1
|
Li YY, Liang GD, Chen ZX, Zhang K, Liang JL, Jiang LR, Yang SZ, Jiang F, Liu SW, Yang J. A small molecule compound targeting hemagglutinin inhibits influenza A virus and exhibits broad-spectrum antiviral activity. Acta Pharmacol Sin 2024; 45:2380-2393. [PMID: 38987389 PMCID: PMC11489770 DOI: 10.1038/s41401-024-01331-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/30/2024] [Indexed: 07/12/2024]
Abstract
Influenza A virus (IAV) is a widespread pathogen that poses a significant threat to human health, causing pandemics with high mortality and pathogenicity. Given the emergence of increasingly drug-resistant strains of IAV, currently available antiviral drugs have been reported to be inadequate to meet clinical demands. Therefore, continuous exploration of safe, effective and broad-spectrum antiviral medications is urgently required. Here, we found that the small molecule compound J1 exhibited low toxicity both in vitro and in vivo. Moreover, J1 exhibits broad-spectrum antiviral activity against enveloped viruses, including IAV, respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), human coronavirus OC43 (HCoV-OC43), herpes simplex virus type 1 (HSV-1) and HSV-2. In this study, we explored the inhibitory effects and mechanism of action of J1 on IAV in vivo and in vitro. The results showed that J1 inhibited infection by IAV strains, including H1N1, H7N9, H5N1 and H3N2, as well as by oseltamivir-resistant strains. Mechanistic studies have shown that J1 blocks IAV infection mainly through specific interactions with the influenza virus hemagglutinin HA2 subunit, thereby blocking membrane fusion. BALB/c mice were used to establish a model of acute lung injury (ALI) induced by IAV. Treatment with J1 increased survival rates and reduced viral titers, lung index and lung inflammatory damage in virus-infected mice. In conclusion, J1 possesses significant anti-IAV effects in vitro and in vivo, providing insights into the development of broad-spectrum antivirals against future pandemics.
Collapse
Affiliation(s)
- Yin-Yan Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guo-Dong Liang
- Key Laboratory for Candidate Drug Design and Screening Based on Chemical Biology, College of Pharmacy, Inner Mongolia Medical University, Huhhot, 010110, China
| | - Zhi-Xuan Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ke Zhang
- Key Laboratory of Microbio and Infectious Disease Prevention & Control in Guizhou Province/Institute of Virology, School of Basic Medicine, Guizhou Medical University, Guiyang, 561113, China
| | - Jin-Long Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lin-Rui Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Si-Zu Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Feng Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shu-Wen Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jie Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Li Y, Huo S, Yin Z, Tian Z, Huang F, Liu P, Liu Y, Yu F. Retracted and republished from: "The current state of research on influenza antiviral drug development: drugs in clinical trial and licensed drugs". mBio 2024; 15:e0017524. [PMID: 38551343 PMCID: PMC11077966 DOI: 10.1128/mbio.00175-24] [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: 05/09/2024] Open
Abstract
Influenza viruses (IVs) threaten global human health due to the high morbidity, infection, and mortality rates. Currently, the influenza drugs recommended by the Food and Drug Administration are oseltamivir, zanamivir, peramivir, and baloxavir marboxil. These recommended antivirals are currently effective for major subtypes of IVs as the compounds target conserved domains in neuraminidase or polymerase acidic (PA) protein. However, this trend may gradually change due to the selection of antiviral drugs and the natural evolution of IVs. Therefore, there is an urgent need to develop drugs related to the treatment of influenza to deal with the next pandemic. Here, we summarized the cutting-edge research in mechanism of action, inhibitory activity, and clinical efficacy of drugs that have been approved and drugs that are still in clinical trials for influenza treatment. We hope this review will provide up-to-date and comprehensive information on influenza antivirals and generate hypotheses for screens and development of new broad-spectrum influenza drugs in the near future.
Collapse
Affiliation(s)
- Yanbai Li
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shanshan Huo
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zhe Yin
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zuguang Tian
- Department of High-Tech Development, Baoding City Science and Technology Bureau, Baoding, China
| | - Fang Huang
- Epidemic Prevention Laboratory, Tongzhou District Center For Animal Disease Control and Prevention, Beijing, China
| | - Peng Liu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Yue Liu
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA
| | - Fei Yu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| |
Collapse
|
3
|
Zeng W, Han C, Mohammed S, Li S, Song Y, Sun F, Du Y. Indole-containing pharmaceuticals: targets, pharmacological activities, and SAR studies. RSC Med Chem 2024; 15:788-808. [PMID: 38516587 PMCID: PMC10953485 DOI: 10.1039/d3md00677h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/29/2024] [Indexed: 03/23/2024] Open
Abstract
Indole is a prestigious heterocyclic skeleton widely found in both naturally-occurring and biologically-active compounds. Pharmaceutical agents containing an indole skeleton in their framework possess a wide range of pharmacological properties, including antiviral, antitumor, analgesic, and other therapeutic activities, and many indole-containing drugs have been proven to have excellent pharmacokinetic and pharmacological effects. Over the past few decades, the FDA has approved over 40 indole-containing drugs for the treatment of various clinical conditions, and the development of indole-related drugs has attracted significant attention from medicinal chemists. This review aims to provide an overview of all the approved drugs that contain an indole nucleus, focusing on their targets, pharmacological activities, and SAR studies.
Collapse
Affiliation(s)
- Wei Zeng
- School of Pharmaceutical Science and Technology, Tianjin University Tianjin 300072 China
| | - Chi Han
- School of Pharmaceutical Science and Technology, Tianjin University Tianjin 300072 China
| | - Sarah Mohammed
- School of Pharmaceutical Science and Technology, Tianjin University Tianjin 300072 China
| | - Shanshan Li
- School of Pharmaceutical Science and Technology, Tianjin University Tianjin 300072 China
| | - Yixuan Song
- Department of Chemical Engineering & Biotechnology, University of Cambridge CB2 3RA Cambridge UK
| | - Fengxia Sun
- Research Center for Chemical Safety & Security and Verification Technology & College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology Shijiazhuang 050018 China
| | - Yunfei Du
- School of Pharmaceutical Science and Technology, Tianjin University Tianjin 300072 China
| |
Collapse
|
4
|
Arman BY, Brun J, Hill ML, Zitzmann N, von Delft A. An Update on SARS-CoV-2 Clinical Trial Results-What We Can Learn for the Next Pandemic. Int J Mol Sci 2023; 25:354. [PMID: 38203525 PMCID: PMC10779148 DOI: 10.3390/ijms25010354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has claimed over 7 million lives worldwide, providing a stark reminder of the importance of pandemic preparedness. Due to the lack of approved antiviral drugs effective against coronaviruses at the start of the pandemic, the world largely relied on repurposed efforts. Here, we summarise results from randomised controlled trials to date, as well as selected in vitro data of directly acting antivirals, host-targeting antivirals, and immunomodulatory drugs. Overall, repurposing efforts evaluating directly acting antivirals targeting other viral families were largely unsuccessful, whereas several immunomodulatory drugs led to clinical improvement in hospitalised patients with severe disease. In addition, accelerated drug discovery efforts during the pandemic progressed to multiple novel directly acting antivirals with clinical efficacy, including small molecule inhibitors and monoclonal antibodies. We argue that large-scale investment is required to prepare for future pandemics; both to develop an arsenal of broad-spectrum antivirals beyond coronaviruses and build worldwide clinical trial networks that can be rapidly utilised.
Collapse
Affiliation(s)
- Benediktus Yohan Arman
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Juliane Brun
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Michelle L. Hill
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK;
| | - Nicole Zitzmann
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Annette von Delft
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
- Centre for Medicine Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
5
|
Zhu M, Anirudhan V, Du R, Rong L, Cui Q. Influenza virus cell entry and targeted antiviral development. J Med Virol 2023; 95:e29181. [PMID: 37930075 DOI: 10.1002/jmv.29181] [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] [Received: 09/04/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
Influenza virus infection is currently one of the most prevalent and transmissible diseases in the world causing local outbreaks every year. It has the potential to cause devastating global pandemics as well. The development of anti-influenza drugs possessing novel mechanisms of action is urgently needed to control the spread of influenza infections; thus, drugs that inhibit influenza virus entry into target cells are emerging as a hot research topic. In addition to discussing the biological significance of hemagglutinin in viral replication, this article provides recent updates on the natural products, small molecules, proteins, peptides, and neutralizing antibody-like proteins that have anti-influenza potency.
Collapse
Affiliation(s)
- Murong Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Varada Anirudhan
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ruikun Du
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Qinghua Cui
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|
6
|
Li Y, Huo S, Yin Z, Tian Z, Huang F, Liu P, Liu Y, Yu F. The current state of research on influenza antiviral drug development: drugs in clinical trial and licensed drugs. mBio 2023; 14:e0127323. [PMID: 37610204 PMCID: PMC10653855 DOI: 10.1128/mbio.01273-23] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023] Open
Abstract
Influenza viruses (IVs) threaten global human health due to the high morbidity, infection, and mortality rates. Currently, the influenza drugs recommended by the FDA are oseltamivir, zanamivir, peramivir, and baloxavir marboxil. Notably, owing to the high variability of IVs, no drug exists that can effectively treat all types and subtypes of IVs. Moreover, the current trend of drug resistance is likely to continue as the viral genome is constantly mutating. Therefore, there is an urgent need to develop drugs related to the treatment of influenza to deal with the next pandemic. Here, we summarized the cutting-edge research in mechanism of action, inhibitory activity, and clinical efficacy of drugs that have been approved and drugs that are still in clinical trials for influenza treatment. We hope this review will provide up-to-date and comprehensive information on influenza antivirals and generate hypotheses for screens and development of new broad-spectrum influenza drugs in the near future.
Collapse
Affiliation(s)
- Yanbai Li
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shanshan Huo
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zhe Yin
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Zuguang Tian
- Baoding City Science and Technology Bureau, Baoding, China
| | - Fang Huang
- Tongzhou District Center For Animal Disease Control and Prevention, Beijing, China
| | - Peng Liu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Yue Liu
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA
| | - Fei Yu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Hebei Wild Animal Health Center, College of Life Sciences, Hebei Agricultural University, Baoding, China
| |
Collapse
|
7
|
Bai X, Xi S, Chen G, Fan X, Wang K, Li Y, Zhao Y, Wang W, Tian Y. Multicenter, randomized controlled, open label evaluation of the efficacy and safety of arbidol hydrochloride tablets in the treatment of influenza-like cases. BMC Infect Dis 2023; 23:585. [PMID: 37674112 PMCID: PMC10483848 DOI: 10.1186/s12879-023-08570-9] [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] [Received: 02/20/2023] [Accepted: 08/28/2023] [Indexed: 09/08/2023] Open
Abstract
OBJECTIVE To study the efficacy and safety of arbidol hydrochloride tablets as a treatment for influenza-like diseases. METHODS In this multicenter, randomized, controlled, open label study, a total of 412 influenza-like cases were collected from 14 hospitals in seven regions of Hebei Province from September 2021 to March 2022. Patients were randomly divided into two groups. The control group (n = 207) were administered oseltamivir phosphate capsules for five days and the experimental group (n = 205) were administered arbidol hydrochloride tablets for five days. The primary endpoint was the time to normal body temperature, and the secondary endpoints included the time to remission of influenza symptoms, incidence of influenza-like complications, and incidence of adverse reactions. RESULTS Before treatment, there was no significant difference between the two groups in general conditions, blood routine, body temperature, or symptom severity. After treatment, there was no significant difference between the groups in the mean time to fever remission (59.24 h ± 25.21 vs. 61.05 h ± 29.47) or the mean time to remission of influenza symptoms (57.31 h ± 30.19 vs. 62.02 h ± 32.08). Survival analyses using Log-rank and Wilcoxon bilateral tests showed that there was no significant difference in fever relief time or influenza symptom relief time between the two groups. Regarding the incidence of complications and adverse events, there was only one case of tracheitis, one case of nausea, one case of vomiting, and one case of dizziness in the control group. In the experimental group, there was one case of nausea, one case of vomiting, and one case of drowsiness. In addition, one patient in the control group was hospitalized for urinary calculi. CONCLUSION There was no significant difference between the patients with influenza-like cases treated with arbidol hydrochloride tablets and those treated with oseltamivir phosphate capsules. Further, the patients treated with arbidol hydrochloride tablets had fewer adverse reactions, and thus, the tablets were safe to use.
Collapse
Affiliation(s)
- Xinfeng Bai
- Emergency Department, Second Hospital of Hebei Medical University, No. 215 Heping West Road, Xinhua District, Shijiazhuang, 050051, Hebei, China
| | - Suya Xi
- Hebei Chest Hospital, Shijiazhuang, Hebei, China
| | - Guiyan Chen
- Chengde Central Hospital, Chengde, Hebei, China
| | | | - Kaiwei Wang
- The Sixth People's Hospital of Hengshui, Hengshui, Hebei, China
| | - Yong Li
- Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yang Zhao
- Qinhuangdao Traditional Chinese Medicine Hospital, Qinhuangdao, Hebei, China
| | - Weizhan Wang
- Harrison International Peace Hospital, Hengshui, Hebei, China
| | - Yingping Tian
- Emergency Department, Second Hospital of Hebei Medical University, No. 215 Heping West Road, Xinhua District, Shijiazhuang, 050051, Hebei, China.
| |
Collapse
|
8
|
Kang Y, Shi Y, Xu S. Arbidol: The current demand, strategies, and antiviral mechanisms. Immun Inflamm Dis 2023; 11:e984. [PMID: 37647451 PMCID: PMC10461429 DOI: 10.1002/iid3.984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND High morbidity and mortality of influenza virus infection have made it become one of the most lethal diseases threatening public health; the lack of drugs with strong antiviral activity against virus strains exacerbates the problem. METHODS Two independent researchers searched relevant studies using Embase, PubMed, Web of Science, Google Scholar, and MEDLINE databases from its inception to December 2022. RESULTS Based on the different antiviral mechanisms, current antiviral strategies can be mainly classified into virus-targeting approaches such as neuraminidase inhibitors, matrix protein 2 ion channel inhibitors, polymerase acidic protein inhibitors and other host-targeting antivirals. However, highly viral gene mutation has underscored the necessity of novel antiviral drug development. Arbidol (ARB) is a Russian-made indole-derivative small molecule licensed in Russia and China for the prevention and treatment of influenza and other respiratory viral infections. ARB also has inhibitory effects on many other viruses such as severe acute respiratory syndrome coronavirus 2, Coxsackie virus, respiratory syncytial virus, Hantaan virus, herpes simplex virus, and hepatitis B and C viruses. ARB is a promising drug which can not only exert activity against virus at different steps of virus replication cycle, but also directly target on hosts before infection to prevent virus invasion. CONCLUSION ARB is a broad-spectrum antiviral drug that inhibits several viruses in vivo and in vitro, with high safety profile and low resistance; the antiviral mechanisms of ARB deserve to be further explored and more high-quality clinical studies are required to establish the efficacy and safety of ARB.
Collapse
Affiliation(s)
- Yue Kang
- Jiangsu Key Laboratory of NeurodegenerationSchool of Pharmacy, Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Yin Shi
- Department of PharmacyJiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Silu Xu
- Department of PharmacyJiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
9
|
Yang X, Sun H, Zhang Z, Ou W, Xu F, Luo L, Liu Y, Chen W, Chen J. Antiviral Effect of Ginsenosides rk1 against Influenza a Virus Infection by Targeting the Hemagglutinin 1-Mediated Virus Attachment. Int J Mol Sci 2023; 24:ijms24054967. [PMID: 36902398 PMCID: PMC10003360 DOI: 10.3390/ijms24054967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Influenza A virus (IAV) infections have been a serious hazard to public health everywhere. With the growing concern of drug-resistant IAV strains, there is an urgent need for novel anti-IAV medications, especially those with alternative mechanisms of action. Hemagglutinin (HA), an IAV glycoprotein, plays critical roles in the early stage of virus infection, including receptor binding and membrane fusion, making it a good target for developing anti-IAV drugs. Panax ginseng is a widely used herb in traditional medicine with extensive biological effects in various disease models, and its extract was reported to show protection in IAV-infected mice. However, the main effective anti-IAV constituents in panax ginseng remain unclear. Here, we report that ginsenoside rk1 (G-rk1) and G-rg5, out of the 23 screened ginsenosides, exhibit significant antiviral effects against 3 different IAV subtypes (H1N1, H5N1, and H3N2) in vitro. Mechanistically, G-rk1 blocked IAV binding to sialic acid in a hemagglutination inhibition (HAI) assay and an indirect ELISA assay; more importantly, we showed that G-rk1 interacted with HA1 in a dose-dependent manner in a surface plasmon resonance (SPR) analysis. Furthermore, G-rk1 treatment by intranasal inoculation effectively reduced the weight loss and mortality of mice challenged with a lethal dose of influenza virus A/Puerto Rico/8/34 (PR8). In conclusion, our findings reveal for the first time that G-rk1 possesses potent anti-IAV effects in vitro and in vivo. We have also identified and characterized with a direct binding assay a novel ginseng-derived IAV HA1 inhibitor for the first time, which could present potential approaches to prevent and treat IAV infections.
Collapse
Affiliation(s)
- Xia Yang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Hailiang Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhening Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Weixin Ou
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Fengxiang Xu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ling Luo
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yahong Liu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
- Correspondence: (W.C.); (J.C.); Tel./Fax: +61-3-9479-3961 (W.C.); +86-20-8528-0234 (J.C.)
| | - Jianxin Chen
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (W.C.); (J.C.); Tel./Fax: +61-3-9479-3961 (W.C.); +86-20-8528-0234 (J.C.)
| |
Collapse
|
10
|
Bao Y, Shi Y, Zhou L, Gao S, Yao R, Guo S, Geng Z, Bao L, Zhao R, Cui X. MicroRNA-205-5p: A potential therapeutic target for influenza A. J Cell Mol Med 2022; 26:5917-5928. [PMID: 36403222 PMCID: PMC9716220 DOI: 10.1111/jcmm.17615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 02/11/2024] Open
Abstract
We are committed to finding host targets for influenza A therapeutics. The nucleoprotein (NP) plays an important role in influenza A virus replication and is an indispensable part of viral transcription and replication. Exploring endogenous substances that can modulate NP is critical for finding host targets. MicroRNAs (miRNAs, miR) are a novel class of powerful, endogenous gene expression regulators. Herein, we used miRanda to analyse the base complementarity between the NP gene and the 14 host miRNAs reported previously by us. MiRanda predicted that miR-431-5p, miR-744-3p and miR-205-5p could complement the NP gene. To understand the effect of these miRNAs on NP expression, we co-transfected 293 T cells with NP gene sequence containing above miRNAs binding site or full sequence of NP gene (transfected into pmirGlo or pcDNA3.1 vectors, respectively), and mimics of miR-205-5p, miR-431-5p and miR-744-3p. Dual luciferase reporter gene or Western blotting assays confirmed that miR-205-5p and miR-431-5p inhibit NP expression by binding with the miRNA binding site of NP gene. Further, we infected Mouse Lung Epithelial (MLE-12) cells overexpressing miR-205-5p and miR-431-5p with influenza A virus and performed Western blotting to examine NP expression. We found that NP expression was significantly reduced in MLE-12 cells overexpressing miR-205-5p during influenza A infection. The miR-205-5p overexpression-induced inhibition of influenza A replication could be attributed to the inhibition of NP expression. Further, we administered oseltamivir and Jinchai Antiviral Capsules (JC, an anti-influenza Chinese medicine) to influenza A virus-infected MLE-12 cells and mice. We found that miR-205-5p was significantly decreased increased in infected cells and lung tissues, and oseltamivir and JC could up-regulate miR-205-5p. In conclusion, we provide new evidence that miR-205-5p plays a role in regulating viral NP protein expression in combating influenza A and may be a potential target for influenza A therapy.
Collapse
Affiliation(s)
- Yanyan Bao
- Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Yujing Shi
- Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Lirun Zhou
- Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Shuangrong Gao
- Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Rongmei Yao
- Institute of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Shanshan Guo
- Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Zihan Geng
- Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Lei Bao
- Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Ronghua Zhao
- Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| | - Xiaolan Cui
- Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingChina
| |
Collapse
|
11
|
Wang WC, Sayedahmed EE, Sambhara S, Mittal SK. Progress towards the Development of a Universal Influenza Vaccine. Viruses 2022; 14:v14081684. [PMID: 36016306 PMCID: PMC9415875 DOI: 10.3390/v14081684] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/22/2022] [Accepted: 07/28/2022] [Indexed: 11/21/2022] Open
Abstract
Influenza viruses are responsible for millions of cases globally and significantly threaten public health. Since pandemic and zoonotic influenza viruses have emerged in the last 20 years and some of the viruses have resulted in high mortality in humans, a universal influenza vaccine is needed to provide comprehensive protection against a wide range of influenza viruses. Current seasonal influenza vaccines provide strain-specific protection and are less effective against mismatched strains. The rapid antigenic drift and shift in influenza viruses resulted in time-consuming surveillance and uncertainty in the vaccine protection efficacy. Most recent universal influenza vaccine studies target the conserved antigen domains of the viral surface glycoproteins and internal proteins to provide broader protection. Following the development of advanced vaccine technologies, several innovative strategies and vaccine platforms are being explored to generate robust cross-protective immunity. This review provides the latest progress in the development of universal influenza vaccines.
Collapse
Affiliation(s)
- Wen-Chien Wang
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.)
| | - Ekramy E. Sayedahmed
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.)
| | - Suryaprakash Sambhara
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
- Correspondence: (S.S.); (S.K.M.)
| | - Suresh K. Mittal
- Department of Comparative Pathobiology, Purdue Institute for Immunology, Inflammation and Infectious Disease, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; (W.-C.W.); (E.E.S.)
- Correspondence: (S.S.); (S.K.M.)
| |
Collapse
|
12
|
Sadeghsoltani F, Mohammadzadeh I, Safari MM, Hassanpour P, Izadpanah M, Qujeq D, Moein S, Vaghari-Tabari M. Zinc and Respiratory Viral Infections: Important Trace Element in Anti-viral Response and Immune Regulation. Biol Trace Elem Res 2022; 200:2556-2571. [PMID: 34368933 PMCID: PMC8349606 DOI: 10.1007/s12011-021-02859-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/28/2021] [Indexed: 12/15/2022]
Abstract
Influenza viruses, respiratory syncytial virus (RSV), and SARS-COV2 are among the most dangerous respiratory viruses. Zinc is one of the essential micronutrients and is very important in the immune system. The aim of this narrative review is to review the most interesting findings about the importance of zinc in the anti-viral immune response in the respiratory tract and defense against influenza, RSV, and SARS-COV2 infections. The most interesting findings on the role of zinc in regulating immunity in the respiratory tract and the relationship between zinc and acute respiratory distress syndrome (ARDS) are reviewed, as well. Besides, current findings regarding the relationship between zinc and the effectiveness of respiratory viruses' vaccines are reviewed. The results of reviewed studies have shown that zinc and some zinc-dependent proteins are involved in anti-viral defense and immune regulation in the respiratory tract. It seems that zinc can reduce the viral titer following influenza infection. Zinc may reduce RSV burden in the lungs. Zinc can be effective in reducing the duration of viral pneumonia symptoms. Zinc may enhance the effectiveness of hydroxychloroquine in reducing mortality rate in COVID-19 patients. Besides, zinc has a positive effect in preventing ARDS and ventilator-induced lung damage. The relationship between zinc levels and the effectiveness of respiratory viruses' vaccines, especially influenza vaccines, is still unclear, and the findings are somewhat contradictory. In conclusion, zinc has anti-viral properties and is important in defending against respiratory viral infections and regulating the immune response in the respiratory tract.
Collapse
Affiliation(s)
- Fatemeh Sadeghsoltani
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Daneshgah Street, P.O. Box 51666-14711, Tabriz, Iran
| | - Iraj Mohammadzadeh
- Non-Communicable Pediatric Diseases Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mir-Meghdad Safari
- Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parisa Hassanpour
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Daneshgah Street, P.O. Box 51666-14711, Tabriz, Iran
| | - Melika Izadpanah
- Department of Anatomy, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Soheila Moein
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mostafa Vaghari-Tabari
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Daneshgah Street, P.O. Box 51666-14711, Tabriz, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Glycomic Analysis Reveals That Sialyltransferase Inhibition Is Involved in the Antiviral Effects of Arbidol. J Virol 2022; 96:e0214121. [PMID: 35044216 PMCID: PMC8941891 DOI: 10.1128/jvi.02141-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Due to the high mutation rate of influenza virus and the rapid increase of drug resistance, it is imperative to discover host-targeting antiviral agents with broad-spectrum antiviral activity. Considering the discrepancy between the urgent demand of antiviral drugs during an influenza pandemic and the long-term process of drug discovery and development, it is feasible to explore host-based antiviral agents and strategies from antiviral drugs on the market. In the current study, the antiviral mechanism of arbidol (ARB), a broad-spectrum antiviral drug with potent activity at early stages of viral replication, was investigated from the aspect of hemagglutinin (HA) receptors of host cells. N-glycans that act as the potential binding receptors of HA on 16-human bronchial epithelial (16-HBE) cells were comprehensively profiled for the first time by using an in-depth glycomic approach based on TiO2-PGC chip-Q-TOF MS. Their relative levels upon the treatment of ARB and virus were carefully examined by employing an ultra-high sensitive qualitative method based on Chip LC-QQQ MS, showing that ARB treatment led to significant and extensive decrease of sialic acid (SA)-linked N-glycans (SA receptors), and thereby impaired the virus utilization on SA receptors for rolling and entry. The SA-decreasing effect of ARB was demonstrated to result from its inhibitory effect on sialyltransferases (ST), ST3GAL4 and ST6GAL1 of 16-HBE cells. Silence of STs, natural ST inhibitors, as well as sialidase treatment of 16-HBE cells, resulted in similar potent antiviral activity, whereas ST-inducing agent led to the diminished antiviral effect of ARB. These observations collectively suggesting the involvement of ST inhibition in the antiviral effect of ARB. IMPORTANCE This study revealed, for the first time, that ST inhibition and the resulted destruction of SA receptors of host cells may be an underlying mechanism for the antiviral activity of ARB. ST inhibition has been proposed as a novel host-targeting antiviral approach recently and several compounds are currently under exploration. ARB is the first antiviral drug on the market that was found to possess ST inhibiting function. This will provide crucial evidence for the clinical usages of ARB, such as in combination with neuraminidase (NA) inhibitors to exert optimized antiviral effect, etc. More importantly, as an agent that can inhibit the expression of STs, ARB can serve as a novel lead compound for the discovery and development of host-targeting antiviral drugs.
Collapse
|
14
|
Leowattana W. COVID-19: Potential Repurposing Drugs. Infect Disord Drug Targets 2022; 22:e110122191924. [PMID: 33645490 DOI: 10.2174/1871526521666210301143441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/12/2020] [Accepted: 01/18/2021] [Indexed: 02/05/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is one of the most infectious diseases which has been caused by coronavirus in 2019 (COVID-19). It has widely spread worldwide and infected more than 28 million people in 215 countries, and more than 920,000 have now died from COVID-19. To date, no effective antiviral drugs or specific vaccines have been discovered yet. Considering this situation, the potential therapeutic antiviral drug targets for the COVID-19 are being repurposed to speed up the discovery of effective treatment. The most potential drug targets that are continuously being recommended include Favipiravir, Chloroquine, Hydroxychloroquine, and Remdesivir. Moreover, the antiviral target proteins and anti-host target proteins are being reported continuously. This review has summarized the current research studies on potential therapeutic drug targets that are being tested against the SARS-CoV-2. It will provide information related to potential repurposing drugs for overcoming COVID-19.
Collapse
Affiliation(s)
- Wattana Leowattana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajavithi Road, Rachatawee, Bangkok 10400, Thailand
| |
Collapse
|
15
|
Shelef O, Gutkin S, Feder D, Ben-Bassat A, Mandelboim M, Haitin Y, Ben-Tal N, Bacharach E, Shabat D. Ultrasensitive chemiluminescent neuraminidase probe for rapid screening and identification of small-molecules with antiviral activity against influenza A virus in mammalian cells. Chem Sci 2022; 13:12348-12357. [PMID: 36382275 PMCID: PMC9629042 DOI: 10.1039/d2sc03460c] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/23/2022] [Indexed: 11/21/2022] Open
Abstract
Influenza A virus is the most virulent influenza subtype and is associated with large-scale global pandemics characterized by high levels of morbidity and mortality. Developing simple and sensitive molecular methods for detecting influenza viruses is critical. Neuraminidase, an exo-glycosidase displayed on the surface of influenza virions, is responsible for the release of the virions and their spread in the infected host. Here, we present a new phenoxy-dioxetane chemiluminescent probe (CLNA) that can directly detect neuraminidase activity. The probe exhibits an effective turn-on response upon reaction with neuraminidase and produces a strong emission signal at 515 nm with an extremely high signal-to-noise ratio. Comparison measurements of our new probe with previously reported analogous neuraminidase optical probes showed superior detection capability in terms of response time and sensitivity. Thus, as far as we know, our probe is the most sensitive neuraminidase probe known to date. The chemiluminescence turn-on response produced by our neuraminidase probe enables rapid screening for small molecules that inhibit viral replication through different mechanisms as validated directly in influenza A-infected mammalian cells using the known inhibitors oseltamivir and amantadine. We expect that our new chemiluminescent neuraminidase probe will prove useful for various applications requiring neuraminidase detection including drug discovery assays against various influenza virus strains in mammalian cells. A new chemiluminescence neuraminidase probe enables rapid screening of small molecules that inhibit viral replication, directly in influenza A-infected mammalian cells.![]()
Collapse
Affiliation(s)
- Omri Shelef
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Sara Gutkin
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Daniel Feder
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ariel Ben-Bassat
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Mandelboim
- Central Virology Laboratory, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52620, Israel
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yoni Haitin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nir Ben-Tal
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Eran Bacharach
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Doron Shabat
- School of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel-Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
16
|
Sette-DE-Souza PH, Costa MJF, Araújo FAC, Alencar EN, Amaral-Machado L. Two phytocompounds from Schinopsis brasiliensis show promising antiviral activity with multiples targets in Influenza A virus. AN ACAD BRAS CIENC 2021; 93:e20210964. [PMID: 34817041 DOI: 10.1590/0001-3765202120210964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/24/2021] [Indexed: 01/02/2023] Open
Abstract
Influenza A virus, the main flu agent, affects billions of people worldwide. Conventional treatments still present limitations related to drug-resistance and severe side effects. As a result, natural product-derived molecules have been increasingly investigated as prospect drug candidates. Therefore, the aim of this study was to investigate the possible anti-flu activity and to evaluate the toxicity and pharmacokinetic parameters, by in silico approaches, of the Schinopsis brasiliensis Engl. phytochemical compounds. Nine phytocompounds and six antiviral drugs (Amantadine, Umifenovir, Favipiravir, Nitazoxanide, Oseltamivir, Zanamivir) were selected for the analyses against four Influenza A proteins: neuraminidase, polymerase basic protein 2, hemagglutinin and M2 ion channel protein. The molecular docking, the predicted antiviral activity, the predicted toxicity and the pharmacokinetics investigations were conducted. The obtained results demonstrated that Syringaresinol and Cycloartenone display promising in silico antiviral activity (binding energy < 5.0 and ≥ 9.0 kcal/mol) and safety (low toxicity than commercial anti-flu drugs). Overall, this study corroborated the hypothesis that S. brasiliensis barks extract has a biological activity against Influenza A virus. Additionally, Syringaresinol and Cycloartenone have multiple targets in Influenza A virus and showed themselves as the most promising phytocompounds to be isolated and considered for the therapeutic arsenal against the flu.
Collapse
Affiliation(s)
- Pedro Henrique Sette-DE-Souza
- Universidade de Pernambuco, Faculdade de Odontologia, Rua Cicero Monteiro de Melo, s/n, São Cristóvão, 56503-146 Arcoverde, PE, Brazil.,Universidade de Pernambuco, Programa de Pós-Graduação em Saúde e Desenvolvimento Socioambiental (PPGSDS), Rua Capitão Pedro Rodrigues, 105, São José, 55294-902 Garanhuns, PE, Brazil.,Universidade de Pernambuco, Programa de Pós-Graduação em Odontologia, Av. General, Newton Cavalcanti, 1650, Tabatinga, 54756-220 Camaragibe, PE, Brazil
| | - Moan J F Costa
- Universidade de Pernambuco, Faculdade de Odontologia, Rua Cicero Monteiro de Melo, s/n, São Cristóvão, 56503-146 Arcoverde, PE, Brazil
| | - Fábio A C Araújo
- Universidade de Pernambuco, Faculdade de Odontologia, Rua Cicero Monteiro de Melo, s/n, São Cristóvão, 56503-146 Arcoverde, PE, Brazil.,Universidade de Pernambuco, Programa de Pós-Graduação em Odontologia, Av. General, Newton Cavalcanti, 1650, Tabatinga, 54756-220 Camaragibe, PE, Brazil
| | - Everton N Alencar
- Universidade Federal do Rio Grande do Norte, Departamento de Farmácia, Rua General Gustavo Cordeiro de Faria, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| | - Lucas Amaral-Machado
- Universidade Federal do Rio Grande do Norte, Departamento de Farmácia, Rua General Gustavo Cordeiro de Faria, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| |
Collapse
|
17
|
Zhao MZ, Guo X, Sun B, Sun XF, Pang GF, Yang LY, Zhao X, Sun LX, Zhang Q. HA of H1N1 enhanced the expression of ICAM-1 and IL-6 in HUVECs and pathological injury in the lungs in mice. Gene 2021; 801:145854. [PMID: 34274468 PMCID: PMC8302257 DOI: 10.1016/j.gene.2021.145854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/28/2021] [Accepted: 07/13/2021] [Indexed: 01/18/2023]
Abstract
Objective Both COVID-19 and influenza are viral respiratory tract infections and the epidemics of viral respiratory tract infections remain highly prevalent with lethal consequences in susceptible individuals. Expression of ICAM-1 on vascular endothelium recruits leukocytes which initiates inflammation. IL-6 induces ICAM-1. Both ICAM-1 and IL-6 can be enhanced in influenza virus infection and COVID-19 patients. Besides initiation of virus entry host cells, whether HA alone, instead of whole virus, of influenza has the effects on expression of ICAM-1 and IL-6 in vascular endothelium with injury in the lungs, remains to be demonstrated. Methods RT-qPCR and Western blot as well as histopathologic examination were used to examine mRNA and protein of ICAM-1 and IL-6 as well as pathological injury in the lung tissues, respectively. Results After incubation of the Human Umbilical Vein Endothelial Cells (HUVECs) with HA of H1N1 for 24 h, the mRNA and protein of ICAM-1 and IL-6 in HUVECs were increased in group of 5 μg/ml concentration with statistical significance (p < 0.05). Pathological injury in lung tissues of the mice was shown 12 h after tail intravenous injection with 100 μl of HA (50 μg/ml and 100 μg/ml in normal saline), including widened alveolar spaces with angiotelectasis in alveolar wall, alveolar luminal and interstitial inflammatory infiltrates, alveolar luminal erythrocyte effusion. Conclusions HA alone, instead of whole H1N1 virus, induced more expression of ICAM-1 and IL-6, two molecules involving in pathological and inflammatory responses, in HUVECs and pathological injury in lung tissues of the mice. This knowledge provides a new HA-targeted potential direction for prevention and treatment of disease related to H1N1 infection.
Collapse
Affiliation(s)
- Ming-Zhen Zhao
- Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Xiang Guo
- Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Bo Sun
- Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Xiao-Fang Sun
- Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Gui-Fen Pang
- Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Lin-Ying Yang
- Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Xing Zhao
- Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Li-Xin Sun
- Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China
| | - Qing Zhang
- Affiliated Hospital of Chengde Medical University, Chengde 067000, Hebei, China.
| |
Collapse
|
18
|
Chen Z, Cui Q, Caffrey M, Rong L, Du R. Small Molecule Inhibitors of Influenza Virus Entry. Pharmaceuticals (Basel) 2021; 14:ph14060587. [PMID: 34207368 PMCID: PMC8234048 DOI: 10.3390/ph14060587] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/16/2022] Open
Abstract
Hemagglutinin (HA) plays a critical role during influenza virus receptor binding and subsequent membrane fusion process, thus HA has become a promising drug target. For the past several decades, we and other researchers have discovered a series of HA inhibitors mainly targeting its fusion machinery. In this review, we summarize the advances in HA-targeted development of small molecule inhibitors. Moreover, we discuss the structural basis and mode of action of these inhibitors, and speculate upon future directions toward more potent inhibitors of membrane fusion and potential anti-influenza drugs.
Collapse
Affiliation(s)
- Zhaoyu Chen
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Z.C.); (Q.C.)
| | - Qinghua Cui
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Z.C.); (Q.C.)
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266122, China
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Correspondence: (L.R.); (R.D.); Tel.: +1-312-355-0203 (L.R.); +86-0531-89628505 (R.D.)
| | - Ruikun Du
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Z.C.); (Q.C.)
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266122, China
- Correspondence: (L.R.); (R.D.); Tel.: +1-312-355-0203 (L.R.); +86-0531-89628505 (R.D.)
| |
Collapse
|
19
|
Du R, Cheng H, Cui Q, Peet NP, Gaisina IN, Rong L. Identification of a novel inhibitor targeting influenza A virus group 2 hemagglutinins. Antiviral Res 2021; 186:105013. [PMID: 33428962 DOI: 10.1016/j.antiviral.2021.105013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/29/2020] [Accepted: 01/02/2021] [Indexed: 10/22/2022]
Abstract
Influenza A virus (IAV) causes seasonal epidemics and occasional but devastating pandemics, which are major public health concerns. The putative antiviral therapeutics are useful for the treatment of influenza, however, the emerging resistant strains necessitate a constant search for new drug candidates. Here we report the discovery of a novel antiviral agent, compound CBS1194, which was identified by a parallel high-throughput screening (HTS) campaign using two retroviral pseudotypes bearing H7 or H5 hemagglutinins (HAs). Subsequent analyses demonstrated that CBS1194 is specific to IAVs of group 2, while it has no effect against those of group 1. In a time-of-addition assay, CBS1194 showed a significant inhibitory effect during the early phase of viral infection. In addition, HA-mediated hemolysis can be inhibited by CBS1194 treatment, indicating that this compound may target the HA stalk region, which is responsible for membrane fusion. Escape mutant analyses and in silico docking further revealed that CBS1194 fits into a pocket near the fusion peptide, causing steric hindrance that blocks the low-pH induced rearrangement of HA. In summary, our study identifies a novel fusion inhibitor of group 2 IAVs, which has the potential as lead compound for further development.
Collapse
Affiliation(s)
- Ruikun Du
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| | - Han Cheng
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Qinghua Cui
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Norton P Peet
- Chicago BioSolutions Inc., 2242 W Harrison Street, Chicago, IL, 60612, United States
| | - Irina N Gaisina
- Chicago BioSolutions Inc., 2242 W Harrison Street, Chicago, IL, 60612, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
20
|
Tsai SC, Lu CC, Bau DT, Chiu YJ, Yen YT, Hsu YM, Fu CW, Kuo SC, Lo YS, Chiu HY, Juan YN, Tsai FJ, Yang JS. Approaches towards fighting the COVID‑19 pandemic (Review). Int J Mol Med 2021; 47:3-22. [PMID: 33236131 PMCID: PMC7723515 DOI: 10.3892/ijmm.2020.4794] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/04/2020] [Indexed: 11/05/2022] Open
Abstract
The coronavirus disease 2019 (COVID‑19) outbreak, which has caused >46 millions confirmed infections and >1.2 million coronavirus related deaths, is one of the most devastating worldwide crises in recent years. Infection with COVID‑19 results in a fever, dry cough, general fatigue, respiratory symptoms, diarrhoea and a sore throat, similar to those of acute respiratory distress syndrome. The causative agent of COVID‑19, SARS‑CoV‑2, is a novel coronavirus strain. To date, remdesivir has been granted emergency use authorization for use in the management of infection. Additionally, several efficient diagnostic tools are being actively developed, and novel drugs and vaccines are being evaluated for their efficacy as therapeutic agents against COVID‑19, or in the prevention of infection. The present review highlights the prevalent clinical manifestations of COVID‑19, characterizes the SARS‑CoV‑2 viral genome sequence and life cycle, highlights the optimal methods for preventing viral transmission, and discusses possible molecular pharmacological mechanisms and approaches in the development of anti‑SARS‑CoV‑2 therapeutic agents. In addition, the use of traditional Chinese medicines for management of COVID‑19 is discussed. It is expected that novel anti‑viral agents, vaccines or an effective combination therapy for treatment/management of SARS‑CoV‑2 infection and spread therapy will be developed and implemented in 2021, and we would like to extend our best regards to the frontline health workers across the world in their fight against COVID‑19.
Collapse
Affiliation(s)
- Shih-Chang Tsai
- Department of Biological Science and Technology, China Medical University
| | - Chi-Cheng Lu
- Department of Sport Performance, National Taiwan University of Sport
| | - Da-Tian Bau
- Graduate Institute of Biomedical Sciences, China Medical University
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354
| | - Yu-Jen Chiu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veteran General Hospital
- Department of Surgery, School of Medicine, National Yang Ming University, Taipei 11217
| | - Yu-Ting Yen
- Drug Development Center, Institute of New Drug Development, China Medical University, Taichung 40402
| | - Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University
| | - Chih-Wei Fu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 310401
| | - Sheng-Chu Kuo
- School of Pharmacy, China Medical University, Taichung 40402
| | - Yu-Shiang Lo
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40447
| | - Hong-Yi Chiu
- Department of Pharmacy, Buddhist Tzu Chi General Hospital, Hualien 97002
- Master and PhD Program in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien 97004
- General Education Center, Tzu Chi University of Science and Technology, Hualien 97005
| | - Yu-Ning Juan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40447
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University
- China Medical University Children's Hospital, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40447
| |
Collapse
|
21
|
Zhang Q, Liang T, Nandakumar KS, Liu S. Emerging and state of the art hemagglutinin-targeted influenza virus inhibitors. Expert Opin Pharmacother 2020; 22:715-728. [PMID: 33327812 DOI: 10.1080/14656566.2020.1856814] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Introduction: Seasonal influenza vaccination, together with FDA-approved neuraminidase (NA) and polymerase acidic (PA) inhibitors, is the most effective way for prophylaxis and treatment of influenza infections. However, the low efficacy of prevailing vaccines to newly emerging influenza strains and increasing resistance to available drugs drives intense research to explore more effective inhibitors. Hemagglutinin (HA), one of the major surface proteins of influenza strains, represents an attractive therapeutic target to develop such new inhibitors.Areas covered: This review summarizes the current progress of HA-based influenza virus inhibitors and their mechanisms of action, which may facilitate further research in developing novel antiviral inhibitors for controlling influenza infections.Expert opinion: HA-mediated entry of influenza virus is an essential step for successful infection of the host, which makes HA a promising target for the development of antiviral drugs. Recent progress in delineating the crystal structures of HA, especially HA-inhibitors complexes, has revealed a number of key residues and conserved binding pockets within HA. This has opened up important insights for developing HA-based antiviral inhibitors that have a high resistance barrier and broad-spectrum activities.
Collapse
Affiliation(s)
- Qiao Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Taizhen Liang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Kutty Selva Nandakumar
- Southern Medical University-Karolinska Institute United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P. R. China.,State Key Laboratory of Organ Failure Research, Institute of Kidney Disease of Guangdong, Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
22
|
Zhou X, Zhao M, Liu Y, Chen Q, Shen L. Statistical Binding Matching between Influenza A Virus and Dynamic Glycan Clusters Determines Its Adhesion onto Lipid Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:15212-15219. [PMID: 33307709 DOI: 10.1021/acs.langmuir.0c02047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The resistance of drugs to the new influenza A virus (IAV) strains and the limited efficiency of vaccines to prevent seasonal flu epidemics underscore the urgency in finding novel strategies to block IAV infection, which is required to gain insights into the mechanism of the initial step of IAV adhesion. While it is well established that IAVs bind to respiratory tract cells by recognizing sialylated glycans on host cell membranes through a multivalency effect, how IAVs dynamically respond to multiple glycan receptors via distinct valencies has not been fully understood, limiting the discovery of novel anti-flu strategies. Using single-particle tracking to record the 2D mobilities and surface residence times of highly pathogenic H5N1 avian IAVs adhered to fluidic membranes containing α2-3 sialylated GM3 glycolipids, we quantified the univalent and multivalent IAV adhesion channels, which provide insights into the mechanism of IAV binding; IAV can guide the clustering of dynamic glycolipids to statistically match the multivalent binding affinities for IAV adhesion. This mechanism can be inhibited by disrupting the dynamic glycan clustering on membranes of varying fluidities, like the gel phase membrane. This work facilitates a deeper fundamental understanding of IAV infection as well as the development of novel anti-flu strategies.
Collapse
Affiliation(s)
- Xin Zhou
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Min Zhao
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yuanyuan Liu
- School of Basic Medical Sciences, Institute of Medical Virology, Wuhan University, Wuhan 430071, China
| | - Quanjiao Chen
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Lei Shen
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| |
Collapse
|
23
|
Pandey A, Nikam AN, Shreya AB, Mutalik SP, Gopalan D, Kulkarni S, Padya BS, Fernandes G, Mutalik S, Prassl R. Potential therapeutic targets for combating SARS-CoV-2: Drug repurposing, clinical trials and recent advancements. Life Sci 2020; 256:117883. [PMID: 32497632 PMCID: PMC7263255 DOI: 10.1016/j.lfs.2020.117883] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/31/2022]
Abstract
The present pandemic of SARS-CoV-2 has been a tough task for the whole world to deal with. With the absence of specific drugs or vaccines against SARS-CoV-2, the situation is very difficult to control. Apart from the absence of specific therapies, the lack of knowledge about potential therapeutic targets and individual perception is adding to the complications. The present review describes the novel SARS-CoV-2 structure, surface proteins, asymptomatic and symptomatic transmission in addition to the genotype and phenotype of SARS-CoV-2 along with genetic strains and similarity between SARS, MERS and SARS-CoV-2. Therapeutic strategies such as inhibition of the endocytic pathway and suppressing RNA polymerase activity by metal ions, which could be quite beneficial for controlling COVID-19, are outlined. The drug repurposing for SARS-CoV-2 is discussed in detail along with therapeutic classes such as antivirals, antibiotics, and amino quinolones and their probable role in suppressing SARS-CoV-2 with reference to case studies. The ongoing clinical trials both with respect to drug repurposing and vaccines are summarized along with a brief description. The recent advancements and future perspective of ongoing research for therapy and detection of SARS-CoV-2 are provided. The review, in brief, summarizes epidemiology, therapy and the current scenario for combating SARS-CoV-2.
Collapse
Affiliation(s)
- Abhjieet Pandey
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India; Gottfried Schatz Research Centre for Cell Signalling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Ajinkya Nitin Nikam
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Ajjappla Basavaraj Shreya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Sadhana P Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Divya Gopalan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Sanjay Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Bharath Singh Padya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Gasper Fernandes
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India.
| | - Ruth Prassl
- Gottfried Schatz Research Centre for Cell Signalling, Metabolism and Aging, Medical University of Graz, Graz, Austria..
| |
Collapse
|
24
|
Al-Horani RA, Kar S, Aliter KF. Potential Anti-COVID-19 Therapeutics that Block the Early Stage of the Viral Life Cycle: Structures, Mechanisms, and Clinical Trials. Int J Mol Sci 2020; 21:E5224. [PMID: 32718020 PMCID: PMC7432953 DOI: 10.3390/ijms21155224] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
The ongoing pandemic of coronavirus disease-2019 (COVID-19) is being caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The disease continues to present significant challenges to the health care systems around the world. This is primarily because of the lack of vaccines to protect against the infection and the lack of highly effective therapeutics to prevent and/or treat the illness. Nevertheless, researchers have swiftly responded to the pandemic by advancing old and new potential therapeutics into clinical trials. In this review, we summarize potential anti-COVID-19 therapeutics that block the early stage of the viral life cycle. The review presents the structures, mechanisms, and reported results of clinical trials of potential therapeutics that have been listed in clinicaltrials.gov. Given the fact that some of these therapeutics are multi-acting molecules, other relevant mechanisms will also be described. The reviewed therapeutics include small molecules and macromolecules of sulfated polysaccharides, polypeptides, and monoclonal antibodies. The potential therapeutics target viral and/or host proteins or processes that facilitate the early stage of the viral infection. Frequent targets are the viral spike protein, the host angiotensin converting enzyme 2, the host transmembrane protease serine 2, and clathrin-mediated endocytosis process. Overall, the review aims at presenting update-to-date details, so as to enhance awareness of potential therapeutics, and thus, to catalyze their appropriate use in combating the pandemic.
Collapse
Affiliation(s)
- Rami A. Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| | - Srabani Kar
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| | - Kholoud F. Aliter
- Department of Chemistry, School of STEM, Dillard University, New Orleans, LA 70122, USA;
| |
Collapse
|
25
|
Hong EH, Song JH, Kim SR, Cho J, Jeong B, Yang H, Jeong JH, Ahn JH, Jeong H, Kim SE, Chang SY, Ko HJ. Morin Hydrate Inhibits Influenza Virus entry into Host Cells and Has Anti-inflammatory Effect in Influenza-infected Mice. Immune Netw 2020; 20:e32. [PMID: 32895619 PMCID: PMC7458794 DOI: 10.4110/in.2020.20.e32] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/30/2020] [Accepted: 07/09/2020] [Indexed: 12/26/2022] Open
Abstract
Influenza virus is the major cause of seasonal and pandemic flu. Currently, oseltamivir, a potent and selective inhibitor of neuraminidase of influenza A and B viruses, is the drug of choice for treating patients with influenza virus infection. However, recent emergence of oseltamivir-resistant influenza viruses has limited its efficacy. Morin hydrate (3,5,7,2′,4′-pentahydroxyflavone) is a flavonoid isolated from Morus alba L. It has antioxidant, anti-inflammatory, neuroprotective, and anticancer effects partly by the inhibition of the NF-кB signaling pathway. However, its effects on influenza virus have not been studied. We evaluated the antiviral activity of morin hydrate against influenza A/Puerto Rico/8/1934 (A/PR/8; H1N1) and oseltamivir-resistant A/PR/8 influenza viruses in vitro. To determine its mode of action, we carried out time course experiments, and time of addition, hemolysis inhibition, and hemagglutination assays. The effects of the co-administration of morin hydrate and oseltamivir were assessed using the murine model of A/PR/8 infection. We found that morin hydrate reduced hemagglutination by A/PR/8 in vitro. It alleviated the symptoms of A/PR/8-infection, and reduced the levels of pro-inflammatory cytokines and chemokines, such as TNF-α and CCL2, in infected mice. Co-administration of morin hydrate and oseltamivir phosphate reduced the virus titers and attenuated pulmonary inflammation. Our results suggest that morin hydrate exhibits antiviral activity by inhibiting the entry of the virus.
Collapse
Affiliation(s)
- Eun-Hye Hong
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Jae-Hyoung Song
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea.,Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon 24341, Korea
| | - Seong-Ryeol Kim
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Jaewon Cho
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Birang Jeong
- Laboratory of Natural Products Chemistry, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Heejung Yang
- Laboratory of Natural Products Chemistry, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Jae-Hyeon Jeong
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Jae-Hee Ahn
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Hyunjin Jeong
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Seong-Eun Kim
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea
| | - Sun-Young Chang
- Laboratory of Microbiology and Immunology, College of Pharmacy, Ajou University, Suwon 16499, Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon 24341, Korea.,Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
26
|
Chen ZR, Zhou Y, Liu J, Peng HW, Zhou J, Zhong HL, Liu LL, Lai MF, Wei XH, Wen JH. Pharmacotherapics Advice in Guidelines for COVID-19. Front Pharmacol 2020; 11:950. [PMID: 32670066 PMCID: PMC7327107 DOI: 10.3389/fphar.2020.00950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
Since December 2019 to May 2020, coronavirus disease 2019 (COVID-19) has infected over 6 million people worldwide. Due to its sudden and rapid outbreak, effective treatment for COVID-19 is scarce. Based on national clinical trials of novel treatments, China, Italy, Germany, and other countries and organizations have published multiple guidelines for COVID-19 and advised many medicines, such as chloroquine and tocilizumab. In this paper, we summarize the pharmacotherapy for COVID-19 according to those guidelines, highlight updates of the pharmacotherapy guidelines, and review the efficacy and safety of the indicated anti-COVID-19 drugs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jin-Hua Wen
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
27
|
SARS-CoV-2: An Update on Potential Antivirals in Light of SARS-CoV Antiviral Drug Discoveries. Vaccines (Basel) 2020; 8:vaccines8020335. [PMID: 32585913 PMCID: PMC7350231 DOI: 10.3390/vaccines8020335] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/04/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Coronaviruses (CoVs) are a group of RNA viruses that are associated with different diseases in animals, birds, and humans. Human CoVs (HCoVs) have long been known to be the causative agents of mild respiratory illnesses. However, two HCoVs associated with severe respiratory diseases are Severe Acute Respiratory Syndrome-CoV (SARS-CoV) and Middle East Respiratory Syndrome-CoV (MERS-CoV). Both viruses resulted in hundreds of deaths after spreading to several countries. Most recently, SARS-CoV-2 has emerged as the third HCoV causing severe respiratory distress syndrome and viral pneumonia (known as COVID-19) in patients from Wuhan, China, in December 2019. Soon after its discovery, SARS-CoV-2 spread to all countries, resulting in millions of cases and thousands of deaths. Since the emergence of SARS-CoV, many research groups have dedicated their resources to discovering effective antivirals that can treat such life-threatening infections. The rapid spread and high fatality rate of SARS-CoV-2 necessitate the quick discovery of effective antivirals to control this outbreak. Since SARS-CoV-2 shares 79% sequence identity with SARS-CoV, several anti-SARS-CoV drugs have shown promise in limiting SARS-CoV-2 replication in vitro and in vivo. In this review, we discuss antivirals described for SARS-CoV and provide an update on therapeutic strategies and antivirals against SARS-CoV-2. The control of the current outbreak will strongly depend on the discovery of effective and safe anti-SARS-CoV-2 drugs.
Collapse
|
28
|
Su X, Wang Q, Wen Y, Jiang S, Lu L. Protein- and Peptide-Based Virus Inactivators: Inactivating Viruses Before Their Entry Into Cells. Front Microbiol 2020; 11:1063. [PMID: 32523582 PMCID: PMC7261908 DOI: 10.3389/fmicb.2020.01063] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 04/29/2020] [Indexed: 12/20/2022] Open
Abstract
Infectious diseases caused by human immunodeficiency virus (HIV) and other highly pathogenic enveloped viruses, have threatened the global public health. Most antiviral drugs act as passive defenders to inhibit viral replication inside the cell, while a few of them function as gate keepers to combat viruses outside the cell, including fusion inhibitors, e.g., enfuvirtide, and receptor antagonists, e.g., maraviroc, as well as virus inactivators (including attachment inhibitors). Different from fusion inhibitors and receptor antagonists that must act in the presence of target cells, virus inactivators can actively inactivate cell-free virions in the blood, through interaction with one or more sites in the envelope glycoproteins (Envs) on virions. Notably, a number of protein- and peptide-based virus inactivators (PPVIs) under development are expected to have a better utilization rate than the current antiviral drugs and be safer for in vivo human application than the chemical-based virus inactivators. Here we have highlighted recent progress in developing PPVIs against several important enveloped viruses, including HIV, influenza virus, Zika virus (ZIKV), dengue virus (DENV), and herpes simplex virus (HSV), and the potential use of PPVIs for urgent treatment of infection by newly emerging or re-emerging viruses.
Collapse
Affiliation(s)
- Xiaojie Su
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qian Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yumei Wen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China.,Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, United States
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Wang L, Xu X, Ruan J, Lin S, Jiang J, Ye H. Quadruple therapy for asymptomatic COVID-19 infection patients. Expert Rev Anti Infect Ther 2020; 18:617-624. [PMID: 32362193 PMCID: PMC7212541 DOI: 10.1080/14787210.2020.1758066] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: The novel coronavirus (COVID-19) is currently in epidemic stage. After large-scale interpersonal infection, asymptomatic patients appear. Whether asymptomatic patients are contagious or not and whether they need medication are the arguments among clinical experts. Areas covered: This paper reports a special asymptomatic couple with COVID-19, of which the male patient is an intercity bus driver but has not induced confirmed infection of his 188 passengers. The patients were treated with four combinations of lopinavir/ritonavir tablets, arbidol tablets, Lianhuaqingwen granules, and recombinant human interferon-α2b (IFN-α2b) injection via aerosol. Their clinical characteristics and medication were summarized and analyzed. Expert opinion: The two asymptomatic patients far away from Wuhan did not seem to be highly contagious. They improved obviously, after treatment with the quadruple therapy, but the effective drug is still unknown. It should be noted that lopinavir/ritonavir tablets have many drug interactions and are the most likely drugs to cause hyperlipidemia and hyperglycemia in these two patients. IFN-α2b is more effective in the early stage of virus infection. Arbidol instruction dose may not be sufficient to inhibit the novel coronavirus in vivo. The evidence-based medicine of Lianhuaqingwen granules for treating various viral infections is just based on Chinese patients.
Collapse
Affiliation(s)
- Ling Wang
- Shengli Clinical Medical College of Fujian Medical University , Fuzhou, Fujian, China.,Department of Pharmacy, Fujian Provincial Hospital , Fuzhou, Fujian, China
| | - Xiaopeng Xu
- Nanping Center for Disease Control and Prevention , Nanping, Fujian, China
| | - Junshan Ruan
- Shengli Clinical Medical College of Fujian Medical University , Fuzhou, Fujian, China.,Department of Pharmacy, Fujian Provincial Hospital , Fuzhou, Fujian, China
| | - Saijin Lin
- Department of Infectious Disease, The First Hospital of Nanping , Nanping, Fujian, China
| | - Jinhua Jiang
- Songxi County Hospital , Songxi County, Nanping, Fujian, China
| | - Hong Ye
- Shengli Clinical Medical College of Fujian Medical University , Fuzhou, Fujian, China.,Department of Pharmacy, Fujian Provincial Hospital , Fuzhou, Fujian, China
| |
Collapse
|
30
|
Host-Virus Interaction: How Host Cells Defend against Influenza A Virus Infection. Viruses 2020; 12:v12040376. [PMID: 32235330 PMCID: PMC7232439 DOI: 10.3390/v12040376] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 02/07/2023] Open
Abstract
Influenza A viruses (IAVs) are highly contagious pathogens infecting human and numerous animals. The viruses cause millions of infection cases and thousands of deaths every year, thus making IAVs a continual threat to global health. Upon IAV infection, host innate immune system is triggered and activated to restrict virus replication and clear pathogens. Subsequently, host adaptive immunity is involved in specific virus clearance. On the other hand, to achieve a successful infection, IAVs also apply multiple strategies to avoid be detected and eliminated by the host immunity. In the current review, we present a general description on recent work regarding different host cells and molecules facilitating antiviral defenses against IAV infection and how IAVs antagonize host immune responses.
Collapse
|
31
|
Sriwilaijaroen N, Suzuki Y. Hemagglutinin Inhibitors are Potential Future Anti-Influenza Drugs for Mono- and Combination Therapies. Methods Mol Biol 2020; 2132:547-565. [PMID: 32306356 DOI: 10.1007/978-1-0716-0430-4_48] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Infections by H1-H16 influenza A viruses require sufficient binding of viral hemagglutinins (HAs) to specific target receptors, glycoconjugates bearing sialyl sugar chains, on the host cell surface. Synthesized sialyl sugar chains targeting sialyl sugar-binding sites in HAs that are immutable as long as the virus does not switch to a different host species might therefore be highly effective candidate drugs for inhibition of the initial required step of virus entry. In this chapter, we describe the following aspects of updated sialyl sugar chains as influenza A virus HA inhibitors (HAIs): (1) mode of terminal sialyl-galactose linkage, (2) molecular length and structure of sialyl glycan receptors, (3) multivalent sialyl sugar chain dimension, (4) clustering of sialyl sugar chains on macromolecular scaffolds, and (5) enhancement of the stability of sialyl sugar chain HA inhibitors. We also discuss about the use of HAI-based combinations that should be considered for future influenza therapy.
Collapse
Affiliation(s)
- Nongluk Sriwilaijaroen
- Department of Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathumthani, Thailand.
- College of Life and Health Sciences, Chubu University, Kasugai, Aichi, Japan.
| | - Yasuo Suzuki
- College of Life and Health Sciences, Chubu University, Kasugai, Aichi, Japan
| |
Collapse
|
32
|
Jeyaram RA, Radha CA, Gromiha MM, Veluraja K. Design of fluorinated sialic acid analog inhibitor to H5 hemagglutinin of H5N1 influenza virus through molecular dynamics simulation study. J Biomol Struct Dyn 2019; 38:3504-3513. [PMID: 31594458 DOI: 10.1080/07391102.2019.1677500] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Influenza epidemics and pandemics are caused by influenza A virus. The cell surface protein of hemagglutinin and neuraminidase is responsible for viral infection and release of progeny virus on the host cell membrane. Now 18 hemagglutinin and 11 neuraminidase subtypes are identified. The avian influenza virus of H5N1 is an emergent threat to public health issues. To control the influenza viral infection it is necessary to develop antiviral inhibitors and vaccination. In the present investigation we carried out 50 ns Molecular Dynamics simulation on H5 hemagglutinin of Influenza A virus H5N1 complexed with fluorinated sialic acid by substituting fluorine atoms at any two hydroxyls of sialic acid by considering combinatorial combination. The binding affinity between the protein-ligand complex system is investigated by calculating pair interaction energy and MM-PBSA binding free energy. All the complex structures are stabilized by hydrogen bonding interactions between the H5 protein and the ligand fluorinated sialic acid. It is concluded from all the analyses that the fluorinated complexes enhance the inhibiting potency against H5 hemagglutinin and the order of inhibiting potency is SIA-F9 ≫ SIA-F2 ≈ SIA-F7 ≈ SIA-F2F4 ≈ SIA-F2F9 ≈ SIA-F7F9 > SIA-F7F8 ≈ SIA-F2F8 ≈ SIA-F8F9 > SIA-F4 ≈ SIA-F4F7 ≈ SIA-F4F8 ≈ SIA-F8 ≈ SIA-F2F7 ≈ SIA > SIA-F4F9. This study suggests that one can design the inhibitor by using the mono fluorinated models SIA-F9, SIA-F2 and SIA-F7 and difluorinated models SIA-F2F4, SIA-F2F9 and SIA-F7F9 to inhibit H5 of H5N1 to avoid Influenza A viral infection.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- R A Jeyaram
- Research Laboratory of Molecular Biophysics, Department of Physics, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - C Anu Radha
- Research Laboratory of Molecular Biophysics, Department of Physics, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - K Veluraja
- Research Laboratory of Molecular Biophysics, Department of Physics, School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
33
|
Novel Small Molecule Targeting the Hemagglutinin Stalk of Influenza Viruses. J Virol 2019; 93:JVI.00878-19. [PMID: 31167918 DOI: 10.1128/jvi.00878-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 06/02/2019] [Indexed: 11/20/2022] Open
Abstract
Combating influenza is one of the perennial global public health issues to be managed. Antiviral drugs are useful for the treatment of influenza in the absence of an appropriate vaccine. However, the appearance of resistant strains necessitates a constant search for new drugs. In this study, we investigated novel anti-influenza drug candidates using in vitro and in vivo assays. We identified anti-influenza hit compounds using a high-throughput screening method with a green fluorescent protein-tagged recombinant influenza virus. Through subsequent analyses of their cytotoxicity and pharmacokinetic properties, one candidate (IY7640) was selected for further evaluation. In a replication kinetics analysis, IY7640 showed greater inhibitory effects during the early phase of viral infection than the viral neuraminidase inhibitor oseltamivir. In addition, we observed that hemagglutinin (HA)-mediated membrane fusion was inhibited by IY7640 treatment, indicating that the HA stalk region, which is highly conserved across various (sub)types of influenza viruses, may be the molecular target of IY7640. In an escape mutant analysis in cells, amino acid mutations were identified at the HA stalk region of the 2009 pandemic H1N1 (pH1N1) virus. Even though the in vivo efficacy of IY7640 did not reach complete protection in a lethal challenge study in mice, these results suggest that IY7640 has potential to be developed as a new type of anti-influenza drug.IMPORTANCE Anti-influenza drugs with broad-spectrum efficacy against antigenically diverse influenza viruses can be highly useful when no vaccines are available. To develop new anti-influenza drugs, we screened a number of small molecules and identified a strong candidate, IY7640. When added at the time of or after influenza virus infection, IY7640 was observed to successfully inhibit or reduce viral replication in cells. We subsequently discovered that IY7640 targets the stalk region of the influenza HA protein, which exhibits a relatively high degree of amino acid sequence conservation across various (sub)types of influenza viruses. Furthermore, IY7640 was observed to block HA-mediated membrane fusion of H1N1, H3N2, and influenza B viruses in cells. Although it appears less effective against strains other than H1N1 subtype viruses in a challenge study in mice, we suggest that the small molecule IY7640 has potential to be optimized as a new anti-influenza drug.
Collapse
|
34
|
Chen F, Liu T, Xu J, Huang Y, Liu S, Yang J. Key amino acid residues of neuraminidase involved in influenza A virus entry. Pathog Dis 2019; 77:5614974. [PMID: 31702775 DOI: 10.1093/femspd/ftz063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 11/07/2019] [Indexed: 11/13/2022] Open
Abstract
Generally, influenza virus neuraminidase (NA) plays a critical role in the release stage of influenza virus. Recently, it has been found that NA may promote influenza virus to access the target cells. However, the mechanism remain unclear. Here, we reported that peramivir indeed possessed anti-influenza A virus (IAV) activity in the stage of viral entry. Importantly, we verified the critical residues of influenza NA involved in the viral entry. As a result, peramivir as an efficient NA inhibitor could suppress the initiation of IAV infection. Furthermore, mutational analysis showed NA might be associated with viral entry via amino acids residues R118, E119, D151, R152, W178, I222, E227, E276, R292 and R371. Our results demonstrated NA must contain the key amino acid residues can involve in IAV entry.
Collapse
Affiliation(s)
- Fangzhao Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Teng Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Jiagui Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Yingna Huang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Jie Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| |
Collapse
|
35
|
Pshenichnaya NY, Bulgakova VA, Lvov NI, Poromov AA, Selkova EP, Grekova AI, Shestakova IV, Maleev VV, Leneva IA. Clinical efficacy of umifenovir in influenza and ARVI (study ARBITR). TERAPEVT ARKH 2019; 91:56-63. [PMID: 31094461 DOI: 10.26442/00403660.2019.03.000127] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
AIM The aim of the study is to obtain additional data on safety and therapeutic efficacy of the antiviral drug Arbidol (umifenovir) in patients with a diagnosis of influenza and common cold. MATERIALS AND METHODS Double-blind, randomized, placebo-controlled clinical study investigating efficacy and safety of Arbidol (umifenovir) in Treatment and Prophylaxis of Influenza and Common Cold (ARBITR) IV phase started in November 2011 and completed in April 2016 on the basis of 15 research centers in various regions of the Russian Federation. A total of 359 patients, aged 18 to 65 years with influenza or acute respiratory tract infection, of no more than 36 hours' duration were enrolled in the study. Patients were randomized into two groups: a group of patients (therapy group) treated by Arbidol (umifenovir) at a dosage of 800 mg/day (2 capsules) for 5 days (n=181), and a group of patients receiving placebo 4 times a day for 5 days (n=178). The primary outcome measures of the study were the duration of clinical illness among patients with common cold and influenza/ARVI, the duration and severity of the main symptoms. Number of clinical complications associated with influenza and common cold was assessed as a secondary outcome. Safety was assessed by analyzing number of adverse events that are probably or definitely related to Arbidol, assessing vital signs, examining the physical condition of patients and general clinical laboratory parameters. RESULTS In the group treated by umifenovir, the number of full recover patients on the 4th day from the disease onset were significantly differed from the number of such cases in the placebo group. The number of cases of complete recovery after 96 hours was 98 patients (54.1%) and 77 (43.3%), p<0.05, and after 108 hours - 117 (64.6%) and 98 (55.1%), p<0.05. Duration of intoxication was reduced with umifenovir compared to placebo, amounted to 77.76 and 88.91 hours, respectively, p=0.013. The duration of all intoxication syndrome symptoms was also lower in the group receiving umifenovir. Thus, in the therapy group and placebo group, these parameters were respectively: fever duration - 67.96 and 75.32 hours (p=0.037), muscle pain - 52.23 and 59.08 hours (p=0.023), headache - 52.78 and 63.28 hours (p=0.013), weakness - 76.90 and 88.89 hours (p=0.008). The incidence of complications in the umifenovir group was 3.8%, in the placebo group 5.62%. Cases of acute tracheobronchitis was an increase in the placebo group (p<0.02). Umifenovir and placebo were well tolerated. A total of 42 cases of adverse events were registered in 11 patients in the treatment group and in 18 patients in the placebo group, which were not associated with umifenovir or placebo. CONCLUSION The results of this study indicate umifenovir safety and confirm its effectiveness to the treatment of influenza and other acute respiratory viral infections in adult patients. It was found that effect of umifenovir in the treatment of influenza in adults is most pronounced in the acute stage of the disease and appears in the reduction of time to resolution of all symptoms of the disease, reducing the severity of symptoms of the disease.
Collapse
Affiliation(s)
- N Yu Pshenichnaya
- Central Research Institute of Epidemiology, Russian Federal Service for Supervision of Consumer Rights Protection and Human Welfare, Moscow, Russia
| | - V A Bulgakova
- Children's Health Research Centre, Ministry of Health of Russia, Moscow, Russia
| | - N I Lvov
- S.M. Kirov Military Medical Academy, Ministry of Defense of the Russian Federation, Saint Petersburg, Russia
| | - A A Poromov
- M.V. Lomonosov Moscow State University, Moscow, Russia
| | - E P Selkova
- G.N. Gabrichevsky Moscow Research Institute of Epidemiology and Microbiology, Russian Federal Service for Supervision of Consumer Rights Protection and Human Welfare, Moscow, Russia
| | - A I Grekova
- Smolensk State Medical University, Ministry of Health of Russia, Smolensk, Russia
| | - I V Shestakova
- A.I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of Russia, Moscow, Russia
| | - V V Maleev
- Central Research Institute of Epidemiology, Russian Federal Service for Supervision of Consumer Rights Protection and Human Welfare, Moscow, Russia
| | - I A Leneva
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia
| |
Collapse
|
36
|
Beigel JH, Nam HH, Adams PL, Krafft A, Ince WL, El-Kamary SS, Sims AC. Advances in respiratory virus therapeutics - A meeting report from the 6th isirv Antiviral Group conference. Antiviral Res 2019; 167:45-67. [PMID: 30974127 PMCID: PMC7132446 DOI: 10.1016/j.antiviral.2019.04.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/06/2019] [Indexed: 12/11/2022]
Abstract
The International Society for Influenza and other Respiratory Virus Diseases held its 6th Antiviral Group (isirv-AVG) conference in Rockville, Maryland, November 13-15, 2018. The three-day program was focused on therapeutics towards seasonal and pandemic influenza, respiratory syncytial virus, coronaviruses including MERS-CoV and SARS-CoV, human rhinovirus, and other respiratory viruses. Updates were presented on several influenza antivirals including baloxavir, CC-42344, VIS410, immunoglobulin, immune plasma, MHAA4549A, pimodivir (JNJ-63623872), umifenovir, and HA minibinders; RSV antivirals including presatovir (GS-5806), ziresovir (AK0529), lumicitabine (ALS-008176), JNJ-53718678, JNJ-64417184, and EDP-938; broad spectrum antivirals such as favipiravir, VH244, remdesivir, and EIDD-1931/EIDD-2801; and host directed strategies including nitazoxanide, eritoran, and diltiazem. Other topics included considerations of novel endpoints such as ordinal scales and patient reported outcomes (PRO), and study design issues, and other regulatory considerations for antiviral drug development. The aim of this report is to provide a summary of the presentations given at this meeting.
Collapse
Affiliation(s)
- John H Beigel
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Hannah H Nam
- (b)Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Peter L Adams
- Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS), Washington, DC, USA
| | - Amy Krafft
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - William L Ince
- Division of Antiviral Products, Office of Antimicrobial Products, Office of New Drugs, Center for Drug Evaluation and Research, U.S Food and Drug Administration, Silver Spring, MD, USA
| | - Samer S El-Kamary
- Division of Antiviral Products, Office of Antimicrobial Products, Office of New Drugs, Center for Drug Evaluation and Research, U.S Food and Drug Administration, Silver Spring, MD, USA
| | - Amy C Sims
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
37
|
Zhang Y, Xu C, Zhang H, Liu GD, Xue C, Cao Y. Targeting Hemagglutinin: Approaches for Broad Protection against the Influenza A Virus. Viruses 2019; 11:v11050405. [PMID: 31052339 PMCID: PMC6563292 DOI: 10.3390/v11050405] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/26/2019] [Accepted: 04/27/2019] [Indexed: 12/13/2022] Open
Abstract
Influenza A viruses are dynamically epidemic and genetically diverse. Due to the antigenic drift and shift of the virus, seasonal vaccines are required to be reformulated annually to match with current circulating strains. However, the mismatch between vaccinal strains and circulating strains occurs frequently, resulting in the low efficacy of seasonal vaccines. Therefore, several “universal” vaccine candidates based on the structure and function of the hemagglutinin (HA) protein have been developed to meet the requirement of a broad protection against homo-/heterosubtypic challenges. Here, we review recent novel constructs and discuss several important findings regarding the broad protective efficacy of HA-based universal vaccines.
Collapse
Affiliation(s)
- Yun Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Cong Xu
- Research Center of Agricultural of Dongguan City, Dongguan 523086, China.
| | - Hao Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - George Dacai Liu
- Firstline Biopharmaceuticals Corporation, 12,050 167th PL NE, Redmond, WA 98052, USA.
| | - Chunyi Xue
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Yongchang Cao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
38
|
Yang J, Huang Y, Liu S. Investigational antiviral therapies for the treatment of influenza. Expert Opin Investig Drugs 2019; 28:481-488. [PMID: 31018720 DOI: 10.1080/13543784.2019.1606210] [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] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Influenza viral ribonucleoprotein complexes (vRNPs) play a key role in viral transcription and replication; hence, the recent development of novel anti-influenza drugs targeting vRNPs has garnered widespread interest. AREAS COVERED We discuss the function of the constituents of vRNPs and summarize those vRNPs-targeted synthetic drugs that are in preclinical and early clinical development. EXPERT OPINION vRNPs contain high-value drug targets; such targets include the subunits PA, PB1, PB2, and NP. Developing a new generation of antiviral therapies with strategies that utilize existing drugs, natural compounds originated from new resources and novel drug combinations may open up new therapeutic approaches to influenza.
Collapse
Affiliation(s)
- Jie Yang
- a Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou , China
| | - Yingna Huang
- a Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou , China
| | - Shuwen Liu
- a Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou , China.,b State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology , Southern Medical University , Guangzhou , China
| |
Collapse
|
39
|
Han L, Chen C, Han X, Lin S, Ao X, Han X, Wang J, Ye H. Structural Insights for Anti-Influenza Vaccine Design. Comput Struct Biotechnol J 2019; 17:475-483. [PMID: 31007873 PMCID: PMC6458449 DOI: 10.1016/j.csbj.2019.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/17/2019] [Accepted: 03/21/2019] [Indexed: 01/29/2023] Open
Abstract
Influenza A virus are a persistent and significant threat to human health, and current vaccines do not provide sufficient protection due to antigenic drift, which allows influenza viruses to easily escape immune surveillance and antiviral drug activity. Influenza hemagglutinin (HA) is a glycoprotein needed for the entry of enveloped influenza viruses into host cells and is a potential target for anti-influenza humoral immune responses. In recent years, a number of broadly neutralizing antibodies (bnAbs) have been isolated, and their relative structural information obtained from the crystallization of influenza antigens in complex with bnAbs has provided some new insights into future influenza vaccine research. Here, we review the current knowledge of the HA-targeted bnAbs and the structure-based mechanisms contributing to neutralization. We also discuss the potential for this structure-based approach to overcome the challenge of obtaining a highly desired "universal" influenza vaccine, especially on small proteins and peptides.
Collapse
Affiliation(s)
- Lifen Han
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, China
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Cong Chen
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Xianlin Han
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Shujin Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, China
| | - Xiulan Ao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, China
| | - Xiao Han
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Jianmin Wang
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, China
| | - Hanhui Ye
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, China
| |
Collapse
|
40
|
Lu W, Pieters RJ. Carbohydrate–protein interactions and multivalency: implications for the inhibition of influenza A virus infections. Expert Opin Drug Discov 2019; 14:387-395. [DOI: 10.1080/17460441.2019.1573813] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Wenjing Lu
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Roland J. Pieters
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
41
|
Li H, Li M, Xu R, Wang S, Zhang Y, Zhang L, Zhou D, Xiao S. Synthesis, structure activity relationship and in vitro anti-influenza virus activity of novel polyphenol-pentacyclic triterpene conjugates. Eur J Med Chem 2019; 163:560-568. [DOI: 10.1016/j.ejmech.2018.12.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/30/2018] [Accepted: 12/02/2018] [Indexed: 01/11/2023]
|
42
|
Szűcs Z, Kelemen V, Le Thai S, Csávás M, Rőth E, Batta G, Stevaert A, Vanderlinden E, Naesens L, Herczegh P, Borbás A. Structure-activity relationship studies of lipophilic teicoplanin pseudoaglycon derivatives as new anti-influenza virus agents. Eur J Med Chem 2018; 157:1017-1030. [PMID: 30170320 PMCID: PMC7115582 DOI: 10.1016/j.ejmech.2018.08.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 02/03/2023]
Abstract
Six series of semisynthetic lipophilic glycopeptide antibiotic derivatives were evaluated for in vitro activity against influenza A and B viruses. The new teicoplanin pseudoaglycon-derived lipoglycopeptides were prepared by coupling one or two side chains to the N-terminus of the glycopeptide core, using various conjugation methods. Three series of derivatives bearing two lipophilic groups were synthesized by attaching bis-alkylthio maleimides directly or through linkers of different lengths to the glycopeptide. Access to the fourth and fifth series of compounds was achieved by click chemistry, introducing single alkyl/aryl chains directly or through a tetraethylene glycol linker to the same position. A sixth group of semisynthetic derivatives was obtained by sulfonylation of the N-terminus. Of the 42 lipophilic teicoplanin pseudoaglycon derivatives tested, about half showed broad activity against influenza A and B viruses, with some of them having reasonable or no cytotoxicity. Minor differences in the side chain length as well as lipophilicity appeared to have significant impact on antiviral activity and cytotoxicity. Several lipoglycopeptides were also found to be active against human coronavirus. Multiple series of lipophilic teicoplanin pseudoaglycon derivatives were prepared. Alkyl or aryl chains were coupled to the N-terminus by various conjugation methods. The activity of new antibiotic derivatives was evaluated against influenza viruses. Half of the 42 derivatives showed high activity against influenza A and B viruses. The length and lipophilicity of the side chains influence the antiviral activity.
Collapse
Affiliation(s)
- Zsolt Szűcs
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Viktor Kelemen
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Son Le Thai
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Magdolna Csávás
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Erzsébet Rőth
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Gyula Batta
- Department of Organic Chemistry, University of Debrecen, H-4032 Debrecen, Hungary
| | - Annelies Stevaert
- Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium
| | | | - Lieve Naesens
- Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium.
| | - Pál Herczegh
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary.
| | - Anikó Borbás
- Department of Pharmaceutical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary.
| |
Collapse
|
43
|
Chen X, Liu S, Goraya MU, Maarouf M, Huang S, Chen JL. Host Immune Response to Influenza A Virus Infection. Front Immunol 2018; 9:320. [PMID: 29556226 PMCID: PMC5845129 DOI: 10.3389/fimmu.2018.00320] [Citation(s) in RCA: 304] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 02/05/2018] [Indexed: 12/25/2022] Open
Abstract
Influenza A viruses (IAVs) are contagious pathogens responsible for severe respiratory infection in humans and animals worldwide. Upon detection of IAV infection, host immune system aims to defend against and clear the viral infection. Innate immune system is comprised of physical barriers (mucus and collectins), various phagocytic cells, group of cytokines, interferons (IFNs), and IFN-stimulated genes, which provide first line of defense against IAV infection. The adaptive immunity is mediated by B cells and T cells, characterized with antigen-specific memory cells, capturing and neutralizing the pathogen. The humoral immune response functions through hemagglutinin-specific circulating antibodies to neutralize IAV. In addition, antibodies can bind to the surface of infected cells and induce antibody-dependent cell-mediated cytotoxicity or complement activation. Although there are neutralizing antibodies against the virus, cellular immunity also plays a crucial role in the fight against IAVs. On the other hand, IAVs have developed multiple strategies to escape from host immune surveillance for successful replication. In this review, we discuss how immune system, especially innate immune system and critical molecules are involved in the antiviral defense against IAVs. In addition, we highlight how IAVs antagonize different immune responses to achieve a successful infection.
Collapse
Affiliation(s)
- Xiaoyong Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Shasha Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mohsan Ullah Goraya
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Mohamed Maarouf
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Ji-Long Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| |
Collapse
|
44
|
Cheng HW, Wang HW, Wong TY, Yeh HW, Chen YC, Liu DZ, Liang PH. Synthesis of S-linked NeuAc-α(2-6)-di-LacNAc bearing liposomes for H1N1 influenza virus inhibition assays. Bioorg Med Chem 2018; 26:2262-2270. [PMID: 29472127 DOI: 10.1016/j.bmc.2018.02.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/07/2018] [Accepted: 02/10/2018] [Indexed: 10/18/2022]
Abstract
S-NeuAc-α(2-6)-di-LacNAc (5) was efficiently synthesized by a [2+2] followed by a [1+4] glycosylation, and later conjugated with 1,2-dilauroyl-sn-glycero-3-phosphoethanolamine (DLPE) to form both single-layer and multi-layer homogeneous liposomes in the presence of dipalmitoyl phosphatidylcholine (DPPC) and cholesterol. These liposomes were found to be weak inhibitors in both the influenza virus entry assay and the hemagglutination inhibition assay. The single layer liposome was found to more efficiently interfere with the entry of the H1N1 influenza virus into MDCK cells than the multilayer liposome containing 5.
Collapse
Affiliation(s)
- Hou-Wen Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Hsiao-Wen Wang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Tsung-Yun Wong
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Hsien-Wei Yeh
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yi-Chun Chen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Der-Zen Liu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan; Medical and Pharmaceutical Industry Technology and Development Center, Academia Sinica, Taipei 128, Taiwan
| | - Pi-Hui Liang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan; The Genomics Research Center, Academia Sinica, Taipei 128, Taiwan.
| |
Collapse
|
45
|
The amyloidogenicity of the influenza virus PB1-derived peptide sheds light on its antiviral activity. Biophys Chem 2018; 234:16-23. [PMID: 29328990 DOI: 10.1016/j.bpc.2018.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/05/2018] [Accepted: 01/06/2018] [Indexed: 01/08/2023]
Abstract
The influenza virus polymerase complex is a promising target for new antiviral drug development. It is known that, within the influenza virus polymerase complex, the PB1 subunit region from the 1st to the 25th amino acid residues has to be is in an alpha-helical conformation for proper interaction with the PA subunit. We have previously shown that PB1(6-13) peptide at low concentrations is able to interact with the PB1 subunit N-terminal region in a peptide model which shows aggregate formation and antiviral activity in cell cultures. In this paper, it was shown that PB1(6-13) peptide is prone to form the amyloid-like fibrillar aggregates. The peptide homo-oligomerization kinetics were examined, and the affinity and characteristic interaction time of PB1(6-13) peptide monomers and the influenza virus polymerase complex PB1 subunit N-terminal region were evaluated by the SPR and TR-SAXS methods. Based on the data obtained, a hypothesis about the PB1(6-13) peptide mechanism of action was proposed: the peptide in its monomeric form is capable of altering the conformation of the PB1 subunit N-terminal region, causing a change from an alpha helix to a beta structure. This conformational change disrupts PB1 and PA subunit interaction and, by that mechanism, the peptide displays antiviral activity.
Collapse
|
46
|
Ustinov NB, Zavyalova EG, Smirnova IG, Kopylov AM. The Power and Limitations of Influenza Virus Hemagglutinin Assays. BIOCHEMISTRY (MOSCOW) 2018; 82:1234-1248. [PMID: 29223151 DOI: 10.1134/s0006297917110025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Influenza virus hemagglutinins (HAs) are surface proteins that bind to sialic acid residues at the host cell surface and ensure further virus internalization. Development of methods for the inhibition of these processes drives progress in the design of new antiviral drugs. The state of the isolated HA (i.e. combining tertiary structure and extent of oligomerization) is defined by multiple factors, like the HA source and purification method, posttranslational modifications, pH, etc. The HA state affects HA functional activity and significantly impacts the results of numerous HA assays. In this review, we analyze the power and limitations of currently used HA assays regarding the state of HA.
Collapse
Affiliation(s)
- N B Ustinov
- Lomonosov Moscow State University, Faculty of Chemistry, Moscow, 119991, Russia.
| | | | | | | |
Collapse
|
47
|
Ding Y, Chen L, Wu W, Yang J, Yang Z, Liu S. Andrographolide inhibits influenza A virus-induced inflammation in a murine model through NF-κB and JAK-STAT signaling pathway. Microbes Infect 2017; 19:605-615. [PMID: 28889969 DOI: 10.1016/j.micinf.2017.08.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 08/22/2017] [Accepted: 08/25/2017] [Indexed: 12/22/2022]
Abstract
Influenza viruses, the main cause of respiratory tract diseases, cause high morbidity and mortality in humans. Excessive inflammation in the lungs is proposed to be a hallmark for the severe influenza virus infection, especially influenza A virus infection. Strategies against inflammation induced by influenza A virus infection could be a potential anti-influenza therapy. Here, lethal dose of mouse-adapted H1N1 strain PR8A/PR/8/34 was inoculated C57BL/6 mice to detect the anti-influenza activity of andrographolide, the active component of traditional Chinese medicinal herb Andrographis paniculata, with or without influenza virus entry inhibitor CL-385319. Treatment was initiated on 4 days after infection. The survival rate, body weight, lung pathology, viral loads, cytokine expression were monitored in 14 days post inoculation. The combination group had the highest survival rate. Andrographolide treatment could increase the survival rate, diminish lung pathology, decrease the virus loads and the inflammatory cytokines expression induced by infection. Mechanism studies showed the NF-κB and JAK-STAT signaling pathway were involved in the activity of andrographolide. In conclusion, combination of virus entry inhibitor with immunomodulator might be a promising therapeutic approach for influenza.
Collapse
Affiliation(s)
- Yi Ding
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Lizhu Chen
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wenjiao Wu
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jie Yang
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, National Clinical Centre of Respiratory Disease, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shuwen Liu
- Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China; State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
48
|
Lactoferrin-derived Peptides Active towards Influenza: Identification of Three Potent Tetrapeptide Inhibitors. Sci Rep 2017; 7:10593. [PMID: 28878220 PMCID: PMC5587682 DOI: 10.1038/s41598-017-10492-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/09/2017] [Indexed: 11/08/2022] Open
Abstract
Bovine lactoferrin is a biglobular multifunctional iron binding glycoprotein that plays an important role in innate immunity against infections. We have previously demonstrated that selected peptides from bovine lactoferrin C-lobe are able to prevent both Influenza virus hemagglutination and cell infection. To deeper investigate the ability of lactoferrin derived peptides to inhibit Influenza virus infection, in this study we identified new bovine lactoferrin C-lobe derived sequences and corresponding synthetic peptides were synthesized and assayed to check their ability to prevent viral hemagglutination and infection. We identified three tetrapeptides endowed with broad anti-Influenza activity and able to inhibit viral infection in a concentration range femto- to picomolar. Our data indicate that these peptides may constitute a non-toxic tool for potential applications as anti-Influenza therapeutics.
Collapse
|