1
|
Lockwood C, Vo AS, Bellafard H, Carter AJR. More evidence for widespread antagonistic pleiotropy in polymorphic disease alleles. Front Genet 2024; 15:1404516. [PMID: 38952711 PMCID: PMC11215129 DOI: 10.3389/fgene.2024.1404516] [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: 03/21/2024] [Accepted: 05/29/2024] [Indexed: 07/03/2024] Open
Abstract
Introduction Many loci segregate alleles classified as "genetic diseases" due to their deleterious effects on health. However, some disease alleles have been reported to show beneficial effects under certain conditions or in certain populations. The beneficial effects of these antagonistically pleiotropic alleles may explain their continued prevalence, but the degree to which antagonistic pleiotropy is common or rare is unresolved. We surveyed the medical literature to identify examples of antagonistic pleiotropy to help determine whether antagonistic pleiotropy appears to be rare or common. Results We identified ten examples of loci with polymorphisms for which the presence of antagonistic pleiotropy is well supported by detailed genetic or epidemiological information in humans. One additional locus was identified for which the supporting evidence comes from animal studies. These examples complement over 20 others reported in other reviews. Discussion The existence of more than 30 identified antagonistically pleiotropic human disease alleles suggests that this phenomenon may be widespread. This poses important implications for both our understanding of human evolutionary genetics and our approaches to clinical treatment and disease prevention, especially therapies based on genetic modification.
Collapse
Affiliation(s)
| | | | | | - Ashley J. R. Carter
- California State University Long Beach, Department of Biological Sciences, Long Beach, CA, United States
| |
Collapse
|
2
|
Tolan AJ, Sanchez KL, Shin SD, White JB, Currais A, Soriano-Castell D, Wilson CG, Maher P, Soriano S. Differential Interferon Signaling Regulation and Oxidative Stress Responses in the Cerebral Cortex and Cerebellum Could Account for the Spatiotemporal Pattern of Neurodegeneration in Niemann-Pick Disease Type C. Genes (Basel) 2024; 15:101. [PMID: 38254990 PMCID: PMC10815326 DOI: 10.3390/genes15010101] [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: 12/18/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Niemann-Pick disease type C (NPC) is a fatal neurodegenerative condition caused by genetic mutations of the NPC1 or NPC2 genes that encode the NPC1 and NPC2 proteins, respectively, which are believed to be responsible for cholesterol efflux from late-endosomes/lysosomes. The pathogenic mechanisms that lead to neurodegeneration in NPC are not well understood. There are, however, well-defined spatiotemporal patterns of neurodegeneration that may provide insight into the pathogenic process. For example, the cerebellum is severely affected from early disease stages, compared with cerebral regions, which remain relatively spared until later stages. Using a genome-wide transcriptome analysis, we have recently identified an aberrant pattern of interferon activation in the cerebella of pre-symptomatic Npc1-/- mice. Here, we carried out a comparative transcriptomic analysis of cerebral cortices and cerebella of pre-symptomatic Npc1-/- mice and age-matched controls to identify differences that may help explain the pathological progression within the NPC brain. We report lower cerebral expression of genes within interferon signaling pathways, and significant differences in the regulation of oxidative stress, compared with the cerebellum. Our findings suggest that a delayed onset of interferon signaling, possibly linked to lower oxidative stress, may account for the slower onset of cerebral cortical pathology in the disease.
Collapse
Affiliation(s)
- Andrew J. Tolan
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA (K.L.S.); (S.D.S.); (J.B.W.)
| | - Kayla L. Sanchez
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA (K.L.S.); (S.D.S.); (J.B.W.)
| | - Samuel D. Shin
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA (K.L.S.); (S.D.S.); (J.B.W.)
| | - Jacob B. White
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA (K.L.S.); (S.D.S.); (J.B.W.)
| | - Antonio Currais
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - David Soriano-Castell
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - Christopher G. Wilson
- Lawrence D. Longo Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA;
| | - Pamela Maher
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - Salvador Soriano
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA (K.L.S.); (S.D.S.); (J.B.W.)
| |
Collapse
|
3
|
Almeida-Pinto F, Pinto R, Rocha J. Navigating the Complex Landscape of Ebola Infection Treatment: A Review of Emerging Pharmacological Approaches. Infect Dis Ther 2024; 13:21-55. [PMID: 38240994 PMCID: PMC10828234 DOI: 10.1007/s40121-023-00913-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/20/2023] [Indexed: 01/31/2024] Open
Abstract
In 1976 Ebola revealed itself to the world, marking the beginning of a series of localized outbreaks. However, it was the Ebola outbreak that began in 2013 that incited fear and anxiety around the globe. Since then, our comprehension of the virus has been steadily expanding. Ebola virus (EBOV), belonging to the Orthoebolavirus genus of the Filoviridae family, possesses a non-segmented, negative single-stranded RNA genome comprising seven genes that encode multiple proteins. These proteins collectively orchestrate the intricate process of infecting host cells. It is not possible to view each protein as monofunctional. Instead, they synergistically contribute to the pathogenicity of the virus. Understanding this multifaceted replication cycle is crucial for the development of effective antiviral strategies. Currently, two antibody-based therapeutics have received approval for treating Ebola virus disease (EVD). In 2022, the first evidence-based clinical practice guideline dedicated to specific therapies for EVD was published. Although notable progress has been made in recent years, deaths still occur. Consequently, there is an urgent need to enhance the therapeutic options available to improve the outcomes of the disease. Emerging therapeutics can target viral proteins as direct-acting antivirals or host factors as host-directed antivirals. They both have advantages and disadvantages. One way to bypass some disadvantages is to repurpose already approved drugs for non-EVD indications to treat EVD. This review offers detailed insight into the role of each viral protein in the replication cycle of the virus, as understanding how the virus interacts with host cells is critical to understanding how emerging therapeutics exert their activity. Using this knowledge, this review delves into the intricate mechanisms of action of current and emerging therapeutics.
Collapse
Affiliation(s)
| | - Rui Pinto
- Faculdade de Farmácia, Universidade de Lisboa, 1649-003, Lisbon, Portugal
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines (iMED.ULisboa), 1649-003, Lisbon, Portugal
- Dr. Joaquim Chaves, Medicine Laboratory, Joaquim Chaves Saúde (JCS), Carnaxide, Portugal
| | - João Rocha
- Faculdade de Farmácia, Universidade de Lisboa, 1649-003, Lisbon, Portugal
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines (iMED.ULisboa), 1649-003, Lisbon, Portugal
| |
Collapse
|
4
|
Coler B, Cervantes O, Li M, Coler C, Li A, Shivakumar M, Every E, Schwartz D, Adams Waldorf KM. Common pathways targeted by viral hemorrhagic fever viruses to infect the placenta and increase the risk of stillbirth. Placenta 2023; 141:2-9. [PMID: 36939178 PMCID: PMC10102255 DOI: 10.1016/j.placenta.2022.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/19/2022] [Accepted: 10/02/2022] [Indexed: 01/06/2023]
Abstract
Viral hemorrhagic fevers (VHF) are endemic to Africa, South America and Asia and contribute to significant maternal and fetal morbidity and mortality. Viruses causing VHFs are typically zoonotic, spreading to humans through livestock, wildlife, or mosquito vectors. Some of the most lethal VHF viruses also impart a high-risk of stillbirth including ebolaviruses, Marburg virus (MARV), Lassa virus (LASV), and Rift Valley Fever Virus (RVFV). Large outbreaks and epidemics are common, though the impact on the mother, fetus and placenta is understudied from a public health, clinical and basic science perspective. Notably, these viruses utilize ubiquitous cellular surface entry receptors critical for normal placental function to enable viral invasion into multiple key cell types of the placenta and set the stage for maternal-fetal transmission and stillbirth. We employ insights from molecular virology and viral immunology to discuss how trophoblast expression of viral entry receptors for VHF viruses may increase the risk for viral transmission to the fetus and stillbirth. As the frequency of VHF outbreaks is expected to increase with worsening climate change, understanding the pathogenesis of VHF-related diseases in the placenta is paramount to predicting the impact of emerging viruses on the placenta and perinatal outcomes.
Collapse
Affiliation(s)
- Brahm Coler
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Orlando Cervantes
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Department of Biological Sciences, Columbia University, New York City, NY, USA
| | | | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Case Western Reserve, Cleveland, OH, USA
| | - Megana Shivakumar
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Emma Every
- School of Medicine, University of Washington, Seattle, WA, USA
| | | | - Kristina M Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
5
|
Osuna-Ramos JF, Farfan-Morales CN, Cordero-Rivera CD, De Jesús-González LA, Reyes-Ruiz JM, Hurtado-Monzón AM, Palacios-Rápalo SN, Jiménez-Camacho R, Meraz-Ríos MA, Del Ángel RM. Cholesterol-Lowering Drugs as Potential Antivirals: A Repurposing Approach against Flavivirus Infections. Viruses 2023; 15:1465. [PMID: 37515153 PMCID: PMC10383882 DOI: 10.3390/v15071465] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Flaviviruses, including Dengue (DENV), Zika (ZIKV), and Yellow Fever (YFV) viruses, represent a significant global health burden. The development of effective antiviral therapies against these viruses is crucial to mitigate their impact. This study investigated the antiviral potential of the cholesterol-lowering drugs atorvastatin and ezetimibe in monotherapy and combination against DENV, ZIKV, and YFV. In vitro results demonstrated a dose-dependent reduction in the percentage of infected cells for both drugs. The combination of atorvastatin and ezetimibe showed a synergistic effect against DENV 2, an additive effect against DENV 4 and ZIKV, and an antagonistic effect against YFV. In AG129 mice infected with DENV 2, monotherapy with atorvastatin or ezetimibe significantly reduced clinical signs and increased survival. However, the combination of both drugs did not significantly affect survival. This study provides valuable insights into the potential of atorvastatin and ezetimibe as antiviral agents against flaviviruses and highlights the need for further investigations into their combined therapeutic effects.
Collapse
Affiliation(s)
- Juan Fidel Osuna-Ramos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
- Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán 80019, Mexico
| | - Carlos Noe Farfan-Morales
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana (UAM), Unidad Cuajimalpa, Mexico City 05348, Mexico
| | - Carlos Daniel Cordero-Rivera
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Luis Adrián De Jesús-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social, Zacatecas 98000, Mexico
| | - José Manuel Reyes-Ruiz
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional "Adolfo Ruiz Cortines", Instituto Mexicano del Seguro Social (IMSS), Veracruz Norte, Veracruz 91810, Mexico
- Facultad de Medicina, Región Veracruz, Universidad Veracruzana (UV), Veracruz 91090, Mexico
| | - Arianna M Hurtado-Monzón
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Selvin Noé Palacios-Rápalo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Ricardo Jiménez-Camacho
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Marco Antonio Meraz-Ríos
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Mexico City 07360, Mexico
| | - Rosa María Del Ángel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City 07360, Mexico
| |
Collapse
|
6
|
Gruber MF, Rubin S, Krause PR. Approaches to demonstrating the effectiveness of filovirus vaccines: Lessons from Ebola and COVID-19. Front Immunol 2023; 14:1109486. [PMID: 36817425 PMCID: PMC9932497 DOI: 10.3389/fimmu.2023.1109486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Zaire ebolavirus (EBOV), Sudan ebolavirus (SUDV) and Marburg virus (MARV), are members of the Filoviridae family that can cause severe disease and death in humans and animals. The reemergence of Ebola, Sudan and Marburg virus disease highlight the need for continued availability of safe and effectives vaccines as well as development of new vaccines. While randomized controlled trials using disease endpoints provide the most robust assessment of vaccine effectiveness, challenges to this approach include the unpredictable size, location, occurrence and duration of filovirus disease outbreaks. Thus, other approaches to demonstrating vaccine effectiveness have been considered. These approaches are discussed using examples of preventive vaccines against other infectious diseases. In addition, this article proposes a clinical immunobridging strategy using licensed EBOV vaccines as comparators for demonstrating the effectiveness of filovirus vaccine candidates that are based on the same licensed vaccine platform technology.
Collapse
Affiliation(s)
- Marion F. Gruber
- International AIDS Vaccine Initiative, New York, NY, United States,*Correspondence: Marion F. Gruber,
| | - Steven Rubin
- GlaxoSmithKline (GSk), Rockville, MD, United States
| | | |
Collapse
|
7
|
Wanninger TG, Millian DE, Saldarriaga OA, Maruyama J, Saito T, Reyna RA, Taniguchi S, Arroyave E, Connolly ME, Stevenson HL, Paessler S. Macrophage infection, activation, and histopathological findings in ebolavirus infection. Front Cell Infect Microbiol 2022; 12:1023557. [PMID: 36310868 PMCID: PMC9597316 DOI: 10.3389/fcimb.2022.1023557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/15/2022] [Indexed: 12/05/2022] Open
Abstract
Macrophages contribute to Ebola virus disease through their susceptibility to direct infection, their multi-faceted response to ebolaviruses, and their association with pathological findings in tissues throughout the body. Viral attachment and entry factors, as well as the more recently described influence of cell polarization, shape macrophage susceptibility to direct infection. Moreover, the study of Toll-like receptor 4 and the RIG-I-like receptor pathway in the macrophage response to ebolaviruses highlight important immune signaling pathways contributing to the breadth of macrophage responses. Lastly, the deep histopathological catalogue of macrophage involvement across numerous tissues during infection has been enriched by descriptions of tissues involved in sequelae following acute infection, including: the eye, joints, and the nervous system. Building upon this knowledge base, future opportunities include characterization of macrophage phenotypes beneficial or deleterious to survival, delineation of the specific roles macrophages play in pathological lesion development in affected tissues, and the creation of macrophage-specific therapeutics enhancing the beneficial activities and reducing the deleterious contributions of macrophages to the outcome of Ebola virus disease.
Collapse
Affiliation(s)
- Timothy G. Wanninger
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Daniel E. Millian
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Omar A. Saldarriaga
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Junki Maruyama
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Takeshi Saito
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Rachel A. Reyna
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Satoshi Taniguchi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Esteban Arroyave
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Melanie E. Connolly
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, United States
| | - Heather L. Stevenson
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
8
|
Wang T, Cao Y, Zhang H, Wang Z, Man CH, Yang Y, Chen L, Xu S, Yan X, Zheng Q, Wang Y. COVID-19 metabolism: Mechanisms and therapeutic targets. MedComm (Beijing) 2022; 3:e157. [PMID: 35958432 PMCID: PMC9363584 DOI: 10.1002/mco2.157] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 01/18/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) dysregulates antiviral signaling, immune response, and cell metabolism in human body. Viral genome and proteins hijack host metabolic network to support viral biogenesis and propagation. However, the regulatory mechanism of SARS-CoV-2-induced metabolic dysfunction has not been elucidated until recently. Multiomic studies of coronavirus disease 2019 (COVID-19) revealed an intensive interaction between host metabolic regulators and viral proteins. SARS-CoV-2 deregulated cellular metabolism in blood, intestine, liver, pancreas, fat, and immune cells. Host metabolism supported almost every stage of viral lifecycle. Strikingly, viral proteins were found to interact with metabolic enzymes in different cellular compartments. Biochemical and genetic assays also identified key regulatory nodes and metabolic dependencies of viral replication. Of note, cholesterol metabolism, lipid metabolism, and glucose metabolism are broadly involved in viral lifecycle. Here, we summarized the current understanding of the hallmarks of COVID-19 metabolism. SARS-CoV-2 infection remodels host cell metabolism, which in turn modulates viral biogenesis and replication. Remodeling of host metabolism creates metabolic vulnerability of SARS-CoV-2 replication, which could be explored to uncover new therapeutic targets. The efficacy of metabolic inhibitors against COVID-19 is under investigation in several clinical trials. Ultimately, the knowledge of SARS-CoV-2-induced metabolic reprogramming would accelerate drug repurposing or screening to combat the COVID-19 pandemic.
Collapse
Affiliation(s)
- Tianshi Wang
- Shanghai Key Laboratory for Tumor Microenvironment and InflammationDepartment of Biochemistry and Molecular Cell BiologyShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ying Cao
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Haiyan Zhang
- Bai Jia Obstetrics and Gynecology HospitalShanghaiChina
| | - Zihao Wang
- Fudan University Shanghai Cancer CenterKey Laboratory of Breast Cancer in ShanghaiShanghai Key Laboratory of Radiation OncologyCancer Instituteand The Shanghai Key Laboratory of Medical EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- The International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghaiChina
| | - Cheuk Him Man
- Division of HematologyDepartment of MedicineUniversity of Hong KongPokfulamHong Kong, China
| | - Yunfan Yang
- Department of Cell BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanChina
| | - Lingchao Chen
- Department of NeurosurgeryHuashan HospitalShanghai Medical CollegeFudan UniversityNational Center for Neurological DisordersShanghai Key Laboratory of Brain Function and Restoration and Neural RegenerationNeurosurgical Institute of Fudan UniversityShanghai Clinical Medical Center of NeurosurgeryShanghaiChina
| | - Shuangnian Xu
- Department of HematologySouthwest HospitalArmy Medical UniversityChongqingChina
| | - Xiaojing Yan
- Department of HematologyThe First Affiliated Hospital of China Medical UniversityShenyangChina
| | - Quan Zheng
- Center for Single‐Cell OmicsSchool of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi‐Ping Wang
- Fudan University Shanghai Cancer CenterKey Laboratory of Breast Cancer in ShanghaiShanghai Key Laboratory of Radiation OncologyCancer Instituteand The Shanghai Key Laboratory of Medical EpigeneticsInstitutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- The International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghaiChina
| |
Collapse
|
9
|
Advances in Antifungal Development: Discovery of New Drugs and Drug Repurposing. Pharmaceuticals (Basel) 2022; 15:ph15070787. [PMID: 35890086 PMCID: PMC9318969 DOI: 10.3390/ph15070787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 06/23/2022] [Indexed: 02/05/2023] Open
Abstract
This Special Issue of Pharmaceuticals describes recent advances accomplished in the field of antifungal development, especially the discovery of new drugs and drug repurposing [...]
Collapse
|
10
|
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
|
11
|
Bradfute SB. The discovery and development of novel treatment strategies for filoviruses. Expert Opin Drug Discov 2021; 17:139-149. [PMID: 34962451 DOI: 10.1080/17460441.2022.2013800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Filoviruses are negative-stranded, enveloped RNA viruses that can cause hemorrhagic fever in humans and include Ebola and Marburg viruses. Lethality rates can reach 90% in isolated outbreaks. The 2013-2016 Ebola virus epidemic demonstrated the global threat of filoviruses and hastened development of vaccines and therapeutics. There are six known filoviruses that cause disease in humans, but still few therapeutics are available for treatment. AREAS COVERED This review summarizes identification, testing, and development of therapeutics based on the peer-reviewed scientific literature beginning with the discovery of filoviruses in 1967. Small molecules, antibodies, cytokines, antisense, post-exposure vaccination, and host-targeted therapeutic approaches are discussed. An emphasis is placed on therapeutics that have shown promise in in vivo studies. EXPERT OPINION Two monoclonal antibody regimens are approved for use in humans for one filovirus (Ebola virus), and preclinical nonhuman primate studies suggest that other monoclonal-based therapies are likely to be effective against other filoviruses. Significant progress has been made in small-molecule antivirals and host-targeted approaches. An important consideration is the necessity of pan-filovirus therapeutics via broadly effective small molecules, antibody cocktails, and cross-reactive antibodies. The use of filovirus therapeutics as prophylactic treatment or in chronically infected individuals should be considered.
Collapse
Affiliation(s)
- Steven B Bradfute
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, USA
| |
Collapse
|
12
|
Kummer S, Lander A, Goretzko J, Kirchoff N, Rescher U, Schloer S. Pharmacologically induced endolysosomal cholesterol imbalance through clinically licensed drugs itraconazole and fluoxetine impairs Ebola virus infection in vitro. Emerg Microbes Infect 2021; 11:195-207. [PMID: 34919035 PMCID: PMC8745396 DOI: 10.1080/22221751.2021.2020598] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Ebola virus disease (EVD) is a severe and frequently lethal disease caused by Ebola virus (EBOV). The latest occasional EVD outbreak (2013–2016) in Western African, which was accompanied by a high fatality rate, showed the great potential of epidemic and pandemic spread. Antiviral therapies against EBOV are very limited, strain-dependent (only antibody therapies are available) and mostly restricted to symptomatic treatment, illustrating the urgent need for novel antiviral strategies. Thus, we evaluated the effect of the clinically widely used antifungal itraconazole and the antidepressant fluoxetine for a repurposing against EBOV infection. While itraconazole, similar to U18666A, directly binds to and inhibits the endosomal membrane protein Niemann-Pick C1 (NPC1), fluoxetine, which belongs to the structurally unrelated group of weakly basic, amphiphile so-called “functional inhibitors of acid sphingomyelinase” (FIASMA) indirectly acts on the lysosome-residing acid sphingomyelinase via enzyme detachment leading to subsequent lysosomal degradation. Both, the drug-induced endolysosomal cholesterol accumulation and the altered endolysosomal pH, might interfere with the fusion of viral and endolysosomal membrane, preventing infection with EBOV. We further provide evidence that cholesterol imbalance is a conserved cross-species mechanism to hamper EBOV infection. Thus, exploring the endolysosomal host–pathogen interface as a suitable antiviral treatment may offer a general strategy to combat EBOV infection.
Collapse
Affiliation(s)
- Susann Kummer
- Center for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Angelika Lander
- Center for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Jonas Goretzko
- Research Group Regulatory Mechanisms of Inflammation, Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation, University of Muenster, Muenster, Germany.,Interdisciplinary Centre for Clinical Research, University of Muenster, Muenster, Germany.,Cluster of Excellence "Cells in Motion", University of Muenster, Muenster, Germany
| | - Norman Kirchoff
- Center for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Ursula Rescher
- Research Group Regulatory Mechanisms of Inflammation, Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation, University of Muenster, Muenster, Germany.,Interdisciplinary Centre for Clinical Research, University of Muenster, Muenster, Germany.,Cluster of Excellence "Cells in Motion", University of Muenster, Muenster, Germany
| | - Sebastian Schloer
- Research Group Regulatory Mechanisms of Inflammation, Institute of Medical Biochemistry, Centre for Molecular Biology of Inflammation, University of Muenster, Muenster, Germany.,Interdisciplinary Centre for Clinical Research, University of Muenster, Muenster, Germany.,Cluster of Excellence "Cells in Motion", University of Muenster, Muenster, Germany
| |
Collapse
|
13
|
Collette N, Dhungel P, Lund SJ, Schwedler JL, Saada EA, Light YK, Sinha A, Schoeniger JS, Negrete OA. Immunocompromised Cas9 transgenic mice for rapid in vivo assessment of host factors involved in highly pathogenic virus infection. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:286-295. [PMID: 34729376 PMCID: PMC8526419 DOI: 10.1016/j.omtm.2021.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/24/2021] [Indexed: 11/26/2022]
Abstract
Targeting host factors for anti-viral development offers several potential advantages over traditional countermeasures that include broad-spectrum activity and prevention of resistance. Characterization of host factors in animal models provides strong evidence of their involvement in disease pathogenesis, but the feasibility of performing high-throughput in vivo analyses on lists of genes is problematic. To begin addressing the challenges of screening candidate host factors in vivo, we combined advances in CRISPR-Cas9 genome editing with an immunocompromised mouse model used to study highly pathogenic viruses. Transgenic mice harboring a constitutively expressed Cas9 allele (Cas9tg/tg) with or without knockout of type I interferon receptors served to optimize in vivo delivery of CRISPR single-guide RNA (sgRNA) using Invivofectamine 3.0, a simple and easy-to-use lipid nanoparticle reagent. Invivofectamine 3.0-mediated liver-specific editing to remove activity of the critical Ebola virus host factor Niemann-Pick disease type C1 in an average of 74% of liver cells protected immunocompromised Cas9tg/tg mice from lethal surrogate Ebola virus infection. We envision that immunocompromised Cas9tg/tg mice combined with straightforward sgRNA in vivo delivery will enable efficient host factor loss-of-function screening in the liver and other organs to rapidly study their effects on viral pathogenesis and help initiate development of broad-spectrum, host-directed therapies against emerging pathogens.
Collapse
Affiliation(s)
- Nicole Collette
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 94550, USA
| | - Pragyesh Dhungel
- Department of Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, CA 94550, USA
| | - Sean J Lund
- Department of Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, CA 94550, USA
| | - Jennifer L Schwedler
- Department of Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, CA 94550, USA
| | - Edwin A Saada
- Department of Systems Biology, Sandia National Laboratories, Livermore, CA 94550, USA
| | - Yooli K Light
- Department of Systems Biology, Sandia National Laboratories, Livermore, CA 94550, USA
| | - Anupama Sinha
- Department of Systems Biology, Sandia National Laboratories, Livermore, CA 94550, USA
| | - Joseph S Schoeniger
- Department of Systems Biology, Sandia National Laboratories, Livermore, CA 94550, USA
| | - Oscar A Negrete
- Department of Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, CA 94550, USA
| |
Collapse
|
14
|
Downs I, Johnson JC, Rossi F, Dyer D, Saunders DL, Twenhafel NA, Esham HL, Pratt WD, Trefry J, Zumbrun E, Facemire PR, Johnston SC, Tompkins EL, Jansen NK, Honko A, Cardile AP. Natural History of Aerosol-Induced Ebola Virus Disease in Rhesus Macaques. Viruses 2021; 13:v13112297. [PMID: 34835103 PMCID: PMC8619410 DOI: 10.3390/v13112297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/03/2021] [Accepted: 11/12/2021] [Indexed: 02/06/2023] Open
Abstract
Ebola virus disease (EVD) is a serious global health concern because case fatality rates are approximately 50% due to recent widespread outbreaks in Africa. Well-defined nonhuman primate (NHP) models for different routes of Ebola virus exposure are needed to test the efficacy of candidate countermeasures. In this natural history study, four rhesus macaques were challenged via aerosol with a target titer of 1000 plaque-forming units per milliliter of Ebola virus. The course of disease was split into the following stages for descriptive purposes: subclinical, clinical, and decompensated. During the subclinical stage, high levels of venous partial pressure of carbon dioxide led to respiratory acidemia in three of four of the NHPs, and all developed lymphopenia. During the clinical stage, all animals had fever, viremia, and respiratory alkalosis. The decompensatory stage involved coagulopathy, cytokine storm, and liver and renal injury. These events were followed by hypotension, elevated lactate, metabolic acidemia, shock and mortality similar to historic intramuscular challenge studies. Viral loads in the lungs of aerosol-exposed animals were not distinctly different compared to previous intramuscularly challenged studies. Differences in the aerosol model, compared to intramuscular model, include an extended subclinical stage, shortened clinical stage, and general decompensated stage. Therefore, the shortened timeframe for clinical detection of the aerosol-induced disease can impair timely therapeutic administration. In summary, this nonhuman primate model of aerosol-induced EVD characterizes early disease markers and additional details to enable countermeasure development.
Collapse
Affiliation(s)
- Isaac Downs
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
- Correspondence: ; Tel.: +1-301-619-0369
| | - Joshua C. Johnson
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
- Moderna, Inc., Cambridge, MA 02139, USA
| | - Franco Rossi
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| | - David Dyer
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| | - David L. Saunders
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| | - Nancy A. Twenhafel
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| | - Heather L. Esham
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| | - William D. Pratt
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| | - John Trefry
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
- Defense Threat Reduction Agency, Fort Belvoir, VA 22060, USA
| | - Elizabeth Zumbrun
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| | - Paul R. Facemire
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| | - Sara C. Johnston
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| | - Erin L. Tompkins
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| | - Nathan K. Jansen
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| | - Anna Honko
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
- Investigator at National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA 02118, USA
| | - Anthony P. Cardile
- US Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, MD 21702, USA; (J.C.J.); (F.R.); (D.D.); (D.L.S.); (N.A.T.); (H.L.E.); (W.D.P.); (J.T.); (E.Z.); (P.R.F.); (S.C.J.); (E.L.T.); (N.K.J.); (A.H.); (A.P.C.)
| |
Collapse
|
15
|
Bokelmann M, Vogel U, Debeljak F, Düx A, Riesle-Sbarbaro S, Lander A, Wahlbrink A, Kromarek N, Neil S, Couacy-Hymann E, Prescott J, Kurth A. Tolerance and Persistence of Ebola Virus in Primary Cells from Mops condylurus, a Potential Ebola Virus Reservoir. Viruses 2021; 13:v13112186. [PMID: 34834992 PMCID: PMC8622823 DOI: 10.3390/v13112186] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/14/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022] Open
Abstract
Although there have been documented Ebola virus disease outbreaks for more than 40 years, the natural reservoir host has not been identified. Recent studies provide evidence that the Angolan free-tailed bat (Mops condylurus), an insectivorous microbat, is a possible ebolavirus reservoir. To investigate the potential role of this bat species in the ecology of ebolaviruses, replication, tolerance, and persistence of Ebola virus (EBOV) were investigated in 10 different primary bat cell isolates from M. condylurus. Varying EBOV replication kinetics corresponded to the expression levels of the integral membrane protein NPC1. All primary cells were highly tolerant to EBOV infection without cytopathic effects. The observed persistent EBOV infection for 150 days in lung primary cells, without resultant selective pressure leading to virus mutation, indicate the intrinsic ability of EBOV to persist in this bat species. These results provide further evidence for this bat species to be a likely reservoir of ebolaviruses.
Collapse
Affiliation(s)
- Marcel Bokelmann
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany; (M.B.); (U.V.); (S.R.-S.); (A.L.); (A.W.); (N.K.); (J.P.)
| | - Uwe Vogel
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany; (M.B.); (U.V.); (S.R.-S.); (A.L.); (A.W.); (N.K.); (J.P.)
| | - Franka Debeljak
- Department of Infectious Diseases, King’s College London, London WC2R 2LS, UK; (F.D.); (S.N.)
| | - Ariane Düx
- Epidemiology of Highly Pathogenic Microorganisms, Robert Koch Institute, 13353 Berlin, Germany;
| | - Silke Riesle-Sbarbaro
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany; (M.B.); (U.V.); (S.R.-S.); (A.L.); (A.W.); (N.K.); (J.P.)
| | - Angelika Lander
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany; (M.B.); (U.V.); (S.R.-S.); (A.L.); (A.W.); (N.K.); (J.P.)
| | - Annette Wahlbrink
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany; (M.B.); (U.V.); (S.R.-S.); (A.L.); (A.W.); (N.K.); (J.P.)
| | - Nicole Kromarek
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany; (M.B.); (U.V.); (S.R.-S.); (A.L.); (A.W.); (N.K.); (J.P.)
| | - Stuart Neil
- Department of Infectious Diseases, King’s College London, London WC2R 2LS, UK; (F.D.); (S.N.)
| | - Emmanuel Couacy-Hymann
- Laboratoire National d’Appui au Développement Agricole, Bingerville BP 206, Côte d’Ivoire;
| | - Joseph Prescott
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany; (M.B.); (U.V.); (S.R.-S.); (A.L.); (A.W.); (N.K.); (J.P.)
| | - Andreas Kurth
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany; (M.B.); (U.V.); (S.R.-S.); (A.L.); (A.W.); (N.K.); (J.P.)
- Correspondence:
| |
Collapse
|
16
|
Farfan-Morales CN, Cordero-Rivera CD, Reyes-Ruiz JM, Hurtado-Monzón AM, Osuna-Ramos JF, González-González AM, De Jesús-González LA, Palacios-Rápalo SN, Del Ángel RM. Anti-flavivirus Properties of Lipid-Lowering Drugs. Front Physiol 2021; 12:749770. [PMID: 34690817 PMCID: PMC8529048 DOI: 10.3389/fphys.2021.749770] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022] Open
Abstract
Although Flaviviruses such as dengue (DENV) and zika (ZIKV) virus are important human pathogens, an effective vaccine or antiviral treatment against them is not available. Hence, the search for new strategies to control flavivirus infections is essential. Several studies have shown that the host lipid metabolism could be an antiviral target because cholesterol and other lipids are required during the replicative cycle of different Flaviviridae family members. FDA-approved drugs with hypolipidemic effects could be an alternative for treating flavivirus infections. However, a better understanding of the regulation between host lipid metabolism and signaling pathways triggered during these infections is required. The metabolic pathways related to lipid metabolism modified during DENV and ZIKV infection are analyzed in this review. Additionally, the role of lipid-lowering drugs as safe host-targeted antivirals is discussed.
Collapse
Affiliation(s)
- Carlos Noe Farfan-Morales
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Carlos Daniel Cordero-Rivera
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - José Manuel Reyes-Ruiz
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 14, Centro Médico Nacional "Adolfo Ruiz Cortines," Instituto Mexicano del Seguro Social, Heroica Veracruz, Mexico
| | - Arianna M Hurtado-Monzón
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Juan Fidel Osuna-Ramos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Arely M González-González
- Laboratorio de Ingeniería Tisular y Medicina Traslacional, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Luis Adrián De Jesús-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Selvin Noé Palacios-Rápalo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| | - Rosa María Del Ángel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
17
|
Wirchnianski AS, Wec AZ, Nyakatura EK, Herbert AS, Slough MM, Kuehne AI, Mittler E, Jangra RK, Teruya J, Dye JM, Lai JR, Chandran K. Two Distinct Lysosomal Targeting Strategies Afford Trojan Horse Antibodies With Pan-Filovirus Activity. Front Immunol 2021; 12:729851. [PMID: 34721393 PMCID: PMC8551868 DOI: 10.3389/fimmu.2021.729851] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Multiple agents in the family Filoviridae (filoviruses) are associated with sporadic human outbreaks of highly lethal disease, while others, including several recently identified agents, possess strong zoonotic potential. Although viral glycoprotein (GP)-specific monoclonal antibodies have demonstrated therapeutic utility against filovirus disease, currently FDA-approved molecules lack antiviral breadth. The development of broadly neutralizing antibodies has been challenged by the high sequence divergence among filovirus GPs and the complex GP proteolytic cleavage cascade that accompanies filovirus entry. Despite this variability in the antigenic surface of GP, all filoviruses share a site of vulnerability-the binding site for the universal filovirus entry receptor, Niemann-Pick C1 (NPC1). Unfortunately, this site is shielded in extracellular GP and only uncovered by proteolytic cleavage by host proteases in late endosomes and lysosomes, which are generally inaccessible to antibodies. To overcome this obstacle, we previously developed a 'Trojan horse' therapeutic approach in which engineered bispecific antibodies (bsAbs) coopt viral particles to deliver GP:NPC1 interaction-blocking antibodies to their endo/lysosomal sites of action. This approach afforded broad protection against members of the genus Ebolavirus but could not neutralize more divergent filoviruses. Here, we describe next-generation Trojan horse bsAbs that target the endo/lysosomal GP:NPC1 interface with pan-filovirus breadth by exploiting the conserved and widely expressed host cation-independent mannose-6-phosphate receptor for intracellular delivery. Our work highlights a new avenue for the development of single therapeutics protecting against all known and newly emerging filoviruses.
Collapse
Affiliation(s)
- Ariel S. Wirchnianski
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Anna Z. Wec
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Elisabeth K. Nyakatura
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Andrew S. Herbert
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
- The Geneva Foundation, Tacoma, WA, United States
| | - Megan M. Slough
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ana I. Kuehne
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Eva Mittler
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Rohit K. Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jonathan Teruya
- Antibody Discovery and Research group, Mapp Biopharmaceutical, San Diego, CA, United States
| | - John M. Dye
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Jonathan R. Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
18
|
García-Dorival I, Cuesta-Geijo MÁ, Barrado-Gil L, Galindo I, Garaigorta U, Urquiza J, Puerto AD, Campillo NE, Martínez A, Gastaminza P, Gil C, Alonso C. Identification of Niemann-Pick C1 protein as a potential novel SARS-CoV-2 intracellular target. Antiviral Res 2021; 194:105167. [PMID: 34450201 PMCID: PMC8382594 DOI: 10.1016/j.antiviral.2021.105167] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 11/30/2022]
Abstract
Niemann-Pick type C1 (NPC1) receptor is an endosomal membrane protein that regulates intracellular cholesterol traffic. This protein has been shown to play an important role for several viruses. It has been reported that SARS-CoV-2 enters the cell through plasma membrane fusion and/or endosomal entry upon availability of proteases. However, the whole process is not fully understood yet and additional viral/host factors might be required for viral fusion and subsequent viral replication. Here, we report a novel interaction between the SARS-CoV-2 nucleoprotein (N) and the cholesterol transporter NPC1. Furthermore, we have found that some compounds reported to interact with NPC1, carbazole SC816 and sulfides SC198 and SC073, were able to reduce SARS-CoV-2 viral infection with a good selectivity index in human cell infection models. These findings suggest the importance of NPC1 for SARS-CoV-2 viral infection and a new possible potential therapeutic target to fight against COVID-19.
Collapse
Affiliation(s)
- Isabel García-Dorival
- Dpt. 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
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Ctra. de la Coruña km 7.5, 28040, Madrid, Spain; Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Lucía Barrado-Gil
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Ctra. de la Coruña km 7.5, 28040, Madrid, Spain; Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Inmaculada Galindo
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Ctra. de la Coruña km 7.5, 28040, Madrid, Spain
| | - Urtzi Garaigorta
- Centro Nacional de Biotecnología CSIC, Calle Darwin 3, 28049, Madrid, Spain
| | - Jesús Urquiza
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Ctra. de la Coruña km 7.5, 28040, Madrid, Spain
| | - Ana Del Puerto
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Ctra. de la Coruña km 7.5, 28040, Madrid, Spain
| | - Nuria E Campillo
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Ana Martínez
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Pablo Gastaminza
- Centro Nacional de Biotecnología CSIC, Calle Darwin 3, 28049, Madrid, Spain
| | - Carmen Gil
- Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Covadonga Alonso
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Ctra. de la Coruña km 7.5, 28040, Madrid, Spain.
| |
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
|
Potential pharmacological strategies targeting the Niemann-Pick C1 receptor and Ebola virus glycoprotein interaction. Eur J Med Chem 2021; 223:113654. [PMID: 34175537 DOI: 10.1016/j.ejmech.2021.113654] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/27/2021] [Accepted: 06/13/2021] [Indexed: 10/21/2022]
Abstract
Niemann-Pick C1 (NPC1) receptor is an intracellular protein located in late endosomes and lysosomes whose main function is to regulate intracellular cholesterol trafficking. Besides being postulated as necessary for the infection of highly pathogenic viruses in which the integrity of cholesterol transport is required, this protein also allows the entry of the Ebola virus (EBOV) into the host cells acting as an intracellular receptor. EBOV glycoprotein (EBOV-GP) interaction with NPC1 at the endosomal membrane triggers the release of the viral material into the host cell, starting the infective cycle. Disruption of the NPC1/EBOV-GP interaction could represent an attractive strategy for the development of drugs aimed at inhibiting viral entry and thus infection. Some of the today available EBOV inhibitors were proposed to interrupt this interaction, but molecular and structural details about their mode of action are still preliminary thus more efforts are needed to properly address these points. Here, we provide a critical discussion of the potential of NPC1 and its interaction with EBOV-GP as a therapeutic target for viral infections.
Collapse
|
21
|
Hansen F, Feldmann H, Jarvis MA. Targeting Ebola virus replication through pharmaceutical intervention. Expert Opin Investig Drugs 2021; 30:201-226. [PMID: 33593215 DOI: 10.1080/13543784.2021.1881061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction. The consistent emergence/reemergence of filoviruses into a world that previously lacked an approved pharmaceutical intervention parallels an experience repeatedly played-out for most other emerging pathogenic zoonotic viruses. Investment to preemptively develop effective and low-cost prophylactic and therapeutic interventions against viruses that have high potential for emergence and societal impact should be a priority.Areas covered. Candidate drugs can be characterized into those that interfere with cellular processes required for Ebola virus (EBOV) replication (host-directed), and those that directly target virally encoded functions (direct-acting). We discuss strategies to identify pharmaceutical interventions for EBOV infections. PubMed/Web of Science databases were searched to establish a detailed catalog of these interventions.Expert opinion. Many drug candidates show promising in vitro inhibitory activity, but experience with EBOV shows the general lack of translation to in vivo efficacy for host-directed repurposed drugs. Better translation is seen for direct-acting antivirals, in particular monoclonal antibodies. The FDA-approved monoclonal antibody treatment, Inmazeb™ is a success story that could be improved in terms of impact on EBOV-associated disease and mortality, possibly by combination with other direct-acting agents targeting distinct aspects of the viral replication cycle. Costs need to be addressed given EBOV emergence primarily in under-resourced countries.
Collapse
Affiliation(s)
- Frederick Hansen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Michael A Jarvis
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.,School of Biomedical Sciences, University of Plymouth, Plymouth, Devon, UK.,The Vaccine Group, Ltd, Plymouth, Devon, UK
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Modeling the efficiency of filovirus entry into cells in vitro: Effects of SNP mutations in the receptor molecule. PLoS Comput Biol 2020; 16:e1007612. [PMID: 32986692 PMCID: PMC7544041 DOI: 10.1371/journal.pcbi.1007612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 10/08/2020] [Accepted: 08/03/2020] [Indexed: 11/27/2022] Open
Abstract
Interaction between filovirus glycoprotein (GP) and the Niemann-Pick C1 (NPC1) protein is essential for membrane fusion during virus entry. Some single-nucleotide polymorphism (SNPs) in two surface-exposed loops of NPC1 are known to reduce viral infectivity. However, the dependence of differences in entry efficiency on SNPs remains unclear. Using vesicular stomatitis virus pseudotyped with Ebola and Marburg virus GPs, we investigated the cell-to-cell spread of viruses in cultured cells expressing NPC1 or SNP derivatives. Eclipse and virus-producing phases were assessed by in vitro infection experiments, and we developed a mathematical model describing spatial-temporal virus spread. This mathematical model fit the plaque radius data well from day 2 to day 6. Based on the estimated parameters, we found that SNPs causing the P424A and D508N substitutions in NPC1 most effectively reduced the entry efficiency of Ebola and Marburg viruses, respectively. Our novel approach could be broadly applied to other virus plaque assays. Ebola (EBOV) and Marburg (MARV) viruses, which are included viruses of the family Filoviridae, cause severe hemorrhagic fever in humans. Filovirus particles is adsorbed to the cell through glycoprotein (GP), which is the only viral surface protein. Interaction between the filovirus sugar protein (GP) and the Niemann-Pick C1 (NPC1) protein plays a key role in membrane fusion during virus entry. Although some single-nucleotide polymorphism (SNPs) in two surface-exposed loops of NPC1 are known to reduce viral infectivity, the dependence of differences in entry efficiency on SNPs has not been studied. We therefore investigated the cell-to-cell spread of viruses in cultured cells expressing NPC1 or SNP derivatives. Using a mathematical model describing spatial-temporal virus spread, we quantitatively analyze viral entry efficiency and how this affected cell-to-cell spread. Our approach may be applied to not only understanding the roles of genetic polymorphisms in human susceptibility to filoviruses, but also other virus plaque assays.
Collapse
|
25
|
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
|
26
|
Engineered Human Cathelicidin Antimicrobial Peptides Inhibit Ebola Virus Infection. iScience 2020; 23:100999. [PMID: 32252021 PMCID: PMC7104201 DOI: 10.1016/j.isci.2020.100999] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/12/2020] [Accepted: 03/18/2020] [Indexed: 01/08/2023] Open
Abstract
The 2014–2016 West Africa Ebola virus (EBOV) outbreak coupled with the most recent outbreaks in Central Africa underscore the need to develop effective treatment strategies against EBOV. Although several therapeutic options have shown great potential, developing a wider breadth of countermeasures would increase our efforts to combat the highly lethal EBOV. Here we show that human cathelicidin antimicrobial peptide (AMP) LL-37 and engineered LL-37 AMPs inhibit the infection of recombinant virus pseudotyped with EBOV glycoprotein (GP) and the wild-type EBOV. These AMPs target EBOV infection at the endosomal cell-entry step by impairing cathepsin B-mediated processing of EBOV GP. Furthermore, two engineered AMPs containing D-amino acids are particularly potent in blocking EBOV infection in comparison with other AMPs, most likely owing to their resistance to intracellular enzymatic degradation. Our results identify AMPs as a novel class of anti-EBOV therapeutics and demonstrate the feasibility of engineering AMPs for improved therapeutic efficacy. Cathelicidin-derived antimicrobial peptides (AMPs) potently inhibit EBOV infection D-form AMPs are more resistant to proteolytic cleavage than L-form AMPs in the cell AMPs prevent cathepsin B-mediated processing of EBOV GP1, 2
Collapse
|
27
|
Bokelmann M, Edenborough K, Hetzelt N, Kreher P, Lander A, Nitsche A, Vogel U, Feldmann H, Couacy-Hymann E, Kurth A. Utility of primary cells to examine NPC1 receptor expression in Mops condylurus, a potential Ebola virus reservoir. PLoS Negl Trop Dis 2020; 14:e0007952. [PMID: 31961874 PMCID: PMC6994141 DOI: 10.1371/journal.pntd.0007952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 01/31/2020] [Accepted: 11/20/2019] [Indexed: 12/17/2022] Open
Abstract
The significance of the integral membrane protein Niemann-Pick C1 (NPC1) in the ebolavirus entry process has been determined using various cell lines derived from humans, non-human primates and fruit bats. Fruit bats have long been purported as the potential reservoir host for ebolaviruses, however several studies provide evidence that Mops condylurus, an insectivorous microbat, is also an ebolavirus reservoir. NPC1 receptor expression in the context of ebolavirus replication in microbat cells remains unstudied. In order to study Ebola virus (EBOV) cellular entry and replication in M. condylurus, we derived primary and immortalized cell cultures from 12 different organs. The NPC1 receptor expression was characterized by confocal microscopy and flow cytometry comparing the expression levels of M. condylurus primary and immortalized cells, HeLa cells, human embryonic kidney cells and cells from a European microbat species. EBOV replication kinetics was studied for four representative cell cultures using qRT-PCR. The aim was to elucidate the suitability of primary and immortalized cells from different tissues for studying NPC1 receptor expression levels and their potential influence on EBOV replication. The NPC1 receptor expression level in M. condylurus primary cells differed depending on the organ they were derived from and was for most cell types significantly lower than in human cell lines. Immortalized cells showed for most cell types higher expression levels than their corresponding primary cells. Concluding from our infection experiments with EBOV we suggest a potential correlation between NPC1 receptor expression level and virus replication rate in vitro. Although there have been Ebola virus (EBOV) outbreaks for more than 40 years, the animal natural reservoir that maintains this virus in nature has not been identified. Viruses and their respective reservoirs coevolve over millions of years, often without causing diseases in the reservoir itself. Upon entering a new host, infection can have devastating consequences, as in the case of EBOV. To gain entry into cells prior to replication, all ebolaviruses utilize the cellular receptor Niemann-Pick C1 (NPC1). In this study the authors focus their work on the Angolan free-tailed bat (Mops condylurus) as a potential reservoir for EBOV. Cells from various organs of this bat were isolated in culture and tested for the presence of NPC1. Most bat cell types had a lower amount of NPC1 compared to the tested human cells. These bat cells were also less efficiently infected by EBOV, indicating adaptation to EBOV. These results suggest low levels of virus replication in the respective tissues of M. condylurus and might be indicative of a virus-natural reservoir relationship.
Collapse
Affiliation(s)
- Marcel Bokelmann
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Kathryn Edenborough
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Nicole Hetzelt
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Petra Kreher
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Angelika Lander
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Andreas Nitsche
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Uwe Vogel
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States of America
| | | | - Andreas Kurth
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
- * E-mail:
| |
Collapse
|
28
|
Chao YK, Chang SY, Grimm C. Endo-Lysosomal Cation Channels and Infectious Diseases. Rev Physiol Biochem Pharmacol 2020; 185:259-276. [PMID: 32748124 DOI: 10.1007/112_2020_31] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Among the infectious diseases caused by pathogenic microorganisms such as bacteria, viruses, parasites, or fungi, the most prevalent ones today are malaria, tuberculosis, influenza, HIV/AIDS, Ebola, dengue fever, and methicillin-resistant Staphylococcus aureus (MRSA) infection, and most recently Covid-19 (SARS-CoV2). Others with a rather devastating history and high fatality rates such as plague, cholera, or typhus seem less threatening today but have not been eradicated, and with a declining efficacy of current antibiotics they ought to be watched carefully. Another emerging issue in this context is health-care associated infection. About 100,000 hospitalized patients in the USA ( www.cdc.gov ) and 33,000 in Europe ( https://www.ecdc.europa.eu ) die each year as a direct consequence of an infection caused by bacteria resistant to antibiotics. Among viral infections, influenza is responsible for about 3-5 million cases of severe illness, and about 250,000 to 500,000 deaths annually ( www.who.int ). About 37 million people are currently living with HIV infection and about one million die from it each year. Coronaviruses such as MERS-CoV, SARS-CoV, but in particular the recent outbreak of Covid-19 (caused by SARS-CoV2) have resulted in large numbers of infections worldwide with an estimated several hundred thousand deaths (anticipated fatality rate: <5%). With a comparatively low mortality rate dengue virus causes between 50 and 100 million infections every year, leading to 50,000 deaths. In contrast, Ebola virus is the causative agent for one of the deadliest viral diseases. The Ebola outbreak in West Africa in 2014 is considered the largest outbreak in history with more than 11,000 deaths. Many of the deadliest pathogens such as Ebola virus, influenza virus, mycobacterium tuberculosis, dengue virus, and cholera exploit the endo-lysosomal trafficking system of host cells for penetration into the cytosol and replication. Defects in endo-lysosomal maturation, trafficking, fusion, or pH homeostasis can efficiently reduce the cytotoxicity caused by these pathogens. Most of these functions critically depend on endo-lysosomal membrane proteins such as transporters and ion channels. In particular, cation channels such as the mucolipins (TRPMLs) or the two-pore channels (TPCs) are involved in all of these aspects of endo-lysosomal integrity. In this review we will discuss the correlations between pathogen toxicity and endo-lysosomal cation channel function, and their potential as drug targets for infectious disease therapy.
Collapse
Affiliation(s)
- Yu-Kai Chao
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Christian Grimm
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität, Munich, Germany.
| |
Collapse
|
29
|
Shin SD, Shin A, Mayagoitia K, Wilson CG, Bellinger DL, Soriano S. Interferon downstream signaling is activated early in pre-symptomatic Niemann-Pick disease type C. Neurosci Lett 2019; 706:43-50. [DOI: 10.1016/j.neulet.2019.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/02/2019] [Accepted: 05/04/2019] [Indexed: 12/16/2022]
|
30
|
RNASeq Analysis of Aedes albopictus Mosquito Midguts after Chikungunya Virus Infection. Viruses 2019; 11:v11060513. [PMID: 31167461 PMCID: PMC6631752 DOI: 10.3390/v11060513] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 06/02/2019] [Indexed: 01/01/2023] Open
Abstract
Chikungunya virus (CHIKV) is an emerging pathogen around the world and causes significant morbidity in patients. A single amino acid mutation in the envelope protein of CHIKV has led to a shift in vector preference towards Aedesalbopictus. While mosquitoes are known to mount an antiviral immune response post-infection, molecular interactions during the course of infection at the tissue level remain largely uncharacterised. We performed whole transcriptome analysis on dissected midguts of Aedes albopictus infected with CHIKV to identify differentially expressed genes. For this, RNA was extracted at two days post-infection (2-dpi) from pooled midguts. We initially identified 25 differentially expressed genes (p-value < 0.05) when mapped to a reference transcriptome. Further, multiple differentially expressed genes were identified from a custom de novo transcriptome, which was assembled using the reads that did not align with the reference genome. Thirteen of the identified transcripts, possibly involved in immunity, were validated by qRT-PCR. Homologues of seven of these genes were also found to be significantly upregulated in Aedes aegypti midguts 2 dpi, indicating a conserved mechanism at play. These results will help us to characterise the molecular interaction between Aedes albopictus and CHIKV and can be utilised to reduce the impact of this viral infection.
Collapse
|
31
|
Olejnik J, Hume AJ, Leung DW, Amarasinghe GK, Basler CF, Mühlberger E. Filovirus Strategies to Escape Antiviral Responses. Curr Top Microbiol Immunol 2019; 411:293-322. [PMID: 28685291 PMCID: PMC5973841 DOI: 10.1007/82_2017_13] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This chapter describes the various strategies filoviruses use to escape host immune responses with a focus on innate immune and cell death pathways. Since filovirus replication can be efficiently blocked by interferon (IFN), filoviruses have evolved mechanisms to counteract both type I IFN induction and IFN response signaling pathways. Intriguingly, marburg- and ebolaviruses use different strategies to inhibit IFN signaling. This chapter also summarizes what is known about the role of IFN-stimulated genes (ISGs) in filovirus infection. These fall into three categories: those that restrict filovirus replication, those whose activation is inhibited by filoviruses, and those that have no measurable effect on viral replication. In addition to innate immunity, mammalian cells have evolved strategies to counter viral infections, including the induction of cell death and stress response pathways, and we summarize our current knowledge of how filoviruses interact with these pathways. Finally, this chapter delves into the interaction of EBOV with myeloid dendritic cells and macrophages and the associated inflammatory response, which differs dramatically between these cell types when they are infected with EBOV. In summary, we highlight the multifaceted nature of the host-viral interactions during filoviral infections.
Collapse
Affiliation(s)
- Judith Olejnik
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA
| | - Adam J Hume
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Christopher F Basler
- Microbial Pathogenesis, Georgia State University, Institute for Biomedical Sciences, Atlanta, GA, 30303, USA
| | - Elke Mühlberger
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, Boston, MA, 02118, USA.
| |
Collapse
|
32
|
Anti-Niemann Pick C1 Single-Stranded Oligonucleotides with Locked Nucleic Acids Potently Reduce Ebola Virus Infection In Vitro. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 16:686-697. [PMID: 31125846 PMCID: PMC6529764 DOI: 10.1016/j.omtn.2019.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/12/2019] [Accepted: 04/10/2019] [Indexed: 12/12/2022]
Abstract
Ebola virus is the causative agent of Ebola virus disease, a severe, often fatal illness in humans. So far, there are no US Food and Drug Administration (FDA)-approved therapeutics directed against Ebola virus. Here, we selected the host factor Niemann-Pick C1 (NPC1), which has been shown to be essential for Ebola virus entry into host cytoplasm, as a therapeutic target for suppression by locked nucleic acid-modified antisense oligonucleotides. Screening of antisense oligonucleotides in human and murine cell lines led to identification of candidates with up to 94% knockdown efficiency and 50% inhibitory concentration (IC50) values in the submicromolar range. Selected candidate oligonucleotides led to efficient NPC1 protein knockdown in vitro without alteration of cell viability. Furthermore, they did not have immune stimulatory activity in cell-based assays. Treatment of Ebola-virus-infected HeLa cells with the most promising candidates resulted in significant (>99%) virus titer reduction, indicating that antisense oligonucleotides against NPC1 are a promising therapeutic approach for treatment of Ebola virus infection.
Collapse
|
33
|
Edwards MR, Basler CF. Current status of small molecule drug development for Ebola virus and other filoviruses. Curr Opin Virol 2019; 35:42-56. [PMID: 31003196 PMCID: PMC6556423 DOI: 10.1016/j.coviro.2019.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/12/2019] [Indexed: 12/16/2022]
Abstract
The filovirus family includes some of the deadliest viruses known, including Ebola virus and Marburg virus. These viruses cause periodic outbreaks of severe disease that can be spread from person to person, making the filoviruses important public health threats. There remains a need for approved drugs that target all or most members of this virus family. Small molecule inhibitors that target conserved functions hold promise as pan-filovirus therapeutics. To date, compounds that effectively target virus entry, genome replication, gene expression, and virus egress have been described. The most advanced inhibitors are nucleoside analogs that target viral RNA synthesis reactions.
Collapse
Affiliation(s)
- Megan R Edwards
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, United States
| | - Christopher F Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, United States.
| |
Collapse
|
34
|
Flint M, Chatterjee P, Lin DL, McMullan LK, Shrivastava-Ranjan P, Bergeron É, Lo MK, Welch SR, Nichol ST, Tai AW, Spiropoulou CF. A genome-wide CRISPR screen identifies N-acetylglucosamine-1-phosphate transferase as a potential antiviral target for Ebola virus. Nat Commun 2019; 10:285. [PMID: 30655525 PMCID: PMC6336797 DOI: 10.1038/s41467-018-08135-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 12/19/2018] [Indexed: 12/28/2022] Open
Abstract
There are no approved therapies for Ebola virus infection. Here, to find potential therapeutic targets, we perform a screen for genes essential for Ebola virus (EBOV) infection. We identify GNPTAB, which encodes the α and β subunits of N-acetylglucosamine-1-phosphate transferase. We show that EBOV infection of a GNPTAB knockout cell line is impaired, and that this is reversed by reconstituting GNPTAB expression. Fibroblasts from patients with mucolipidosis II, a disorder associated with mutations in GNPTAB, are refractory to EBOV, whereas cells from their healthy parents support infection. Impaired infection correlates with loss of the expression of cathepsin B, known to be essential for EBOV entry. GNPTAB activity is dependent upon proteolytic cleavage by the SKI-1/S1P protease. Inhibiting this protease with the small-molecule PF-429242 blocks EBOV entry and infection. Disruption of GNPTAB function may represent a strategy for a host-targeted therapy for EBOV. Genetic screens are important tools to identify host factors associated with viral infections. Here, Flint et al. perform a genome-wide CRISPR screen using infectious Ebola virus (EBOV) and show that the host transferase GNPTAB is required for EBOV infection and a potential target for antiviral therapies
Collapse
Affiliation(s)
- Mike Flint
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA.
| | - Payel Chatterjee
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - David L Lin
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Laura K McMullan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Punya Shrivastava-Ranjan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Éric Bergeron
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Michael K Lo
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Stephen R Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA
| | - Andrew W Tai
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road, MS G-14, Atlanta, GA, 30329, USA.
| |
Collapse
|
35
|
Gordon TB, Hayward JA, Marsh GA, Baker ML, Tachedjian G. Host and Viral Proteins Modulating Ebola and Marburg Virus Egress. Viruses 2019; 11:v11010025. [PMID: 30609802 PMCID: PMC6357148 DOI: 10.3390/v11010025] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/21/2018] [Accepted: 01/01/2019] [Indexed: 12/11/2022] Open
Abstract
The filoviruses Ebolavirus and Marburgvirus are among the deadliest viral pathogens known to infect humans, causing emerging diseases with fatality rates of up to 90% during some outbreaks. The replication cycles of these viruses are comprised of numerous complex molecular processes and interactions with their human host, with one key feature being the means by which nascent virions exit host cells to spread to new cells and ultimately to a new host. This review focuses on our current knowledge of filovirus egress and the viral and host factors and processes that are involved. Within the virus, these factors consist of the major matrix protein, viral protein 40 (VP40), which is necessary and sufficient for viral particle release, and nucleocapsid and glycoprotein that interact with VP40 to promote egress. In the host cell, some proteins are hijacked by filoviruses in order to enhance virion budding capacity that include members of the family of E3 ubiquitin ligase and the endosomal sorting complexes required for transport (ESCRT) pathway, while others such as tetherin inhibit viral egress. An understanding of these molecular interactions that modulate viral particle egress provides an important opportunity to identify new targets for the development of antivirals to prevent and treat filovirus infections.
Collapse
Affiliation(s)
- Tamsin B Gordon
- Health Security Program, Life Sciences Discipline, Burnet Institute, Melbourne, VIC 3004, Australia.
- Department of Microbiology, Monash University, Clayton, VIC 3168, Australia.
| | - Joshua A Hayward
- Health Security Program, Life Sciences Discipline, Burnet Institute, Melbourne, VIC 3004, Australia.
- Department of Microbiology, Monash University, Clayton, VIC 3168, Australia.
| | - Glenn A Marsh
- Department of Microbiology, Monash University, Clayton, VIC 3168, Australia.
- CSIRO Australian Animal Health Laboratory, Health and Biosecurity Business Unit, Geelong, VIC 3220, Australia.
| | - Michelle L Baker
- CSIRO Australian Animal Health Laboratory, Health and Biosecurity Business Unit, Geelong, VIC 3220, Australia.
| | - Gilda Tachedjian
- Health Security Program, Life Sciences Discipline, Burnet Institute, Melbourne, VIC 3004, Australia.
- Department of Microbiology, Monash University, Clayton, VIC 3168, Australia.
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne VIC 3010, Australia.
- School of Science, College of Science, Engineering and Health, RMIT University, Melbourne, VIC 3000, Australia.
| |
Collapse
|
36
|
Do GWAS and studies of heterozygotes for NPC1 and/or NPC2 explain why NPC disease cases are so rare? J Appl Genet 2018; 59:441-447. [PMID: 30209687 DOI: 10.1007/s13353-018-0465-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/31/2018] [Accepted: 09/04/2018] [Indexed: 12/15/2022]
Abstract
Early onset Niemann-Pick C diseases are extremely rare, especially Niemann-Pick C2. Perhaps unusually for autosomal recessive diseases, heterozygotes for mutations in NPC1 manifest many biological variations. NPC2 deficiency has large effects on fertility. These features of NPC1 and NPC2 are reviewed in regard to possible negative selection for heterozygotes carrying null and hypomorphic alleles.
Collapse
|
37
|
Shrivastava-Ranjan P, Flint M, Bergeron É, McElroy AK, Chatterjee P, Albariño CG, Nichol ST, Spiropoulou CF. Statins Suppress Ebola Virus Infectivity by Interfering with Glycoprotein Processing. mBio 2018; 9:e00660-18. [PMID: 29717011 PMCID: PMC5930306 DOI: 10.1128/mbio.00660-18] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 12/18/2022] Open
Abstract
Ebola virus (EBOV) infection is a major public health concern due to high fatality rates and limited effective treatments. Statins, widely used cholesterol-lowering drugs, have pleiotropic mechanisms of action and were suggested as potential adjunct therapy for Ebola virus disease (EVD) during the 2013-2016 outbreak in West Africa. Here, we evaluated the antiviral effects of statin (lovastatin) on EBOV infection in vitro Statin treatment decreased infectious EBOV production in primary human monocyte-derived macrophages and in the hepatic cell line Huh7. Statin treatment did not interfere with viral entry, but the viral particles released from treated cells showed reduced infectivity due to inhibition of viral glycoprotein processing, as evidenced by decreased ratios of the mature glycoprotein form to precursor form. Statin-induced inhibition of infectious virus production and glycoprotein processing was reversed by exogenous mevalonate, the rate-limiting product of the cholesterol biosynthesis pathway, but not by low-density lipoprotein. Finally, statin-treated cells produced EBOV particles devoid of the surface glycoproteins required for virus infectivity. Our findings demonstrate that statin treatment inhibits EBOV infection and suggest that the efficacy of statin treatment should be evaluated in appropriate animal models of EVD.IMPORTANCE Treatments targeting Ebola virus disease (EVD) are experimental, expensive, and scarce. Statins are inexpensive generic drugs that have been used for many years for the treatment of hypercholesterolemia and have a favorable safety profile. Here, we show the antiviral effects of statins on infectious Ebola virus (EBOV) production. Our study reveals a novel molecular mechanism in which statin regulates EBOV particle infectivity by preventing glycoprotein processing and incorporation into virus particles. Additionally, statins have anti-inflammatory and immunomodulatory effects. Since inflammation and dysregulation of the immune system are characteristic features of EVD, statins could be explored as part of EVD therapeutics.
Collapse
Affiliation(s)
- Punya Shrivastava-Ranjan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Mike Flint
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Éric Bergeron
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Anita K McElroy
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania, USA
| | - Payel Chatterjee
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - César G Albariño
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
38
|
Duy J, Honko AN, Altamura LA, Bixler SL, Wollen-Roberts S, Wauquier N, O'Hearn A, Mucker EM, Johnson JC, Shamblin JD, Zelko J, Botto MA, Bangura J, Coomber M, Pitt ML, Gonzalez JP, Schoepp RJ, Goff AJ, Minogue TD. Virus-encoded miRNAs in Ebola virus disease. Sci Rep 2018; 8:6480. [PMID: 29691416 PMCID: PMC5915558 DOI: 10.1038/s41598-018-23916-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/15/2018] [Indexed: 12/31/2022] Open
Abstract
Ebola virus (EBOV) is a negative-strand RNA virus that replicates in the cytoplasm and causes an often-fatal hemorrhagic fever. EBOV, like other viruses, can reportedly encode its own microRNAs (miRNAs) to subvert host immune defenses. miRNAs are short noncoding RNAs that can regulate gene expression by hybridizing to multiple mRNAs, and viral miRNAs can enhance viral replication and infectivity by regulating host or viral genes. To date, only one EBOV miRNA has been examined in human infection. Here, we assayed mouse, rhesus macaque, cynomolgus macaque, and human samples infected with three EBOV variants for twelve computationally predicted viral miRNAs using RT-qPCR. Ten miRNAs aligned to EBOV variants and were detectable in the four species during disease with several viral miRNAs showing presymptomatic amplification in animal models. miRNA abundances in both the mouse and nonhuman primate models mirrored the human cohort, with miR-1-5p, miR-1-3p, and miR-T3-3p consistently at the highest levels. These striking similarities in the most abundant miRNAs during infection with different EBOV variants and hosts indicate that these miRNAs are potential valuable diagnostic markers and key effectors of EBOV pathogenesis.
Collapse
Affiliation(s)
- Janice Duy
- Diagnostic Systems Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Anna N Honko
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Louis A Altamura
- Diagnostic Systems Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Sandra L Bixler
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Suzanne Wollen-Roberts
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Nadia Wauquier
- Metabiota, Kenema, Sierra Leone.,MRIGlobal - Global Health Surveillance and Diagnostics, Gaithersburg, MD, USA
| | - Aileen O'Hearn
- Diagnostic Systems Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Eric M Mucker
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Joshua C Johnson
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Joshua D Shamblin
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Justine Zelko
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Miriam A Botto
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | | | | | - M Louise Pitt
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Jean-Paul Gonzalez
- Metabiota, Washington, DC, USA.,Center of Excellence for Emerging & Zoonotic Animal Disease, Kansas State University, Manhattan, KS, USA
| | - Randal J Schoepp
- Diagnostic Systems Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Arthur J Goff
- Virology Division, U.S. Army Medical Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Timothy D Minogue
- Diagnostic Systems Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA.
| |
Collapse
|
39
|
Capuzzi SJ, Sun W, Muratov EN, Martínez-Romero C, He S, Zhu W, Li H, Tawa G, Fisher EG, Xu M, Shinn P, Qiu X, García-Sastre A, Zheng W, Tropsha A. Computer-Aided Discovery and Characterization of Novel Ebola Virus Inhibitors. J Med Chem 2018; 61:3582-3594. [PMID: 29624387 DOI: 10.1021/acs.jmedchem.8b00035] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The Ebola virus (EBOV) causes severe human infection that lacks effective treatment. A recent screen identified a series of compounds that block EBOV-like particle entry into human cells. Using data from this screen, quantitative structure-activity relationship models were built and employed for virtual screening of a ∼17 million compound library. Experimental testing of 102 hits yielded 14 compounds with IC50 values under 10 μM, including several sub-micromolar inhibitors, and more than 10-fold selectivity against host cytotoxicity. These confirmed hits include FDA-approved drugs and clinical candidates with non-antiviral indications, as well as compounds with novel scaffolds and no previously known bioactivity. Five selected hits inhibited BSL-4 live-EBOV infection in a dose-dependent manner, including vindesine (0.34 μM). Additional studies of these novel anti-EBOV compounds revealed their mechanisms of action, including the inhibition of NPC1 protein, cathepsin B/L, and lysosomal function. Compounds identified in this study are among the most potent and well-characterized anti-EBOV inhibitors reported to date.
Collapse
Affiliation(s)
- Stephen J Capuzzi
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry , UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Wei Sun
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry , UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States.,Department of Chemical Technology , Odessa National Polytechnic University , Odessa 65000 , Ukraine
| | - Carles Martínez-Romero
- Department of Microbiology , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Global Health and Emerging Pathogens Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Shihua He
- Special Pathogens Program, National Microbiology Laboratory , Public Health Agency of Canada , 1015 Arlington Street , Winnipeg , Manitoba R3E 3R2 , Canada
| | - Wenjun Zhu
- Special Pathogens Program, National Microbiology Laboratory , Public Health Agency of Canada , 1015 Arlington Street , Winnipeg , Manitoba R3E 3R2 , Canada.,Department of Medical Microbiology , University of Manitoba , 745 Bannatyne Avenue , Winnipeg , Manitoba R3E 0J9 , Canada
| | - Hao Li
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Gregory Tawa
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Ethan G Fisher
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Miao Xu
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Paul Shinn
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory , Public Health Agency of Canada , 1015 Arlington Street , Winnipeg , Manitoba R3E 3R2 , Canada.,Department of Medical Microbiology , University of Manitoba , 745 Bannatyne Avenue , Winnipeg , Manitoba R3E 0J9 , Canada
| | - Adolfo García-Sastre
- Department of Microbiology , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Global Health and Emerging Pathogens Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Department of Medicine, Division of Infectious Diseases , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Wei Zheng
- National Center for Advancing Translational Sciences , National Institutes of Health , Bethesda , Maryland 20892 , United States
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry , UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| |
Collapse
|
40
|
McElroy AK, Mühlberger E, Muñoz-Fontela C. Immune barriers of Ebola virus infection. Curr Opin Virol 2018; 28:152-160. [PMID: 29452995 PMCID: PMC5886007 DOI: 10.1016/j.coviro.2018.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/23/2018] [Accepted: 01/26/2018] [Indexed: 01/10/2023]
Abstract
Since its initial emergence in 1976 in northern Democratic Republic of Congo (DRC), Ebola virus (EBOV) has been a global health concern due to its virulence in humans, the mystery surrounding the identity of its host reservoir and the unpredictable nature of Ebola virus disease (EVD) outbreaks. Early after the first clinical descriptions of a disease resembling a 'septic-shock-like syndrome', with coagulation abnormalities and multi-system organ failure, researchers began to evaluate the role of the host immune response in EVD pathophysiology. In this review, we summarize how data gathered during the last 40 years in the laboratory as well as in the field have provided insight into EBOV immunity. From molecular mechanisms involved in EBOV recognition in infected cells, to antigen processing and adaptive immune responses, we discuss current knowledge on the main immune barriers of infection as well as outstanding research questions.
Collapse
Affiliation(s)
- Anita K McElroy
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, 3501 Fifth Ave, Pittsburgh, PA 15261, USA
| | - Elke Mühlberger
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, 02118 Boston, MA, USA
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Strasse 74, 20359 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg, Germany.
| |
Collapse
|
41
|
Honko AN, Johnson JC, Marchand JS, Huzella L, Adams RD, Oberlander N, Torzewski LM, Bennett RS, Hensley LE, Jahrling PB, Olinger GG. High dose sertraline monotherapy fails to protect rhesus macaques from lethal challenge with Ebola virus Makona. Sci Rep 2017; 7:5886. [PMID: 28725019 PMCID: PMC5517626 DOI: 10.1038/s41598-017-06179-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/07/2017] [Indexed: 11/09/2022] Open
Abstract
The recent epidemic of Ebola virus disease in West Africa resulted in an unprecedented number of cases and deaths. Due to the scope of the outbreak combined with the lack of available approved treatment options, there was strong motivation to investigate any potential drug which had existing data reporting anti-Ebola activity. Drugs with demonstrated antiviral activity in the nonhuman primate models already approved for another indication or for which there was existing safety data were considered to be priorities for evaluation by the World Health Organization. Sertraline hydrochloride was reported to have anti-Ebola activity in vitro alone and in combination with other approved drugs. Although the efficacy was less than 100% in the murine model, the established safety profile of this product, the potential benefit alone and in combination, as well as the lack of other available options prioritized this compound for testing in the Ebola virus intramuscular rhesus macaque challenge model. Using a blinded dosing strategy, we demonstrated that high dose sertraline monotherapy provided no benefit for the prevention of Ebola virus disease in rhesus macaques with regards to reduction of viral load, morbidity, or survival highlighting the challenges of translating results between in vitro and in vivo models.
Collapse
Affiliation(s)
- Anna N Honko
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, 21702, Maryland, USA.
| | - Joshua C Johnson
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, 21702, Maryland, USA
| | - Jonathan S Marchand
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, 21702, Maryland, USA
| | - Louis Huzella
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, 21702, Maryland, USA
| | - Ricky D Adams
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, 21702, Maryland, USA
| | - Nicholas Oberlander
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, 21702, Maryland, USA
- BD Technologies/Charles River Labs, Research Triangle Park, 27709, North Carolina, USA
| | - Lisa M Torzewski
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, 21702, Maryland, USA
| | - Richard S Bennett
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, 21702, Maryland, USA
| | - Lisa E Hensley
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, 21702, Maryland, USA
| | - Peter B Jahrling
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, 21702, Maryland, USA
| | - Gene G Olinger
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, 21702, Maryland, USA
- MRIGlobal-Global Health Surveillance and Diagnostics, Gaithersburg, Maryland, 20878, USA
| |
Collapse
|
42
|
Imipramine Inhibits Chikungunya Virus Replication in Human Skin Fibroblasts through Interference with Intracellular Cholesterol Trafficking. Sci Rep 2017; 7:3145. [PMID: 28600536 PMCID: PMC5466638 DOI: 10.1038/s41598-017-03316-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 04/26/2017] [Indexed: 02/06/2023] Open
Abstract
Chikungunya virus (CHIKV) is an emerging arbovirus of the Togaviridae family that poses a present worldwide threat to human in the absence of any licensed vaccine or antiviral treatment to control viral infection. Here, we show that compounds interfering with intracellular cholesterol transport have the capacity to inhibit CHIKV replication in human skin fibroblasts, a major viral entry site in the human host. Pretreatment of these cells with the class II cationic amphiphilic compound U18666A, or treatment with the FDA-approved antidepressant drug imipramine resulted in a near total inhibition of viral replication and production at the highest concentration used without any cytotoxic effects. Imipramine was found to affect both the fusion and replication steps of the viral life cycle. The key contribution of cholesterol availability to the CHIKV life cycle was validated further by the use of fibroblasts from Niemann-Pick type C (NPC) patients in which the virus was unable to replicate. Interestingly, imipramine also strongly inhibited the replication of several Flaviviridae family members, including Zika, West Nile and Dengue virus. Together, these data show that this compound is a potential drug candidate for anti-arboviral treatment.
Collapse
|
43
|
Nelson EA, Dyall J, Hoenen T, Barnes AB, Zhou H, Liang JY, Michelotti J, Dewey WH, DeWald LE, Bennett RS, Morris PJ, Guha R, Klumpp-Thomas C, McKnight C, Chen YC, Xu X, Wang A, Hughes E, Martin S, Thomas C, Jahrling PB, Hensley LE, Olinger GG, White JM. The phosphatidylinositol-3-phosphate 5-kinase inhibitor apilimod blocks filoviral entry and infection. PLoS Negl Trop Dis 2017; 11:e0005540. [PMID: 28403145 PMCID: PMC5402990 DOI: 10.1371/journal.pntd.0005540] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/24/2017] [Accepted: 03/30/2017] [Indexed: 12/12/2022] Open
Abstract
Phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) is a lipid kinase involved in endosome maturation that emerged from a haploid genetic screen as being required for Ebola virus (EBOV) infection. Here we analyzed the effects of apilimod, a PIKfyve inhibitor that was reported to be well tolerated in humans in phase 2 clinical trials, for its effects on entry and infection of EBOV and Marburg virus (MARV). We first found that apilimod blocks infections by EBOV and MARV in Huh 7, Vero E6 and primary human macrophage cells, with notable potency in the macrophages (IC50, 10 nM). We next observed that similar doses of apilimod block EBOV-glycoprotein-virus like particle (VLP) entry and transcription-replication competent VLP infection, suggesting that the primary mode of action of apilimod is as an entry inhibitor, preventing release of the viral genome into the cytoplasm to initiate replication. After providing evidence that the anti-EBOV action of apilimod is via PIKfyve, we showed that it blocks trafficking of EBOV VLPs to endolysosomes containing Niemann-Pick C1 (NPC1), the intracellular receptor for EBOV. Concurrently apilimod caused VLPs to accumulate in early endosome antigen 1-positive endosomes. We did not detect any effects of apilimod on bulk endosome acidification, on the activity of cathepsins B and L, or on cholesterol export from endolysosomes. Hence by antagonizing PIKfyve, apilimod appears to block EBOV trafficking to its site of fusion and entry into the cytoplasm. Given the drug's observed anti-filoviral activity, relatively unexplored mechanism of entry inhibition, and reported tolerability in humans, we propose that apilimod be further explored as part of a therapeutic regimen to treat filoviral infections.
Collapse
Affiliation(s)
- Elizabeth A. Nelson
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Julie Dyall
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Thomas Hoenen
- Laboratory of Virology, Division of Intramural Research, National Institutes of Health, Hamilton, Montana, United States of America
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald–Insel Riems, Germany
| | - Alyson B. Barnes
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Huanying Zhou
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Janie Y. Liang
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Julia Michelotti
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - William H. Dewey
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Lisa Evans DeWald
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Richard S. Bennett
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Patrick J. Morris
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Carleen Klumpp-Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Crystal McKnight
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yu-Chi Chen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Xin Xu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amy Wang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Emma Hughes
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Scott Martin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Craig Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Peter B. Jahrling
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Lisa E. Hensley
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Gene G. Olinger
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Judith M. White
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
44
|
Sweiti H, Ekwunife O, Jaschinski T, Lhachimi SK. Repurposed Therapeutic Agents Targeting the Ebola Virus: A Systematic Review. CURRENT THERAPEUTIC RESEARCH 2017; 84:10-21. [PMID: 28761574 PMCID: PMC5522984 DOI: 10.1016/j.curtheres.2017.01.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 01/30/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND The Ebola virus has been responsible for numerous outbreaks since the 1970s, with the most recent outbreak taking place between 2014 and 2016 and causing an international public health emergency. Ebola virus disease (EVD) has a high mortality rate and no approved targeted treatment exists to date. A number of established drugs are being considered as potential therapeutic agents for the treatment of EVD. OBJECTIVE We aimed to identify potential drug repositioning candidates and to assess the scientific evidence available on their efficacy. METHODS We conducted a systematic literature search in MEDLINE, Embase, and other relevant trial registry platforms for studies published between January 1976 and January 2017. We included drug screening, preclinical studies, and clinical studies on repurposed drugs for the treatment of EVD. The risk of bias for animal studies and nonrandomized clinical studies was assessed. The quality of reporting for case series and case reports was evaluated. Finally, we selected drugs approved by established regulatory authorities, which have positive in vitro study outcomes and at least one additional animal or clinical trial. RESULTS We identified 3301 publications, of which 37 studies fulfilled our inclusion criteria. Studies were highly heterogeneous in terms of study type, methodology, and intervention. The risk of bias was high for 13 out of 14 animal studies. We selected 11 drugs with potential anti-EVD therapeutic effects and summarized their evidence. CONCLUSIONS Several established drugs may have therapeutic effects on EVD, but the quality and quantity of current scientific evidence is lacking. This review highlights the need for well-designed and conducted preclinical and clinical research to establish the efficacy of potential repurposed drugs against EVD.
Collapse
Affiliation(s)
- Hussein Sweiti
- Institute of Health Services Research and Health Economics, School of Medicine, Heinrich-Heine University Dû¥sseldorf, Dû¥sseldorf, Germany
- Surgical Department, Klinikum Frankfurt HûÑchst, Frankfurt, Germany
| | - Obinna Ekwunife
- Cooperative Research Group for Evidence-Based Public Health, Department of Prevention and Evaluation, Leibniz Institute for Prevention Research and Epidemiology-BIPS, Bremen, Germany
- Department of Clinical Pharmacy and Pharmacy Management, Nnamdi Azikiwe University, Awka, Nigeria
| | - Thomas Jaschinski
- Department for Evidence-based Health Services Research, Institute for Research in Operative Medicine, Witten/Herdecke University, Witten, Germany
| | - Stefan K. Lhachimi
- Institute of Health Services Research and Health Economics, School of Medicine, Heinrich-Heine University Dû¥sseldorf, Dû¥sseldorf, Germany
- Cooperative Research Group for Evidence-Based Public Health, Department of Prevention and Evaluation, Leibniz Institute for Prevention Research and Epidemiology-BIPS, Bremen, Germany
- Institute for Public Health, Health Sciences Bremen, University of Bremen, Bremen, Germany
| |
Collapse
|
45
|
Davey RA, Shtanko O, Anantpadma M, Sakurai Y, Chandran K, Maury W. Mechanisms of Filovirus Entry. Curr Top Microbiol Immunol 2017; 411:323-352. [PMID: 28601947 DOI: 10.1007/82_2017_14] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Filovirus entry into cells is complex, perhaps as complex as any viral entry mechanism identified to date. However, over the past 10 years, the important events required for filoviruses to enter into the endosomal compartment and fuse with vesicular membranes have been elucidated (Fig. 1). Here, we highlight the important steps that are required for productive entry of filoviruses into mammalian cells.
Collapse
Affiliation(s)
- R A Davey
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - O Shtanko
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - M Anantpadma
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Y Sakurai
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - K Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - W Maury
- Department of Microbiology, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
46
|
Sun W, He S, Martínez-Romero C, Kouznetsova J, Tawa G, Xu M, Shinn P, Fisher E, Long Y, Motabar O, Yang S, Sanderson PE, Williamson PR, García-Sastre A, Qiu X, Zheng W. Synergistic drug combination effectively blocks Ebola virus infection. Antiviral Res 2017; 137:165-172. [PMID: 27890675 PMCID: PMC5182099 DOI: 10.1016/j.antiviral.2016.11.017] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/21/2016] [Accepted: 11/24/2016] [Indexed: 11/24/2022]
Abstract
Although a group of FDA-approved drugs were previously identified with activity against Ebola virus (EBOV), most of them are not clinically useful because their human blood concentrations are not high enough to inhibit EBOV infection. We screened 795 unique three-drug combinations in an EBOV entry assay. Two sets of three-drug combinations, toremifene-mefloquine-posaconazole and toremifene-clarithromycin-posaconazole, were identified that effectively blocked EBOV entry and were further validated for inhibition of live EBOV infection. The individual drug concentrations in the combinations were reduced to clinically relevant levels. We identified mechanisms of action of these drugs: functional inhibitions of Niemann-Pick C1, acid sphingomyelinase, and lysosomal calcium release. Our findings identify the drug combinations with potential to treat EBOV infection.
Collapse
Affiliation(s)
- Wei Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda MD 20892, USA
| | - Shihua He
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| | - Carles Martínez-Romero
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jennifer Kouznetsova
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda MD 20892, USA
| | - Gregory Tawa
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda MD 20892, USA
| | - Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda MD 20892, USA
| | - Paul Shinn
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda MD 20892, USA
| | - Ethan Fisher
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda MD 20892, USA
| | - Yan Long
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda MD 20892, USA
| | - Omid Motabar
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda MD 20892, USA
| | - Shu Yang
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda MD 20892, USA
| | - Philip E. Sanderson
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda MD 20892, USA
| | - Peter R. Williamson
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda MD 20892, USA
| |
Collapse
|
47
|
|
48
|
Wec AZ, Nyakatura EK, Herbert AS, Howell KA, Holtsberg FW, Bakken RR, Mittler E, Christin JR, Shulenin S, Jangra RK, Bharrhan S, Kuehne AI, Bornholdt ZA, Flyak AI, Saphire EO, Crowe JE, Aman MJ, Dye JM, Lai JR, Chandran K. A "Trojan horse" bispecific-antibody strategy for broad protection against ebolaviruses. Science 2016; 354:350-354. [PMID: 27608667 PMCID: PMC5647781 DOI: 10.1126/science.aag3267] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/25/2016] [Indexed: 12/26/2022]
Abstract
There is an urgent need for monoclonal antibody (mAb) therapies that broadly protect against Ebola virus and other filoviruses. The conserved, essential interaction between the filovirus glycoprotein, GP, and its entry receptor Niemann-Pick C1 (NPC1) provides an attractive target for such mAbs but is shielded by multiple mechanisms, including physical sequestration in late endosomes. Here, we describe a bispecific-antibody strategy to target this interaction, in which mAbs specific for NPC1 or the GP receptor-binding site are coupled to a mAb against a conserved, surface-exposed GP epitope. Bispecific antibodies, but not parent mAbs, neutralized all known ebolaviruses by coopting viral particles themselves for endosomal delivery and conferred postexposure protection against multiple ebolaviruses in mice. Such "Trojan horse" bispecific antibodies have potential as broad antifilovirus immunotherapeutics.
Collapse
Affiliation(s)
- Anna Z Wec
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Elisabeth K Nyakatura
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Andrew S Herbert
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Katie A Howell
- Integrated Biotherapeutics Inc., Gaithersburg, MD 20878, USA
| | | | - Russell R Bakken
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Eva Mittler
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John R Christin
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sergey Shulenin
- Integrated Biotherapeutics Inc., Gaithersburg, MD 20878, USA
| | - Rohit K Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sushma Bharrhan
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ana I Kuehne
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Zachary A Bornholdt
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 10550, USA
| | - Andrew I Flyak
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37235, USA
| | - Erica Ollmann Saphire
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 10550, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 10550, USA
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN 37235, USA.
- Department of Pediatrics, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University, Nashville, TN 37232, USA
| | - M Javad Aman
- Integrated Biotherapeutics Inc., Gaithersburg, MD 20878, USA.
| | - John M Dye
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
49
|
|
50
|
Nelson EA, Barnes AB, Wiehle RD, Fontenot GK, Hoenen T, White JM. Clomiphene and Its Isomers Block Ebola Virus Particle Entry and Infection with Similar Potency: Potential Therapeutic Implications. Viruses 2016; 8:v8080206. [PMID: 27490565 PMCID: PMC4997570 DOI: 10.3390/v8080206] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/08/2016] [Accepted: 07/19/2016] [Indexed: 01/08/2023] Open
Abstract
The 2014 outbreak of Ebola virus (EBOV) in Western Africa highlighted the need for anti-EBOV therapeutics. Clomiphene is a U.S. Food and Drug Administration (FDA)-approved drug that blocks EBOV entry and infection in cells and significantly protects EBOV-challenged mice. As provided, clomiphene is, approximately, a 60:40 mixture of two stereoisomers, enclomiphene and zuclomiphene. The pharmacokinetic properties of the two isomers vary, but both accumulate in the eye and male reproductive tract, tissues in which EBOV can persist. Here we compared the ability of clomiphene and its isomers to inhibit EBOV using viral-like particle (VLP) entry and transcription/replication-competent VLP (trVLP) assays. Clomiphene and its isomers inhibited the entry and infection of VLPs and trVLPs with similar potencies. This was demonstrated with VLPs bearing the glycoproteins from three filoviruses (EBOV Mayinga, EBOV Makona, and Marburg virus) and in two cell lines (293T/17 and Vero E6). Visual problems have been noted in EBOV survivors, and viral RNA has been isolated from semen up to nine months post-infection. Since the clomiphene isomers accumulate in these affected tissues, clomiphene or one of its isomers warrants consideration as an anti-EBOV agent, for example, to potentially help ameliorate symptoms in EBOV survivors.
Collapse
Affiliation(s)
- Elizabeth A Nelson
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA.
| | - Alyson B Barnes
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | - Thomas Hoenen
- Laboratory of Virology, Division of Intramural Research, National Institutes of Health, Hamilton, MT 59840, USA.
- Friedrich-Loeffler-Institut, D-17493, Greifswald-Insel Riems, Germany.
| | - Judith M White
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|