
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Gilead Sciences and IOCB Research Center, Flemingovo n. 2, 166 10 Prague 6, Czech Republic; Department of Biochemistry, Faculty of Science, Charles University, Hlavova 8, 128 43 Prague 2, Czech Republic
| | - Aleš Machara
- Department of Organic Chemistry, Faculty of Science, Charles University, Hlavova 8, 128 43 Prague 2, Czech Republic.
| |
Collapse
|