1
|
Devantier K, Kjær VMS, Griffin S, Kragelund BB, Rosenkilde MM. Advancing the field of viroporins-Structure, function and pharmacology: IUPHAR Review 39. Br J Pharmacol 2024; 181:4450-4490. [PMID: 39224966 DOI: 10.1111/bph.17317] [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: 02/15/2024] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 09/04/2024] Open
Abstract
Viroporins possess important potential as antiviral targets due to their critical roles during virus life cycles, spanning from virus entry to egress. Although the antiviral amantadine targets the M2 viroporin of influenza A virus, successful progression of other viroporin inhibitors into clinical use remains challenging. These challenges relate in varying proportions to a lack of reliable full-length 3D-structures, difficulties in functionally characterising individual viroporins, and absence of verifiable direct binding between inhibitor and viroporin. This review offers perspectives to help overcome these challenges. We provide a comprehensive overview of the viroporin family, including their structural and functional features, highlighting the moldability of their energy landscapes and actions. To advance the field, we suggest a list of best practices to aspire towards unambiguous viroporin identification and characterisation, along with considerations of potential pitfalls. Finally, we present current and future scenarios of, and prospects for, viroporin targeting drugs.
Collapse
Affiliation(s)
- Kira Devantier
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Viktoria M S Kjær
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stephen Griffin
- Leeds Institute of Medical Research, St James' University Hospital, School of Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Birthe B Kragelund
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mette M Rosenkilde
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Mtambo SE, Amoako DG, Somboro AM, Agoni C, Lawal MM, Gumede NS, Khan RB, Kumalo HM. Influenza Viruses: Harnessing the Crucial Role of the M2 Ion-Channel and Neuraminidase toward Inhibitor Design. Molecules 2021; 26:880. [PMID: 33562349 PMCID: PMC7916051 DOI: 10.3390/molecules26040880] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 02/01/2021] [Accepted: 02/01/2021] [Indexed: 12/18/2022] Open
Abstract
As a member of the Orthomyxoviridae family of viruses, influenza viruses (IVs) are known causative agents of respiratory infection in vertebrates. They remain a major global threat responsible for the most virulent diseases and global pandemics in humans. The virulence of IVs and the consequential high morbidity and mortality of IV infections are primarily attributed to the high mutation rates in the IVs' genome coupled with the numerous genomic segments, which give rise to antiviral resistant and vaccine evading strains. Current therapeutic options include vaccines and small molecule inhibitors, which therapeutically target various catalytic processes in IVs. However, the periodic emergence of new IV strains necessitates the continuous development of novel anti-influenza therapeutic options. The crux of this review highlights the recent studies on the biology of influenza viruses, focusing on the structure, function, and mechanism of action of the M2 channel and neuraminidase as therapeutic targets. We further provide an update on the development of new M2 channel and neuraminidase inhibitors as an alternative to existing anti-influenza therapy. We conclude by highlighting therapeutic strategies that could be explored further towards the design of novel anti-influenza inhibitors with the ability to inhibit resistant strains.
Collapse
Affiliation(s)
- Sphamadla E. Mtambo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.M.S.); (C.A.); (M.M.L.); (N.S.G.); (R.B.K.)
| | - Daniel G. Amoako
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.M.S.); (C.A.); (M.M.L.); (N.S.G.); (R.B.K.)
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, Johannesburg 2131, South Africa
| | - Anou M. Somboro
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.M.S.); (C.A.); (M.M.L.); (N.S.G.); (R.B.K.)
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, Johannesburg 2131, South Africa
| | - Clement Agoni
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.M.S.); (C.A.); (M.M.L.); (N.S.G.); (R.B.K.)
| | - Monsurat M. Lawal
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.M.S.); (C.A.); (M.M.L.); (N.S.G.); (R.B.K.)
| | - Nelisiwe S. Gumede
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.M.S.); (C.A.); (M.M.L.); (N.S.G.); (R.B.K.)
| | - Rene B. Khan
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.M.S.); (C.A.); (M.M.L.); (N.S.G.); (R.B.K.)
| | - Hezekiel M. Kumalo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4000, South Africa; (S.E.M.); (A.M.S.); (C.A.); (M.M.L.); (N.S.G.); (R.B.K.)
| |
Collapse
|
3
|
Zak AJ, Hill BD, Rizvi SM, Smith MR, Yang M, Wen F. Enhancing the Yield and Quality of Influenza Virus-like Particles (VLPs) Produced in Insect Cells by Inhibiting Cytopathic Effects of Matrix Protein M2. ACS Synth Biol 2019; 8:2303-2314. [PMID: 31487465 DOI: 10.1021/acssynbio.9b00111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
To provide broader protection and eliminate the need for annual update of influenza vaccines, biomolecular engineering of influenza virus-like particles (VLPs) to display more conserved influenza proteins such as the matrix protein M2 has been explored. However, achieving high surface density of full-length M2 in influenza VLPs has been left unrealized. In this study, we show that the ion channel activity of M2 induces significant cytopathic effects in Spodoptera frugiperda (Sf9) insect cells when expressed using M2-encoding baculovirus. These effects include altered Sf9 cell morphology and reduced baculovirus replication, resulting in impaired influenza protein expression and thus VLP production. On the basis of the function of M2, we hypothesized that blocking its ion channel activity could potentially relieve these cytopathic effects, and thus restore influenza protein expression to improve VLP production. The use of the M2 inhibitor amantadine indeed improves Sf9 cellular expression not only of M2 (∼3-fold), but also of hemagglutinin (HA) (∼7-fold) and of matrix protein M1 (∼3-fold) when coexpressed to produce influenza VLPs. This increased cellular expression of all three influenza proteins further leads to ∼2-fold greater VLP yield. More importantly, the quality of the resulting influenza VLPs is significantly improved, as demonstrated by the ∼2-fold, ∼50-fold, and ∼2-fold increase in the antigen density to approximately 53 HA, 48 M1, and 156 M2 per influenza VLP, respectively. Taken together, this study represents a novel approach to enable the efficient incorporation of full-length M2 while enhancing both the yield and quality of influenza VLPs produced by Sf9 cells.
Collapse
Affiliation(s)
- Andrew J. Zak
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Brett D. Hill
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Syed M. Rizvi
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mason R. Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Madeleine Yang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Fei Wen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
4
|
Chua SCJH, Tan HQ, Engelberg D, Lim LHK. Alternative Experimental Models for Studying Influenza Proteins, Host-Virus Interactions and Anti-Influenza Drugs. Pharmaceuticals (Basel) 2019; 12:E147. [PMID: 31575020 PMCID: PMC6958409 DOI: 10.3390/ph12040147] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022] Open
Abstract
Ninety years after the discovery of the virus causing the influenza disease, this malady remains one of the biggest public health threats to mankind. Currently available drugs and vaccines only partially reduce deaths and hospitalizations. Some of the reasons for this disturbing situation stem from the sophistication of the viral machinery, but another reason is the lack of a complete understanding of the molecular and physiological basis of viral infections and host-pathogen interactions. Even the functions of the influenza proteins, their mechanisms of action and interaction with host proteins have not been fully revealed. These questions have traditionally been studied in mammalian animal models, mainly ferrets and mice (as well as pigs and non-human primates) and in cell lines. Although obviously relevant as models to humans, these experimental systems are very complex and are not conveniently accessible to various genetic, molecular and biochemical approaches. The fact that influenza remains an unsolved problem, in combination with the limitations of the conventional experimental models, motivated increasing attempts to use the power of other models, such as low eukaryotes, including invertebrate, and primary cell cultures. In this review, we summarized the efforts to study influenza in yeast, Drosophila, zebrafish and primary human tissue cultures and the major contributions these studies have made toward a better understanding of the disease. We feel that these models are still under-utilized and we highlight the unique potential each model has for better comprehending virus-host interactions and viral protein function.
Collapse
Affiliation(s)
- Sonja C J H Chua
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore.
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore.
- CREATE-NUS-HUJ Molecular Mechanisms of Inflammatory Diseases Programme, National University of Singapore, Singapore 138602, Singapore.
| | - Hui Qing Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore.
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore.
| | - David Engelberg
- CREATE-NUS-HUJ Molecular Mechanisms of Inflammatory Diseases Programme, National University of Singapore, Singapore 138602, Singapore.
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.
- Department of Biological Chemistry, The Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Lina H K Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore.
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore.
| |
Collapse
|
5
|
Li Z, Zou Z, Jiang Z, Huang X, Liu Q. Biological Function and Application of Picornaviral 2B Protein: A New Target for Antiviral Drug Development. Viruses 2019; 11:v11060510. [PMID: 31167361 PMCID: PMC6630369 DOI: 10.3390/v11060510] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 05/31/2019] [Accepted: 06/02/2019] [Indexed: 12/22/2022] Open
Abstract
Picornaviruses are associated with acute and chronic diseases. The clinical manifestations of infections are often mild, but infections may also lead to respiratory symptoms, gastroenteritis, myocarditis, meningitis, hepatitis, and poliomyelitis, with serious impacts on human health and economic losses in animal husbandry. Thus far, research on picornaviruses has mainly focused on structural proteins such as VP1, whereas the non-structural protein 2B, which plays vital roles in the life cycle of the viruses and exhibits a viroporin or viroporin-like activity, has been overlooked. Viroporins are viral proteins containing at least one amphipathic α-helical structure, which oligomerizes to form transmembrane hydrophilic pores. In this review, we mainly summarize recent research data on the viroporin or viroporin-like activity of 2B proteins, which affects the biological function of the membrane, regulates cell death, and affects the host immune response. Considering these mechanisms, the potential application of the 2B protein as a candidate target for antiviral drug development is discussed, along with research challenges and prospects toward realizing a novel treatment strategy for picornavirus infections.
Collapse
Affiliation(s)
- Zengbin Li
- School of Public Health, Nanchang University, Nanchang 330006, China.
| | - Zixiao Zou
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang 330006, China.
| | - Zeju Jiang
- Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang 330006, China.
| | - Qiong Liu
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
6
|
Yu Y, Tazeem, Xu Z, Du L, Jin M, Dong C, Zhou HB, Wu S. Design and synthesis of heteroaromatic-based benzenesulfonamide derivatives as potent inhibitors of H5N1 influenza A virus. MEDCHEMCOMM 2019; 10:89-100. [PMID: 31559005 PMCID: PMC6735340 DOI: 10.1039/c8md00474a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 11/23/2018] [Indexed: 01/04/2023]
Abstract
Influenza A virus is an enveloped negative single-stranded RNA virus that causes febrile respiratory infection and represents a clinically challenging threat to human health and even lives worldwide. Even more alarming is the emergence of highly pathogenic avian influenza (HPAI) strains such as H5N1, which possess much higher mortality rate (60%) than seasonal influenza strains in human infection. In this study, a novel series of heteroaromatic-based benzenesulfonamide derivatives were identified as M2 proton channel inhibitors. A systematic investigation of the structure-activity relationships and a molecular docking study demonstrated that the sulfonamide moiety and 2,5-dimethyl-substituted thiophene as the core structure played significant roles in the anti-influenza activity. Among the derivatives, compound 11k exhibited excellent antiviral activity against H5N1 virus with an EC50 value of 0.47 μM and selectivity index of 119.9, which are comparable to those of the reference drug amantadine.
Collapse
Affiliation(s)
- Yongshi Yu
- State Key Laboratory of Virology , College of Life Sciences , Wuhan University , Wuhan 430072 , China .
- Hubei Province Key Laboratory of Allergy and Immunology , Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Tazeem
- Hubei Province Key Laboratory of Allergy and Immunology , Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
- Department of Chemistry , Shia P. G. College (University of Lucknow) , Lucknow , Uttar Pradesh 226020 , India
| | - Zhichao Xu
- Hubei Province Key Laboratory of Allergy and Immunology , Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Liaoqi Du
- State Key Laboratory of Virology , College of Life Sciences , Wuhan University , Wuhan 430072 , China .
| | - Mengyu Jin
- Hubei Province Key Laboratory of Allergy and Immunology , Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Chune Dong
- Hubei Province Key Laboratory of Allergy and Immunology , Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Hai-Bing Zhou
- Hubei Province Key Laboratory of Allergy and Immunology , Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals , Wuhan University School of Pharmaceutical Sciences , Wuhan 430071 , China .
| | - Shuwen Wu
- State Key Laboratory of Virology , College of Life Sciences , Wuhan University , Wuhan 430072 , China .
| |
Collapse
|
7
|
Denny PW. Yeast: bridging the gap between phenotypic and biochemical assays for high-throughput screening. Expert Opin Drug Discov 2018; 13:1153-1160. [DOI: 10.1080/17460441.2018.1534826] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Paul W. Denny
- Department of Biosciences and Centre for Global Infectious Disease, Durham University, Durham, UK
| |
Collapse
|
8
|
Synthesis and structure-activity relationship study of arylsulfonamides as novel potent H5N1 inhibitors. Eur J Med Chem 2018; 159:206-216. [PMID: 30292897 DOI: 10.1016/j.ejmech.2018.09.065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 09/22/2018] [Accepted: 09/25/2018] [Indexed: 12/13/2022]
Abstract
H5N1 virus, one subtype of highly pathogenic influenza A virus in human infection, has recently received attention due to its unpredictable and high mortality. In this study, a series of arylsulfonamide derivatives were identified as improved H5N1 inhibitors for the influenza treatment by systematic structure-activity relationship investigation. Among them, the most potent H5N1 inhibitor 3h exhibited excellent antiviral activity against H5N1 virus with EC50 value of 0.006 μM and selectivity index 33543.3. Moreover, the molecular docking of 3h with M2 proton channel protein provides practical way for understanding the inhibition of H5N1 with this kind of compounds.
Collapse
|
9
|
Influenza A Virus M2 Protein: Roles from Ingress to Egress. Int J Mol Sci 2017; 18:ijms18122649. [PMID: 29215568 PMCID: PMC5751251 DOI: 10.3390/ijms18122649] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 11/17/2022] Open
Abstract
Influenza A virus (IAV) matrix protein 2 (M2) is among the smallest bona fide, hence extensively studied, ion channel proteins. The M2 ion channel activity is not only essential for virus replication, but also involved in modulation of cellular homeostasis in a variety of ways. It is also the target for ion channel inhibitors, i.e., anti-influenza drugs. Thus far, several studies have been conducted to elucidate its biophysical characteristics, structure-function relationships of the ion channel, and the M2-host interactome. In this review, we discuss M2 protein synthesis and assembly into an ion channel, its roles in IAV replication, and the pathophysiological impact on the host cell.
Collapse
|
10
|
Sornpet B, Potha T, Tragoolpua Y, Pringproa K. Antiviral activity of five Asian medicinal pant crude extracts against highly pathogenic H5N1 avian influenza virus. ASIAN PAC J TROP MED 2017; 10:871-876. [PMID: 29080615 DOI: 10.1016/j.apjtm.2017.08.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/16/2017] [Accepted: 08/17/2017] [Indexed: 10/18/2022] Open
Abstract
OBJECTIVE To study the antiviral properties of the five Asian medicinal plants against in vitro infection by the highly pathogenic avian influenza virus (H5N1). METHODS Crude extracts of Andrographis paniculata, Curcuma longa (C. longa), Gynostemma pentaphyllum, Kaempferia parviflora (K. parviflora), and Psidium guajava obtained by both water and ethanol extractions were investigated for their cytotoxicity in the Madin-Darby canine kidney cells. Thereafter, they were investigated in vitro for antiviral activity and cytokine response upon H5N1 virus infection. RESULTS The results revealed that both water and ethanol extracts of all the five studied plants showed significant antiviral activity against H5N1 virus. Among these plants, C. longa and K. parviflora showed strong anti-H5N1 activity. Thus, they were selected for further studies on their cytokine response upon virus infection. It was found that ethanol and water crude extracts of C. longa and K. parviflora induced significant upregulation of TNF-α and IFN-β mRNA expressions, suggesting their roles in the inhibition of H5N1 virus replication. CONCLUSIONS To the best of the authors' knowledge, this study is among the earliest reports to illustrate the antiviral property of these Asian medicinal plants against the highly pathogenic avian H5N1 influenza virus. The results of this study shed light on alternative therapeutic sources for treatment of H5N1 influenza virus infection in the future.
Collapse
Affiliation(s)
- Benjaporn Sornpet
- Central Veterinary Diagnostic Laboratory, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Teerapong Potha
- Central Veterinary Diagnostic Laboratory, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Yingmanee Tragoolpua
- Department of Biology, Faculty of Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kidsadagon Pringproa
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand.
| |
Collapse
|
11
|
Wang J, Li F, Ma C. Recent progress in designing inhibitors that target the drug-resistant M2 proton channels from the influenza A viruses. Biopolymers 2016; 104:291-309. [PMID: 25663018 DOI: 10.1002/bip.22623] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 01/24/2015] [Indexed: 12/15/2022]
Abstract
Influenza viruses are the causative agents for seasonal influenza, which results in thousands of deaths and millions of hospitalizations each year. Moreover, sporadic transmission of avian or swan influenza viruses to humans often leads to an influenza pandemic, as there is no preimmunity in the human body to fight against such novel strains. The metastable genome of the influenza viruses, coupled with the reassortment of different strains from a wide range of host origins, leads to the continuous evolution of the influenza virus diversity. Such characteristics of influenza viruses present a grand challenge in devising therapeutic strategies to combat influenza virus infection. This review summarizes recent progress in designing small molecule inhibitors that target the drug-resistant influenza A virus M2 proton channels and highlights the contribution of mechanistic studies of proton conductance to drug discovery. The lessons learned throughout the course of M2 drug discovery might provide insights for designing inhibitors that target other therapeutically important ion channels.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721.,BIO5 Institute, University of Arizona, Tucson, AZ, 85721
| | - Fang Li
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721
| | - Chunlong Ma
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721
| |
Collapse
|
12
|
Arns S, Balgi AD, Shimizu Y, Pfeifer TA, Kumar N, Shidmoossavee FS, Sun S, Tai SSH, Agafitei O, Jaquith JB, Bourque E, Niikura M, Roberge M. Novel spirothiazamenthane inhibitors of the influenza A M2 proton channel. Eur J Med Chem 2016; 120:64-73. [PMID: 27187859 DOI: 10.1016/j.ejmech.2016.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 05/04/2016] [Accepted: 05/05/2016] [Indexed: 01/19/2023]
Abstract
The development of treatments for influenza that inhibit the M2 proton channel without being susceptible to the widespread resistance mechanisms associated with the adamantanes is an ongoing challenge. Using a yeast high-throughput yeast growth restoration assay designed to identify M2 channel inhibitors, a single screening hit was uncovered. This compound (3), whose structure was incorrectly identified in the literature, is an inhibitor with similar potency to amantadine against WT M2. A library of derivatives of 3 was prepared and activity against WT M2 and the two principal mutant strains (V27A and S31N) was assessed in the yeast assay. The best compounds were further evaluated in an antiviral plaque reduction assay using engineered WT, V27A and S31N M2 influenza A strains with otherwise identical genetic background. Compound 63 was found to inhibit all three virus strains in this cell based antiviral assay at micromolar concentrations, possibly through a mechanism other than M2 inhibition.
Collapse
Affiliation(s)
- Steve Arns
- The Centre for Drug Research and Development, Vancouver, BC, Canada
| | - Aruna D Balgi
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Yoko Shimizu
- The Centre for Drug Research and Development, Vancouver, BC, Canada
| | - Tom A Pfeifer
- The Centre for Drug Research and Development, Vancouver, BC, Canada
| | - Nag Kumar
- The Centre for Drug Research and Development, Vancouver, BC, Canada
| | | | - Sharon Sun
- The Centre for Drug Research and Development, Vancouver, BC, Canada
| | - Sheldon S-H Tai
- Faculty of Heath Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Olga Agafitei
- Faculty of Heath Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - James B Jaquith
- The Centre for Drug Research and Development, Vancouver, BC, Canada
| | - Elyse Bourque
- The Centre for Drug Research and Development, Vancouver, BC, Canada
| | - Masahiro Niikura
- Faculty of Heath Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Michel Roberge
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
13
|
Abstract
Since the discovery that certain small viral membrane proteins, collectively termed as viroporins, can permeabilize host cellular membranes and also behave as ion channels, attempts have been made to link this feature to specific biological roles. In parallel, most viroporins identified so far are virulence factors, and interest has focused toward the discovery of channel inhibitors that would have a therapeutic effect, or be used as research tools to understand the biological roles of viroporin ion channel activity. However, this paradigm is being shifted by the difficulties inherent to small viral membrane proteins, and by the realization that protein-protein interactions and other diverse roles in the virus life cycle may represent an equal, if not, more important target. Therefore, although targeting the channel activity of viroporins can probably be therapeutically useful in some cases, the focus may shift to their other functions in following years. Small-molecule inhibitors have been mostly developed against the influenza A M2 (IAV M2 or AM2). This is not surprising since AM2 is the best characterized viroporin to date, with a well-established biological role in viral pathogenesis combined the most extensive structural investigations conducted, and has emerged as a validated drug target. For other viroporins, these studies are still mostly in their infancy, and together with those for AM2, are the subject of the present review.
Collapse
|
14
|
Shen Z, Lou K, Wang W. New small-molecule drug design strategies for fighting resistant influenza A. Acta Pharm Sin B 2015; 5:419-30. [PMID: 26579472 PMCID: PMC4629447 DOI: 10.1016/j.apsb.2015.07.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 07/05/2015] [Indexed: 11/24/2022] Open
Abstract
Influenza A virus is the major cause of seasonal or pandemic flu worldwide. Two main treatment strategies-vaccination and small molecule anti-influenza drugs are currently available. As an effective vaccine usually takes at least 6 months to develop, anti-influenza small molecule drugs are more effective for the first line of protection against the virus during an epidemic outbreak, especially in the early stage. Two major classes of anti-influenza drugs currently available are admantane-based M2 protein blockers (amantadine and rimantadine) and neuraminidase (NA) inhibitors (oseltamivir, zanamivir, and peramivir). However, the continuous evolvement of influenza A virus and the rapid emergence of resistance to current drugs, particularly to amantadine, rimantadine, and oseltamivir, have raised an urgent need for developing new anti-influenza drugs against resistant forms of influenza A virus. In this review, we first give a brief introduction of the molecular mechanisms behind resistance, and then discuss new strategies in small-molecule drug development to overcome influenza A virus resistance targeting mutant M2 proteins and neuraminidases, and other viral proteins not associated with current drugs.
Collapse
Affiliation(s)
- Zuyuan Shen
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Kaiyan Lou
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
| | - Wei Wang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, and State Key Laboratory of Bioengineering Reactor, East China University of Science and Technology, Shanghai 200237, China
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM 87131-0001, USA
| |
Collapse
|
15
|
"Too little, too late?" Will inhibitors of the hepatitis C virus p7 ion channel ever be used in the clinic? Future Med Chem 2015; 6:1893-907. [PMID: 25495983 DOI: 10.4155/fmc.14.121] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hepatitis C virus (HCV) p7 is a virus-coded ion channel, or 'viroporin'. p7 is an essential HCV protein, promoting infectious virion production, and this process can be blocked by prototypic p7 inhibitors. However, prototype potency is weak and effects in clinical trials are unsatisfactory. Nevertheless, recent structural studies render p7 amenable to modern drug discovery, with studies supporting that effective drug-like molecules should be achievable. However, burgeoning HCV therapies clear infection in the majority of treated patients. This perspective summarizes current understanding of p7 channel function and structure, pertaining to the development of improved p7 inhibitors. We ask, 'is this too little, too late', or could p7 inhibitors play a role in the long-term management of HCV disease?
Collapse
|
16
|
Scott C, Griffin S. Viroporins: structure, function and potential as antiviral targets. J Gen Virol 2015; 96:2000-2027. [PMID: 26023149 DOI: 10.1099/vir.0.000201] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The channel-forming activity of a family of small, hydrophobic integral membrane proteins termed 'viroporins' is essential to the life cycles of an increasingly diverse range of RNA and DNA viruses, generating significant interest in targeting these proteins for antiviral development. Viroporins vary greatly in terms of their atomic structure and can perform multiple functions during the virus life cycle, including those distinct from their role as oligomeric membrane channels. Recent progress has seen an explosion in both the identification and understanding of many such proteins encoded by highly significant pathogens, yet the prototypic M2 proton channel of influenza A virus remains the only example of a viroporin with provenance as an antiviral drug target. This review attempts to summarize our current understanding of the channel-forming functions for key members of this growing family, including recent progress in structural studies and drug discovery research, as well as novel insights into the life cycles of many viruses revealed by a requirement for viroporin activity. Ultimately, given the successes of drugs targeting ion channels in other areas of medicine, unlocking the therapeutic potential of viroporins represents a valuable goal for many of the most significant viral challenges to human and animal health.
Collapse
Affiliation(s)
- Claire Scott
- Leeds Institute of Cancer & Pathology and Leeds CRUK Clinical Centre, Faculty of Medicine and Health, St James's University Hospital, University of Leeds, Beckett Street, Leeds LS9 7TF, UK
| | - Stephen Griffin
- Leeds Institute of Cancer & Pathology and Leeds CRUK Clinical Centre, Faculty of Medicine and Health, St James's University Hospital, University of Leeds, Beckett Street, Leeds LS9 7TF, UK
| |
Collapse
|
17
|
Taube R, Alhadeff R, Assa D, Krugliak M, Arkin IT. Bacteria-based analysis of HIV-1 Vpu channel activity. PLoS One 2014; 9:e105387. [PMID: 25272035 PMCID: PMC4182682 DOI: 10.1371/journal.pone.0105387] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 07/21/2014] [Indexed: 02/04/2023] Open
Abstract
HIV-1 Vpu is a small, single-span membrane protein with two attributed functions that increase the virus' pathogenicity: degradation of CD4 and inactivation of BST-2. Vpu has also been shown to posses ion channel activity, yet no correlation has been found between this attribute and Vpu's role in viral release. In order to gain further insight into the channel activity of Vpu we devised two bacteria-based assays that can examine this function in detail. In the first assay Vpu was over-expressed, such that it was deleterious to bacterial growth due to membrane permeabilization. In the second and more sensitive assay, the channel was expressed at low levels in K+ transport deficient bacteria. Consequently, Vpu expression enabled the bacteria to grow at otherwise non permissive low K+ concentrations. Hence, Vpu had the opposite impact on bacterial growth in the two assays: detrimental in the former and beneficial in the latter. Furthermore, we show that channel blockers also behave reciprocally in the two assays, promoting growth in the first assay and hindering it in the second assay. Taken together, we investigated Vpu's channel activity in a rapid and quantitative approach that is amenable to high-throughput screening, in search of novel blockers.
Collapse
Affiliation(s)
- Robert Taube
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmund J. Safra Campus, Jerusalem, Israel
- Institue of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Raphael Alhadeff
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmund J. Safra Campus, Jerusalem, Israel
| | - Dror Assa
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmund J. Safra Campus, Jerusalem, Israel
| | - Miriam Krugliak
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmund J. Safra Campus, Jerusalem, Israel
| | - Isaiah T. Arkin
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmund J. Safra Campus, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
18
|
Denny PW, Steel PG. Yeast as a potential vehicle for neglected tropical disease drug discovery. ACTA ACUST UNITED AC 2014; 20:56-63. [PMID: 25121554 DOI: 10.1177/1087057114546552] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
High-throughput screening (HTS) efforts for neglected tropical disease (NTD) drug discovery have recently received increased attention because several initiatives have begun to attempt to reduce the deficit in new and clinically acceptable therapies for this spectrum of infectious diseases. HTS primarily uses two basic approaches, cell-based and in vitro target-directed screening. Both of these approaches have problems; for example, cell-based screening does not reveal the target or targets that are hit, whereas in vitro methodologies lack a cellular context. Furthermore, both can be technically challenging, expensive, and difficult to miniaturize for ultra-HTS [(u)HTS]. The application of yeast-based systems may overcome some of these problems and offer a cost-effective platform for target-directed screening within a eukaryotic cell context. Here, we review the advantages and limitations of the technologies that may be used in yeast cell-based, target-directed screening protocols, and we discuss how these are beginning to be used in NTD drug discovery.
Collapse
Affiliation(s)
- P W Denny
- Biophysical Sciences Institute, Department of Chemistry and School of Biological Sciences, University Science Laboratories, Durham, UK School of Medicine, Pharmacy and Health, Durham University, Durham, UK
| | - P G Steel
- Biophysical Sciences Institute, Department of Chemistry and School of Biological Sciences, University Science Laboratories, Durham, UK
| |
Collapse
|
19
|
Barberis A, Gunde T, Berset C, Audetat S, Lüthi U. Yeast as a screening tool. DRUG DISCOVERY TODAY. TECHNOLOGIES 2014; 2:187-92. [PMID: 24981847 DOI: 10.1016/j.ddtec.2005.05.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The versatile genetic malleability of yeast, and the high degree of conservation between its cellular processes and those of human cells, have made it the model of choice for pioneering research in molecular and cell biology over the past four decades. These characteristics of yeast, taken together with technical advantages such as simple growth conditions, rapid cell division and the development of a wealth of genetic tools for analysis of biological functions, have expanded the application of yeast as screening tool to the field of drug discovery.:
Collapse
Affiliation(s)
- Alcide Barberis
- ESBATech AG, Wagistrasse 21, CH-8952 Zürich-Schlieren, Switzerland.
| | - Tea Gunde
- ESBATech AG, Wagistrasse 21, CH-8952 Zürich-Schlieren, Switzerland
| | - Catherine Berset
- ESBATech AG, Wagistrasse 21, CH-8952 Zürich-Schlieren, Switzerland
| | - Stephan Audetat
- ESBATech AG, Wagistrasse 21, CH-8952 Zürich-Schlieren, Switzerland
| | - Urs Lüthi
- ESBATech AG, Wagistrasse 21, CH-8952 Zürich-Schlieren, Switzerland.
| |
Collapse
|
20
|
Kolocouris A, Tzitzoglaki C, Johnson FB, Zell R, Wright AK, Cross TA, Tietjen I, Fedida D, Busath DD. Aminoadamantanes with persistent in vitro efficacy against H1N1 (2009) influenza A. J Med Chem 2014; 57:4629-39. [PMID: 24793875 PMCID: PMC4127532 DOI: 10.1021/jm500598u] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
![]()
A series of 2-adamantanamines with
alkyl adducts of various lengths
were examined for efficacy against strains of influenza A including
those having an S31N mutation in M2 proton channel that confer resistance
to amantadine and rimantadine. The addition of as little as one CH2 group to the methyl adduct of the amantadine/rimantadine
analogue, 2-methyl-2-aminoadamantane, led to activity in vitro against
two M2 S31N viruses A/Calif/07/2009 (H1N1) and A/PR/8/34 (H1N1) but
not to a third A/WS/33 (H1N1). Solid state NMR of the transmembrane
domain (TMD) with a site mutation corresponding to S31N shows evidence
of drug binding. But electrophysiology using the full length S31N
M2 protein in HEK cells showed no blockade. A wild type strain, A/Hong
Kong/1/68 (H3N2) developed resistance to representative drugs within
one passage with mutations in M2 TMD, but A/Calif/07/2009 S31N was
slow (>8 passages) to develop resistance in vitro, and the resistant
virus had no mutations in M2 TMD. The results indicate that 2-alkyl-2-aminoadamantane
derivatives with sufficient adducts can persistently block p2009 influenza
A in vitro through an alternative mechanism. The observations of an
HA1 mutation, N160D, near the sialic acid binding site in both 6-resistant A/Calif/07/2009(H1N1) and the broadly resistant
A/WS/33(H1N1) and of an HA1 mutation, I325S, in the 6-resistant virus at a cell-culture stable site suggest that the drugs
tested here may block infection by direct binding near these critical
sites for virus entry to the host cell.
Collapse
Affiliation(s)
- Antonios Kolocouris
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens , Athens 15771, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Recent progress and challenges in the computer-aided design of inhibitors for influenza A M2 channel proteins. Med Chem Res 2014. [DOI: 10.1007/s00044-014-0964-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
22
|
Strategies for the Development of Influenza Drugs: Basis for New Efficient Combination Therapies. TOPICS IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1007/7355_2014_84] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
23
|
Tarasenko IV, Taranov AI, Firsov AP, Dolgov SV. Expression of the nucleotide sequence for the M2e peptide of avian influenza virus in transgenic tobacco plants. APPL BIOCHEM MICRO+ 2013. [DOI: 10.1134/s0003683813080061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
24
|
Gu RX, Liu LA, Wei DQ. Structural and energetic analysis of drug inhibition of the influenza A M2 proton channel. Trends Pharmacol Sci 2013; 34:571-80. [PMID: 24011996 DOI: 10.1016/j.tips.2013.08.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/09/2013] [Accepted: 08/12/2013] [Indexed: 01/07/2023]
Abstract
The type A influenza virus matrix protein 2 (M2) is a highly selective proton channel in the viral envelope. Because of its crucial role in viral infection and replication, the M2 channel has been a target of anti-influenza drugs. Due to the occurrence of drug-resistant mutations in the M2 channel, existing anti-influenza drugs that block the M2 channel, such as amantadine and rimantadine, have lost their efficacy against these mutant channels. Recent experimental and computational efforts have made great progress in understanding the drug resistance mechanisms of these mutations as well as designing novel drug candidates to block the mutant M2 channels. In this review, we briefly summarize the structural characteristics of the M2 channel, and then we discuss these recent studies on drug resistance and drug design of the mutant channels, focusing on the structures and energetics. We show that structural biology experiments and molecular modeling have led to the successful design of novel drugs targeting mutant M2 channels.
Collapse
Affiliation(s)
- Ruo-Xu Gu
- State Key Laboratory of Microbial Metabolism, and College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai Minhang District, 200240, China
| | | | | |
Collapse
|
25
|
Beyleveld G, White KM, Ayllon J, Shaw ML. New-generation screening assays for the detection of anti-influenza compounds targeting viral and host functions. Antiviral Res 2013; 100:120-32. [PMID: 23933115 DOI: 10.1016/j.antiviral.2013.07.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Revised: 07/23/2013] [Accepted: 07/26/2013] [Indexed: 01/08/2023]
Abstract
Current options for influenza antiviral therapy are limited to the neuraminidase inhibitors, and knowledge that high levels of oseltamivir resistance have been seen among previously circulating H1N1 viruses increases the urgency to find new influenza therapeutics. To feed this pipeline, assays that are appropriate for use in high-throughput screens are being developed and are discussed in this review. Particular emphasis is placed on cell-based assays that capture both inhibitors of viral functions as well as the host functions that facilitate optimal influenza virus replication. Success in this area has been fueled by a greater understanding of the genome structure of influenza viruses and the ability to generate replication-competent recombinant viruses that carry a reporter gene, allowing for easy monitoring of viral infection in a high-throughput setting. This article forms part of a symposium in Antiviral Research on "Treatment of influenza: targeting the virus or the host."
Collapse
Affiliation(s)
- Grant Beyleveld
- Department of Microbiology and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | | |
Collapse
|
26
|
The utility of yeast as a tool for cell-based, target-directed high-throughput screening. Parasitology 2013; 141:8-16. [PMID: 23611102 DOI: 10.1017/s0031182013000425] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Many Neglected Tropical Diseases (NTDs) have recently been subject of increased focus, particularly with relation to high-throughput screening (HTS) initiatives. These vital endeavours largely rely of two approaches, in vitro target-directed screening using biochemical assays or cell-based screening which takes no account of the target or targets being hit. Despite their successes both of these approaches have limitations; for example, the production of soluble protein and a lack of cellular context or the problems and expense of parasite cell culture. In addition, both can be challenging to miniaturize for ultra (u)HTS and expensive to utilize. Yeast-based systems offer a cost-effective approach to study and screen protein targets in a direct-directed manner within a eukaryotic cellular context. In this review, we examine the utility and limitations of yeast cell-based, target-directed screening. In particular we focus on the currently under-explored possibility of using such formats in uHTS screening campaigns for NTDs.
Collapse
|
27
|
Wang J, Ma C, Wang J, Jo H, Canturk B, Fiorin G, Pinto LH, Lamb RA, Klein ML, DeGrado WF. Discovery of novel dual inhibitors of the wild-type and the most prevalent drug-resistant mutant, S31N, of the M2 proton channel from influenza A virus. J Med Chem 2013; 56:2804-12. [PMID: 23437766 DOI: 10.1021/jm301538e] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Anti-influenza drugs, amantadine and rimantadine, targeting the M2 channel from influenza A virus are no longer effective because of widespread drug resistance. S31N is the predominant and amantadine-resistant M2 mutant, present in almost all of the circulating influenza A strains as well as in the pandemic 2009 H1N1 and the highly pathogenic H5N1 flu strains. Thus, there is an urgent need to develop second-generation M2 inhibitors targeting the S31N mutant. However, the S31N mutant presents a huge challenge to drug discovery, and it has been considered undruggable for several decades. Using structural information, classical medicinal chemistry approaches, and M2-specific biological testing, we discovered benzyl-substituted amantadine derivatives with activity against both S31N and WT, among which 4-(adamantan-1-ylaminomethyl)-benzene-1,3-diol (44) is the most potent dual inhibitor. These inhibitors demonstrate that S31N is a druggable target and provide a new starting point to design novel M2 inhibitors that address the problem of drug-resistant influenza A infections.
Collapse
Affiliation(s)
- Jizhou Wang
- Influmedix Inc, 170 North Radnor-Chester Road, Suite 300, Radnor, Pennsylvania 19087, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Balgi AD, Wang J, Cheng DYH, Ma C, Pfeifer TA, Shimizu Y, Anderson HJ, Pinto LH, Lamb RA, DeGrado WF, Roberge M. Inhibitors of the influenza A virus M2 proton channel discovered using a high-throughput yeast growth restoration assay. PLoS One 2013; 8:e55271. [PMID: 23383318 PMCID: PMC3562233 DOI: 10.1371/journal.pone.0055271] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/22/2012] [Indexed: 11/18/2022] Open
Abstract
The M2 proton channel of the influenza A virus is the target of the anti-influenza drugs amantadine and rimantadine. The effectiveness of these drugs has been dramatically limited by the rapid spread of drug resistant mutations, mainly at sites S31N, V27A and L26F in the pore of the channel. Despite progress in designing inhibitors of V27A and L26F M2, there are currently no drugs targeting these mutated channels in clinical trials. Progress in developing new drugs has been hampered by the lack of a robust assay with sufficient throughput for discovery of new active chemotypes among chemical libraries and sufficient sensitivity to provide the SAR data essential for their improvement and development as drugs. In this study we adapted a yeast growth restoration assay, in which expression of the M2 channel inhibits yeast growth and exposure to an M2 channel inhibitor restores growth, into a robust and sensitive high-throughput screen for M2 channel inhibitors. A screen of over 250,000 pure chemicals and semi-purified fractions from natural extracts identified 21 active compounds comprising amantadine, rimantadine, 13 related adamantanes and 6 non-adamantanes. Of the non-adamantanes, hexamethylene amiloride and a triazine derivative represented new M2 inhibitory chemotypes that also showed antiviral activity in a plaque reduction assay. Of particular interest is the fact that the triazine derivative was not sufficiently potent for detection as an inhibitor in the traditional two electrode voltage clamp assay for M2 channel activity, but its discovery in the yeast assay led to testing of analogues of which one was as potent as amantadine.
Collapse
Affiliation(s)
- Aruna D. Balgi
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jun Wang
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Daphne Y. H. Cheng
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Chunlong Ma
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois, United States of America
| | - Tom A. Pfeifer
- The Centre for Drug Research and Development, Vancouver, British Columbia, Canada
| | - Yoko Shimizu
- The Centre for Drug Research and Development, Vancouver, British Columbia, Canada
| | - Hilary J. Anderson
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lawrence H. Pinto
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois, United States of America
| | - Robert A. Lamb
- Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Evanston, Illinois, United States of America
- Howard Hughes Medical Institute, Northwestern University, Evanston, Illinois, United States of America
| | - William F. DeGrado
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, United States of America
| | - Michel Roberge
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- The Centre for Drug Research and Development, Vancouver, British Columbia, Canada
| |
Collapse
|
29
|
Herrero L, Monroy N, González ME. HIV-1 Vpu Protein Mediates the Transport of Potassium in Saccharomyces cerevisiae. Biochemistry 2012; 52:171-7. [DOI: 10.1021/bi3011175] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Laura Herrero
- Unidad de Expresión Viral, Centro
Nacional de
Microbiología, Instituto de Salud Carlos III, Carretera de
Majadahonda-Pozuelo Km 2, 28220 Majadahonda, Madrid, Spain
| | - Noemí Monroy
- Unidad de Expresión Viral, Centro
Nacional de
Microbiología, Instituto de Salud Carlos III, Carretera de
Majadahonda-Pozuelo Km 2, 28220 Majadahonda, Madrid, Spain
| | - María Eugenia González
- Unidad de Expresión Viral, Centro
Nacional de
Microbiología, Instituto de Salud Carlos III, Carretera de
Majadahonda-Pozuelo Km 2, 28220 Majadahonda, Madrid, Spain
| |
Collapse
|
30
|
Lee SMY, Yen HL. Targeting the host or the virus: current and novel concepts for antiviral approaches against influenza virus infection. Antiviral Res 2012; 96:391-404. [PMID: 23022351 PMCID: PMC7132421 DOI: 10.1016/j.antiviral.2012.09.013] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 09/11/2012] [Accepted: 09/17/2012] [Indexed: 12/22/2022]
Abstract
Influenza epidemics and pandemics are constant threats to human health. The application of antiviral drugs provides an immediate and direct control of influenza virus infection. At present, the major strategy for managing patients with influenza is through targeting conserved viral proteins critical for viral replication. Two classes of conventional antiviral drugs, the M2 ion channel blockers and the neuraminidase inhibitors, are frequently used. In recent years, increasing levels of resistance to both drug classes has become a major public health concern, highlighting the urgent need for the development of alternative treatments. Novel classes of antiviral compounds or biomolecules targeting viral replication mechanism are under development, using approaches including high-throughput small-molecule screening platforms and structure-based designs. In response to influenza virus infection, host cellular mechanisms are triggered to defend against the invaders. At the same time, viruses as obligate intracellular pathogens have evolved to exploit cellular responses in support of their efficient replication, including antagonizing the host type I interferon response as well as activation of specific cellular pathways at different stages of the replication cycle. Numerous studies have highlighted the possibility of targeting virus-host interactions and host cellular mechanisms to develop new treatment regimens. This review aims to give an overview of current and novel concepts targeting the virus and the host for managing influenza.
Collapse
Affiliation(s)
- Suki Man-Yan Lee
- Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong
| | | |
Collapse
|
31
|
Du J, Cross TA, Zhou HX. Recent progress in structure-based anti-influenza drug design. Drug Discov Today 2012; 17:1111-20. [PMID: 22704956 DOI: 10.1016/j.drudis.2012.06.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 05/21/2012] [Accepted: 06/06/2012] [Indexed: 01/22/2023]
Abstract
Seasonal and pandemic influenza have caused high morbidity and mortality worldwide. Recent emergence of influenza A H5N1 and H1N1 strains has heightened concern, especially as a result of their drug resistance. The life cycle of influenza viruses has been well studied and nearly all the viral proteins are becoming potential therapeutic targets. In this review, we present an overview of recent progress in structure-based anti-influenza drug design, paying close attention to the increasing role of computation and strategies for overcoming drug resistance.
Collapse
Affiliation(s)
- Juan Du
- Department of Physics, Florida State University, Tallahassee, FL 32306, USA
| | | | | |
Collapse
|
32
|
Affiliation(s)
- Kalyan Das
- Department of Chemistry and Chemical Biology, Center for Advanced Biotechnology and Medicine, Rutgers University, 679 Hoes Lane, Piscataway, NJ 08854, USA.
| |
Collapse
|
33
|
Wei G, Meng W, Guo H, Pan W, Liu J, Peng T, Chen L, Chen CY. Potent neutralization of influenza A virus by a single-domain antibody blocking M2 ion channel protein. PLoS One 2011; 6:e28309. [PMID: 22164266 PMCID: PMC3229572 DOI: 10.1371/journal.pone.0028309] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 11/06/2011] [Indexed: 11/19/2022] Open
Abstract
Influenza A virus poses serious health threat to humans. Neutralizing antibodies against the highly conserved M2 ion channel is thought to offer broad protection against influenza A viruses. Here, we screened synthetic Camel single-domain antibody (VHH) libraries against native M2 ion channel protein. One of the isolated VHHs, M2-7A, specifically bound to M2-expressed cell membrane as well as influenza A virion, inhibited replication of both amantadine-sensitive and resistant influenza A viruses in vitro, and protected mice from a lethal influenza virus challenge. Moreover, M2-7A showed blocking activity for proton influx through M2 ion channel. These pieces of evidence collectively demonstrate for the first time that a neutralizing antibody against M2 with broad specificity is achievable, and M2-7A may have potential for cross protection against a number of variants and subtypes of influenza A viruses.
Collapse
Affiliation(s)
- Guowei Wei
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Weixu Meng
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Haijiang Guo
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Weiqi Pan
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Jinsong Liu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Tao Peng
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Chang-You Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- * E-mail:
| |
Collapse
|
34
|
Wang J, Ma C, Fiorin G, Carnevale V, Wang T, Hu F, Lamb RA, Pinto LH, Hong M, Klein ML, DeGrado WF. Molecular dynamics simulation directed rational design of inhibitors targeting drug-resistant mutants of influenza A virus M2. J Am Chem Soc 2011; 133:12834-41. [PMID: 21744829 DOI: 10.1021/ja204969m] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Influenza A virus M2 (A/M2) forms a homotetrameric proton selective channel in the viral membrane. It has been the drug target of antiviral drugs such as amantadine and rimantadine. However, most of the current virulent influenza A viruses carry drug-resistant mutations alongside the drug binding site, such as S31N, V27A, and L26F, etc., each of which might be dominant in a given flu season. Among these mutations, the V27A mutation was prevalent among transmissible viruses under drug selection pressure. Until now, V27A has not been successfully targeted by small molecule inhibitors, despite years of extensive medicinal chemistry research efforts and high throughput screening. Guided by molecular dynamics (MD) simulation of drug binding and the influence of drug binding on the dynamics of A/M2 from earlier experimental studies, we designed a series of potent spirane amine inhibitors targeting not only WT, but also both A/M2-27A and L26F mutants with IC(50)s similar to that seen for amantadine's inhibition of the WT channel. The potencies of these inhibitors were further demonstrated in experimental binding and plaque reduction assays. These results demonstrate the power of MD simulations to probe the mechanism of drug binding as well as the ability to guide design of inhibitors of targets that had previously appeared to be undruggable.
Collapse
Affiliation(s)
- Jun Wang
- Department of Chemistry, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6059, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Quantitative analysis of influenza M2 channel blockers. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1808:394-8. [DOI: 10.1016/j.bbamem.2010.08.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 08/26/2010] [Accepted: 08/31/2010] [Indexed: 01/19/2023]
|
36
|
Padkina MV, Parfenova LV, Gradoboeva AE, Sambuk EV. Heterologous interferons synthesis in yeast Pichia pastoris. APPL BIOCHEM MICRO+ 2010. [DOI: 10.1134/s0003683810040083] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
37
|
Hu W, Zeng S, Li C, Jie Y, Li Z, Chen L. Identification of hits as matrix-2 protein inhibitors through the focused screening of a small primary amine library. J Med Chem 2010; 53:3831-4. [PMID: 20394375 DOI: 10.1021/jm901664a] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although amantadine derivatives are the only M2 drugs for influenza virus A, their use is limited in the U.S. because of drug resistance. Here we report the identification of multiple M2 inhibitors that were rapidly generated through focused screening of a small primary amine library that was designed using a scaffold-hopping strategy based on amantadine. These compounds are as active as amantadine and might be hits for further lead generation processes.
Collapse
Affiliation(s)
- Wenhui Hu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Science Park, Guangdong 510530, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
38
|
Cady SD, Luo W, Hu F, Hong M. Structure and function of the influenza A M2 proton channel. Biochemistry 2009; 48:7356-64. [PMID: 19601584 DOI: 10.1021/bi9008837] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The M2 protein of influenza A viruses forms a tetrameric pH-activated proton-selective channel that is targeted by the amantadine class of antiviral drugs. Its ion channel function has been extensively studied by electrophysiology and mutagenesis; however, the molecular mechanism of proton transport is still elusive, and the mechanism of inhibition by amantadine is controversial. We review the functional data on proton channel activity, molecular dynamics simulations of the proton conduction mechanism, and high-resolution structural and dynamical information of this membrane protein in lipid bilayers and lipid-mimetic detergents. These studies indicate that elucidation of the structural basis of M2 channel activity and inhibition requires thorough examination of the complex dynamics and conformational plasticity of the protein in different lipid bilayers and lipid-mimetic environments.
Collapse
Affiliation(s)
- Sarah D Cady
- Department of Chemistry, Iowa State University, Ames, Iowa 50011, USA
| | | | | | | |
Collapse
|
39
|
Wang J, Cady SD, Balannik V, Pinto LH, DeGrado WF, Hong M. Discovery of spiro-piperidine inhibitors and their modulation of the dynamics of the M2 proton channel from influenza A virus. J Am Chem Soc 2009; 131:8066-76. [PMID: 19469531 PMCID: PMC3807247 DOI: 10.1021/ja900063s] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Amantadine has been used for decades as an inhibitor of the influenza A virus M2 protein (AM2) in the prophylaxis and treatment of influenza A infections, but its clinical use has been limited by its central nervous system (CNS) side effects as well as emerging drug-resistant strains of the virus. With the goal of searching for new classes of M2 inhibitors, a structure-activity relation study based on 2-[3-azaspiro(5,5)undecanol]-2-imidazoline (BL-1743) was initiated. The first generation BL-1743 series of compounds has been synthesized and tested by two-electrode voltage-clamp (TEV) assays. The most active compound from this library, 3-azaspiro[5,5]undecane hydrochloride (9), showed an IC(50) as low as 0.92 +/- 0.11 microM against AM2, more than an order of magnitude more potent than amantadine (IC(50) = 16 microM). (15)N and (13)C solid-state NMR was employed to determine the effect of compound 9 on the structure and dynamics of the transmembrane domain of AM2 (AM2-TM) in phospholipid bilayers. Compared to amantadine, spiro-piperidine 9 (1) induces a more homogeneous conformation of the peptide, (2) reduces the dynamic disorder of the G34-I35 backbone near the water-filled central cavity of the helical bundle, and (3) influences the dynamics and magnetic environment of more residues within the transmembrane helices. These data suggest that spiro-piperidine 9 binds more extensively with the AM2 channel, thus leading to stronger inhibitory potency.
Collapse
Affiliation(s)
- Jun Wang
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6059, USA
| | | | | | | | | | | |
Collapse
|
40
|
Balgi AD, Roberge M. Screening for chemical inhibitors of heterologous proteins expressed in yeast using a simple growth-restoration assay. Methods Mol Biol 2009; 486:125-37. [PMID: 19347620 DOI: 10.1007/978-1-60327-545-3_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Overexpression of heterologous proteins in the yeast Saccharomyces cerevisiae often inhibits its growth, while inhibitors of the overexpressed proteins can restore growth. These simple observations form the basis of a technically easy, inexpensive, scalable, and widely applicable assay to identify inhibitors of such proteins. An expression plasmid for the inducible expression of a gene of interest is introduced into a yeast strain rendered more sensitive to chemicals by deletion of efflux pumps. Protein expression is induced, cells are exposed to test chemicals, and growth is measured by A (600) reading. The chemicals that relieve growth inhibition are subjected to secondary assays to establish their selectivity toward the protein of interest. This assay has been used successfully to identify inhibitors of proteins of viral, microbial, and mammalian origin.
Collapse
Affiliation(s)
- Aruna D Balgi
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
41
|
Abstract
Annual epidemics of influenza virus infection are responsible for considerable morbidity and mortality, and pandemics are much more devastating. Considerable knowledge of viral infectivity and replication has been acquired, but many details still have to be elucidated and the virus remains a challenging target for drug design and development. This review provides an overview of the antiviral drugs targeting the influenza viral replicative cycle. Included are a brief description of their chemical syntheses and biological activities. For other reviews, see References1-9.
Collapse
Affiliation(s)
- Irene M. Lagoja
- Laboratory of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Rega Institute for Medical Research, Minderbroedersstraat 10, B‐3000 Leuven, Belgium
| | - Erik De Clercq
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, Minderbroedersstraat 10, B‐3000 Leuven, Belgium
| |
Collapse
|
42
|
Galao RP, Scheller N, Alves-Rodrigues I, Breinig T, Meyerhans A, Díez J. Saccharomyces cerevisiae: a versatile eukaryotic system in virology. Microb Cell Fact 2007; 6:32. [PMID: 17927824 PMCID: PMC2148055 DOI: 10.1186/1475-2859-6-32] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Accepted: 10/10/2007] [Indexed: 02/07/2023] Open
Abstract
The yeast Saccharomyces cerevisiae is a well-established model system for understanding fundamental cellular processes relevant to higher eukaryotic organisms. Less known is its value for virus research, an area in which Saccharomyces cerevisiae has proven to be very fruitful as well. The present review will discuss the main achievements of yeast-based studies in basic and applied virus research. These include the analysis of the function of individual proteins from important pathogenic viruses, the elucidation of key processes in viral replication through the development of systems that allow the replication of higher eukayotic viruses in yeast, and the use of yeast in antiviral drug development and vaccine production.
Collapse
Affiliation(s)
- Rui P Galao
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|
43
|
Vottero E, Balgi A, Woods K, Tugendreich S, Melese T, Andersen RJ, Mauk AG, Roberge M. Inhibitors of human indoleamine 2,3-dioxygenase identified with a target-based screen in yeast. Biotechnol J 2006; 1:282-8. [PMID: 16897708 DOI: 10.1002/biot.200600001] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Indoleamine 2,3-dioxygenase (IDO) is a tryptophan degradation enzyme that is emerging as an important drug target. IDO is expressed by many human tumors to help them escape immune detection, and it has been implicated in depression and in the formation of senile nuclear cataracts. There is a need for potent and selective IDO inhibitors for use in research and as lead compounds for drug development. We show that expression of human IDO in a Saccharomyces cerevisiae tryptophan auxotroph restricts yeast growth in the presence of low tryptophan concentrations and that inhibition of IDO activity can restore growth. We use this assay to screen for IDO inhibitors in collections of pure chemicals and crude natural extracts. We identify NSC 401366 (imidodicarbonimidic diamide, N-methyl-N'-9-phenanthrenyl-, monohydrochloride) as a potent nonindolic IDO inhibitor (Ki=1.5 +/- 0.2 microM) that is competitive with respect to tryptophan. We also use this assay to identify the active compound caulerpin from a crude algal extract. The yeast growth restoration assay is simple and inexpensive. It combines desirable attributes of cell- and target-based screens and is an attractive tool for chemical biology and drug screening.
Collapse
Affiliation(s)
- Eduardo Vottero
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Venkataraman P, Lamb RA, Pinto LH. Chemical rescue of histidine selectivity filter mutants of the M2 ion channel of influenza A virus. J Biol Chem 2005; 280:21463-72. [PMID: 15784624 DOI: 10.1074/jbc.m412406200] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The influenza virus M2 proton-selective ion channel activity facilitates virus uncoating, a process that occurs in the acidic environment of the endosome. The M2 channel causes acidification of the interior of the virus particle, which results in viral protein-protein dissociation. The M2 protein is a homotetramer that contains in its aqueous pore a histidine residue (His-37) that acts as a selectivity filter and a tryptophan residue (Trp-41) that acts as a channel gate. Substitution of His-37 modifies M2 ion channel properties drastically. However, the results of such experiments are difficult to interpret because substitution of His-37 could cause gross structural changes to the channel pore. We described here experiments in which partial or, in some cases, full rescue of specific M2 ion channel properties of His-37 substitution mutants was achieved by addition of imidazole to the bathing medium. Chemical rescue was demonstrated for three histidine substitution mutant ion channels (M2-H37G, M2-H37S, and M2-H37T) and for two double mutants in which the Trp-41 channel gate was also mutated (H37G/W41Y and H37G/W41A). Currents of the M2-H37G mutant ion channel were inhibited by Cu(II), which has been shown to coordinate with His-37 in the wild-type channel. Chemical rescue was very specific for imidazole. Buffer molecules that were neutral when protonated (4-morpholineethanesulfonic acid and 3-morpholino-2-hydroxypropanesulfonic acid) did not rescue ion channel activity of the M2-H37G mutant ion channel, but 1-methylimidazole did provide partial rescue of function. These results were consistent with a model for proton transport through the pore of the wild-type channel in which the imidazole side chain of His-37 acted as an intermediate proton acceptor/donor group.
Collapse
Affiliation(s)
- Padmavati Venkataraman
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208-3500, USA
| | | | | |
Collapse
|
45
|
Schroeder C, Heider H, Möncke-Buchner E, Lin TI. The influenza virus ion channel and maturation cofactor M2 is a cholesterol-binding protein. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2004; 34:52-66. [PMID: 15221235 DOI: 10.1007/s00249-004-0424-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2003] [Revised: 03/06/2004] [Accepted: 05/19/2004] [Indexed: 11/26/2022]
Abstract
The influenza-virus M2 protein has proton channel activity required for virus uncoating and maturation of hemagglutinin (HA) through low-pH compartments. The proton channel is cytotoxic in heterologous expression systems and can be blocked with rimantadine. In an independent, rimantadine-resistant function, M2, interacting with the M1 protein, controls the shape of virus particles. These bud from cholesterol-rich membrane rafts where viral glycoproteins and matrix (M1)/RNP complexes assemble. We demonstrate that M2 preparations from influenza virus-infected cells and from a baculovirus expression system contain 0.5-0.9 molecules of cholesterol per monomer. Sequence analyses of the membrane-proximal M2 endodomain reveal interfacial hydrophobicity, a cholesterol-binding motif first identified in peripheral benzodiazepine receptor and human immunodeficiency virus gp41, and an overlapping phosphatidylinositol 4,5-bisphosphate-binding motif. M2 induced rimantadine-reversible cytotoxicity in intrinsically cholesterol-free E. coli, and purified E. coli-expressed M2 functionally reconstituted into cholesterol-free liposomes supported rimantadine-sensitive proton translocation. Therefore, cholesterol was nonessential for M2 ion-channel function and cytotoxicity and for the effect of rimantadine. Only about 5-8% of both M2 preparations, regardless of cholesterol content, associated with detergent-resistant membranes. Cholesterol affinity and palmitoylation, in combination with a short transmembrane segment suggest M2 is a peripheral raft protein. Preference for the raft/non-raft interface may determine colocalization with HA during apical transport, the low level of M2 incorporated into the viral envelope and its undisclosed role in virus budding for which a model is presented. M2 may promote clustering and merger of rafts and the pinching-off (fission) of virus particles.
Collapse
Affiliation(s)
- Cornelia Schroeder
- Abteilung Virologie, Institut für Mikrobiologie und Hygiene, Universität des Saarlandes, Homburg/Saar, 66421 Homburg, Germany
| | | | | | | |
Collapse
|
46
|
Shinjoh M, Yoshikawa T, Li Y, Shiraishi K, Ueki H, Nerome K. Prophylaxis and treatment of influenza encephalitis in an experimental mouse model. J Med Virol 2002; 67:406-17. [PMID: 12116035 DOI: 10.1002/jmv.10088] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A mouse model study using mouse brain-adapted influenza A virus was performed to establish the prophylaxis and treatment of influenza encephalitis and encephalopathy. All mice died after intranasal inoculation of the brain-adapted influenza A virus (H7N3), and the pathological findings indicated the presence of significant encephalitis. Viral antigen was also detected in the brain, both pathologically and virologically. By contrast, infected mice immunized with inactivated vaccine of the same strain did not lose weight, which is an indicator of the overall condition of the mice, and all of them survived. Similarly, antiserum treatment in the early period (0-1 day post-infection) resulted in 100% survival, and no pathological findings were observed in the brain. However, mice treated with antiserum 3 days post-infection showed encephalitis with viral antigens in both glial cells and neurocytes. Although amantadine treatment for 4 days delayed weight loss, it did not prevent death from encephalitis. These results show vaccination and early antiserum treatment to be highly effective, whereas 4-day treatment of amantadine was not very effective in treating or preventing influenza encephalitis. The life-prolonging effect of amantadine, however, suggests that use of amantadine together with other treatments may inhibit the progression of encephalitis.
Collapse
Affiliation(s)
- Masayoshi Shinjoh
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Viral ion channels are short auxiliary membrane proteins with a length of ca. 100 amino acids. They are found in enveloped viruses from influenza A, influenza B and influenza C (Orthomyxoviridae), and the human immunodeficiency virus type 1 (HIV-1, Retroviridae). The channels are called M2 (influenza A), NB (influenza B), CM2 (influenza C) and Vpu (HIV-1). Recently, in Paramecium bursaria chlorella virus (PBCV-1, Phycodnaviridae), a K+ selective ion channel has been discovered. The viral channels form homo oligomers to allow an ion flux and represent miniaturised systems. Proton conductivity of M2 is established; NB, Vpu and the potassium channel from PBC-1 conduct ions; for CM2 ion conductivity is still under proof. This review summarises the current knowledge of these short viral membrane proteins. Their discovery is outlined and experimental evidence for their structure and function is discussed. Studies using computational methods are presented as well as investigations of drug-protein interactions.
Collapse
Affiliation(s)
- Wolfgang B Fischer
- Department of Biochemistry, University of Oxford, South Parks Road, OX1 3QU, UK.
| | | |
Collapse
|
48
|
Tugendreich S, Perkins E, Couto J, Barthmaier P, Sun D, Tang S, Tulac S, Nguyen A, Yeh E, Mays A, Wallace E, Lila T, Shivak D, Prichard M, Andrejka L, Kim R, Melese T. A streamlined process to phenotypically profile heterologous cDNAs in parallel using yeast cell-based assays. Genome Res 2001; 11:1899-912. [PMID: 11691855 PMCID: PMC311162 DOI: 10.1101/gr.191601] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
To meet the demands of developing lead drugs for the profusion of human genes being sequenced as part of the human genome project, we developed a high-throughput assay construction method in yeast. A set of optimized techniques allows us to rapidly transfer large numbers of heterologous cDNAs from nonyeast plasmids into yeast expression vectors. These high- or low-copy yeast expression plasmids are then converted quickly into integration-competent vectors for phenotypic profiling of the heterologous gene products. The process was validated first by testing proteins of diverse function, such as p38, poly(ADP-ribose) polymerase-1, and PI 3-kinase, by making active-site mutations and using existing small molecule inhibitors of these proteins. For less well-characterized genes, a novel random mutagenesis scheme was developed that allows a combination selection/screen for mutations that retain full-length expression and yet reverse a growth phenotype in yeast. A broad range of proteins in different functional classes has been profiled, with an average yield for growth interference phenotypes of approximately 30%. The ease of manipulation of the yeast genome affords us the opportunity to approach drug discovery and exploratory biology on a genomic scale and shortens assay development time significantly.
Collapse
Affiliation(s)
- S Tugendreich
- Iconix Pharmaceuticals, Mountain View, California 94043, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
|
50
|
Lin TI, Schroeder C. Definitive assignment of proton selectivity and attoampere unitary current to the M2 ion channel protein of influenza A virus. J Virol 2001; 75:3647-56. [PMID: 11264354 PMCID: PMC114856 DOI: 10.1128/jvi.75.8.3647-3656.2001] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The viral ion channel protein M2 supports the transit of influenza virus and its glycoproteins through acidic compartments of the cell. M2 conducts endosomal protons into the virion to initiate uncoating and, by equilibrating the pH at trans-Golgi membranes, preserves the native conformation of acid-sensitive viral hemagglutinin. The exceptionally low conductance of the M2 channel thwarted resolution of single channels by electrophysiological techniques. Assays of liposome-reconstituted M2 yielded the average unitary channel current of the M2 tetramer--1.2 aA (1.2 x 10(-18) A) at neutral pH and 2.7 to 4.1 aA at pH 5.7--which activates the channel. Extrapolation to physiological temperature predicts 4.8 and 40 aA, respectively, and a unitary conductance of 0.03 versus 0.4 fS. This minute activity, below previous estimates, appears sufficient for virus reproduction, but low enough to avert abortive cytotoxicity. The unitary permeability of M2 was within the range reported for other proton channels. To address the ion selectivity of M2, we exploited the coupling of ionic influx and efflux in sealed liposomes. Metal ion fluxes were monitored by proton counterflow, employing a pH probe 1,000 times more sensitive than available Na+ or K+ probes. Even low-pH-activated M2 did not conduct Na+ and K+. The proton selectivity of M2 was estimated to be at least 3 x 10(6) (over sodium or potassium ions), in agreement with electrophysiological studies. The stringent proton selectivity of M2 suggests that the cytopathology of influenza virus does not involve direct perturbation of cellular sodium or potassium gradients.
Collapse
Affiliation(s)
- T I Lin
- Institut für Virologie, Universitätsklinikum Charité der Humboldt-Universität zu Berlin, D-10098 Berlin, Germany
| | | |
Collapse
|