1
|
Hebditch M, Warwicker J. Protein-sol pKa: prediction of electrostatic frustration, with application to coronaviruses. Bioinformatics 2020; 36:5112-5114. [PMID: 32683439 PMCID: PMC7454282 DOI: 10.1093/bioinformatics/btaa646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/18/2020] [Accepted: 07/13/2020] [Indexed: 12/02/2022] Open
Abstract
Motivation Evolution couples differences in ambient pH to biological function through protonatable groups, in particular, those that switch from buried to exposed and alter protonation state in doing so. We present a tool focusing on structure-based discovery and display of these groups. Results Since prediction of buried group pKas is computationally intensive, solvent accessibility of ionizable groups is displayed, from which the user can iteratively select pKa calculation centers. Results are color-coded, with emphasis on buried groups. Utility is demonstrated with benchmarking against known pH sensing sites in influenza virus hemagglutinin and in variants of murine hepatitis virus, a coronavirus. A pair of histidine residues, which are conserved in coronavirus spike proteins, are predicted to be electrostatically frustrated at acidic pH in both pre- and post-fusion conformations. We suggest that an intermediate expanded conformation at endosomal pH could relax the frustration, allowing histidine protonation and facilitating conformational conversion of coronavirus spike protein. Availability and implementation This tool is available at http://www.protein-sol.manchester.ac.uk/pka/.
Collapse
Affiliation(s)
- Max Hebditch
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Institute of Biotechnology, Manchester M1 7DN, UK
| | - Jim Warwicker
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Institute of Biotechnology, Manchester M1 7DN, UK
| |
Collapse
|
2
|
Pattnaik GP, Chakraborty H. Entry Inhibitors: Efficient Means to Block Viral Infection. J Membr Biol 2020; 253:425-444. [PMID: 32862236 PMCID: PMC7456447 DOI: 10.1007/s00232-020-00136-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022]
Abstract
The emerging and re-emerging viral infections are constant threats to human health and wellbeing. Several strategies have been explored to develop vaccines against these viral diseases. The main effort in the journey of development of vaccines is to neutralize the fusion protein using antibodies. However, significant efforts have been made in discovering peptides and small molecules that inhibit the fusion between virus and host cell, thereby inhibiting the entry of viruses. This class of inhibitors is called entry inhibitors, and they are extremely efficient in reducing viral infection as the entry of the virus is considered as the first step of infection. Nevertheless, these inhibitors are highly selective for a particular virus as antibody-based vaccines. The recent COVID-19 pandemic lets us ponder to shift our attention towards broad-spectrum antiviral agents from the so-called ‘one bug-one drug’ approach. This review discusses peptide and small molecule-based entry inhibitors against class I, II, and III viruses and sheds light on broad-spectrum antiviral agents.
Collapse
Affiliation(s)
| | - Hirak Chakraborty
- School of Chemistry, Sambalpur University, Jyoti Vihar, Burla, Odisha, 768 019, India. .,Centre of Excellence in Natural Products and Therapeutics, Sambalpur University, Jyoti Vihar, Burla, Odisha, 768 019, India.
| |
Collapse
|
3
|
Göktaş F, Özbil M, Cesur N, Vanderlinden E, Naesens L, Cesur Z. Novel
N
‐(1‐thia‐4‐azaspiro[4.5]decan‐4‐yl)carboxamide derivatives as potent and selective influenza virus fusion inhibitors. Arch Pharm (Weinheim) 2019; 352:e1900028. [DOI: 10.1002/ardp.201900028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 11/07/2022]
Affiliation(s)
- Füsun Göktaş
- Department of Pharmaceutical Chemistry Faculty of Pharmacy, Istanbul University Istanbul Turkey
| | - Mehmet Özbil
- Department of Molecular Biology and Genetics Istanbul Arel University Istanbul Turkey
| | - Nesrin Cesur
- Department of Pharmaceutical Chemistry Faculty of Pharmacy, Istanbul University Istanbul Turkey
| | - Evelien Vanderlinden
- Department of Microbiology, Immunology and Transplantation Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research KU Leuven Leuven Belgium
| | - Lieve Naesens
- Department of Microbiology, Immunology and Transplantation Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research KU Leuven Leuven Belgium
| | - Zafer Cesur
- Department of Pharmaceutical Chemistry Faculty of Pharmacy, Istanbul University Istanbul Turkey
| |
Collapse
|
4
|
Kannan S, Shankar R, Kolandaivel P. Insights into structural and inhibitory mechanisms of low pH-induced conformational change of influenza HA2 protein: a computational approach. J Mol Model 2019; 25:99. [PMID: 30904969 DOI: 10.1007/s00894-019-3982-y] [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] [Received: 10/10/2018] [Accepted: 03/05/2019] [Indexed: 01/26/2023]
Abstract
Though oseltamivir and zanamivir are the active anti-influenza drugs, the emergence of different strains of influenza A virus with mutations creates drug-resistance to these drugs. Therefore, it is essential to find a suitable approach to stop the viral infection. The present study focuses on understanding the conformational changes of the HA2 protein at different pH levels (pH 7, pH 6, pH 5) and on blocking the low pH-induced conformational changes of the HA2 protein with a suitable ligand using molecular docking and molecular dynamics (MD) simulation methods. As the pH value decreases to pH 5, the protein undergoes large conformational changes with less stability in the order of pH 7 > pH 6 > pH 5. The fusion peptide (residues 1-20) and the extended loop (residues 58-75) deviate more at pH 5. The ligand stachyflin bound between the N- and C-terminal helix regions retains the stability of the HA2 protein at pH 5 and blocks the low pH-induced conformational transition. The performance of stachyflin is increased when it directly interacts with residues at the intramonomer binding site rather than the intermonomer binding site. The susceptibility of the HA2 protein of different subtypes to stachyflin is in the order of H1 > H7 > H5 > H2 > H3. Stachflin has a higher binding affinity for H1 (at pH 7, pH 6, pH 5) and H7 subtypes than others. Lys47, Lys58, and Glu103 are the key residues that favor the binding and highly stabilize the HA2 protein at low pH. Graphical abstract Low pH-induced conformational change of influenza HA2 protein.
Collapse
Affiliation(s)
- S Kannan
- Department of Physics, Bharathiar University, Coimbatore, 641 046, India
| | - R Shankar
- Department of Physics, Bharathiar University, Coimbatore, 641 046, India
| | - P Kolandaivel
- Department of Physics, Bharathiar University, Coimbatore, 641 046, India. .,Periyar University, Salem, 636 011, India.
| |
Collapse
|
5
|
Abstract
Various viral diseases, such as acquired immunodeficiency syndrome, influenza, and hepatitis, have emerged as leading causes of human death worldwide. Scientific endeavor since invention of DNA-dependent RNA polymerase of pox virus in 1967 resulted in better understanding of virus replication and development of various novel therapeutic strategies. Despite considerable advancement in every facet of drug discovery process, development of commercially viable, safe, and effective drugs for these viruses still remains a big challenge. Decades of intense research yielded a handful of natural and synthetic therapeutic options. But emergence of new viruses and drug-resistant viral strains had made new drug development process a never-ending battle. Small-molecule fungal metabolites due to their vast diversity, stereochemical complexity, and preapproved biocompatibility always remain an attractive source for new drug discovery. Though, exploration of therapeutic importance of fungal metabolites has started early with discovery of penicillin, recent prediction asserted that only a small percentage (5-10%) of fungal species have been identified and much less have been scientifically investigated. Therefore, exploration of new fungal metabolites, their bioassay, and subsequent mechanistic study bears huge importance in new drug discovery endeavors. Though no fungal metabolites so far approved for antiviral treatment, many of these exhibited high potential against various viral diseases. This review comprehensively discussed about antiviral activities of fungal metabolites of diverse origin against some important viral diseases. This also highlighted the mechanistic details of inhibition of viral replication along with structure-activity relationship of some common and important classes of fungal metabolites.
Collapse
Affiliation(s)
- Biswajit G Roy
- Department of Chemistry, Sikkim University, Gangtok, India
| |
Collapse
|
6
|
Wu X, Wu X, Sun Q, Zhang C, Yang S, Li L, Jia Z. Progress of small molecular inhibitors in the development of anti-influenza virus agents. Am J Cancer Res 2017; 7:826-845. [PMID: 28382157 PMCID: PMC5381247 DOI: 10.7150/thno.17071] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/18/2016] [Indexed: 02/05/2023] Open
Abstract
The influenza pandemic is a major threat to human health, and highly aggressive strains such as H1N1, H5N1 and H7N9 have emphasized the need for therapeutic strategies to combat these pathogens. Influenza anti-viral agents, especially active small molecular inhibitors play important roles in controlling pandemics while vaccines are developed. Currently, only a few drugs, which function as influenza neuraminidase (NA) inhibitors and M2 ion channel protein inhibitors, are approved in clinical. However, the acquired resistance against current anti-influenza drugs and the emerging mutations of influenza virus itself remain the major challenging unmet medical needs for influenza treatment. It is highly desirable to identify novel anti-influenza agents. This paper reviews the progress of small molecular inhibitors act as antiviral agents, which include hemagglutinin (HA) inhibitors, RNA-dependent RNA polymerase (RdRp) inhibitors, NA inhibitors and M2 ion channel protein inhibitors etc. Moreover, we also summarize new, recently reported potential targets and discuss strategies for the development of new anti-influenza virus drugs.
Collapse
|
7
|
Zeng LY, Yang J, Liu S. Investigational hemagglutinin-targeted influenza virus inhibitors. Expert Opin Investig Drugs 2016; 26:63-73. [PMID: 27918208 DOI: 10.1080/13543784.2017.1269170] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Seasonal influenza and pandemic outbreaks typically result in high mortality and morbidity associated with severe economic burdens. Vaccines and anti-influenza drugs have made great contributions to control the infection. However, antigenic drifts and shifts allow influenza viruses to easily escape immune neutralization and antiviral drug activity. Hemagglutinin (HA)is an important envelope protein for the entry of influenza viruses into host cells, thus, HA-targeted agents may be potential anti-influenza drugs. Areas covered: In this review, we describe arbidol, a unique licensed drug targeting HA; discuss and summarize HA-targeted anti-influenza agents been tested before or being tested currently in clinical trials, including monoclonal antibodies, small molecule inhibitors, proteins and peptides. Other small molecule inhibitors are also briefly introduced. Expert opinion: Exploring new clinical applications for existing drugs can provide additional anti-influenza candidates with promising safety and bioavailability, and largely shortened time and costs. To enhance therapeutic efficacy and avoid drug-resistance, combination therapy involving in HA-targeted anti-influenza agent is reasonable and attractive. For drug discovery, it is helpful to keep an eye on the development of methodology in organic synthesis and probe into the co-crystal structure of HA in complex with small molecule.
Collapse
Affiliation(s)
- Li-Yan Zeng
- a Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences , Southern Medical University , Guangzhou , China
| | - Jie Yang
- 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
|
8
|
Igarashi M. [Antiviral Drugs Targeting Influenza Virus Surface Proteins: A Computational Structural Biology Approach]. YAKUGAKU ZASSHI 2016; 135:1015-21. [PMID: 26329546 DOI: 10.1248/yakushi.15-00175-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
For the prevention and control of infectious viral diseases, vaccines and antiviral drugs targeting viral proteins are of great importance. Amino acid substitutions in viral proteins occasionally cause the emergence of antibody-escape and drug-resistant mutants. With regard to this, we have studied the proteins of several viruses, especially the influenza A virus, by using techniques of computational chemistry and biology such as molecular modeling, molecular docking, and molecular dynamics simulations. Influenza A virus is a zoonotic pathogen that is transmitted from animals to humans. This virus has two surface glycoproteins, hemagglutinin (HA) and neuraminidase (NA). The HA of influenza viruses plays a key role in the initiation of viral infection. And HA is also the major target of antibodies that neutralize viral infectivity. Some amino acid substitutions in the antigenic epitope on HA could decrease the interaction between HA and antibodies, leading to the generation of antigenic variants with novel antigenic structures of HA. In addition, HA protein seems to be a favorable target for anti-influenza drugs, but effective HA inhibitors have not been developed due to the emergence of drug-resistant viruses with amino acid substitutions on the HA. To understand how amino acid substitutions affect changes in drug susceptibility, we have been computationally analyzing the three-dimensional structures of influenza virus proteins. In this paper, we review the results obtained through our current analysis.
Collapse
Affiliation(s)
- Manabu Igarashi
- Division of Global Epidemiology, Hokkaido University Research Center for Zoonosis Control
| |
Collapse
|
9
|
Yang J, Liu S, Du L, Jiang S. A new role of neuraminidase (NA) in the influenza virus life cycle: implication for developing NA inhibitors with novel mechanism of action. Rev Med Virol 2016; 26:242-50. [PMID: 27061123 PMCID: PMC7169148 DOI: 10.1002/rmv.1879] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 02/13/2016] [Accepted: 02/17/2016] [Indexed: 11/17/2022]
Abstract
The entire life cycle of influenza virus involves viral attachment, entry, replication, and release. Previous studies have demonstrated that neuraminidase (NA) is an essential glycoprotein on the surface of influenza virus and that it is responsible for release of progeny virions from the host cell to infect new cells. However, recent studies have also suggested that NA may play other roles in the early stages of the viral life cycle, that is, viral attachment and entry. This review focuses on the new role of NA in the early stages of influenza life cycle and the corresponding development of novel NA inhibitors. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jie Yang
- Key Lab of New Drug Screening of Guangdong Province, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Shuwen Liu
- Key Lab of New Drug Screening of Guangdong Province, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Shibo Jiang
- Key Lab of Medical Molecular Virology of MOE/MOH, Shanghai Medical College, Fudan University, Shanghai, China.,Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| |
Collapse
|
10
|
Quercetin as an Antiviral Agent Inhibits Influenza A Virus (IAV) Entry. Viruses 2015; 8:v8010006. [PMID: 26712783 PMCID: PMC4728566 DOI: 10.3390/v8010006] [Citation(s) in RCA: 242] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 11/25/2015] [Accepted: 11/30/2015] [Indexed: 12/22/2022] Open
Abstract
Influenza A viruses (IAVs) cause seasonal pandemics and epidemics with high morbidity and mortality, which calls for effective anti-IAV agents. The glycoprotein hemagglutinin of influenza virus plays a crucial role in the initial stage of virus infection, making it a potential target for anti-influenza therapeutics development. Here we found that quercetin inhibited influenza infection with a wide spectrum of strains, including A/Puerto Rico/8/34 (H1N1), A/FM-1/47/1 (H1N1), and A/Aichi/2/68 (H3N2) with half maximal inhibitory concentration (IC50) of 7.756 ± 1.097, 6.225 ± 0.467, and 2.738 ± 1.931 μg/mL, respectively. Mechanism studies identified that quercetin showed interaction with the HA2 subunit. Moreover, quercetin could inhibit the entry of the H5N1 virus using the pseudovirus-based drug screening system. This study indicates that quercetin showing inhibitory activity in the early stage of influenza infection provides a future therapeutic option to develop effective, safe and affordable natural products for the treatment and prophylaxis of IAV infections.
Collapse
|
11
|
Rowse M, Qiu S, Tsao J, Xian T, Khawaja S, Yamauchi Y, Yang Z, Wang G, Luo M. Characterization of potent fusion inhibitors of influenza virus. PLoS One 2015; 10:e0122536. [PMID: 25803288 PMCID: PMC4372562 DOI: 10.1371/journal.pone.0122536] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/15/2015] [Indexed: 12/16/2022] Open
Abstract
New inhibitors of influenza viruses are needed to combat the potential emergence of novel human influenza viruses. We have identified a class of small molecules that inhibit replication of influenza virus at picomolar concentrations in plaque reduction assays. The compound also inhibits replication of vesicular stomatitis virus. Time of addition and dilution experiments with influenza virus indicated that an early time point of infection was blocked and that inhibitor 136 tightly bound to virions. Using fluorescently labeled influenza virus, inhibition of viral fusion to cellular membranes by blocked lipid mixing was established as the mechanism of action for this class of inhibitors. Stabilization of the neutral pH form of hemagglutinin (HA) was ruled out by trypsin digestion studies in vitro and with conformation specific HA antibodies within cells. Direct visualization of 136 treated influenza virions at pH 7.5 or acidified to pH 5.0 showed that virions remain intact and that glycoproteins become disorganized as expected when HA undergoes a conformational change. This suggests that exposure of the fusion peptide at low pH is not inhibited but lipid mixing is inhibited, a different mechanism than previously reported fusion inhibitors. We hypothesize that this new class of inhibitors intercalate into the virus envelope altering the structure of the viral envelope required for fusion to cellular membranes.
Collapse
Affiliation(s)
- Michael Rowse
- Department of Microbiology, University of Alabama at Birmingham, 1025 18 Street South, Birmingham, Alabama, 35294, United States of America
| | - Shihong Qiu
- Department of Microbiology, University of Alabama at Birmingham, 1025 18 Street South, Birmingham, Alabama, 35294, United States of America
| | - Jun Tsao
- Department of Microbiology, University of Alabama at Birmingham, 1025 18 Street South, Birmingham, Alabama, 35294, United States of America
| | - Tongmei Xian
- Laboratory of Structural Biology, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Sarah Khawaja
- Institute of Biochemistry, ETH Zurich HPM E8.2, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Yohei Yamauchi
- Institute of Biochemistry, ETH Zurich HPM E8.2, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
- * E-mail: (YY); (ZY); (GXW); (ML)
| | - Zhen Yang
- Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
- * E-mail: (YY); (ZY); (GXW); (ML)
| | - Guoxin Wang
- Laboratory of Structural Biology, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
- * E-mail: (YY); (ZY); (GXW); (ML)
| | - Ming Luo
- Laboratory of Structural Biology, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
- Department of Chemistry, College of Arts and Sciences, Georgia State University, Atlanta, Georgia, 30302, United States of America
- * E-mail: (YY); (ZY); (GXW); (ML)
| |
Collapse
|
12
|
New small molecule entry inhibitors targeting hemagglutinin-mediated influenza a virus fusion. J Virol 2013; 88:1447-60. [PMID: 24198411 DOI: 10.1128/jvi.01225-13] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Influenza viruses are a major public health threat worldwide, and options for antiviral therapy are limited by the emergence of drug-resistant virus strains. The influenza virus glycoprotein hemagglutinin (HA) plays critical roles in the early stage of virus infection, including receptor binding and membrane fusion, making it a potential target for the development of anti-influenza drugs. Using pseudotype virus-based high-throughput screens, we have identified several new small molecules capable of inhibiting influenza virus entry. We prioritized two novel inhibitors, MBX2329 and MBX2546, with aminoalkyl phenol ether and sulfonamide scaffolds, respectively, that specifically inhibit HA-mediated viral entry. The two compounds (i) are potent (50% inhibitory concentration [IC50] of 0.3 to 5.9 μM); (ii) are selective (50% cytotoxicity concentration [CC(50)] of >100 μM), with selectivity index (SI) values of >20 to 200 for different influenza virus strains; (iii) inhibit a wide spectrum of influenza A viruses, which includes the 2009 pandemic influenza virus A/H1N1/2009, highly pathogenic avian influenza (HPAI) virus A/H5N1, and oseltamivir-resistant A/H1N1 strains; (iv) exhibit large volumes of synergy with oseltamivir (36 and 331 μM(2) % at 95% confidence); and (v) have chemically tractable structures. Mechanism-of-action studies suggest that both MBX2329 and MBX2546 bind to HA in a nonoverlapping manner. Additional results from HA-mediated hemolysis of chicken red blood cells (cRBCs), competition assays with monoclonal antibody (MAb) C179, and mutational analysis suggest that the compounds bind in the stem region of the HA trimer and inhibit HA-mediated fusion. Therefore, MBX2329 and MBX2546 represent new starting points for chemical optimization and have the potential to provide valuable future therapeutic options and research tools to study the HA-mediated entry process.
Collapse
|
13
|
Shen X, Zhang X, Liu S. Novel hemagglutinin-based influenza virus inhibitors. J Thorac Dis 2013; 5 Suppl 2:S149-59. [PMID: 23977436 DOI: 10.3978/j.issn.2072-1439.2013.06.14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 06/17/2013] [Indexed: 12/22/2022]
Abstract
Influenza virus has caused seasonal epidemics and worldwide pandemics, which caused tremendous loss of human lives and socioeconomics. Nowadays, only two classes of anti-influenza drugs, M2 ion channel inhibitors and neuraminidase inhibitors respectively, are used for prophylaxis and treatment of influenza virus infection. Unfortunately, influenza virus strains resistant to one or all of those drugs emerge frequently. Hemagglutinin (HA), the glycoprotein in influenza virus envelope, plays a critical role in viral binding, fusion and entry processes. Therefore, HA is a promising target for developing anti-influenza drugs, which block the initial entry step of viral life cycle. Here we reviewed recent understanding of conformational changes of HA in protein folding and fusion processes, and the discovery of HA-based influenza entry inhibitors, which may provide more choices for preventing and controlling potential pandemics caused by multi-resistant influenza viruses.
Collapse
Affiliation(s)
- Xintian Shen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; ; Department of Physiology, Huaihua Medical College, Huaihua 418000, China
| | | | | |
Collapse
|
14
|
Motohashi Y, Igarashi M, Okamatsu M, Noshi T, Sakoda Y, Yamamoto N, Ito K, Yoshida R, Kida H. Antiviral activity of stachyflin on influenza A viruses of different hemagglutinin subtypes. Virol J 2013; 10:118. [PMID: 23587221 PMCID: PMC3648499 DOI: 10.1186/1743-422x-10-118] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 04/11/2013] [Indexed: 01/21/2023] Open
Abstract
Background The hemagglutinin (HA) of influenza viruses is a possible target for antiviral drugs because of its key roles in the initiation of infection. Although it was found that a natural compound, Stachyflin, inhibited the growth of H1 and H2 but not H3 influenza viruses in MDCK cells, inhibitory activity of the compound has not been assessed against H4-H16 influenza viruses and the precise mechanism of inhibition has not been clarified. Methods Inhibitory activity of Stachyflin against H4-H16 influenza viruses, as well as H1-H3 viruses was examined in MDCK cells. To identify factors responsible for the susceptibility of the viruses to this compound, Stachyflin-resistant viruses were selected in MDCK cells and used for computer docking simulation. Results It was found that in addition to antiviral activity of Stachyflin against influenza viruses of H1 and H2 subtypes, it inhibited replication of viruses of H5 and H6 subtypes, as well as A(H1N1)pdm09 virus in MDCK cells. Stachyflin also inhibited the virus growth in the lungs of mice infected with A/WSN/1933 (H1N1) and A/chicken/Ibaraki/1/2005 (H5N2). Substitution of amino acid residues was found on the HA2 subunit of Stachyflin-resistant viruses. Docking simulation indicated that D37, K51, T107, and K121 are responsible for construction of the cavity for the binding of the compound. In addition, 3-dimensional structure of the cavity of the HA of Stachyflin-susceptible virus strains was different from that of insusceptible virus strains. Conclusion Antiviral activity of Stachyflin was found against A(H1N1)pdm09, H5, and H6 viruses, and identified a potential binding pocket for Stachyflin on the HA. The present results should provide us with useful information for the development of HA inhibitors with more effective and broader spectrum.
Collapse
Affiliation(s)
- Yurie Motohashi
- Department of Disease Control, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Influenza A virus entry inhibitors targeting the hemagglutinin. Viruses 2013; 5:352-73. [PMID: 23340380 PMCID: PMC3564125 DOI: 10.3390/v5010352] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 01/17/2013] [Accepted: 01/21/2013] [Indexed: 01/13/2023] Open
Abstract
Influenza A virus (IAV) has caused seasonal influenza epidemics and influenza pandemics, which resulted in serious threat to public health and socioeconomic impacts. Until now, only 5 drugs belong to two categories are used for prophylaxis and treatment of IAV infection. Hemagglutinin (HA), the envelope glycoprotein of IAV, plays a critical role in viral binding, fusion and entry. Therefore, HA is an attractive target for developing anti‑IAV drugs to block the entry step of IAV infection. Here we reviewed the recent progress in the study of conformational changes of HA during viral fusion process and the development of HA-based IAV entry inhibitors, which may provide a new choice for controlling future influenza pandemics.
Collapse
|
16
|
Zhu Z, Li R, Xiao G, Chen Z, Yang J, Zhu Q, Liu S. Design, synthesis and structure-activity relationship of novel inhibitors against H5N1 hemagglutinin-mediated membrane fusion. Eur J Med Chem 2012; 57:211-6. [PMID: 23059548 DOI: 10.1016/j.ejmech.2012.08.041] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 08/29/2012] [Accepted: 08/29/2012] [Indexed: 11/18/2022]
Abstract
We reported previously that a small molecule named CL-385319 could inhibit H5N1 influenza virus infection by targeting hemagglutinin, the envelope protein mediating virus entry. In the present study, a novel series of derivatives focused on the structural variation of CL-385319 were synthesized as specific inhibitors against the H5 subtype of influenza A viruses. These small molecules inhibited the low pH-induced conformational change of hemagglutinin, thereby blocking viral entry into host cells. Compound 1l was the most active inhibitor in this series with an IC(50) of 0.22 μM. The structure-activity relationships analysis of these compounds showed that the 3-fluoro-5-(trifluoromethyl)benzamide moiety was very important for activity, and the -F group was a better substituent group than -CF(3) group in the phenyl ring. The inhibitory activity was sensitive to the benzamide because the oxygen and hydrogen of the amide served as H-bond acceptor and donor, respectively.
Collapse
Affiliation(s)
- Zhibo Zhu
- School of Pharmaceutical Sciences, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Yanagita H, Yamamoto N, Fuji H, Liu X, Ogata M, Yokota M, Takaku H, Hasegawa H, Odagiri T, Tashiro M, Hoshino T. Mechanism of drug resistance of hemagglutinin of influenza virus and potent scaffolds inhibiting its function. ACS Chem Biol 2012; 7:552-62. [PMID: 22217114 DOI: 10.1021/cb200332k] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Highly pathogenic influenza viruses have become a global threat to humans. It is important to select an effective therapeutic option suitable for the subtypes in an epidemic or pandemic. To increase the options, the development of novel antiviral agents acting on targets different from those of the currently approved drugs is required. In this study, we performed molecular dynamics simulations on a spike protein on the viral envelop, hemagglutinin, for the wild-type and three kinds of mutants using a model system consisting of a trimeric hemagglutinin complex, viral lipid membrane, solvation waters, and ions. A natural product, stachyflin, which shows a high level of antiviral activity specific to some subtypes of influenza viruses, was examined on binding to the wild-type hemagglutinin by docking simulation. The compound potency of stachyflin is, however, easily lost due to resistant mutations. From a comparison of simulation results between the wild-type and the resistant mutants, the reason for the drug resistance of hemagglutinin was clarified. Next, 8 compounds were selected from a chemical database by in silico screening, considering the findings from the simulations. Inhibitory activities to suppress the proliferation of influenza virus were measured by cell-based antiviral assays, and two chemical scaffolds were found to be potent for an inhibitor. More than 30 derivatives bearing either of these two chemical scaffolds were synthesized, and cell culture assays were carried out to evaluate the compound potency. Several derivatives displayed a high compound potency, and 50% effective concentrations of two synthesized compounds were below 1 μM.
Collapse
Affiliation(s)
- Hiroshi Yanagita
- Graduate School of Pharmaceutical
Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675,
Japan
| | - Norio Yamamoto
- Department of General
Medicine,
Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo
113-8421, Japan
| | - Hideyoshi Fuji
- Graduate School of Pharmaceutical
Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675,
Japan
| | - Xinli Liu
- Graduate School of Pharmaceutical
Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675,
Japan
| | - Masakazu Ogata
- Graduate School of Pharmaceutical
Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675,
Japan
| | - Mizuho Yokota
- Graduate School of Pharmaceutical
Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675,
Japan
| | - Hiroshi Takaku
- Department
of Life and Environmental
Science, Chiba Institute of Technology, 2-17-1 Tsudanuma, Narashino-shi,
Chiba 275-0016, Japan
| | | | | | | | - Tyuji Hoshino
- Graduate School of Pharmaceutical
Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8675,
Japan
| |
Collapse
|
18
|
Park SY, Kim S, Yoon H, Kim KB, Kalme SS, Oh S, Song CS, Kim DE. Selection of an antiviral RNA aptamer against hemagglutinin of the subtype H5 avian influenza virus. Nucleic Acid Ther 2011; 21:395-402. [PMID: 22017542 DOI: 10.1089/nat.2011.0321] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Avian influenza is an acute viral respiratory disease caused by RNA viruses of the family Orthomyxoviridae. The influenza A virus subtype H5 can cause severe illness and results in almost 100% mortality rate among livestock. Hemagglutinin (HA) present in the virus envelope plays an essential role in the initiation of viral infection. In this study, we investigated the efficacy of using HA as a target for antiviral therapy through nucleic acid aptamers. After purification of the receptor binding domain (HA1) of HA protein, activity of recombinant HA1 was confirmed by using hemagglutination assay. We selected RNA aptamer candidates after 15 rounds of iterative Systematic Evolution of Ligands by EXponential enrichment (SELEX) targeting the biologically active HA protein. The selected RNA aptamer HAS15-5, which specifically binds to HA1, exhibited significant antiviral efficacy according to the results of a hemagglutination inhibition assay using egg allantoic fluids harboring the virus. Thus, the RNA aptamer HAS15-5, which acts by blocking and inhibiting the receptor-binding domain of viral HA, can be developed as a novel antiviral agent against type H5 avian influenza virus.
Collapse
Affiliation(s)
- Sun Young Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Liu S, Li R, Zhang R, Chan CCS, Xi B, Zhu Z, Yang J, Poon VKM, Zhou J, Chen M, Münch J, Kirchhoff F, Pleschka S, Haarmann T, Dietrich U, Pan C, Du L, Jiang S, Zheng B. CL-385319 inhibits H5N1 avian influenza A virus infection by blocking viral entry. Eur J Pharmacol 2011; 660:460-7. [PMID: 21536025 DOI: 10.1016/j.ejphar.2011.04.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 03/16/2011] [Accepted: 04/06/2011] [Indexed: 12/09/2022]
Abstract
CL-385319, an N-substituted piperidine, is effective in inhibiting infection of H1-, H2-, and to a lesser extent, H3-typed influenza A viruses by interfering with the fusogenic function of the viral hemagglutinin. Here we show that CL-385319 is effective in inhibiting infection of highly pathogenic H5N1 influenza A virus in Madin-Darby Canine Kidney (MDCK) cells with an IC50 of 27.03±2.54 μM. This compound with low cytotoxicity (CC50=1.48±0.01 mM) could also inhibit entry of pseudoviruses carrying hemagglutinins from H5N1 strains that were isolated from different places at different times, while it had no inhibitory activity on the entry of VSV-G pseudotyped particles. CL385319 could not inhibit N1-typed neuraminidase activity and the adsorption of H5-typed HA to chicken erythrocytes at the concentration as high as 1 mg/ml (2.8 mM). Computer-aid molecular docking analysis suggested that CL-385319 might bind to the cavity of HA2 stem region which was known to undergo significant rearrangement during membrane fusion. Pseudoviruses with M24A mutation in HA1 or F110S mutation in HA2 were resistant to CL-385319, indicating that these two residues in the cavity region may be critical for CL-385319 bindings. These findings suggest that CL-385319 can serve as a lead for development of novel virus entry inhibitors for preventing and treating H5N1 influenza A virus infection.
Collapse
Affiliation(s)
- Shuwen Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Sakurai J, Kikuchi T, Takahashi O, Watanabe K, Katoh T. Enantioselective Total Synthesis of (+)-Stachyflin: A Potential Anti-Influenza A Virus Agent Isolated from a Microorganism. European J Org Chem 2011. [DOI: 10.1002/ejoc.201100173] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
21
|
Jiang S, Li R, Du L, Liu S. Roles of the hemagglutinin of influenza A virus in viral entry and development of antiviral therapeutics and vaccines. Protein Cell 2010; 1:342-354. [PMID: 21203946 PMCID: PMC4728157 DOI: 10.1007/s13238-010-0054-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Accepted: 03/31/2010] [Indexed: 11/25/2022] Open
Abstract
Seasonal influenza epidemics and influenza pandemics caused by influenza A virus (IAV) has resulted in millions of deaths in the world. The development of anti-IAV vaccines and therapeutics is urgently needed for prevention and treatment of IAV infection and for controlling future influenza pandemics. Hemagglutinin (HA) of IAV plays a critical role in viral binding, fusion and entry, and contains the major neutralizing epitopes. Therefore, HA is an attractive target for developing anti-IAV drugs and vaccines. Here we have reviewed the recent progress in study of conformational changes of HA during viral fusion process and development of HA-based antiviral therapeutics and vaccines.
Collapse
Affiliation(s)
- Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA. .,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Runming Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA
| | - Shuwen Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
22
|
Watanabe K, Sakurai J, Abe H, Katoh T. Total synthesis of (+)-stachyflin: a potential anti-influenza A virus agent. Chem Commun (Camb) 2010; 46:4055-7. [PMID: 20361095 DOI: 10.1039/c000193g] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The first enantioselective total synthesis of (+)-stachyflin, a potential anti-influenza A virus agent, was achieved; the method features a BF(3).Et(2)O-induced domino epoxide-opening/rearrangement/cyclization reaction to stereoselectively form the requisite pentacyclic ring system in one step.
Collapse
Affiliation(s)
- Kazuhiro Watanabe
- Laboratory of Synthetic Medicinal Chemistry, Department of Chemical Pharmaceutical Science, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, 981-8558, Aoba-ku, Sendai, Japan
| | | | | | | |
Collapse
|
23
|
Nakatani M, Nakamura M, Suzuki A, Inoue M, Katoh T. A new strategy toward the total synthesis of stachyflin, a potent anti-influenza A virus agent: concise route to the tetracyclic core structure. Org Lett 2002; 4:4483-6. [PMID: 12465918 DOI: 10.1021/ol0271032] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
[reaction: see text] A new strategy directed toward the total synthesis of stachyflin, a potent and novel anti-influenza A virus agent isolated from a microorganism, has been presented through the enantioselective synthesis of the tetracyclic core structure. The synthetic method features a BF(3) x Et(2)O-induced domino epoxide-opening/rearrangement/cyclization reaction as the key step.
Collapse
Affiliation(s)
- Mari Nakatani
- Sagami Chemical Research Center, Hayakawa 2743-1, Ayase, Kanagawa 252-1193, Japan
| | | | | | | | | |
Collapse
|