1
|
Subramaniyan B, Falcon EC, Moore AR, Larabee JL, Nimmo SL, Berrios-Rivera JL, Reddig WJ, Blewett EL, Papin JF, Walters MS, Burgett AWG. Anti-SARS-CoV-2 Small Molecule Targeting of Oxysterol-Binding Protein (OSBP) Activates Cellular Antiviral Innate Immunity. ACS Infect Dis 2025. [PMID: 40255103 DOI: 10.1021/acsinfecdis.4c00631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Human oxysterol-binding protein (OSBP) is a potentially druggable mediator in the replication of a broad spectrum of positive-sense (+) single-stranded RNA (ssRNA) viruses, including members of the Picornaviridae, Flaviviridae, and Coronaviridae. OSBP is a cytoplasmic lipid transporting protein capable of moving cholesterol and phosphoinositides between the endoplasmic reticulum (ER) and Golgi, and the ER and lysosome. Several structurally diverse antiviral compounds have been reported to function through targeting OSBP, including the natural product compound OSW-1. Our prior work shows that transient OSW-1 treatment induces a reduction in OSBP protein levels over multiple successive cell generations (i.e., multigenerational), with no apparent cellular toxicity, and the OSW-1-induced reduction of OSBP has antiviral activity against multiple (+)ssRNA viruses. This study extends these findings and establishes that OSW-1 has in vitro antiviral activity against multiple pathogenic (+)ssRNA viruses, including human rhinovirus (HRV1B), the feline coronavirus peritonitis virus (FIPV), human coronavirus 229E (HCoV-229E), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We also demonstrate that OSW-1 treatment in human airway epithelial cells alters the expression of multiple antiviral innate immune mediators, including the interferon (IFN) related genes IFNB1, IFNL3, CXCL10, ISG15, and MX1. Furthermore, OSW-1 enhances the induction of specific components of type I and III IFN antiviral responses triggered by the RNA viral mimetic polyinosinic-polycytidylic acid (Poly IC). In summary, this study further demonstrates the importance of OSBP in (+)ssRNA virus replication and presents OSBP as a potential regulator of cellular antiviral innate immune responses.
Collapse
Affiliation(s)
- Bharathiraja Subramaniyan
- Department of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Emily C Falcon
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Andrew R Moore
- Department of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Jason L Larabee
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Susan L Nimmo
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Jorge L Berrios-Rivera
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - William J Reddig
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma 74107, United States
| | - Earl L Blewett
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, Oklahoma 74107, United States
| | - James F Papin
- Department of Pathology, Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma 73104, United States
| | - Matthew S Walters
- Department of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Anthony W G Burgett
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| |
Collapse
|
2
|
Jézéquel G, Grimanelli Z, Guimard C, Bigay J, Haddad J, Bignon J, Apel C, Steinmetz V, Askenatzis L, Levaïque H, Pradelli C, Pham VC, Huong DTM, Litaudon M, Gautier R, El Kalamouni C, Antonny B, Desrat S, Mesmin B, Roussi F. Minimalist Natural ORPphilin Macarangin B Delineates OSBP Biological Function. J Med Chem 2025; 68:196-211. [PMID: 39704626 DOI: 10.1021/acs.jmedchem.4c01705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
OSBP ligands from the ORPphilin family are chemically complex natural products with promising anticancer properties. Here, we describe macarangin B, a natural racemic flavonoid selective for OSBP, which stands out from other ORPphilins due to its structural simplicity and distinct biological activity. Using a bioinspired strategy, we synthesized both (R,R,R) and (S,S,S)-macarangin B enantiomers, enabling us to study their interaction with OSBP based on their unique optical properties. Experimental and computational analyzes revealed that (R,R,R)-macarangin B has the highest affinity for OSBP. Importantly, both enantiomers showed significantly decreased cytotoxicity compared to other ORPphilins, suggesting OSBP is not the primary target in ORPphilin-induced cell death. Yet, OSBP is an attractive antiviral target, as it is hijacked by many positive-strand RNA viruses. Remarkably, (R,R,R)-macarangin B significantly inhibited Zika virus replication in human cells, highlighting its potential as a lead compound for antiviral drug development.
Collapse
Affiliation(s)
- Gwenaëlle Jézéquel
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Zoé Grimanelli
- Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne 06560, France
| | - Carole Guimard
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Joëlle Bigay
- Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne 06560, France
| | - Juliano Haddad
- Inserm U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de la Réunion, Sainte Clotilde 94791, France
| | - Jérôme Bignon
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Cécile Apel
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Vincent Steinmetz
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Laurie Askenatzis
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Hélène Levaïque
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Clara Pradelli
- Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne 06560, France
| | - Van Cuong Pham
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Caugiay, Hanoi 10000, Vietnam
| | - Doan T M Huong
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Caugiay, Hanoi 10000, Vietnam
| | - Marc Litaudon
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Romain Gautier
- Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne 06560, France
| | - Chaker El Kalamouni
- Inserm U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de la Réunion, Sainte Clotilde 94791, France
| | - Bruno Antonny
- Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne 06560, France
| | - Sandy Desrat
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| | - Bruno Mesmin
- Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne 06560, France
| | - Fanny Roussi
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-sur-Yvette 91198, France
| |
Collapse
|
3
|
Nigro F, Civra A, Porporato D, Costantino M, Francese R, Poli G, Romani A, Lembo D, Marinozzi M. Cholenamide-based, antiviral fluorescent probes targeting oxysterol-binding protein. Bioorg Chem 2024; 153:107922. [PMID: 39486114 DOI: 10.1016/j.bioorg.2024.107922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/25/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
Oxysterols (OSs) represent a large family of cholesterol-derived molecules, involved in several physiological and pathological processes. Recently, we reported the remarkable antiviral activity against herpes simplex virus 2 (HSV-2) infection of three cholenamide or homocholenamide derivatives, namely PFM067, PFM064, and PFM069, identified by the screening of an in-house library of OS derivatives. With the aim to shed light on the antiviral mechanism of action of this class of molecules, we assumed to exploit the use of cholenamide-based fluorescent probes. Herein, we report that PFM120 and PFM124, two fluorescent tagged version of PFM067 maintain the same antiviral properties against HSV-2 as the parent compound and localize intracellularly inside the endoplasmic reticulum and the cis-Golgi network. Moreover, we also demonstrate that both tagged molecules co-localize with oxysterol-binding protein (OSBP) and are able to induce its re-localization. Finally, we report that PFM120 and PFM124 are endowed with antiviral activity against another OSBP-dependent viral pathogen, i.e. the human rhinovirus (HRV), different in structure and replication strategy from HSV-2. Taken together, these results candidate PFM120 and PFM124 as useful tools to investigate the actual mechanism of action and molecular target(s) of cholenamide-based antivirals and provide a proof of principle to explore them as a promising broad-spectrum class of antiviral agents.
Collapse
Affiliation(s)
- Fatima Nigro
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Perugia, Via del Liceo, 1-06123 Perugia, Italy
| | - Andrea Civra
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Regione Gonzole, 1-10043 Orbassano, Turin, Italy
| | - Domiziana Porporato
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Regione Gonzole, 1-10043 Orbassano, Turin, Italy; National PhD Programme in One Health approaches to infectious diseases and life science research, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, 27100, Italy
| | - Matteo Costantino
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Regione Gonzole, 1-10043 Orbassano, Turin, Italy
| | - Rachele Francese
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Regione Gonzole, 1-10043 Orbassano, Turin, Italy
| | - Giuseppe Poli
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Regione Gonzole, 1-10043 Orbassano, Turin, Italy
| | - Aldo Romani
- Dipartimento di Chimica, Biologia e Biotecnologie, Università degli Studi di Perugia, Via Elce di Sotto, 8-06123 Perugia, Italy
| | - David Lembo
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Regione Gonzole, 1-10043 Orbassano, Turin, Italy
| | - Maura Marinozzi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Perugia, Via del Liceo, 1-06123 Perugia, Italy.
| |
Collapse
|
4
|
Granela H, Perez AB, Morier L, Alvarez M, Guzmán MG, Sierra B. Possible effects of ancestry-related oxysterol-binding protein-like 10 genetic polymorphisms on dengue virus replication and anti-dengue immune response. Heliyon 2024; 10:e37535. [PMID: 39444395 PMCID: PMC11497451 DOI: 10.1016/j.heliyon.2024.e37535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024] Open
Abstract
Purpose Oxysterol-binding protein-like 10 (OSBPL10) gene has been associated with reduced susceptibility to severe dengue in individuals of African descent. The aim of this study was to determine the possible effect of OSBPL10 on dengue virus (DENV) replication as well as the impact of African and European haplotypes of six OSBPL10 small nuclear polymorphisms (SNPs) on dengue multiplication and innate immune response. Methods We conducted gene knockdown experiments targeting OSBPL10 in THP-1 and Huh-7D12 cell lines, followed by a DENV-2 replication assay. Extracellular viral load was determined using qRT-PCR. To investigate the impact of SNPs haplotypes on viral replication and gene expression we cultured peripheral blood mononuclear cells (PBMC) from individuals with homozygous African and European haplotypes of OSBPL10 with DENV-2. Individual genotyping was performed using High Resolution Melt (HRM) analysis. The level of viral replication was assessed through plaque assay, while RT-PCR was employed to determine the expression levels of RXR-α, IFN-γ, IL-10 and IL-8 genes. Results In vitro OSBPL10 knockdown significantly reduced DENV-2 replication. Individuals carrying European haplotypes showed higher DENV titers along with elevated levels of RXR-α and IL-8 mRNA compared to those carrying African haplotypes, who exhibited lower viral loads alongside increased IFN-γ and IL-10 expression. Conclusions Our findings further explore the role of OSBPL10 in DENV multiplication, immune response to infection. The European haplotypes of OSBPL10 appear to increase DENV replication and promote RXR-α and IL-8 mRNA expression which correlates with the suppressive effect of these mediators on type I IFN, promoting viral replication and a deficient antiviral response. In contrast, the African haplotype showed a reduction in DENV replication and enhanced IFN-γ and IL-10 mRNA expression, which could be related to the better management of dengue infection and the low frequency of severe disease in this ethnic groupe.
Collapse
Affiliation(s)
- Hector Granela
- Virology Department, “Pedro Kourí” Tropical Medicine Institute (IPK). Autopista Novia del Mediodía, km 61/2.Havana, Cuba
| | - Ana B. Perez
- Virology Department, “Pedro Kourí” Tropical Medicine Institute (IPK). Autopista Novia del Mediodía, km 61/2.Havana, Cuba
| | - Luis Morier
- Virology Department, Biology Faculty, University of Havana. 455, 25th Street. Havana, Cuba
| | - Mayling Alvarez
- Virology Department, “Pedro Kourí” Tropical Medicine Institute (IPK). Autopista Novia del Mediodía, km 61/2.Havana, Cuba
| | - Maria G. Guzmán
- Virology Department, “Pedro Kourí” Tropical Medicine Institute (IPK). Autopista Novia del Mediodía, km 61/2.Havana, Cuba
| | - Beatriz Sierra
- Virology Department, “Pedro Kourí” Tropical Medicine Institute (IPK). Autopista Novia del Mediodía, km 61/2.Havana, Cuba
| |
Collapse
|
5
|
Li JQ, Zhang J, Chen Y, Le T, Chang MX. Coordination of oxysterol binding protein 1 and VAP-A/B modulates the generation of cholesterol and viral inclusion bodies to promote grass carp reovirus replication. Front Immunol 2024; 15:1419321. [PMID: 39081319 PMCID: PMC11286474 DOI: 10.3389/fimmu.2024.1419321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024] Open
Abstract
Similar to other RNA viruses, grass carp reovirus, the causative agent of the hemorrhagic disease, replicates in cytoplasmic viral inclusion bodies (VIBs), orchestrated by host proteins and lipids. The host pathways that facilitate the formation and function of GCRV VIBs are poorly understood. This work demonstrates that GCRV manipulates grass carp oxysterol binding protein 1 (named as gcOSBP1) and vesicle-associated membrane protein-associated protein A/B (named as gcVAP-A/B), 3 components of cholesterol transport pathway, to generate VIBs. By siRNA-mediated knockdown, we demonstrate that gcOSBP1 is an essential host factor for GCRV replication. We reveal that the nonstructural proteins NS80 and NS38 of GCRV interact with gcOSBP1, and that the gcOSBP1 is recruited by NS38 and NS80 for promoting the generation of VIBs. gcOSBP1 increases the expression of gcVAP-A/B and promotes the accumulation of intracellular cholesterol. gcOSBP1 also interacts with gcVAP-A/B for forming gcOSBP1-gcVAP-A/B complexes, which contribute to enhance the accumulation of intracellular cholesterol and gcOSBP1-mediated generation of VIBs. Inhibiting cholesterol accumulation by lovastatin can completely abolish the effects of gcOSBP1 and/or gcVAP-A/B in promoting GCRV infection, suggesting that cholesterol accumulation is vital for gcOSBP1- and/or gcVAP-A/B-mediated GCRV replication. Thus, our results, which highlight that gcOSBP1 functions in the replication of GCRV via its interaction with essential viral proteins for forming VIBs and with host gcVAP-A/B, provide key molecular targets for obtaining anti-hemorrhagic disease grass carp via gene editing technology.
Collapse
Affiliation(s)
- Jia Qi Li
- Chongqing Key Laboratory of Conservation and Utilization of Freshwater Fishes, College of Life Sciences, Chongqing Normal University, Chongqing, China
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Jie Zhang
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yang Chen
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tao Le
- Chongqing Key Laboratory of Conservation and Utilization of Freshwater Fishes, College of Life Sciences, Chongqing Normal University, Chongqing, China
| | - Ming Xian Chang
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
6
|
Sotorilli GE, Gravina HD, de Carvalho AC, Shimizu JF, Fontoura MA, Melo-Hanchuk TD, Cordeiro AT, Marques RE. Phenotypical Screening of an MMV Open Box Library and Identification of Compounds with Antiviral Activity against St. Louis Encephalitis Virus. Viruses 2023; 15:2416. [PMID: 38140657 PMCID: PMC10747599 DOI: 10.3390/v15122416] [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: 10/14/2023] [Revised: 12/02/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
St. Louis encephalitis virus (SLEV) is a neglected mosquito-borne Flavivirus that may cause severe neurological disease in humans and other animals. There are no specific treatments against SLEV infection or disease approved for human use, and drug repurposing may represent an opportunity to accelerate the development of treatments against SLEV. Here we present a scalable, medium-throughput phenotypic cell culture-based screening assay on Vero CCL81 cells to identify bioactive compounds that could be repurposed against SLEV infection. We screened eighty compounds from the Medicines for Malaria Venture (MMV) COVID Box library to identify nine (11%) compounds that protected cell cultures from SLEV-induced cytopathic effects, with low- to mid-micromolar potencies. We validated six hit compounds using viral plaque-forming assays to find that the compounds ABT-239, Amiodarone, Fluphenazine, Posaconazole, Triparanol, and Vidofludimus presented varied levels of antiviral activity and selectivity depending on the mammalian cell type used for testing. Importantly, we identified and validated the antiviral activity of the anti-flavivirus nucleoside analog 7DMA against SLEV. Triparanol and Fluphenazine reduced infectious viral loads in both Vero CCL81 and HBEC-5i cell cultures and, similar to the other validated compounds, are likely to exert antiviral activity through a molecular target in the host.
Collapse
Affiliation(s)
- Giuliana Eboli Sotorilli
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
- Department of Genetics, Microbiology and Immunology, Institute of Biology, State University of Campinas (UNICAMP), Campinas 13083-100, Brazil
| | - Humberto Doriguetto Gravina
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
| | - Ana Carolina de Carvalho
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
- Department of Genetics, Microbiology and Immunology, Institute of Biology, State University of Campinas (UNICAMP), Campinas 13083-100, Brazil
| | - Jacqueline Farinha Shimizu
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
| | - Marina Alves Fontoura
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
- Department of Cellular and Structural Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas 13083-100, Brazil
| | - Talita Diniz Melo-Hanchuk
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
| | - Artur Torres Cordeiro
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
| |
Collapse
|
7
|
Lin Y, Ran L, Du X, Yang H, Wu Y. Oxysterol-Binding Protein: new insights into lipid transport functions and human diseases. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159365. [PMID: 37455011 DOI: 10.1016/j.bbalip.2023.159365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Oxysterol-binding protein (OSBP) mediates lipid exchange between organelles at membrane contact sites, thereby regulating lipid dynamics and homeostasis. How OSBP's lipid transfer function impacts health and disease remain to be elucidated. In this review, we first summarize the structural characteristics and lipid transport functions of OSBP, and then focus on recent progresses linking OSBP with fatty liver disease, diabetes, lysosome-related diseases, cancer and viral infections, with the aim of discovering novel therapeutic strategies for common human diseases.
Collapse
Affiliation(s)
- Yani Lin
- Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250021, China
| | - Liyuan Ran
- Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250021, China; Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Ximing Du
- School of Biotechnology and Biomolecular Sciences, the University of New South Wales, Sydney, NSW 2052, Australia
| | - Hongyuan Yang
- School of Biotechnology and Biomolecular Sciences, the University of New South Wales, Sydney, NSW 2052, Australia.
| | - Yingjie Wu
- Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250021, China; Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning 116044, China; Department of Molecular Pathobiology, New York University College of Dentistry, New York 10010, USA.
| |
Collapse
|
8
|
Subra M, Antonny B, Mesmin B. New insights into the OSBP‒VAP cycle. Curr Opin Cell Biol 2023; 82:102172. [PMID: 37245352 DOI: 10.1016/j.ceb.2023.102172] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/16/2023] [Accepted: 04/24/2023] [Indexed: 05/30/2023]
Abstract
VAP-A is a major endoplasmic reticulum (ER) receptor that allows this organelle to engage numerous membrane contact sites with other organelles. One highly studied example is the formation of contact sites through VAP-A interaction with Oxysterol-binding protein (OSBP). This lipid transfer protein transports cholesterol from the ER to the trans-Golgi network owing to the counter-exchange of the phosphoinositide PI(4)P. In this review, we highlight recent studies that advance our understanding of the OSBP cycle and extend the model of lipid exchange to other cellular contexts and other physiological and pathological conditions.
Collapse
Affiliation(s)
- Mélody Subra
- Université Côte d'Azur, Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 660 Route des Lucioles, 06560, Valbonne, France
| | - Bruno Antonny
- Université Côte d'Azur, Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 660 Route des Lucioles, 06560, Valbonne, France.
| | - Bruno Mesmin
- Université Côte d'Azur, Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 660 Route des Lucioles, 06560, Valbonne, France.
| |
Collapse
|
9
|
Severance ZC, Nuñez JI, Le-McClain AT, Malinky CA, Bensen RC, Fogle RS, Manginelli GW, Sakers SH, Falcon EC, Bui RH, Snead KJ, Bourne CR, Burgett AWG. Structure-Activity Relationships of Ligand Binding to Oxysterol-Binding Protein (OSBP) and OSBP-Related Protein 4. J Med Chem 2023; 66:3866-3875. [PMID: 36916802 PMCID: PMC10786236 DOI: 10.1021/acs.jmedchem.2c01025] [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] [Indexed: 03/16/2023]
Abstract
Oxysterol-binding protein (OSBP) and OSBP-related protein 4 (ORP4) have emerged as potentially druggable targets in antiviral and precision cancer drug development. Multiple structurally diverse small molecules function through targeting the OSBP/ORP family of proteins, including the antiviral steroidal compounds OSW-1 and T-00127-HEV2. Here, the structure-activity relationships of oxysterols and related compound binding to human OSBP and ORP4 are characterized. Oxysterols with hydroxylation at various side chain positions (i.e., C-20, C-24, C-25, and C-27)─but not C-22─confer high affinity interactions with OSBP and ORP4. A library of 20(S)-hydroxycholesterol analogues with varying sterol side chains reveal that side chain length modifications are not well tolerated for OSBP and ORP4 interactions. This side chain requirement is contradicted by the high affinity binding of T-00127-HEV2, a steroidal compound lacking the side chain. The binding results, in combination with docking studies using homology models of OSBP and ORP4, suggest multiple modes of steroidal ligand binding to OSBP and ORP4.
Collapse
Affiliation(s)
- Zachary C Severance
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Juan I Nuñez
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Anh T Le-McClain
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Cori A Malinky
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Ryan C Bensen
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Robert S Fogle
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Gianni W Manginelli
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Sophia H Sakers
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Emily C Falcon
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Richard Hoang Bui
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Kevin J Snead
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Christina R Bourne
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Anthony W G Burgett
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| |
Collapse
|
10
|
Chen Y, Chi X, Zhang H, Zhang Y, Qiao L, Ding J, Han Y, Lin Y, Jiang J. Identification of Potent Zika Virus NS5 RNA-Dependent RNA Polymerase Inhibitors Combining Virtual Screening and Biological Assays. Int J Mol Sci 2023; 24:ijms24031900. [PMID: 36768218 PMCID: PMC9915956 DOI: 10.3390/ijms24031900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
The Zika virus (ZIKV) epidemic poses a significant threat to human health globally. Thus, there is an urgent need for developing effective anti-ZIKV agents. ZIKV non-structural protein 5 RNA-dependent RNA polymerase (RdRp), a viral enzyme for viral replication, has been considered an attractive drug target. In this work, we screened an anti-infection compound library and a natural product library by virtual screening to identify potential candidates targeting RdRp. Then, five selected candidates were further applied for RdRp enzymatic analysis, cytotoxicity, and binding examination by SPR. Finally, posaconazole (POS) was confirmed to effectively inhibit both RdRp activity with an IC50 of 4.29 μM and the ZIKV replication with an EC50 of 0.59 μM. Moreover, POS was shown to reduce RdRp activity by binding with the key amino acid D666 through molecular docking and site-directed mutation analysis. For the first time, our work found that POS could inhibit ZIKV replication with a stronger inhibitory activity than chloroquine. This work also demonstrated fast anti-ZIKV screening for inhibitors of RdRp and provided POS as a potential anti-ZIKV agent.
Collapse
|
11
|
Linking Late Endosomal Cholesterol with Cancer Progression and Anticancer Drug Resistance. Int J Mol Sci 2022; 23:ijms23137206. [PMID: 35806209 PMCID: PMC9267071 DOI: 10.3390/ijms23137206] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer cells undergo drastic metabolic adaptions to cover increased bioenergetic needs, contributing to resistance to therapies. This includes a higher demand for cholesterol, which often coincides with elevated cholesterol uptake from low-density lipoproteins (LDL) and overexpression of the LDL receptor in many cancers. This implies the need for cancer cells to accommodate an increased delivery of LDL along the endocytic pathway to late endosomes/lysosomes (LE/Lys), providing a rapid and effective distribution of LDL-derived cholesterol from LE/Lys to other organelles for cholesterol to foster cancer growth and spread. LDL-cholesterol exported from LE/Lys is facilitated by Niemann–Pick Type C1/2 (NPC1/2) proteins, members of the steroidogenic acute regulatory-related lipid transfer domain (StARD) and oxysterol-binding protein (OSBP) families. In addition, lysosomal membrane proteins, small Rab GTPases as well as scaffolding proteins, including annexin A6 (AnxA6), contribute to regulating cholesterol egress from LE/Lys. Here, we summarize current knowledge that links upregulated activity and expression of cholesterol transporters and related proteins in LE/Lys with cancer growth, progression and treatment outcomes. Several mechanisms on how cellular distribution of LDL-derived cholesterol from LE/Lys influences cancer cell behavior are reviewed, some of those providing opportunities for treatment strategies to reduce cancer progression and anticancer drug resistance.
Collapse
|
12
|
The Antifungal Itraconazole Is a Potent Inhibitor of Chikungunya Virus Replication. Viruses 2022; 14:v14071351. [PMID: 35891332 PMCID: PMC9317443 DOI: 10.3390/v14071351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 12/04/2022] Open
Abstract
Chikungunya virus (CHIKV) is the causative agent of chikungunya fever, a disabling disease that can cause long-term severe arthritis. Since the last large CHIKV outbreak in 2015, the reemergence of the virus represents a serious public health concern. The morbidity associated with viral infection emphasizes the need for the development of specific anti-CHIKV drugs. Herein, we describe the development and characterization of a CHIKV reporter replicon cell line and its use in replicon-based screenings. We tested 960 compounds from MMV/DNDi Open Box libraries and identified four candidates with interesting antiviral activities, which were confirmed in viral infection assays employing CHIKV-nanoluc and BHK-21 cells. The most noteworthy compound identified was itraconazole (ITZ), an orally available, safe, and cheap antifungal, that showed high selectivity indexes of >312 and >294 in both replicon-based and viral infection assays, respectively. The antiviral activity of this molecule has been described against positive-sense single stranded RNA viruses (+ssRNA) and was related to cholesterol metabolism that could affect the formation of the replication organelles. Although its precise mechanism of action against CHIKV still needs to be elucidated, our results demonstrate that ITZ is a potent inhibitor of the viral replication that could be repurposed as a broad-spectrum antiviral.
Collapse
|
13
|
Abdulaziz L, Elhadi E, Abdallah EA, Alnoor FA, Yousef BA. Antiviral Activity of Approved Antibacterial, Antifungal, Antiprotozoal and Anthelmintic Drugs: Chances for Drug Repurposing for Antiviral Drug Discovery. J Exp Pharmacol 2022; 14:97-115. [PMID: 35299994 PMCID: PMC8922315 DOI: 10.2147/jep.s346006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 02/28/2022] [Indexed: 11/29/2022] Open
Abstract
Drug repurposing process aims to identify new uses for the existing drugs to overcome traditional de novo drug discovery and development challenges. At the same time, as viral infections became a serious threat to humans and the viral organism itself has a high ability to mutate genetically, and due to serious adverse effects that result from antiviral drugs, there are crucial needs for the discovery of new antiviral drugs, and to identify new antiviral effects for the exciting approved drugs towards different types of viral infections depending on the observed antiviral activity in preclinical studies or clinical findings is one of the approaches to counter the viral infections problems. This narrative review article summarized mainly the published preclinical studies that evaluated the antiviral activity of drugs that are approved and used mainly as antibacterial, antifungal, antiprotozoal, and anthelmintic drugs, and the preclinical studies included the in silico, in vitro, and in vivo findings, additionally some clinical observations were also included while trying to relate them to the preclinical findings. Finally, the structure used for writing about the antiviral activity of the drugs was according to the families of the viruses used in the studies to form a better image for the target of antiviral activity of different drugs in the different kinds of viruses and to relate between the antiviral activity of the drugs against different strains of viruses within the same viral family.
Collapse
Affiliation(s)
- Leena Abdulaziz
- Department of Pharmacology, Faculty of Pharmacy, Omdurman Islamic University, Khartoum, 14415, Sudan
| | - Esraa Elhadi
- Department of Pharmacology, Faculty of Pharmacy, Omdurman Islamic University, Khartoum, 14415, Sudan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, 210009, People’s Republic of China
| | - Ejlal A Abdallah
- Department of Pharmacology and Pharmacy Practice, Faculty of Pharmacy, Sudan University of Science and Technology, Khartoum, 11111, Sudan
| | - Fadlalbaseer A Alnoor
- Department of Pharmacology, Faculty of Pharmacy, National University, Khartoum, 11111, Sudan
| | - Bashir A Yousef
- Department of Pharmacology, Faculty of Pharmacy, University of Khartoum, Khartoum, 11111, Sudan
- Correspondence: Bashir A Yousef, Department of Pharmacology, Faculty of Pharmacy, University of Khartoum, Al-Qasr Ave, Khartoum, 11111, Sudan, Tel +249 912932418, Fax +249 183780696, Email
| |
Collapse
|
14
|
Bezemer B, van Cleef KW, Overheul GJ, Miesen P, van Rij RP. The calcium channel inhibitor lacidipine inhibits Zika virus replication in neural progenitor cells. Antiviral Res 2022; 202:105313. [DOI: 10.1016/j.antiviral.2022.105313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/21/2022] [Accepted: 03/27/2022] [Indexed: 01/04/2023]
|
15
|
Repurposing Antifungals for Host-Directed Antiviral Therapy? Pharmaceuticals (Basel) 2022; 15:ph15020212. [PMID: 35215323 PMCID: PMC8878022 DOI: 10.3390/ph15020212] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
Because of their epidemic and pandemic potential, emerging viruses are a major threat to global healthcare systems. While vaccination is in general a straightforward approach to prevent viral infections, immunization can also cause escape mutants that hide from immune cell and antibody detection. Thus, other approaches than immunization are critical for the management and control of viral infections. Viruses are prone to mutations leading to the rapid emergence of resistant strains upon treatment with direct antivirals. In contrast to the direct interference with pathogen components, host-directed therapies aim to target host factors that are essential for the pathogenic replication cycle or to improve the host defense mechanisms, thus circumventing resistance. These relatively new approaches are often based on the repurposing of drugs which are already licensed for the treatment of other unrelated diseases. Here, we summarize what is known about the mechanisms and modes of action for a potential use of antifungals as repurposed host-directed anti-infectives for the therapeutic intervention to control viral infections.
Collapse
|
16
|
Arora A, Taskinen JH, Olkkonen VM. Coordination of inter-organelle communication and lipid fluxes by OSBP-related proteins. Prog Lipid Res 2022; 86:101146. [PMID: 34999137 DOI: 10.1016/j.plipres.2022.101146] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/10/2021] [Accepted: 01/03/2022] [Indexed: 12/31/2022]
Abstract
Oxysterol-binding protein (OSBP) and OSBP-related proteins (ORPs) constitute one of the largest families of lipid-binding/transfer proteins (LTPs) in eukaryotes. The current view is that many of them mediate inter-organelle lipid transfer over membrane contact sites (MCS). The transfer occurs in several cases in a 'counter-current' fashion: A lipid such as cholesterol or phosphatidylserine (PS) is transferred against its concentration gradient driven by transport of a phosphoinositide in the opposite direction. In this way ORPs are envisioned to maintain the distinct organelle lipid compositions, with impacts on multiple organelle functions. However, the functions of ORPs extend beyond lipid homeostasis to regulation of processes such as cell survival, proliferation and migration. Important expanding areas of mammalian ORP research include their roles in viral and bacterial infections, cancers, and neuronal function. The yeast OSBP homologue (Osh) proteins execute multifaceted functions in sterol and glycerophospholipid homeostasis, post-Golgi vesicle transport, phosphatidylinositol-4-phosphate, sphingolipid and target of rapamycin (TOR) signalling, and cell cycle control. These observations identify ORPs as lipid transporters and coordinators of signals with an unforeseen variety of cellular processes. Understanding their activities not only enlightens the biology of the living cell but also allows their employment as targets of new therapeutic approaches for disease.
Collapse
Affiliation(s)
- Amita Arora
- Minerva Foundation Institute for Medical Research, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland
| | - Juuso H Taskinen
- Minerva Foundation Institute for Medical Research, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, and Department of Anatomy, Faculty of Medicine, University of Helsinki, Finland.
| |
Collapse
|
17
|
Varghese FS, Meutiawati F, Teppor M, Jacobs S, de Keyzer C, Taşköprü E, van Woudenbergh E, Overheul GJ, Bouma E, Smit JM, Delang L, Merits A, van Rij RP. Posaconazole inhibits multiple steps of the alphavirus replication cycle. Antiviral Res 2021; 197:105223. [PMID: 34856248 DOI: 10.1016/j.antiviral.2021.105223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/22/2021] [Accepted: 11/27/2021] [Indexed: 11/28/2022]
Abstract
Repurposing drugs is a promising strategy to identify therapeutic interventions against novel and re-emerging viruses. Posaconazole is an antifungal drug used to treat invasive aspergillosis and candidiasis. Recently, posaconazole and its structural analog, itraconazole were shown to inhibit replication of multiple viruses by modifying intracellular cholesterol homeostasis. Here, we show that posaconazole inhibits replication of the alphaviruses Semliki Forest virus (SFV), Sindbis virus and chikungunya virus with EC50 values ranging from 1.4 μM to 9.5 μM. Posaconazole treatment led to a significant reduction of virus entry in an assay using a temperature-sensitive SFV mutant, but time-of-addition and RNA transfection assays indicated that posaconazole also inhibits post-entry stages of the viral replication cycle. Virus replication in the presence of posaconazole was partially rescued by the addition of exogenous cholesterol. A transferrin uptake assay revealed that posaconazole considerably slowed down cellular endocytosis. A single point mutation in the SFV E2 glycoprotein, H255R, provided partial resistance to posaconazole as well as to methyl-β-cyclodextrin, corroborating the effect of posaconazole on cholesterol and viral entry. Our results indicate that posaconazole inhibits multiple steps of the alphavirus replication cycle and broaden the spectrum of viruses that can be targeted in vitro by posaconazole, which could be further explored as a therapeutic agent against emerging viruses.
Collapse
Affiliation(s)
- Finny S Varghese
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Febrina Meutiawati
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mona Teppor
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Sofie Jacobs
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Carolien de Keyzer
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Ezgi Taşköprü
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Esther van Woudenbergh
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands; Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Gijs J Overheul
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ellen Bouma
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jolanda M Smit
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Leen Delang
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
18
|
Nagaraj S, Manivannan S, Narayan S. Potent antifungal agents and use of nanocarriers to improve delivery to the infected site: A systematic review. J Basic Microbiol 2021; 61:849-873. [PMID: 34351655 DOI: 10.1002/jobm.202100204] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/29/2021] [Accepted: 08/01/2021] [Indexed: 01/30/2023]
Abstract
There are four major classes of antifungals with the predominant mechanism of action being targeting of cell wall or cell membrane. As in other drugs, low solubility of these compounds has led to low bioavailability in target tissues. Enhanced drug dosages have effects such as toxicity, drug-drug interactions, and increased drug resistance by fungi. This article reviews the current state-of-the-art of antifungals, structure, mechanism of action, other usages, and toxic side effects. The emergence of nanoformulations to transport and uniformly release cargo at the target site is a boon in antifungal treatment. The article details research that lead to the development of nanoformulations of antifungals and potential advantages and avoidance of the lacunae characterizing conventional drugs. A range of nanoformulations based on liposomes, polymers are in various stages of research and their potential advantages have been brought out. It could be observed that under similar dosages, test models, and duration, nanoformulations provided enhanced activity, reduced toxicity, higher uptake and higher immunostimulatory effects. In most instances, the mechanism of antifungal activity of nanoformulations was similar to that of regular antifungal. There are possibilities of coupling multiple antifungals on the same nano-platform. Increased activity coupled with multiple mechanisms of action presents for nanoformulations a tremendous opportunity to overcome antifungal resistance. In the years to come, robust methods for the preparation of nanoformulations taking into account the repeatability and reproducibility in action, furthering the studies on nanoformulation toxicity and studies of human models are required before extensive use of nanoformulations as a prescribed drug.
Collapse
Affiliation(s)
- Saraswathi Nagaraj
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamilnadu, India
| | - Sivakami Manivannan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamilnadu, India
| | - Shoba Narayan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamilnadu, India
| |
Collapse
|
19
|
Avula K, Singh B, Kumar PV, Syed GH. Role of Lipid Transfer Proteins (LTPs) in the Viral Life Cycle. Front Microbiol 2021; 12:673509. [PMID: 34248884 PMCID: PMC8260984 DOI: 10.3389/fmicb.2021.673509] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/17/2021] [Indexed: 12/14/2022] Open
Abstract
Viruses are obligate parasites that depend on the host cell machinery for their replication and dissemination. Cellular lipids play a central role in multiple stages of the viral life cycle such as entry, replication, morphogenesis, and egress. Most viruses reorganize the host cell membranes for the establishment of viral replication complex. These specialized structures allow the segregation of replicating viral RNA from ribosomes and protect it from host nucleases. They also facilitate localized enrichment of cellular components required for viral replication and assembly. The specific composition of the lipid membrane governs its ability to form negative or positive curvature and possess a rigid or flexible form, which is crucial for membrane rearrangement and establishment of viral replication complexes. In this review, we highlight how different viruses manipulate host lipid transfer proteins and harness their functions to enrich different membrane compartments with specific lipids in order to facilitate multiple aspects of the viral life cycle.
Collapse
Affiliation(s)
- Kiran Avula
- Virus-Host Interaction Lab, Institute of Life Sciences, Bhubaneshwar, India.,Regional Centre for Biotechnology, Faridabad, India
| | - Bharati Singh
- Virus-Host Interaction Lab, Institute of Life Sciences, Bhubaneshwar, India.,School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneshwar, India
| | - Preethy V Kumar
- Virus-Host Interaction Lab, Institute of Life Sciences, Bhubaneshwar, India.,School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneshwar, India
| | - Gulam H Syed
- Virus-Host Interaction Lab, Institute of Life Sciences, Bhubaneshwar, India
| |
Collapse
|
20
|
Dos Santos Nascimento IJ, de Aquino TM, da Silva-Júnior EF. Drug Repurposing: A Strategy for Discovering Inhibitors against Emerging Viral Infections. Curr Med Chem 2021; 28:2887-2942. [PMID: 32787752 DOI: 10.2174/0929867327666200812215852] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Viral diseases are responsible for several deaths around the world. Over the past few years, the world has seen several outbreaks caused by viral diseases that, for a long time, seemed to possess no risk. These are diseases that have been forgotten for a long time and, until nowadays, there are no approved drugs or vaccines, leading the pharmaceutical industry and several research groups to run out of time in the search for new pharmacological treatments or prevention methods. In this context, drug repurposing proves to be a fast and economically viable technique, considering the fact that it uses drugs that have a well-established safety profile. Thus, in this review, we present the main advances in drug repurposing and their benefit for searching new treatments against emerging viral diseases. METHODS We conducted a search in the bibliographic databases (Science Direct, Bentham Science, PubMed, Springer, ACS Publisher, Wiley, and NIH's COVID-19 Portfolio) using the keywords "drug repurposing", "emerging viral infections" and each of the diseases reported here (CoV; ZIKV; DENV; CHIKV; EBOV and MARV) as an inclusion/exclusion criterion. A subjective analysis was performed regarding the quality of the works for inclusion in this manuscript. Thus, the selected works were those that presented drugs repositioned against the emerging viral diseases presented here by means of computational, high-throughput screening or phenotype-based strategies, with no time limit and of relevant scientific value. RESULTS 291 papers were selected, 24 of which were CHIKV; 52 for ZIKV; 43 for DENV; 35 for EBOV; 10 for MARV; and 56 for CoV and the rest (72 papers) related to the drugs repurposing and emerging viral diseases. Among CoV-related articles, most were published in 2020 (31 papers), updating the current topic. Besides, between the years 2003 - 2005, 10 articles were created, and from 2011 - 2015, there were 7 articles, portraying the outbreaks that occurred at that time. For ZIKV, similar to CoV, most publications were during the period of outbreaks between the years 2016 - 2017 (23 articles). Similarly, most CHIKV (13 papers) and DENV (14 papers) publications occur at the same time interval. For EBOV (13 papers) and MARV (4 papers), they were between the years 2015 - 2016. Through this review, several drugs were highlighted that can be evolved in vivo and clinical trials as possible used against these pathogens showed that remdesivir represent potential treatments against CoV. Furthermore, ribavirin may also be a potential treatment against CHIKV; sofosbuvir against ZIKV; celgosivir against DENV, and favipiravir against EBOV and MARV, representing new hopes against these pathogens. CONCLUSION The conclusions of this review manuscript show the potential of the drug repurposing strategy in the discovery of new pharmaceutical products, as from this approach, drugs could be used against emerging viral diseases. Thus, this strategy deserves more attention among research groups and is a promising approach to the discovery of new drugs against emerging viral diseases and also other diseases.
Collapse
|
21
|
Kumar S, Singh B, Kumari P, Kumar PV, Agnihotri G, Khan S, Kant Beuria T, Syed GH, Dixit A. Identification of multipotent drugs for COVID-19 therapeutics with the evaluation of their SARS-CoV2 inhibitory activity. Comput Struct Biotechnol J 2021; 19:1998-2017. [PMID: 33841751 PMCID: PMC8025584 DOI: 10.1016/j.csbj.2021.04.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/04/2021] [Accepted: 04/04/2021] [Indexed: 12/12/2022] Open
Abstract
The SARS-CoV2 is a highly contagious pathogen that causes COVID-19 disease. It has affected millions of people globally with an average lethality of ~3%. There is an urgent need of drugs for the treatment of COVID-19. In the current studies, we have used bioinformatics techniques to screen the FDA approved drugs against nine SARS-CoV2 proteins to identify drugs for repurposing. Additionally, we analyzed if the identified molecules can also affect the human proteins whose expression in lung changed during SARS-CoV2 infection. Targeting such genes may also be a beneficial strategy to curb disease manifestation. We have identified 74 molecules that can bind to various SARS-CoV2 and human host proteins. We experimentally validated our in-silico predictions using vero E6 cells infected with SARS-CoV2 virus. Interestingly, many of our predicted molecules viz. capreomycin, celecoxib, mefloquine, montelukast, and nebivolol showed good activity (IC50) against SARS-CoV2. We hope that these studies may help in the development of new therapeutic options for the treatment of COVID-19.
Collapse
Affiliation(s)
- Sugandh Kumar
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha 751023, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) University, Bhubaneswar, Odisha 751024, India
| | - Bharati Singh
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha 751023, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) University, Bhubaneswar, Odisha 751024, India
| | - Pratima Kumari
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha 751023, India
- Regional Centre for Biotechnology (RCB), 3rd Milestone, Faridabad-Gurgaon, Haryana 121001, India
| | - Preethy V. Kumar
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha 751023, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) University, Bhubaneswar, Odisha 751024, India
| | - Geetanjali Agnihotri
- School of Chemical Technology, Kalinga Institute of Industrial Technology (KIIT) University, Bhubaneswar, Odisha 751024, India
| | - Shaheerah Khan
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha 751023, India
- Regional Centre for Biotechnology (RCB), 3rd Milestone, Faridabad-Gurgaon, Haryana 121001, India
| | - Tushar Kant Beuria
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha 751023, India
| | - Gulam Hussain Syed
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha 751023, India
| | - Anshuman Dixit
- Institute of Life Science, Nalco Square, Bhubaneswar, Odisha 751023, India
| |
Collapse
|
22
|
Mercorelli B, Celegato M, Luganini A, Gribaudo G, Lepesheva GI, Loregian A. The antifungal drug isavuconazole inhibits the replication of human cytomegalovirus (HCMV) and acts synergistically with anti-HCMV drugs. Antiviral Res 2021; 189:105062. [PMID: 33722615 DOI: 10.1016/j.antiviral.2021.105062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/29/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
We recently reported that some clinically approved antifungal drugs are potent inhibitors of human cytomegalovirus (HCMV). Here, we report the broad-spectrum activity against HCMV of isavuconazole (ICZ), a new extended-spectrum triazolic antifungal drug. ICZ inhibited the replication of clinical isolates of HCMV as well as strains resistant to the currently available DNA polymerase inhibitors. The antiviral activity of ICZ against HCMV could be linked to the inhibition of human cytochrome P450 51 (hCYP51), an enzyme whose activity we previously demonstrated to be required for productive HCMV infection. Moreover, time-of-addition studies indicated that ICZ might have additional inhibitory effects during the first phase of HCMV replication. Importantly, ICZ showed synergistic antiviral activity in vitro when administered in combination with different approved anti-HCMV drugs at clinically relevant doses. Together, these results pave the way to possible future clinical studies aimed at evaluating the repurposing potential of ICZ in the treatment of HCMV-associated diseases.
Collapse
Affiliation(s)
| | - Marta Celegato
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Anna Luganini
- Department of Life Sciences and Systems Biology, University of Turin, 10123, Turin, Italy
| | - Giorgio Gribaudo
- Department of Life Sciences and Systems Biology, University of Turin, 10123, Turin, Italy
| | - Galina I Lepesheva
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
| |
Collapse
|
23
|
Dong S, Dimopoulos G. Antiviral Compounds for Blocking Arboviral Transmission in Mosquitoes. Viruses 2021; 13:v13010108. [PMID: 33466915 PMCID: PMC7830659 DOI: 10.3390/v13010108] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/06/2021] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Mosquito-borne arthropod-borne viruses (arboviruses) such as the dengue virus (DENV), Zika virus (ZIKV), and chikungunya virus (CHIKV) are important human pathogens that are responsible for significant global morbidity and mortality. The recent emergence and re-emergence of mosquito-borne viral diseases (MBVDs) highlight the urgent need for safe and effective vaccines, therapeutics, and vector-control approaches to prevent MBVD outbreaks. In nature, arboviruses circulate between vertebrate hosts and arthropod vectors; therefore, disrupting the virus lifecycle in mosquitoes is a major approach for combating MBVDs. Several strategies were proposed to render mosquitoes that are refractory to arboviral infection, for example, those involving the generation of genetically modified mosquitoes or infection with the symbiotic bacterium Wolbachia. Due to the recent development of high-throughput screening methods, an increasing number of drugs with inhibitory effects on mosquito-borne arboviruses in mammalian cells were identified. These antivirals are useful resources that can impede the circulation of arboviruses between arthropods and humans by either rendering viruses more vulnerable in humans or suppressing viral infection by reducing the expression of host factors in mosquitoes. In this review, we summarize recent advances in small-molecule antiarboviral drugs in mammalian and mosquito cells, and discuss how to use these antivirals to block the transmission of MBVDs.
Collapse
|
24
|
Obayes Al-Khikani F. Evaluation of the role of itraconazole and posaconazole in viral infection as immunomodulatory drugs. MATRIX SCIENCE MEDICA 2021. [DOI: 10.4103/mtsm.mtsm_34_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
25
|
AL-Khikani FO. Evaluation of the role of itraconazole and posaconazole in viral infection as immunomodulatory drugs. MGM JOURNAL OF MEDICAL SCIENCES 2021. [DOI: 10.4103/mgmj.mgmj_59_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
26
|
Luquain-Costaz C, Rabia M, Hullin-Matsuda F, Delton I. Bis(monoacylglycero)phosphate, an important actor in the host endocytic machinery hijacked by SARS-CoV-2 and related viruses. Biochimie 2020; 179:247-256. [PMID: 33159981 PMCID: PMC7642752 DOI: 10.1016/j.biochi.2020.10.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022]
Abstract
Viruses, including the novel coronavirus SARS-CoV-2, redirect infected cell metabolism to their own purposes. After binding to its receptor angiotensin-converting enzyme 2 (ACE2) on the cell surface, the SARS-CoV-2 is taken up by receptor-mediated endocytosis ending in the acidic endolysosomal compartment. The virus hijacks the endosomal machinery leading to fusion of viral and endosomal membranes and release of the viral RNA into the cytosol. This mini-review specifically highlights the membrane lipid organization of the endosomal system focusing on the unconventional and late endosome/lysosome-specific phospholipid, bis(monoacylglycero)phosphate (BMP). BMP is enriched in alveolar macrophages of lung, one of the target tissue of SARS-CoV-2. This review details the BMP structure, its unsaturated fatty acid composition and fusogenic properties that are essential for the highly dynamic formation of the intraluminal vesicles inside the endosomes. Interestingly, BMP is necessary for infection and replication of enveloped RNA virus such as SARS-CoV-1 and Dengue virus. We also emphasize the role of BMP in lipid sorting and degradation, especially cholesterol transport in cooperation with Niemann Pick type C proteins (NPC 1 and 2) and with some oxysterol-binding protein (OSBP)-related proteins (ORPs) as well as in sphingolipid degradation. Interestingly, numerous virus infection required NPC1 as well as ORPs along the endocytic pathway. Furthermore, BMP content is increased during pathological endosomal lipid accumulation in various lysosomal storage disorders. This is particularly important knowing the high percentage of patients with metabolic disorders among the SARS-CoV-2 infected patients presenting severe forms of COVID-19.
Collapse
Affiliation(s)
- Céline Luquain-Costaz
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | - Maxence Rabia
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France
| | | | - Isabelle Delton
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAe U1397, INSA Lyon, Villeurbanne, France.
| |
Collapse
|
27
|
Khan N, Chen X, Geiger JD. Role of Endolysosomes in Severe Acute Respiratory Syndrome Coronavirus-2 Infection and Coronavirus Disease 2019 Pathogenesis: Implications for Potential Treatments. Front Pharmacol 2020; 11:595888. [PMID: 33324224 PMCID: PMC7723437 DOI: 10.3389/fphar.2020.595888] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is an enveloped, single-stranded RNA virus. Humans infected with SARS-CoV-2 develop a disease known as coronavirus disease 2019 (COVID-19) with symptoms and consequences including acute respiratory distress syndrome (ARDS), cardiovascular disorders, and death. SARS-CoV-2 appears to infect cells by first binding viral spike proteins with host protein angiotensin-converting enzyme 2 (ACE2) receptors; the virus is endocytosed following priming by transmembrane protease serine 2 (TMPRSS2). The process of virus entry into endosomes and its release from endolysosomes are key features of enveloped viruses. Thus, it is important to focus attention on the role of endolysosomes in SARS-CoV-2 infection. Indeed, coronaviruses are now known to hijack endocytic machinery to enter cells such that they can deliver their genome at replication sites without initiating host detection and immunological responses. Hence, endolysosomes might be good targets for developing therapeutic strategies against coronaviruses. Here, we focus attention on the involvement of endolysosomes in SARS-CoV-2 infection and COVID-19 pathogenesis. Further, we explore endolysosome-based therapeutic strategies to restrict SARS-CoV-2 infection and COVID-19 pathogenesis.
Collapse
Affiliation(s)
| | | | - Jonathan D. Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| |
Collapse
|
28
|
The Clinically Approved Antifungal Drug Posaconazole Inhibits Human Cytomegalovirus Replication. Antimicrob Agents Chemother 2020; 64:AAC.00056-20. [PMID: 32690644 DOI: 10.1128/aac.00056-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/11/2020] [Indexed: 12/14/2022] Open
Abstract
Posaconazole (PCZ) is a clinically approved drug used predominantly for prophylaxis and salvage therapy of fungal infections. Here, we report its previously undescribed anti-human cytomegalovirus (HCMV) activity. By using antiviral assays, we demonstrated that PCZ, along with other azolic antifungals, has a broad anti-HCMV activity, being active against different strains, including low-passage-number clinical isolates and strains resistant to viral DNA polymerase inhibitors. Using a pharmacological approach, we identified the inhibition of human cytochrome P450 51 (hCYP51), or lanosterol 14α demethylase, a cellular target of posaconazole in infected cells, as a mechanism of anti-HCMV activity of the drug. Indeed, hCYP51 expression was stimulated upon HCMV infection, and the inhibition of its enzymatic activity by either the lanosterol analog VFV {(R)-N-(1-(3,4'-difluoro-[1,1'-biphenyl]-4-yl)-2-(1H-imidazol-1-yl)ethyl)-4-(5-phenyl-1,3,4-oxadiazol-2-yl)benzamide} or PCZ decreased HCMV yield and infectivity of released virus particles. Importantly, we observed that the activity of the first-line anti-HCMV drug ganciclovir was boosted tenfold by PCZ and that ganciclovir (GCV) and PCZ act synergistically in inhibiting HCMV replication. Taken together, these findings suggest that this clinically approved drug deserves further investigation in the development of host-directed antiviral strategies as a candidate anti-HCMV drug with a dual antimicrobial effect.
Collapse
|
29
|
Sturley SL, Rajakumar T, Hammond N, Higaki K, Márka Z, Márka S, Munkacsi AB. Potential COVID-19 therapeutics from a rare disease: weaponizing lipid dysregulation to combat viral infectivity. J Lipid Res 2020; 61:972-982. [PMID: 32457038 PMCID: PMC7328045 DOI: 10.1194/jlr.r120000851] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/21/2020] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus (SARS-CoV)-2 has resulted in the death of more than 328,000 persons worldwide in the first 5 months of 2020. Herculean efforts to rapidly design and produce vaccines and other antiviral interventions are ongoing. However, newly evolving viral mutations, the prospect of only temporary immunity, and a long path to regulatory approval pose significant challenges and call for a common, readily available, and inexpensive treatment. Strategic drug repurposing combined with rapid testing of established molecular targets could provide a pause in disease progression. SARS-CoV-2 shares extensive structural and functional conservation with SARS-CoV-1, including engagement of the same host cell receptor (angiotensin-converting enzyme 2) localized in cholesterol-rich microdomains. These lipid-enveloped viruses encounter the endosomal/lysosomal host compartment in a critical step of infection and maturation. Niemann-Pick type C (NP-C) disease is a rare monogenic neurodegenerative disease caused by deficient efflux of lipids from the late endosome/lysosome (LE/L). The NP-C disease-causing gene (NPC1) has been strongly associated with viral infection, both as a filovirus receptor (e.g., Ebola) and through LE/L lipid trafficking. This suggests that NPC1 inhibitors or NP-C disease mimetics could serve as anti-SARS-CoV-2 agents. Fortunately, there are such clinically approved molecules that elicit antiviral activity in preclinical studies, without causing NP-C disease. Inhibition of NPC1 may impair viral SARS-CoV-2 infectivity via several lipid-dependent mechanisms, which disturb the microenvironment optimum for viral infectivity. We suggest that known mechanistic information on NPC1 could be utilized to identify existing and future drugs to treat COVID-19.
Collapse
MESH Headings
- Androstenes/therapeutic use
- Angiotensin-Converting Enzyme 2
- Anticholesteremic Agents/therapeutic use
- Antiviral Agents/therapeutic use
- Betacoronavirus/drug effects
- Betacoronavirus/metabolism
- Betacoronavirus/pathogenicity
- COVID-19
- Cholesterol/metabolism
- Coronavirus Infections/diagnosis
- Coronavirus Infections/drug therapy
- Coronavirus Infections/epidemiology
- Drug Repositioning/methods
- Humans
- Hydroxychloroquine/therapeutic use
- Intracellular Signaling Peptides and Proteins/antagonists & inhibitors
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Lysosomes/drug effects
- Lysosomes/metabolism
- Lysosomes/virology
- Niemann-Pick C1 Protein
- Niemann-Pick Disease, Type C/drug therapy
- Niemann-Pick Disease, Type C/genetics
- Niemann-Pick Disease, Type C/metabolism
- Niemann-Pick Disease, Type C/pathology
- Pandemics
- Peptidyl-Dipeptidase A/genetics
- Peptidyl-Dipeptidase A/metabolism
- Pneumonia, Viral/diagnosis
- Pneumonia, Viral/drug therapy
- Pneumonia, Viral/epidemiology
- Protein Binding
- Receptors, Virus/antagonists & inhibitors
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- SARS-CoV-2
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/metabolism
Collapse
Affiliation(s)
| | - Tamayanthi Rajakumar
- School of Biological Sciences and Centre for
Biodiscovery, Victoria University of Wellington,
Wellington 6012, New Zealand
| | - Natalie Hammond
- School of Biological Sciences and Centre for
Biodiscovery, Victoria University of Wellington,
Wellington 6012, New Zealand
| | - Katsumi Higaki
- Division of Functional Genomics,
Tottori University, Yonago 683-8503,
Japan
| | - Zsuzsa Márka
- Department of Physics,
Columbia University, New York,
NY 10027
| | - Szabolcs Márka
- Department of Physics,
Columbia University, New York,
NY 10027
| | - Andrew B. Munkacsi
- School of Biological Sciences and Centre for
Biodiscovery, Victoria University of Wellington,
Wellington 6012, New Zealand
| |
Collapse
|
30
|
Antifungal Triazole Posaconazole Targets an Early Stage of the Parechovirus A3 Life Cycle. Antimicrob Agents Chemother 2020; 64:AAC.02372-19. [PMID: 31818821 DOI: 10.1128/aac.02372-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022] Open
Abstract
Viruses in species Parechovirus A (Picornaviridae) are associated with a wide variety of clinical manifestations. Parechovirus A3 (PeV-A3) is known to cause sepsis-like illness, meningitis, and encephalitis in infants and young children. To date, no specific therapies are available to treat PeV-A3-infected children. We had previously identified two FDA-cleared antifungal drugs, itraconazole (ITC) and posaconazole (POS), with potent and specific antiviral activity against PeV-A3. Time-of-addition and synchronized infection assays revealed that POS targets an early stage of the PeV-A3 life cycle. POS exerts an antiviral effect, evidenced by a reduction in viral titer following the addition of POS to Vero-P cells before infection, coaddition of POS and PeV-A3 to Vero-P cells, incubation of POS and PeV-A3 prior to Vero-P infection, and at attachment. POS exerts less of an effect on virus entry. A PeV-A3 enzyme-linked immunosorbent assay inhibition experiment, using an anti-PeV-A3 monoclonal antibody, suggested that POS binds directly to the PeV-A3 capsid. POS-resistant PeV-A3 strains developed by serial passage in the presence of POS acquired substitutions in multiple regions of the genome, including the capsid. Reverse genetics confirmed substitutions in capsid proteins VP0, VP3, and VP1 and nonstructural proteins 2A and 3A. Single mutants VP0_K66R, VP0_A124T, VP3_N88S, VP1_Y224C, 2A_S78L, and 3A_T1I were 4-, 9-, 12-, 34-, 51-, and 119-fold more resistant to POS, respectively, than the susceptible prototype strain. Our studies demonstrate that POS may be a valuable tool in developing an antiviral therapy for PeV-A3.
Collapse
|
31
|
Roberts BL, Severance ZC, Bensen RC, Le-McClain AT, Malinky CA, Mettenbrink EM, Nuñez JI, Reddig WJ, Blewett EL, Burgett AWG. Differing activities of oxysterol-binding protein (OSBP) targeting anti-viral compounds. Antiviral Res 2019; 170:104548. [PMID: 31271764 PMCID: PMC10786240 DOI: 10.1016/j.antiviral.2019.104548] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/25/2019] [Accepted: 06/29/2019] [Indexed: 11/27/2022]
Abstract
Oxysterol-binding Protein (OSBP) is a human lipid-transport protein required for the cellular replication of many types of viruses, including several human pathogens. The structurally-diverse small molecule compounds OSW-1, itraconazole (ITZ), T-00127-HEV2 (THEV) and TTP-8307 (TTP) inhibit viral replication through interaction with the OSBP protein. The OSW-1 compound reduces intracellular OSBP, and the reduction of OSBP protein levels persists multiple days after the OSW-1-compound treatment is stopped. The OSW-1-induced reduction of OSBP levels inhibited Enterovirus replication prophylactically in cells. In this report, the OSBP-interacting compounds ITZ, THEV, and TTP are shown not to reduce OSBP levels in cells, unlike the OSW-1-compound, and the OSW-1 compound is determined to be the only compound capable of providing prophylactic antiviral activity in cells. Furthermore, OSW-1 and THEV inhibit the binding of 25-hydroxycholesterol (25-OHC) to OSBP indicating that these compounds bind at the conserved sterol ligand binding site. The ITZ and TTP compounds do not inhibit 25-hydroxycholesterol binding to OSBP, and therefore ITZ and TTP interact with OSBP through other, unidentified binding sites. Co-administration of the THEV compound partially blocks the cellular activity of OSW-1, including the reduction of cellular OSBP protein levels; co-administration of the ITZ and TTP compounds have minimal effect on OSW-1 cellular activity further supporting different modes of interaction with these compounds to OSBP. OSW-1, ITZ, THEV, and TTP treatment alter OSBP cellular localization and levels, but in four distinct ways. Co-administration of OSW-1 and ITZ induced OSBP cellular localization patterns with features similar to the effects of ITZ and OSW-1 treatment alone. Based on these results, OSBP is capable of interacting with multiple structural classes of antiviral small molecule compounds at different binding sites, and the different compounds have distinct effects on OSBP cellular activity.
Collapse
Affiliation(s)
- Brett L Roberts
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Zachary C Severance
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Ryan C Bensen
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Anh T Le-McClain
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Cori A Malinky
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Evan M Mettenbrink
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - Juan I Nuñez
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States
| | - William J Reddig
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Earl L Blewett
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Anthony W G Burgett
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States.
| |
Collapse
|
32
|
Kovanich D, Saisawang C, Sittipaisankul P, Ramphan S, Kalpongnukul N, Somparn P, Pisitkun T, Smith DR. Analysis of the Zika and Japanese Encephalitis Virus NS5 Interactomes. J Proteome Res 2019; 18:3203-3218. [PMID: 31199156 DOI: 10.1021/acs.jproteome.9b00318] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Mosquito-borne flaviviruses, including dengue virus (DENV), Japanese encephalitis virus (JEV), and Zika virus (ZIKV), are major human pathogens. Among the flaviviral proteins, the nonstructural protein 5 (NS5) is the largest, most conserved, and major enzymatic component of the viral replication complex. Disruption of the common key NS5-host protein-protein interactions critical for viral replication could aid in the development of broad-spectrum antiflaviviral therapeutics. Hundreds of NS5 interactors have been identified, but these are mostly DENV-NS5 interactors. To this end, we sought to investigate the JEV- and ZIKV-NS5 interactomes using EGFP immunoprecipitation with label-free quantitative mass spectrometry analysis. We report here a total of 137 NS5 interactors with a significant enrichment of spliceosomal and spliceosomal-associated proteins. The transcription complex Paf1C and phosphatase 6 were identified as common NS5-associated complexes. PAF1 was shown to play opposite roles in JEV and ZIKV infections. Additionally, we validated several NS5 targets and proposed their possible roles in infection. These include lipid-shuttling proteins OSBPL9 and OSBPL11, component of RNAP3 transcription factor TFIIIC, minichromosome maintenance, and cochaperone PAQosome. Mining this data set, our study expands the current interaction landscape of NS5 and uncovers several NS5 targets that are new to flavivirus biology.
Collapse
Affiliation(s)
- Duangnapa Kovanich
- Institute of Molecular Biosciences, Mahidol University , Nakhon Pathom , Thailand
| | - Chonticha Saisawang
- Institute of Molecular Biosciences, Mahidol University , Nakhon Pathom , Thailand
| | | | - Suwipa Ramphan
- Institute of Molecular Biosciences, Mahidol University , Nakhon Pathom , Thailand
| | - Nuttiya Kalpongnukul
- Center of Excellence in Systems Biology, Research affairs, Faculty of Medicine , Chulalongkorn University , Bangkok , Thailand
| | - Poorichaya Somparn
- Center of Excellence in Systems Biology, Research affairs, Faculty of Medicine , Chulalongkorn University , Bangkok , Thailand
| | - Trairak Pisitkun
- Center of Excellence in Systems Biology, Research affairs, Faculty of Medicine , Chulalongkorn University , Bangkok , Thailand
| | - Duncan R Smith
- Institute of Molecular Biosciences, Mahidol University , Nakhon Pathom , Thailand
| |
Collapse
|
33
|
Dighe SN, Ekwudu O, Dua K, Chellappan DK, Katavic PL, Collet TA. Recent update on anti-dengue drug discovery. Eur J Med Chem 2019; 176:431-455. [PMID: 31128447 DOI: 10.1016/j.ejmech.2019.05.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/12/2019] [Accepted: 05/06/2019] [Indexed: 01/27/2023]
Abstract
Dengue is the most important arthropod-borne viral disease of humans, with more than half of the global population living in at-risk areas. Despite the negative impact on public health, there are no antiviral therapies available, and the only licensed vaccine, Dengvaxia®, has been contraindicated in children below nine years of age. In an effort to combat dengue, several small molecules have entered into human clinical trials. Here, we review anti-DENV molecules and their drug targets that have been published within the past five years (2014-2018). Further, we discuss their probable mechanisms of action and describe a role for classes of clinically approved drugs and also an unclassified class of anti-DENV agents. This review aims to enhance our understanding of novel agents and their cognate targets in furthering innovations in the use of small molecules for dengue drug therapies.
Collapse
Affiliation(s)
- Satish N Dighe
- Innovative Medicines Group, Institute of Health & Biomedical Innovation, School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia.
| | - O'mezie Ekwudu
- Innovative Medicines Group, Institute of Health & Biomedical Innovation, School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Bukit Jalil, Kuala Lumpur, 57000, Malaysia
| | - Peter L Katavic
- Innovative Medicines Group, Institute of Health & Biomedical Innovation, School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Trudi A Collet
- Innovative Medicines Group, Institute of Health & Biomedical Innovation, School of Clinical Sciences, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
34
|
Galindo I, Cuesta-Geijo MÁ, Del Puerto A, Soriano E, Alonso C. Lipid Exchange Factors at Membrane Contact Sites in African Swine Fever Virus Infection. Viruses 2019; 11:v11030199. [PMID: 30813555 PMCID: PMC6466349 DOI: 10.3390/v11030199] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 01/23/2023] Open
Abstract
African swine fever (ASF) is a hemorrhagic fever of wild and domestic pigs with a high rate of mortality. Originally endemic in Africa, this disease is currently disseminating in Europe and China, causing a large socioeconomic impact. ASF is caused by a DNA virus, African swine fever virus (ASFV). There is no vaccine available against ASFV, limiting the options for disease control. ASFV reorganizes intracellular membranes to generate viral factories (VFs) in order to amplify its genome. However, little is known about the process involved in the formation of these viral replication organelles. Membrane contact sites (MCSs) allow nonvesicular lipids and ion exchange between organelles. Lipid exchange to form VFs apparently requires a number of proteins at MCSs, such as the oxysterol-binding protein (OSBP), the acyl-coenzyme A binding domain containing 3 (ACBD3) and the phosphatidylinositol-phosphate-4-kinase III beta (PI4Kβ). Itraconazole (ITZ) is an antifungal agent that targets sterol-transport molecules such as OSBP and OSBP-related protein 4 (ORP4). 25-Hydroxycholesterol (25-HC) inhibits lipid transport by high affinity binding OSBP. In this work, we analyzed the antiviral function of ITZ and 25-HC against ASFV in Vero cell cultures using the cell-adapted Ba71V isolate. ITZ and 25-HC decreased significantly ASFV replication. Our study revealed OSBP distribution in cytoplasmic membranes in uninfected Vero cells and to the periphery of VFs in infected cells. In addition, we showed that OSBP and OSBP-related proteins, PI4Kβ and ACBD3 were recruited to VFs in the context ASFV infection.
Collapse
Affiliation(s)
- Inmaculada Galindo
- Department Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Ctra. de la Coruña km 7.5, 28040 Madrid, Spain.
| | - Miguel Ángel Cuesta-Geijo
- Department Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Ctra. de la Coruña km 7.5, 28040 Madrid, Spain.
| | - Ana Del Puerto
- Department Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Ctra. de la Coruña km 7.5, 28040 Madrid, Spain.
| | - Eva Soriano
- Department Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Ctra. de la Coruña km 7.5, 28040 Madrid, Spain.
| | - Covadonga Alonso
- Department Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Ctra. de la Coruña km 7.5, 28040 Madrid, Spain.
| |
Collapse
|
35
|
Roberts BL, Severance ZC, Bensen RC, Le AT, Kothapalli NR, Nuñez JI, Ma H, Wu S, Standke SJ, Yang Z, Reddig WJ, Blewett EL, Burgett AWG. Transient Compound Treatment Induces a Multigenerational Reduction of Oxysterol-Binding Protein (OSBP) Levels and Prophylactic Antiviral Activity. ACS Chem Biol 2019; 14:276-287. [PMID: 30576108 PMCID: PMC6379863 DOI: 10.1021/acschembio.8b00984] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Oxysterol-binding
protein (OSBP) is a lipid transport and regulatory
protein required for the replication of Enterovirus genus viruses, which includes many significant human pathogens.
Short-term exposure (i.e., 1–6 h) to a low dose (i.e., 1 nM)
of the natural product compound OSW-1 induces a reduction of cellular
OSBP levels by ∼90% in multiple different cell lines with no
measurable cytotoxicity, defect in cellular proliferation, or global
proteome reduction. Interestingly, the reduction of OSBP levels persists
multiple days after the low-dose, transient OSW-1 compound treatment
is ended and the intracellular OSW-1 compound levels drop to undetectable
levels. The reduction in OSBP levels is inherited in multiple generations
of cells that are propagated after the OSW-1 compound treatment is
stopped. The enduring multiday, multigenerational reduction of OSBP
levels triggered by the OSW-1 compound is not due to proteasome degradation
of OSBP or due to a reduction in OSBP mRNA levels. OSW-1 compound
treatment induces transient autophagy in cells, but blocking autophagy
does not rescue OSBP levels. Although the specific cellular mechanism
of long-term OSBP repression is not yet identified, these results
clearly show the existence of an OSBP specific cellular regulation
process that is triggered upon treatment with an OSBP-binding compound.
The stable reduction of OSBP levels upon short-term, transient OSW-1
compound treatment will be a powerful tool to understand OSBP regulation
and cellular function. Additionally, the persistent reduction in OSBP
levels triggered by the transient OSW-1 compound treatment substantially
reduces viral replication in treated cells. Therefore, the long-term,
compound-induced reduction of OSBP in cells presents a new route to
broad spectrum anti-Enterovirus activity, including
as a novel route to antiviral prophylactic treatment through small
molecule targeting a human host protein.
Collapse
Affiliation(s)
- Brett L. Roberts
- Department of Chemistry and Biochemistry, The University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Zachary C. Severance
- Department of Chemistry and Biochemistry, The University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Ryan C. Bensen
- Department of Chemistry and Biochemistry, The University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Anh T. Le
- Department of Chemistry and Biochemistry, The University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Naga Rama Kothapalli
- Department of Chemistry and Biochemistry, The University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Juan I. Nuñez
- Department of Chemistry and Biochemistry, The University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Hongyan Ma
- Department of Chemistry and Biochemistry, The University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Si Wu
- Department of Chemistry and Biochemistry, The University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Shawna J. Standke
- Department of Chemistry and Biochemistry, The University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Zhibo Yang
- Department of Chemistry and Biochemistry, The University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - William J. Reddig
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, 1111 West 17th Street, Tulsa, Oklahoma 74107, United States
| | - Earl L. Blewett
- Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, 1111 West 17th Street, Tulsa, Oklahoma 74107, United States
| | - Anthony W. G. Burgett
- Department of Chemistry and Biochemistry, The University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| |
Collapse
|