1
|
Zhao L, Shen G, Luo J, Zhang Y, Yao Y, Cui L, Yang B. Effects of Steroidal Compounds on Viruses. Viral Immunol 2025; 38:44-52. [PMID: 39912855 DOI: 10.1089/vim.2024.0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025] Open
Abstract
Viral infections are ubiquitous, and their prevention and treatment have become a great challenge. Steroids have different biological activities, including antiviral activity, which is related to steroid structural diversity. With the intensive study of steroids, it has been found that steroids can interfere with almost any step of the viral life cycle to exert antiviral activity. In this article, we review the antiviral activity and mechanism of action of steroids and their derivatives against a range of human viruses and conclude that natural steroids and their derivatives are very promising antiviral drug candidates that deserve further study to elucidate their pharmacological potential.
Collapse
Affiliation(s)
- Li Zhao
- College of Pharmacy, Harbin University of Commerce, Harbin, China
- Heilongjiang Provincial Key Laboratory of Drug Prevention and Treatment for Senile Diseases, Harbin, China
| | - Guanghuan Shen
- College of Pharmacy, Harbin University of Commerce, Harbin, China
- Heilongjiang Provincial Key Laboratory of Drug Prevention and Treatment for Senile Diseases, Harbin, China
| | - Jianghan Luo
- College of Pharmacy, Harbin University of Commerce, Harbin, China
- Heilongjiang Provincial Key Laboratory of Drug Prevention and Treatment for Senile Diseases, Harbin, China
| | - Yingyu Zhang
- College of Pharmacy, Harbin University of Commerce, Harbin, China
- Heilongjiang Provincial Key Laboratory of Drug Prevention and Treatment for Senile Diseases, Harbin, China
| | - Ying Yao
- College of Pharmacy, Harbin University of Commerce, Harbin, China
- Heilongjiang Provincial Key Laboratory of Drug Prevention and Treatment for Senile Diseases, Harbin, China
| | - Linlin Cui
- College of Pharmacy, Harbin University of Commerce, Harbin, China
- Heilongjiang Provincial Key Laboratory of Drug Prevention and Treatment for Senile Diseases, Harbin, China
| | - Bo Yang
- College of Pharmacy, Harbin University of Commerce, Harbin, China
- Heilongjiang Provincial Key Laboratory of Drug Prevention and Treatment for Senile Diseases, Harbin, China
| |
Collapse
|
2
|
Shu J, Cao K, Fei C, Dai H, Li Y, Cao Y, Zhou T, Yu M, Xia Z, An M, Wu Y. Antiviral Mechanisms of Anisomycin Produced by Streptomyces albulus SN40 on Potato Virus Y. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:3506-3519. [PMID: 38346922 DOI: 10.1021/acs.jafc.3c07732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Microbial secondary metabolites produced by Streptomyces have diverse application prospects in the control of plant diseases. Herein, the fermentation filtrate of Streptomyces SN40 effectively inhibited the infection of tobacco mosaic virus (TMV) in Nicotiana glutinosa and systemic infection of potato virus Y (PVY) in Nicotiana benthamiana. Additionally, metabolomic analysis indicated that anisomycin (C14H19NO4) and trans-3-indoleacrylic acid (C11H9NO2) were highly abundant in the crude extract and that anisomycin effectively suppressed the infection of TMV as well as PVY. Subsequently, transcriptomic analysis was conducted to elucidate its mechanisms on the induction of host defense responses. Furthermore, the results of molecular docking suggested that anisomycin can potentially bind with the helicase domain (Hel) of TMV replicase, TMV coat protein (CP), and PVY helper component proteinase (HC-Pro). This study demonstrates new functions of anisomycin in virus inhibition and provides important theoretical significance for the development of new biological pesticides to control diverse plant viruses.
Collapse
Affiliation(s)
- Jing Shu
- Liaoning Key Laboratory of Plant Pathology, College of Plant Protection, Shenyang Agricultural University, No. 120 Dongling, Shenyang 110866, China
| | - Kexin Cao
- Liaoning Key Laboratory of Plant Pathology, College of Plant Protection, Shenyang Agricultural University, No. 120 Dongling, Shenyang 110866, China
| | - Chuanjiang Fei
- Guizhou Qianxinan Prefectural Tobacco Company, Xingyi 562400, China
| | - Hui Dai
- Guizhou Qianxinan Prefectural Tobacco Company, Xingyi 562400, China
| | - Yuhang Li
- Guizhou Qianxinan Prefectural Tobacco Company, Xingyi 562400, China
| | - Yi Cao
- Guizhou Academy of Tobacco Science, Guiyang 550081, China
| | - Tao Zhou
- Liaoning Key Laboratory of Plant Pathology, College of Plant Protection, Shenyang Agricultural University, No. 120 Dongling, Shenyang 110866, China
| | - Miao Yu
- Liaoning Key Laboratory of Plant Pathology, College of Plant Protection, Shenyang Agricultural University, No. 120 Dongling, Shenyang 110866, China
| | - Zihao Xia
- Liaoning Key Laboratory of Plant Pathology, College of Plant Protection, Shenyang Agricultural University, No. 120 Dongling, Shenyang 110866, China
| | - Mengnan An
- Liaoning Key Laboratory of Plant Pathology, College of Plant Protection, Shenyang Agricultural University, No. 120 Dongling, Shenyang 110866, China
| | - Yuanhua Wu
- Liaoning Key Laboratory of Plant Pathology, College of Plant Protection, Shenyang Agricultural University, No. 120 Dongling, Shenyang 110866, China
| |
Collapse
|
3
|
Nguyen AMT, Shalev-Benami M, Rosa-Teijeiro C, Ibarra-Meneses AV, Yonath A, Bashan A, Jaffe CL, Olivier M, Fernandez-Prada C, Lubell WD. Systematic Exploration of Functional Group Relevance for Anti-Leishmanial Activity of Anisomycin. Biomedicines 2023; 11:2541. [PMID: 37760981 PMCID: PMC10526209 DOI: 10.3390/biomedicines11092541] [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: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Assessment of structure-activity relationships for anti-protozoan activity revealed a strategy for preparing potent anisomycin derivatives with reduced host toxicity. Thirteen anisomycin analogs were synthesized by modifying the alcohol, amine, and aromatic functional groups. Examination of anti-protozoal activity against various strains of Leishmania and cytotoxicity against leucocytes with comparison against the parent natural product demonstrated typical losses of activity with modifications of the alcohol, amine, and aromatic meta-positions. On the other hand, the para-phenol moiety of anisomycin proved an effective location for introducing substituents without significant loss of anti-protozoan potency. An entry point for differentiating activity against Leishmania versus host has been uncovered by this systematic study.
Collapse
Affiliation(s)
| | - Moran Shalev-Benami
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel; (M.S.-B.); (A.Y.); (A.B.)
| | - Chloé Rosa-Teijeiro
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (C.R.-T.); (A.V.I.-M.); (C.F.-P.)
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, Université de Montréal, Montreal, QC J2S 2M2, Canada
| | - Ana Victoria Ibarra-Meneses
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (C.R.-T.); (A.V.I.-M.); (C.F.-P.)
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, Université de Montréal, Montreal, QC J2S 2M2, Canada
| | - Ada Yonath
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel; (M.S.-B.); (A.Y.); (A.B.)
| | - Anat Bashan
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel; (M.S.-B.); (A.Y.); (A.B.)
| | - Charles L. Jaffe
- Department of Microbiology & Molecular Genetics, Kuvin Center for the Study of Tropical & Infectious Diseases, Institute for Medical Research (IMRIC), Hadassah Hebrew University Medical Center, Jerusalem 9112102, Israel;
| | - Martin Olivier
- Departments of Medicine, and of Microbiology and Immunology, Faculty of Medicine, McGill University, Montreal, QC H3A 2B4, Canada;
- The Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC H4A 3J1, Canada
| | - Christopher Fernandez-Prada
- Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada; (C.R.-T.); (A.V.I.-M.); (C.F.-P.)
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, Université de Montréal, Montreal, QC J2S 2M2, Canada
| | - William D. Lubell
- Department of Chemistry, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
4
|
Srivastava KS, Jeswani V, Pal N, Bohra B, Vishwakarma V, Bapat AA, Patnaik YP, Khanna N, Shukla R. Japanese Encephalitis Virus: An Update on the Potential Antivirals and Vaccines. Vaccines (Basel) 2023; 11:vaccines11040742. [PMID: 37112654 PMCID: PMC10146181 DOI: 10.3390/vaccines11040742] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/17/2023] [Accepted: 02/24/2023] [Indexed: 03/29/2023] Open
Abstract
Japanese encephalitis virus (JEV) is the causal agent behind Japanese encephalitis (JE), a potentially severe brain infection that spreads through mosquito bites. JE is predominant over the Asia-Pacific Region and has the potential to spread globally with a higher rate of morbidity and mortality. Efforts have been made to identify and select various target molecules essential in JEV’s progression, but until now, no licensed anti-JEV drug has been available. From a prophylactic point of view, a few licensed JE vaccines are available, but various factors, viz., the high cost and different side effects imposed by them, has narrowed their global use. With an average occurrence of >67,000 cases of JE annually, there is an urgent need to find a suitable antiviral drug to treat patients at the acute phase, as presently only supportive care is available to mitigate infection. This systematic review highlights the current status of efforts put in to develop antivirals against JE and the available vaccines, along with their effectiveness. It also summarizes epidemiology, structure, pathogenesis, and potential drug targets that can be explored to develop a new range of anti-JEV drugs to combat JEV infection globally.
Collapse
|
5
|
Molecular Mechanism and Role of Japanese Encephalitis Virus Infection in Central Nervous System-Mediated Diseases. Viruses 2022; 14:v14122686. [PMID: 36560690 PMCID: PMC9781168 DOI: 10.3390/v14122686] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
The Japanese encephalitis virus (JEV) is the most common cause of neurodegenerative disease in Southeast Asia and the Western Pacific region; approximately 1.15 billion people are at risk, and thousands suffer from permanent neurological disorders across Asian countries, with 10-15 thousand people dying each year. JEV crosses the blood-brain barrier (BBB) and forms a complex with receptors on the surface of neurons. GRP78, Src, TLR7, caveolin-1, and dopamine receptor D2 are involved in JEV binding and entry into the neurons, and these receptors also play a role in carcinogenic activity in cells. JEV binds to GRP78, a member of the HSP70 overexpressed on malignant cells to enter neurons, indicating a higher chance of JEV infection in cancer patients. However, JEV enters human brain microvascular endothelial cells via an endocytic pathway mediated by caveolae and the ezrin protein and also targets dopamine-rich areas for infection of the midbrain via altering dopamine levels. In addition, JEV complexed with CLEC5A receptor of macrophage cells is involved in the breakdown of the BBB and central nervous system (CNS) inflammation. CLEC5A-mediated infection is also responsible for the influx of cytokines into the CNS. In this review, we discuss the neuronal and macrophage surface receptors involved in neuronal death.
Collapse
|
6
|
Yadav P, El-Kafrawy SA, El-Day MM, Alghafari WT, Faizo AA, Jha SK, Dwivedi VD, Azhar EI. Discovery of Small Molecules from Echinacea angustifolia Targeting RNA-Dependent RNA Polymerase of Japanese Encephalitis Virus. Life (Basel) 2022; 12:life12070952. [PMID: 35888042 PMCID: PMC9324244 DOI: 10.3390/life12070952] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 05/28/2023] Open
Abstract
The Japanese encephalitis virus (JEV), a mosquito-borne flavivirus that causes viral encephalitis leading to neural damage, is a major threat in most Asian countries. The RNA-dependent RNA polymerase (RdRp) present in the viral genome is the key component for genome replication, making it an attractive target for antiviral drug development. In this study, the natural products from Echinacea angustifolia were retrieved for structure-based virtual screening against JEV-RdRp. The top six compounds (Echinacoside, Echinacin, Rutin, Cynaroside, Quercetagetin 7-glucoside, and Kaempferol-3-glucoside) were obtained based on the highest negative docking score, ADMET (absorption, distribution, metabolism, excretion, and toxicity), and molecular interaction. The computational analysis of these selected compounds against the co-crystallized ligands, i.e., ATP and GTP, were performed. Further, 100 ns molecular dynamic simulation and post-free binding energy calculation of all the selected compounds complexed with JEV-RdRP were performed to check the stability of the complexes. The obtained results showed considerable stability and intermolecular interaction with native ligand-binding site residues of JEV-RdRp. Hence, selected natural compounds are admissible inhibitors of JEV-RdRp protein and can be considered for future antiviral drug development studies.
Collapse
Affiliation(s)
- Pardeep Yadav
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida 201310, India; (P.Y.); (S.K.J.)
- Center for Bioinformatics, Computational and Systems Biology, Pathfinder Research and Training Foundation, Greater Noida 201308, India
| | - Sherif A. El-Kafrawy
- Special Infectious Agents Unit-BSL-3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia; (S.A.E.-K.); (M.M.E.-D.); (W.T.A.); (A.A.F.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Mai M. El-Day
- Special Infectious Agents Unit-BSL-3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia; (S.A.E.-K.); (M.M.E.-D.); (W.T.A.); (A.A.F.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Wejdan T. Alghafari
- Special Infectious Agents Unit-BSL-3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia; (S.A.E.-K.); (M.M.E.-D.); (W.T.A.); (A.A.F.)
- Clinical Nutrition Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Arwa A. Faizo
- Special Infectious Agents Unit-BSL-3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia; (S.A.E.-K.); (M.M.E.-D.); (W.T.A.); (A.A.F.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida 201310, India; (P.Y.); (S.K.J.)
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India
| | - Vivek Dhar Dwivedi
- Center for Bioinformatics, Computational and Systems Biology, Pathfinder Research and Training Foundation, Greater Noida 201308, India
- Institute of Advanced Materials, IAAM, 59053 Ulrika, Sweden
| | - Esam I. Azhar
- Special Infectious Agents Unit-BSL-3, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia; (S.A.E.-K.); (M.M.E.-D.); (W.T.A.); (A.A.F.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| |
Collapse
|
7
|
Antiviral drug research for Japanese encephalitis: an updated review. Pharmacol Rep 2022; 74:273-296. [PMID: 35182390 PMCID: PMC8964565 DOI: 10.1007/s43440-022-00355-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/21/2022] [Accepted: 01/28/2022] [Indexed: 12/23/2022]
Abstract
Japanese encephalitis (JE) caused by the Japanese encephalitis virus (JEV) is one of Asia's most common viral encephalitis. JEV is a flavivirus, common in rural and sub-urban regions of Asian countries. Although only 1% of JEV-infected individuals develop JE, there is a 20-30% chance of death among these individuals and possible neurological sequelae post-infection. No licensed anti-JE drugs are currently available, despite extensive efforts to develop them. Literature search was performed using databases such as PubMed Central, Google Scholar, Wiley Online Library, etc. using keywords such as Japanese encephalitis virus, antiviral drugs, antiviral drug screening, antiviral drug targets, etc. From around 230 papers/abstracts and research reviews retrieved and reviewed for this study, approximately 180 most relevant and important ones have been cited. Different approaches in drug testing and various antiviral drug targets explored so far have been thoroughly searched from the literature and compiled, besides addressing the future perspectives of the antiviral drug development strategies. Although the development of effective anti-JE drugs is an urgent issue, only supportive care is currently available. Recent advancements in understanding the biology of infection and new drug targets have been promising improvements. Despite hindrances such as the unavailability of a proper drug delivery system or a treatment regimen irrespective of the stage of infection, several promising anti-JE candidate molecules are in different phases of clinical trials. Nonetheless, efficient therapy against JEV is expected to be achieved with drug combinations and a highly targeted drug delivery system soon.
Collapse
|
8
|
Zhang L, Zhou D, Li Q, Zhu S, Imran M, Duan H, Cao S, Ke S, Ye J. The Antiviral Effect of Novel Steroidal Derivatives on Flaviviruses. Front Microbiol 2021; 12:727236. [PMID: 34690968 PMCID: PMC8527100 DOI: 10.3389/fmicb.2021.727236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/09/2021] [Indexed: 11/13/2022] Open
Abstract
Flaviviruses are the major emerging arthropod-borne pathogens globally. However, there is still no practical anti-flavivirus approach. Therefore, existing and emerging flaviviruses desperately need active broad-spectrum drugs. In the present study, the antiviral effect of steroidal dehydroepiandrosterone (DHEA) and 23 synthetic derivatives against flaviviruses such as Japanese encephalitis virus (JEV), Zika virus (ZIKV), and Dengue virus (DENV) were appraised by examining the characteristics of virus infection both in vitro and in vivo. Our results revealed that AV1003, AV1004 and AV1017 were the most potent inhibitors of flavivirus propagation in cells. They mainly suppress the viral infection in the post-invasion stage in a dose-dependent manner. Furthermore, orally administered compound AV1004 protected mice from lethal JEV infection by increasing the survival rate and reducing the viral load in the brain of infected mice. These results indicate that the compound AV1004 might be a potential therapeutic drug against JEV infection. These DHEA derivatives may provide lead scaffolds for further design and synthesis of potential anti-flavivirus potential drugs.
Collapse
Affiliation(s)
- Luping Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Dengyuan Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Qiuyan Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Shuo Zhu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Muhammad Imran
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Hongyu Duan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Shengbo Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Shaoyong Ke
- National Biopesticide Engineering Research Center, Hubei Biopesticide Engineering Research Center, Hubei Academy of Agricultural Sciences, Wuhan, China
| | - Jing Ye
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
9
|
Mechanistic insights into the Japanese encephalitis virus RNA dependent RNA polymerase protein inhibition by bioflavonoids from Azadirachta indica. Sci Rep 2021; 11:18125. [PMID: 34518560 PMCID: PMC8437980 DOI: 10.1038/s41598-021-96917-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/18/2021] [Indexed: 02/05/2023] Open
Abstract
Japanese encephalitis (JE) virus is a flavivirus causing encephalitis causing neurological damage. RNA-dependent-RNA-polymerase (RdRp) is responsible for genome replication making it excellent anti-viral target. In this study, the crystal structure of JE RdRp (jRdRp) and bioflavonoids reported in Azadirachta indica were retrieved from specific databases. Structure-based virtual screening was employed using MTiOpenScreen server and top four compounds selected with the most negative docking scores. Conformations were redocked using AutoDock Vina; these complexes showed mechanistic interactions with Arg474, Gly605, Asp668, and Trp800 residues in the active site of jRdRp, i.e., guanosine-5′-triphosphate. Furthermore, 100 ns classical molecular dynamics simulation and binding free energy calculation showed stability of docked bioflavonoids in the active jRdRp pocket and significant contribution of van-der-Waals interactions for docked complex stability during simulation. Therefore, this study predicted the anti-viral activity of Gedunin, Nimbolide, Ohchinin acetate, and Kulactone against jRdRp and can be considered for further antiviral drug development.
Collapse
|
10
|
Cava C, Bertoli G, Castiglioni I. A protein interaction map identifies existing drugs targeting SARS-CoV-2. BMC Pharmacol Toxicol 2020; 21:65. [PMID: 32883368 PMCID: PMC7470683 DOI: 10.1186/s40360-020-00444-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus (SARS-CoV-2), an emerging Betacoronavirus, is the causative agent of COVID-19. Angiotensin converting enzyme 2 (ACE2), being the main cell receptor of SARS-CoV-2, plays a role in the entry of the virus into the cell. Currently, there are neither specific antiviral drugs for the treatment or preventive drugs such as vaccines. METHODS We proposed a bioinformatics analysis to test in silico existing drugs as a fast way to identify an efficient therapy. We performed a differential expression analysis in order to identify differentially expressed genes in COVID-19 patients correlated with ACE-2 and we explored their direct relations with a network approach integrating also drug-gene interactions. The drugs with a central role in the network were also investigated with a molecular docking analysis. RESULTS We found 825 differentially expressed genes correlated with ACE2. The protein-protein interactions among differentially expressed genes identified a network of 474 genes and 1130 interactions. CONCLUSIONS The integration of drug-gene interactions in the network and molecular docking analysis allows us to obtain several drugs with antiviral activity that, alone or in combination with other treatment options, could be considered as therapeutic approaches against COVID-19.
Collapse
Affiliation(s)
- Claudia Cava
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Via F. Cervi 93, 20090 Segrate-Milan, Milan, Italy.
| | - Gloria Bertoli
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Via F. Cervi 93, 20090 Segrate-Milan, Milan, Italy
| | - Isabella Castiglioni
- Department of Physics "Giuseppe Occhialini", University of Milan-Bicocca Piazza dell'Ateneo Nuovo, 1 - 20126, Milan, Italy
| |
Collapse
|
11
|
Screening of Natural Extracts for Inhibitors against Japanese Encephalitis Virus Infection. Antimicrob Agents Chemother 2020; 64:AAC.02373-19. [PMID: 31871089 PMCID: PMC7038234 DOI: 10.1128/aac.02373-19] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 12/18/2019] [Indexed: 12/17/2022] Open
Abstract
The mosquito-borne Japanese encephalitis virus (JEV) causes serious illness worldwide that is associated with high morbidity and mortality. Currently, there are no effective drugs approved for the treatment of JEV infection. Drug-repurposing screening is an alternative approach to discover potential antiviral agents. In this study, high-content screening (HCS) of a natural extracts library was performed, and two hit FDA-approved Na+/K+-ATPase inhibitors, ouabain and digoxin, were identified as having robust efficiency against JEV infection with the selectivity indexes over 1,000. The results indicated that ouabain and digoxin blocked the JEV infection at the replication stage by targeting the Na+/K+-ATPase. Furthermore, it was proven that ouabain significantly reduced the morbidity and mortality caused by JEV in a BALB/c mouse model. This work demonstrated that Na+/K+-ATPase could serve as the target of treatment of JEV infection, and ouabain has the potential to be developed as an effective anti-JEV drug.
Collapse
|
12
|
Quintana VM, Selisko B, Brunetti JE, Eydoux C, Guillemot JC, Canard B, Damonte EB, Julander JG, Castilla V. Antiviral activity of the natural alkaloid anisomycin against dengue and Zika viruses. Antiviral Res 2020; 176:104749. [PMID: 32081740 DOI: 10.1016/j.antiviral.2020.104749] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 01/21/2020] [Accepted: 02/15/2020] [Indexed: 12/26/2022]
Abstract
Flaviviruses constitute a public health concern because of their global burden and the lack of specific antiviral treatment. Here we investigated the antiviral activity of the alkaloid anisomycin against dengue (DENV) and Zika (ZIKV) viruses. At non-cytotoxic concentrations, anisomycin strongly inhibited the replication of reference strains and clinical isolates of all DENV serotypes and Asian and African strains of ZIKV in Vero cells. Anisomycin also prevented DENV and ZIKV multiplication in human cell lines. While initial steps of DENV and ZIKV replicative cycle were unaffected, a high inhibition of viral protein expression was demonstrated after treatment with anisomycin. DENV RNA synthesis was strongly reduced in anisomycin treated cultures, but the compound did not exert a direct inhibitory effect on 2' O-methyltransferase or RNA polymerase activities of DENV NS5 protein. Furthermore, anisomycin-mediated activation of p38 signaling was not related to the antiviral action of the compound. The evaluation of anisomycin efficacy in a mouse model of ZIKV morbidity and mortality revealed that animals treated with a low dose of anisomycin exhibited a significant reduction in viremia levels and died significantly later than the control group. This protective effect was lost at higher doses, though. In conclusion, anisomycin is a potent and selective in vitro inhibitor of DENV and ZIKV that impairs a post-entry step of viral replication; and a low-dose anisomycin treatment may provide some minimal benefit in a mouse model.
Collapse
Affiliation(s)
- V M Quintana
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires/ IQUIBICEN (CONICET), Ciudad Universitaria, Pabellón 2, Piso 4, Buenos Aires, 1428, Argentina.
| | - B Selisko
- Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université (AMU), Laboratoire d'Architecture et Fonction des Macromolécules Biologiques (AFMB) UMR 7257, 163 Avenue de Luminy, 13288, Marseille Cedex 9, France.
| | - J E Brunetti
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires/ IQUIBICEN (CONICET), Ciudad Universitaria, Pabellón 2, Piso 4, Buenos Aires, 1428, Argentina.
| | - C Eydoux
- Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université (AMU), Laboratoire d'Architecture et Fonction des Macromolécules Biologiques (AFMB) UMR 7257, 163 Avenue de Luminy, 13288, Marseille Cedex 9, France.
| | - J C Guillemot
- Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université (AMU), Laboratoire d'Architecture et Fonction des Macromolécules Biologiques (AFMB) UMR 7257, 163 Avenue de Luminy, 13288, Marseille Cedex 9, France.
| | - B Canard
- Centre National de la Recherche Scientifique (CNRS) and Aix-Marseille Université (AMU), Laboratoire d'Architecture et Fonction des Macromolécules Biologiques (AFMB) UMR 7257, 163 Avenue de Luminy, 13288, Marseille Cedex 9, France.
| | - E B Damonte
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires/ IQUIBICEN (CONICET), Ciudad Universitaria, Pabellón 2, Piso 4, Buenos Aires, 1428, Argentina.
| | - J G Julander
- Institute for Antiviral Research, Utah State University, Logan, UT, USA.
| | - V Castilla
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires/ IQUIBICEN (CONICET), Ciudad Universitaria, Pabellón 2, Piso 4, Buenos Aires, 1428, Argentina.
| |
Collapse
|
13
|
Dehydroepiandrosterone resisted E. Coli O157:H7-induced inflammation via blocking the activation of p38 MAPK and NF-κB pathways in mice. Cytokine 2019; 127:154955. [PMID: 31864092 DOI: 10.1016/j.cyto.2019.154955] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/24/2019] [Accepted: 12/10/2019] [Indexed: 11/23/2022]
Abstract
Dehydroepiandrosterone (DHEA), a critical metabolite in cholesterol metabolism, can regulate the inflammatory responses in humans or animals. However, the precise mechanisms of these beneficial actions remains poorly understood. Present study aims to clarify the anti-inflammatory function of DHEA and its possible mechanisms in the E. coli O157:H7-stimulated mice. The results indicated that DHEA reduced the mortality of mice and bacterial concentration in the peritoneal fluid in the E. coli-stimulated mice. DHEA increased the spleen index, the activity of lactate dehydrogenase and acid phosphatase; while it decreased the nitric oxide (NO) content and inducible nitric oxide synthase (iNOS) activity in mice. The mRNA levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6) and interferon gamma (IFN-γ) were decreased, whereas the interleukin-4 (IL-4) and interleukin-10 (IL-10) mRNA levels were increased in the E. coli-stimulated mice treated with DHEA. Moreover, DHEA treatment reversed the increasing of IFN-γ/IL-4 ratio in mice caused by E. coli infection. Importantly, DHEA blocked the nuclear translocation of p65 through down-regulation the IκB-α protein phosphorylation level in the mice stimulated with E. coli O157:H7. No statically changes were showed on the phospho (p)-ERK1/2 and p-JNK1/2 protein level, while DHEA significantly suppressed the p-p38 protein level in mice. The above results indicated that DHEA alleviated inflammatory responses by suppressing NO secretion and promoting Th2-associated anti-inflammatory cytokines production in mice; and this action might relate to the blocking of p38 MAPK and NF-κB signaling pathways activation. All the above results provide substantial information for understanding the anti-inflammatory function of DHEA and further support it as a potential immunomodulatory in prevention inflammatory and bacterial infection diseases.
Collapse
|
14
|
Effect of dehydroepiandrosterone on the immune function of mice in vivo and in vitro. Mol Immunol 2019; 112:283-290. [PMID: 31228660 DOI: 10.1016/j.molimm.2019.06.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/29/2019] [Accepted: 06/05/2019] [Indexed: 01/17/2023]
Abstract
Dehydroepiandrosterone (DHEA) has anti-inflammatory, anti-oxidant and immune-regulating properties, while the mechanism of DHEA actions remains unclear. The present study aims to investigate the effect and possible mechanism of DHEA on immune function of mice in vivo and in vitro. In vivo, a lipopolysaccharide (LPS)-induced experimental inflammation model was constructed to analyze the regulation of DHEA on anti-oxidative and immune function in ICR mice; In vitro, the effects of DHEA on the biological functions of lymphocytes and macrophages were studied. The results showed that DHEA increased the activity of total antioxidant capacity and superoxide dismutase, while it decreased the level of reactive oxygen species in LPS-induced mice. Meanwhile, DHEA increased the proportion of T lymphocytes and decreased that of B lymphocytes in primary cultured spleen lymphocytes, and markedly enhanced the Th1/Th2 ratio in spleen T lymphocytes. Furthermore, DHEA significantly increased the Th1 type cytokine (IL-2 and IFN-α) and decreased the Th2 type cytokine (IL-4 and IL-10) levels in LPS-induced mice or in primary cultured spleen T lymphocytes. In addition, DHEA improved the phagocytic ability, enhanced the NO production and increased the iNOS activity in peritoneal macrophages. Our data indicates that DHEA increases the macrophages function via improving NO content and up-regulating TNF-α expression levels; and it evoked a Th1 immuno-response and repressed a Th2 immuno-response through promoting a shift in Th1/Th2 balance toward Th1-dominant immunity in vivo and in vitro. These results provide substantial evidence on the mechanism of DHEA-mediated immune function and the efficient protection against infectious and inflammatory response in animals and humans.
Collapse
|
15
|
Synthesis and evaluation of steroidal thiazoline conjugates as potential antiviral agents. Future Med Chem 2018; 10:2589-2605. [PMID: 30499701 DOI: 10.4155/fmc-2018-0075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Many heterocyclic compounds derived from natural steroids exhibited broad activities, so this work focused on the investigations on a series of steroidal thiazoline conjugates as antiviral agents. Materials & methods: A series of steroid derivatives containing thiazoline heterocycles were designed and synthesized via a convenient condensation procedure. The compounds were screened for their potential antivirus activities against Enterovirus 71 (EV71) and Coxsackie Virus Type B (CVB3). Results and Conclusion: The in vitro bioassay indicated that compounds 5b, 5g and 5i exhibited excellent antiviral effects on EV71, and compounds 5b, 5e, 6c and 6g presented better antiviral activities against CVB3 compared with the controls ribavirin or pirodavir. These results indicate that these steroidal thiazoline conjugates might be feasible therapeutic candidates against EV71 infection, which might also be considered as promising compounds for optimization of potential antivirus agents.
Collapse
|
16
|
The inhibitory effect of dehydroepiandrosterone and its derivatives against influenza A virus in vitro and in vivo. Arch Virol 2016; 161:3061-72. [DOI: 10.1007/s00705-016-2993-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 07/16/2016] [Indexed: 10/21/2022]
|
17
|
Cook BWM, Ranadheera C, Nikiforuk AM, Cutts TA, Kobasa D, Court DA, Theriault SS. Limited Effects of Type I Interferons on Kyasanur Forest Disease Virus in Cell Culture. PLoS Negl Trop Dis 2016; 10:e0004871. [PMID: 27479197 PMCID: PMC4968803 DOI: 10.1371/journal.pntd.0004871] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/30/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The tick-borne flavivirus, Kyasanur Forest disease virus (KFDV) causes seasonal infections and periodic outbreaks in south-west India. The current vaccine offers poor protection with reported issues of coverage and immunogenicity. Since there are no approved prophylactic therapeutics for KFDV, type I IFN-α/β subtypes were assessed for antiviral potency against KFDV in cell culture. METHODOLOGY/PRINCIPAL FINDINGS The continued passage of KFDV-infected cells with re-administered IFN-α2a treatment did not eliminate KFDV and had little effect on infectious particle production whereas the IFN-sensitive, green fluorescent protein-expressing vesicular stomatitis virus (VSV-GFP) infection was controlled. Further evaluation of the other IFN-α/β subtypes versus KFDV infection indicated that single treatments of either IFN-αWA and IFN-αΚ appeared to be more effective than IFN-α2a at reducing KFDV titres. Concentration-dependent analysis of these IFN-α/β subtypes revealed that regardless of subtype, low concentrations of IFN were able to limit cytopathic effects (CPE), while significantly higher concentrations were needed for inhibition of virion release. Furthermore, expression of the KFDV NS5 in cell culture before IFN addition enabled VSV-GFP to overcome the effects of IFN-α/β signalling, producing a robust infection. CONCLUSIONS/SIGNIFICANCE Treatment of cell culture with IFN does not appear to be suitable for KFDV eradication and the assay used for such studies should be carefully considered. Further, it appears that the NS5 protein is sufficient to permit KFDV to bypass the antiviral properties of IFN. We suggest that other prophylactic therapeutics should be evaluated in place of IFN for treatment of individuals with KFDV disease.
Collapse
Affiliation(s)
- Bradley W. M. Cook
- Applied Biosafety Research Program, National Microbiology Laboratory at the Canadian Science Centre for Human and Animal Health and National Microbiology Laboratory at the J. C. Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Microbiology, The University of Manitoba, Winnipeg, Manitoba, Canada
| | - Charlene Ranadheera
- High Containment Respiratory Viruses Group, Special Pathogens Program, National Microbiology Laboratory at the Canadian Science Centre for Human and Animal Health, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Aidan M. Nikiforuk
- Applied Biosafety Research Program, National Microbiology Laboratory at the Canadian Science Centre for Human and Animal Health and National Microbiology Laboratory at the J. C. Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Microbiology, The University of Manitoba, Winnipeg, Manitoba, Canada
| | - Todd A. Cutts
- Applied Biosafety Research Program, National Microbiology Laboratory at the Canadian Science Centre for Human and Animal Health and National Microbiology Laboratory at the J. C. Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Darwyn Kobasa
- High Containment Respiratory Viruses Group, Special Pathogens Program, National Microbiology Laboratory at the Canadian Science Centre for Human and Animal Health, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, The University of Manitoba, Winnipeg, Manitoba, Canada
| | - Deborah A. Court
- Department of Microbiology, The University of Manitoba, Winnipeg, Manitoba, Canada
| | - Steven S. Theriault
- Applied Biosafety Research Program, National Microbiology Laboratory at the Canadian Science Centre for Human and Animal Health and National Microbiology Laboratory at the J. C. Wilt Infectious Diseases Research Centre, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Microbiology, The University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
18
|
Fang J, Sun L, Peng G, Xu J, Zhou R, Cao S, Chen H, Song Y. Identification of three antiviral inhibitors against Japanese encephalitis virus from library of pharmacologically active compounds 1280. PLoS One 2013; 8:e78425. [PMID: 24348901 PMCID: PMC3857149 DOI: 10.1371/journal.pone.0078425] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 09/19/2013] [Indexed: 12/24/2022] Open
Abstract
Japanese encephalitis virus (JEV) can cause severe central nervous disease with a high mortality rate. There is no antiviral drug available for JEV-specific treatment. In this study, a cytopathic-effect-based, high-throughput screening assay was developed and applied to screen JEV inhibitors from Library of Pharmacologically Active Compounds 1280. The antiviral effects of three hit compounds including FGIN-1-27, cilnidipine, and niclosamide were evaluated in cells by western blotting, indirect immunofluorescence assay, and plaque reduction assay. A time-of-addition assay proved that all three compounds inhibited JEV at the stage of replication. The EC50s of FGIN-1-27, cilnidipine, and niclosamide were 3.21, 6.52, and 5.80 µM, respectively, while the selectivity indexes were 38.79, 30.67, and 7.49. FGIN-1-27 and cilnidipine have high efficiency and selectivity against JEV. This study provided two JEV antiviral inhibitors as candidates for treatment of JEV infection.
Collapse
Affiliation(s)
- Jin'e Fang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Leqiang Sun
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Guiqing Peng
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jia Xu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rui Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shengbo Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yunfeng Song
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- * E-mail:
| |
Collapse
|
19
|
Haridas V, Rajgokul KS, Sadanandan S, Agrawal T, Sharvani V, Gopalakrishna MVS, Bijesh MB, Kumawat KL, Basu A, Medigeshi GR. Bispidine-amino acid conjugates act as a novel scaffold for the design of antivirals that block Japanese encephalitis virus replication. PLoS Negl Trop Dis 2013; 7:e2005. [PMID: 23350007 PMCID: PMC3547849 DOI: 10.1371/journal.pntd.0002005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 11/28/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Japanese encephalitis virus (JEV) is a major cause of viral encephalitis in South and South-East Asia. Lack of antivirals and non-availability of affordable vaccines in these endemic areas are a major setback in combating JEV and other closely related viruses such as West Nile virus and dengue virus. Protein secondary structure mimetics are excellent candidates for inhibiting the protein-protein interactions and therefore serve as an attractive tool in drug development. We synthesized derivatives containing the backbone of naturally occurring lupin alkaloid, sparteine, which act as protein secondary structure mimetics and show that these compounds exhibit antiviral properties. METHODOLOGY/PRINCIPAL FINDINGS In this study we have identified 3,7-diazabicyclo[3.3.1]nonane, commonly called bispidine, as a privileged scaffold to synthesize effective antiviral agents. We have synthesized derivatives of bispidine conjugated with amino acids and found that hydrophobic amino acid residues showed antiviral properties against JEV. We identified a tryptophan derivative, Bisp-W, which at 5 µM concentration inhibited JEV infection in neuroblastoma cells by more than 100-fold. Viral inhibition was at a stage post-entry and prior to viral protein translation possibly at viral RNA replication. We show that similar concentration of Bisp-W was capable of inhibiting viral infection of two other encephalitic viruses namely, West Nile virus and Chandipura virus. CONCLUSIONS/SIGNIFICANCE We have demonstrated that the amino-acid conjugates of 3,7-diazabicyclo[3.3.1]nonane can serve as a molecular scaffold for development of potent antivirals against encephalitic viruses. Our findings will provide a novel platform to develop effective inhibitors of JEV and perhaps other RNA viruses causing encephalitis.
Collapse
Affiliation(s)
- V. Haridas
- Department of Chemistry, Indian Institute of Technology, New Delhi, India
| | | | - Sandhya Sadanandan
- Department of Chemistry, Indian Institute of Technology, New Delhi, India
| | - Tanvi Agrawal
- Vaccine and Infectious Disease Research Center, Translational Health Science and Technology Institute, Gurgaon, India
| | - Vats Sharvani
- Vaccine and Infectious Disease Research Center, Translational Health Science and Technology Institute, Gurgaon, India
| | | | - M. B. Bijesh
- Department of Chemistry, Indian Institute of Technology, New Delhi, India
| | | | - Anirban Basu
- National Brain Research Center, Manesar, Haryana, India
| | - Guruprasad R. Medigeshi
- Vaccine and Infectious Disease Research Center, Translational Health Science and Technology Institute, Gurgaon, India
| |
Collapse
|
20
|
Torres NI, Castilla V, Wachsman M. DHEA inhibits measles virus through a mechanism independent of its ability to modulate the Raf/MEK/ERK signaling pathway. Future Virol 2012. [DOI: 10.2217/fvl.12.107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: Despite the existence of an effective vaccine, measles infection is still frequent in many developing countries with reduced health infrastructure, and it is one of the major causes of child death globally. In the past decade numerous outbreaks have occurred in developed countries, giving a fresh impetus to antiviral research against measles virus. The aim of this study was to investigate the antiviral activity of the natural steroid hormone DHEA against measles virus and the role of the Raf/MEK/ERK signaling pathway in viral multiplication and DHEA’s antiviral activity. Materials & methods: The antiviral activity of DHEA and two ERK modulators, UO126 and anisomycin, was determined using a virus yield reduction assay. Furthermore, we studied DHEA’s virucidal activity and the viral multiplication step affected by the compound. The effect of virus infection on the Raf/MEK/ERK pathway and the activity of those compounds against measles virus spread and induced cytopathic effect were studied using western blot and indirect immunofluorescence. Results & conclusion: We found that DHEA and UO126 are active against measles virus and that they are able to diminish virus-induced cytopathic effects. Also, our study showed that early events in the viral multiplication cycle trigger ERK activation, suggesting that DHEA, a Raf/MEK/ERK modulator, may not exert its antiviral activity through the modulation of this pathway. Our results may provide a first step in the development of new antiviral agents against measles virus.
Collapse
Affiliation(s)
- Nicolás I Torres
- Laboratorio de Virología. Departamento de Química Biológica. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Piso 4, 1428, Buenos Aires, Argentina
| | - Viviana Castilla
- Laboratorio de Virología. Departamento de Química Biológica. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Piso 4, 1428, Buenos Aires, Argentina
| | - Mónica Wachsman
- Laboratorio de Virología. Departamento de Química Biológica. Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Piso 4, 1428, Buenos Aires, Argentina
| |
Collapse
|
21
|
In vitro antiviral activity of dehydroepiandrosterone, 17 synthetic analogs and ERK modulators against herpes simplex virus type 1. Antiviral Res 2012; 95:37-48. [PMID: 22584352 DOI: 10.1016/j.antiviral.2012.05.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 04/24/2012] [Accepted: 05/01/2012] [Indexed: 11/22/2022]
Abstract
In the present study the in vitro antiviral activity of dehydroepiandrosterone (DHEA) and 17 synthetic derivatives against herpes simplex type 1 (HSV-1) was determined. DHEA, epiandrosterone (EA), two synthetic DHEA analogs and three synthetic EA analogs showed a selective inhibitory effect on HSV in vitro multiplication. DHEA and E2, a synthetic derivative of EA, were not found to be virucidal to cell-free HSV-1 and did not impair virus adsorption or penetration. We determined that treatment with both compounds decreased viral protein synthesis. Moreover, inhibitory effect of DHEA and E2 on extracellular viral titer was stronger than the inhibition found on total viral infectivity, suggesting that the antiherpetic activity of these compounds may also be in part due to an inhibition in virus formation and release. Since DHEA is a known Raf/MEK/ERK signaling pathway activator, we studied the role of this pathway on HSV-1 infection. ERK1/2 phosphorylation was stimulated in HSV-1 infected cultures. UO126, a Raf/MEK/ERK signaling pathway inhibitor, impaired viral multiplication, while anisomycin, an activator of this pathway, enhanced it. Treatment with DHEA 6 h before infection enhanced HSV-1 multiplication. On the contrary, pre-treatment with E2, which does not modulate Raf/MEK/ERK signaling pathway, did not produce an increase of viral replication. Taking together these results, the antiviral activity of DHEA seems to occur via a mechanism independent of its ability to modulate ERK phosphorylation.
Collapse
|
22
|
Ishikawa T, Konishi E. Combating Japanese encephalitis: Vero-cell derived inactivated vaccines and the situation in Japan. Future Virol 2010. [DOI: 10.2217/fvl.10.55] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Japanese encephalitis (JE) is a major public health threat in Asia, because of its high mortality and high incidence of psychoneurological sequelae in survivors. It is caused by JE virus (JEV) infection, transmitted by vector mosquitoes. The disease is vaccine preventable and has been well controlled in some countries. Since no specific antivirals have been approved, prevention with vaccine is important in this disease. This article provides a general overview of JE and JEV, but special focus has been put on recently developed Vero cell-derived formalin-inactivated JE vaccines, and the situation in Japan relating to these vaccines. In Japan, where JE has been well controlled, the strong governmental recommendation of the mouse brain-derived vaccine for routine immunization was suspended in 2005, owing to a patient suffering severe postvaccination events. In 2010, the recommendation was reinstated, targeting a limited population utilizing a Vero cell-derived vaccine.
Collapse
Affiliation(s)
- Tomohiro Ishikawa
- Department of International Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe 654-0142, Japan
| | | |
Collapse
|
23
|
Acrani GO, Gomes R, Proença-Módena JL, da Silva AF, Oliveira Carminati P, Silva ML, Santos RIM, Arruda E. Apoptosis induced by Oropouche virus infection in HeLa cells is dependent on virus protein expression. Virus Res 2010; 149:56-63. [DOI: 10.1016/j.virusres.2009.12.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 12/19/2009] [Accepted: 12/22/2009] [Indexed: 01/31/2023]
|
24
|
Romanutti C, Bruttomesso AC, Castilla V, Bisceglia JA, Galagovsky LR, Wachsman MB. In vitro antiviral activity of dehydroepiandrosterone and its synthetic derivatives against vesicular stomatitis virus. Vet J 2008; 182:327-35. [PMID: 18682333 DOI: 10.1016/j.tvjl.2008.06.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Revised: 06/19/2008] [Accepted: 06/20/2008] [Indexed: 11/26/2022]
Abstract
In this work the antiviral activity of 20 dehydroepiandrosterone (DHEA) analogs with different substituents at positions C-3, C-15, C-16 and C-17 were evaluated against vesicular stomatitis virus (VSV) in Vero cell cultures. The selectivity indexes (SI) obtained with DHEA and epiandrosterone (EA) were 50 and 72.6, respectively. The work showed that the compounds 21-norpregna-5,17(20)-dien-3beta,16alpha-diyl-diacetate, 17,17-ethylendioxyandrostan-5,15-dien-3beta-ol and 3beta-hydroxypregn-17(20)-en-16-one had higher SI values than ribavirin, which was used as a reference drug. The antiviral mode of action of DHEA was also investigated against VSV replication in Vero cells, and time of addition experiments showed that DHEA mainly affected a late event in the virus growth cycle. Analysis of RNA and protein synthesis indicated that DHEA adversely affected positive strand RNA synthesis and viral mature particle formation.
Collapse
Affiliation(s)
- Carina Romanutti
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Piso 4, C1428EGA, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
25
|
Acosta EG, Bruttomesso AC, Bisceglia JA, Wachsman MB, Galagovsky LR, Castilla V. Dehydroepiandrosterone, epiandrosterone and synthetic derivatives inhibit Junin virus replication in vitro. Virus Res 2008; 135:203-12. [PMID: 18462821 DOI: 10.1016/j.virusres.2008.03.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Revised: 03/17/2008] [Accepted: 03/24/2008] [Indexed: 10/22/2022]
Abstract
In the present paper the in vitro antiviral activity of dehydroepiandrosterone (DHEA), epiandrosterone (EA) and 16 synthetic derivatives against Junin virus (JUNV) replication in Vero cells was studied. DHEA and EA caused a selective inhibition of the replication of JUNV and other members of the Arenaviridae family such as Pichinde virus and Tacaribe virus. The compounds were not virucidal to cell-free JUNV. The impairment of viral replication was not due to an inhibitory effect of the steroids on virus adsorption or internalization. An inhibitory effect of the compounds on JUNV protein synthesis and both intracellular and extracellular virus production was demonstrated. A partial inhibitory action on cell surface expression of JUNV glycoprotein G1 was also detected on DHEA- and EA-treated cultures. Like DHEA and EA, three compounds obtained from EA by chemical synthesis showed selectivity indexes higher than ribavirin, the only antiviral compound that has shown partial efficacy against arenavirus infections.
Collapse
Affiliation(s)
- Eliana G Acosta
- Laboratorio de Virología, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, Piso 4, C1428EGA Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
26
|
Gould EA, Solomon T, Mackenzie JS. Does antiviral therapy have a role in the control of Japanese encephalitis? Antiviral Res 2008; 78:140-9. [PMID: 18053589 DOI: 10.1016/j.antiviral.2007.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2007] [Revised: 09/27/2007] [Accepted: 10/15/2007] [Indexed: 01/01/2023]
Abstract
Approximately 2 billion people live in countries where Japanese encephalitis (JE) presents a significant risk to humans and animals, particularly in China and India, with at least 700 million potentially susceptible children. The combined effects of climate change, altered bird migratory patterns, increasing movement of humans, animals and goods, increasing deforestation and development of irrigation projects will inevitably lead to further geographic dispersal of the virus and an enhanced threat. Although most human infections are mild or asymptomatic, some 50% of patients who develop encephalitis suffer permanent neurologic defects, and 25% die. Vaccines have reduced the incidence of JE in some countries. No specific antiviral therapy is currently available. Interferon alpha-2a was tested in a double-blind placebo-controlled trial on children with Japanese encephalitis, but with negative results. There is thus a real need for antivirals that can reduce the toll of death and neurological sequelae resulting from infection with JE virus. Here we briefly review the epidemiological problems presented by this virus, the present state of drug development and the contributory role that antiviral therapy might play in developing future control strategies for JE.
Collapse
Affiliation(s)
- E A Gould
- Unité des Virus Emergents, Faculté de Médecine, 27 Boulevard Jean Moulin, 13005 Marseille, France.
| | | | | |
Collapse
|
27
|
Ou YC, Yang CR, Cheng CL, Raung SL, Hung YY, Chen CJ. Indomethacin induces apoptosis in 786-O renal cell carcinoma cells by activating mitogen-activated protein kinases and AKT. Eur J Pharmacol 2007; 563:49-60. [PMID: 17341418 DOI: 10.1016/j.ejphar.2007.01.071] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Revised: 01/24/2007] [Accepted: 01/24/2007] [Indexed: 12/21/2022]
Abstract
Studies on chemoprevention of cancer are generating increasing interest. The anti-neoplastic effect of nonsteroidal anti-inflammatory drugs (NSAIDs) involves cyclooxygenase (COX)-dependent and COX-independent mechanisms. Evidence suggests that mitogen-activated protein kinases (MAPKs) may mediate apoptotic signaling induced by anti-neoplastic agents. While many reports have revealed the existence of MAPK activation in apoptosis induced by various stimuli, the signaling transduction pathways used by NSAIDs to trigger apoptosis in human renal cell carcinoma (RCC) remain largely unknown. Treatment of RCC 786-O cells with indomethacin resulted in growth regression and apoptosis. Caspase-dependent apoptosis was evidenced by the detection of enzymatic activities of caspase-3, caspase-6, and caspase-9 and suppression of toxicity using a caspase inhibitor. Indomethacin treatment was associated with increased expression of glucose-regulated protein 78 (GRP78) and C/EBP homologus protein (CHOP) and activation of ATF-6, characteristics of endoplasmic reticulum stress. In addition, the concomitant induction of peroxisome proliferator-activated receptor (PPAR), especially PPAR-beta, was apparent in treated cells. Western blotting revealed the activation of extracellular signal-regulated kinase (ERK), p38 MAPK, and c-Jun N-terminal kinase (JNK) with indomethacin treatment. Selective inhibitors of ERK, p38 MAPK, and JNK suppressed the induction of GRP78, CHOP, and PPAR-beta, attenuated indomethacin-induced cytotoxicity and reduced increased caspase activity. LY294002, a phosphoinositide-3 kinase (PI3K)/AKT inhibitor, and Trolox, an antioxidant, suppressed indomethacin-induced cytotoxicity and caspase activation. Furthermore, Trolox attenuated indomethacin-induced increased phosphorylation in ERK, p38 MAPK, JNK, and AKT. In conclusion, our findings establish a mechanistic link between the oxidative stress, PI3K/AKT pathway, MAPK pathway and indomethacin-induced cellular alterations and apoptosis in 786-O cells.
Collapse
Affiliation(s)
- Yen-Chuan Ou
- Division of Urology, Department of Education and Research, Taichung Veterans General Hospital, and Institute of Medical Technology, National Chung-Hsing University, Taichung, Taiwan
| | | | | | | | | | | |
Collapse
|
28
|
Inhibition of apoptosis prevents West Nile virus induced cell death. BMC Microbiol 2007; 7:49. [PMID: 17535425 PMCID: PMC1891299 DOI: 10.1186/1471-2180-7-49] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Accepted: 05/29/2007] [Indexed: 12/16/2022] Open
Abstract
Background West Nile virus (WNV) infection can cause severe meningitis and encephalitis in humans. Apoptosis was recently shown to contribute to the pathogenesis of WNV encephalitis. Here, we used WNV-infected glioma cells to study WNV-replication and WNV-induced apoptosis in human brain-derived cells. Results T98G cells are highly permissive for lytic WNV-infection as demonstrated by the production of infectious virus titre and the development of a characteristic cytopathic effect. WNV replication decreased cell viability and induced apoptosis as indicated by the activation of the effector caspase-3, the initiator caspases-8 and -9, poly(ADP-ribose)polymerase (PARP) cleavage and the release of cytochrome c from the mitochondria. Truncation of BID indicated cross-talk between the extrinsic and intrinsic apoptotic pathways. Inhibition of the caspases-8 or -9 inhibited PARP cleavage, demonstrating that both caspases are involved in WNV-induced apoptosis. Pan-caspase inhibition prevented WNV-induced apoptosis without affecting virus replication. Conclusion We found that WNV infection induces cell death in the brain-derived tumour cell line T98G by apoptosis under involvement of constituents of the extrinsic as well as the intrinsic apoptotic pathways. Our results illuminate the molecular mechanism of WNV-induced neural cell death.
Collapse
|
29
|
Mackenzie JS, Williams DT, Smith DW. Japanese Encephalitis Virus: The Geographic Distribution, Incidence, and Spread of a Virus with a Propensity to Emerge in New Areas. PERSPECTIVES IN MEDICAL VIROLOGY 2006. [DOI: 10.1016/s0168-7069(06)16010-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|