1
|
Prokop JW, Alberta S, Witteveen-Lane M, Pell S, Farag HA, Bhargava D, Vaughan RM, Frisch A, Bauss J, Bhatti H, Arora S, Subrahmanya C, Pearson D, Goodyke A, Westgate M, Cook TW, Mitchell JT, Zieba J, Sims MD, Underwood A, Hassouna H, Rajasekaran S, Tamae Kakazu MA, Chesla D, Olivero R, Caulfield AJ. SARS-CoV-2 Genotyping Highlights the Challenges in Spike Protein Drift Independent of Other Essential Proteins. Microorganisms 2024; 12:1863. [PMID: 39338537 PMCID: PMC11433680 DOI: 10.3390/microorganisms12091863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
As of 2024, SARS-CoV-2 continues to propagate and drift as an endemic virus, impacting healthcare for years. The largest sequencing initiative for any species was initiated to combat the virus, tracking changes over time at a full virus base-pair resolution. The SARS-CoV-2 sequencing represents a unique opportunity to understand selective pressures and viral evolution but requires cross-disciplinary approaches from epidemiology to functional protein biology. Within this work, we integrate a two-year genotyping window with structural biology to explore the selective pressures of SARS-CoV-2 on protein insights. Although genotype and the Spike (Surface Glycoprotein) protein continue to drift, most SARS-CoV-2 proteins have had few amino acid alterations. Within Spike, the high drift rate of amino acids involved in antibody evasion also corresponds to changes within the ACE2 binding pocket that have undergone multiple changes that maintain functional binding. The genotyping suggests selective pressure for receptor specificity that could also confer changes in viral risk. Mapping of amino acid changes to the structures of the SARS-CoV-2 co-transcriptional complex (nsp7-nsp14), nsp3 (papain-like protease), and nsp5 (cysteine protease) proteins suggest they remain critical factors for drug development that will be sustainable, unlike those strategies targeting Spike.
Collapse
Affiliation(s)
- Jeremy W. Prokop
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA; (M.W.-L.); (H.A.F.); (S.A.); (C.S.); (D.P.); (A.G.); (M.W.); (S.R.); (D.C.)
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
| | - Sheryl Alberta
- Advanced Technology Lab, Corewell Health, Grand Rapids, MI 49503, USA; (S.A.); (S.P.)
| | - Martin Witteveen-Lane
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA; (M.W.-L.); (H.A.F.); (S.A.); (C.S.); (D.P.); (A.G.); (M.W.); (S.R.); (D.C.)
| | - Samantha Pell
- Advanced Technology Lab, Corewell Health, Grand Rapids, MI 49503, USA; (S.A.); (S.P.)
| | - Hosam A. Farag
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA; (M.W.-L.); (H.A.F.); (S.A.); (C.S.); (D.P.); (A.G.); (M.W.); (S.R.); (D.C.)
| | - Disha Bhargava
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
| | - Robert M. Vaughan
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
| | - Austin Frisch
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
| | - Jacob Bauss
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
| | - Humza Bhatti
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
| | - Sanjana Arora
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA; (M.W.-L.); (H.A.F.); (S.A.); (C.S.); (D.P.); (A.G.); (M.W.); (S.R.); (D.C.)
| | - Charitha Subrahmanya
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA; (M.W.-L.); (H.A.F.); (S.A.); (C.S.); (D.P.); (A.G.); (M.W.); (S.R.); (D.C.)
| | - David Pearson
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA; (M.W.-L.); (H.A.F.); (S.A.); (C.S.); (D.P.); (A.G.); (M.W.); (S.R.); (D.C.)
| | - Austin Goodyke
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA; (M.W.-L.); (H.A.F.); (S.A.); (C.S.); (D.P.); (A.G.); (M.W.); (S.R.); (D.C.)
| | - Mason Westgate
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA; (M.W.-L.); (H.A.F.); (S.A.); (C.S.); (D.P.); (A.G.); (M.W.); (S.R.); (D.C.)
| | - Taylor W. Cook
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
| | - Jackson T. Mitchell
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
| | - Jacob Zieba
- Genetics and Genome Sciences Program, BioMolecular Science, Michigan State University, East Lansing, MI 48824, USA;
| | - Matthew D. Sims
- Section of Infectious Diseases, Corewell Health, Royal Oak, MI 48073, USA;
- Department of Internal Medicine, Oakland University William Beaumont School of Medicine, Auburn Hills, MI 48309, USA
| | - Adam Underwood
- Division of Mathematics and Science, Walsh University, North Canton, OH 44720, USA;
| | - Habiba Hassouna
- Adult Infectious Disease, Corewell Health, Grand Rapids, MI 49503, USA;
| | - Surender Rajasekaran
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA; (M.W.-L.); (H.A.F.); (S.A.); (C.S.); (D.P.); (A.G.); (M.W.); (S.R.); (D.C.)
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
| | - Maximiliano A. Tamae Kakazu
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
- Division of Pulmonary and Critical Care Medicine, Corewell Health, Grand Rapids, MI 49503, USA
| | - Dave Chesla
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA; (M.W.-L.); (H.A.F.); (S.A.); (C.S.); (D.P.); (A.G.); (M.W.); (S.R.); (D.C.)
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
| | - Rosemary Olivero
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (D.B.); (R.M.V.); (A.F.); (J.B.); (H.B.); (T.W.C.); (J.T.M.); (M.A.T.K.); (R.O.)
- Pediatric Infectious Disease, Helen DeVos Children’s Hospital, Corewell Health, Grand Rapids, MI 49503, USA
| | | |
Collapse
|
2
|
Mousavi MJ, Arefinia N, Azarsa M, Hoseinnezhad T, Behboudi E. MicroRNA profiles in Zika virus infection: Insights from diverse sources. Indian J Med Microbiol 2024; 51:100697. [PMID: 39103054 DOI: 10.1016/j.ijmmb.2024.100697] [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: 11/08/2023] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Zika virus (ZIKV) stands as one of the most significant reemerging viral pathogens, linked to neurological diseases such as meningoencephalitis and congenital microcephaly. Today there are no effective therapies for treating ZIKV-infected patients. MiRNAs play a critical role in regulating cellular signaling and physiological conditions, and alterations in their profiles can bear great significance in disease progression. OBJECTIVES Despite significant progress in understanding the interaction between the ZIKV and its host since the outbreak, a more comprehensive understanding on these interactions is imperative. This review aims to summarize the studies in the field and shed light on the intricate relationship between ZIKV and its host at the molecular level. CONTENT We found that in ZIKV-infected humans, over-expression of miR-431-5p and miR-30e-5p plays a crucial role in innate immune responses and contributes to neurological damage. Additionally, in ZIKA-infected mice, we observed upregulated expression of all the targets of miR-124-3p including CCL2, IL7, IRF1, and SBNO2. Notably, other targets of this miRNA include TLR6, TNF, STAT3, and NF-kB also exhibited upregulation in the central nervous system (CNS) of infected mice. Conversely, miR-654-3p levels were reduced, correlating with the upregulation of its predicted targets including FLT3LG, LITAF, CD69, and TLR2. In the case of insects, aae-miR-286a/b-3p was predicted to target all ZIKV genotypes. This specific miRNA is typically found in ovaries and can be transferred to embryos. In conclusion, our findings suggest that host microRNAs and ZIKV-encoded microRNAs hold promise as potential targets for the diagnosis of ZIKV infections and may even serve as a therapeutic approach for managing this infectious disease.
Collapse
Affiliation(s)
- Mohammad Javad Mousavi
- Department of Hematology, School of Para-Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Nasir Arefinia
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Mohammad Azarsa
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Taraneh Hoseinnezhad
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Emad Behboudi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran.
| |
Collapse
|
3
|
Ezzemani W, Altawalah H, Windisch M, Ouladlahsen A, Saile R, Kettani A, Ezzikouri S. Identification of Zika virus NS2B-NS3 protease and NS5 polymerase inhibitors by structure-based virtual screening of FDA-approved drugs. J Biomol Struct Dyn 2024; 42:8073-8088. [PMID: 37528667 DOI: 10.1080/07391102.2023.2242963] [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: 03/01/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023]
Abstract
Zika virus (ZIKV) is a mosquito-borne human flavivirus responsible that causing emergency outbreaks in Brazil. ZIKV is suspected of causing Guillain-Barre syndrome in adults and microcephaly. The NS2B-NS3 protease and NS5 RNA-dependent RNA polymerase (RdRp), central to ZIKV multiplication, have been identified as attractive molecular targets for drugs. We performed a structure-based virtual screening of 2,659 FDA-approved small molecule drugs in the DrugBank database using AutoDock Vina in PyRx v0.8. Accordingly, 15 potential drugs were selected as ZIKV inhibitors because of their high values (binding affinity - binding energy) and we analyzed the molecular interactions between the active site amino acids and the compounds. Among these drugs, tamsulosin was found to interact most efficiently with NS2B/NS3 protease, as indicated by the lowest binding energy value (-8.27 kJ/mol), the highest binding affinity (-5.7 Kcal/mol), and formed H-bonds with amino acid residues TYRB130, SERB135, TYRB150. Furthermore, biotin was found to interact most efficiently with NS5 RdRp with a binding energy of -150.624 kJ/mol, a binding affinity of -5.6 Kcal/mol, and formed H-bonds with the amino acid residues ASPA665 and ASPA540. In vitro, in vivo, and clinical studies are needed to demonstrate anti-ZIKV safety and the efficacy of these FDA-approved drug candidates.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Wahiba Ezzemani
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
- Laboratoire de Biologie et Santé (URAC34), Départment de Biologie, Faculté des Sciences Ben Msik, Hassan II University of Casablanca, Morocco
| | - Haya Altawalah
- Department of Microbiology, Faculty of Medicine, Kuwait University, Kuwait
- Virology Unit, Yacoub Behbehani Center, Sabah Hospital, Ministry of Health, Kuwait
| | - Marc Windisch
- Applied Molecular Virology Laboratory, Discovery Biology Department, Institut Pasteur Korea, Gyeonggi-do, South Korea
| | - Ahd Ouladlahsen
- Faculté de médecine et de pharmacie, Université Hassan II, Casablanca, Morocco
- Service des maladies Infectieuses, CHU Ibn Rochd, Casablanca, Morocco
| | - Rachid Saile
- Laboratoire de Biologie et Santé (URAC34), Départment de Biologie, Faculté des Sciences Ben Msik, Hassan II University of Casablanca, Morocco
| | - Anass Kettani
- Laboratoire de Biologie et Santé (URAC34), Départment de Biologie, Faculté des Sciences Ben Msik, Hassan II University of Casablanca, Morocco
| | - Sayeh Ezzikouri
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
4
|
Roy A, Liu Q, Yang Y, Debnath AK, Du L. Envelope Protein-Targeting Zika Virus Entry Inhibitors. Int J Mol Sci 2024; 25:9424. [PMID: 39273370 PMCID: PMC11394925 DOI: 10.3390/ijms25179424] [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: 07/01/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Zika virus (ZIKV; family, Flaviviridae), which causes congenital Zika syndrome, Guillain-Barré Syndrome, and other severe diseases, is transmitted mainly by mosquitoes; however, the virus can be transmitted through other routes. Among the three structural and seven nonstructural proteins, the surface envelope (E) protein of ZIKV plays a critical role in viral entry and pathogenesis, making it a key target for the development of effective entry inhibitors. This review article describes the life cycle, genome, and encoded proteins of ZIKV, illustrates the structure and function of the ZIKV E protein, summarizes E protein-targeting entry inhibitors (with a focus on those based on natural products and small molecules), and highlights challenges that may potentially hinder the development of effective inhibitors of ZIKV infection. Overall, the article will provide useful guidance for further development of safe and potent ZIKV entry inhibitors targeting the viral E protein.
Collapse
Affiliation(s)
- Abhijeet Roy
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Qian Liu
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Yang Yang
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| | - Asim K. Debnath
- Lindsey F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Lanying Du
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
5
|
Gupta Y, Baranwal M, Chudasama B. Immunoinformatics-Based Identification of the Conserved Immunogenic Peptides Targeting of Zika Virus Precursor Membrane Protein. Viral Immunol 2023; 36:503-519. [PMID: 37486711 DOI: 10.1089/vim.2023.0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023] Open
Abstract
Zika virus infections lead to neurological complications such as congenital Zika syndrome and Guillain-Barré syndrome. Rising Zika infections in newborns and adults have triggered the need for vaccine development. In the current study, the precursor membrane (prM) protein of the Zika virus is explored for its functional importance and design of epitopes enriched conserved peptides with the usage of different immunoinformatics approach. Phylogenetic and mutational analyses inferred that the prM protein is highly conserved. Three conserved peptides containing multiple T and B cell epitopes were designed by employing different epitope prediction algorithms. IEDB population coverage analysis of selected peptides in six different continents has shown the population coverage of 60-99.8% (class I HLA) and 80-100% (class II HLA). Molecular docking of selected peptides/epitopes was carried out with each of class I and II HLA alleles using HADDOCK. A majority of peptide-HLA complex (pHLA) have HADDOCK scores found to be comparable and more than native-HLA complex representing the good binding interaction of peptides to HLA. Molecular dynamics simulation with best docked pHLA complexes revealed that pHLA complexes are stable with RMSD <5.5Å. Current work highlights the importance of prM as a strong antigenic protein and selected peptides have the potential to elicit humoral and cell-mediated immune responses.
Collapse
Affiliation(s)
- Yogita Gupta
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Bhupendra Chudasama
- School of Physics & Materials Science, Thapar Institute of Engineering and Technology, Patiala, India
| |
Collapse
|
6
|
Zhou P, Ma B, Gao Y, Xu Y, Li Z, Jin H, Luo R. Epidemiology, genetic diversity, and evolutionary dynamics of Tembusu virus. Arch Virol 2023; 168:262. [PMID: 37773423 DOI: 10.1007/s00705-023-05885-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 08/02/2023] [Indexed: 10/01/2023]
Abstract
Tembusu virus (TMUV) is an emerging pathogenic flavivirus associated with acute egg-drop and fatal encephalitis in domestic waterfowl. Since its initial identification in mosquitoes in 1955, TMUV has been confirmed to infect ducks, pigeons, sparrows, geese, and chickens, posing a significant threat to the poultry industry. Here, we sequenced two DTMUV strains isolated in 2019 and systematically investigated the possible origin, genetic relationships, evolutionary dynamics, and transmission patterns of TMUV based on complete virus genome sequences in the public database. We found that TMUV can be divided into four major clusters: TMUV, cluster 1, cluster 2, and cluster 3. Interestingly, we found that cluster 2.2 (within cluster 2) is the most commonly involved in interspecies transmission events, and subcluster 2.1.2 (within cluster 2.1) is currently the most prevalent cluster circulating in Asia. Notably, we also identified three positively selected sites in the E and NS1 proteins, which may be involved in virus replication, immune evasion, and host adaptation. Finally, phylogeographic analysis revealed that cluster dispersal originated in Southeast Asia and that short-distance transmission events have occurred frequently. Altogether, these data provide novel insights into the evolution and dispersal of TMUV, facilitating the development of rapid diagnostics, vaccines, and therapeutics against TMUV infection.
Collapse
Affiliation(s)
- Peng Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Road, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
| | - Bin Ma
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Road, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
| | - Yuan Gao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Road, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
| | - Yumin Xu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Road, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
| | - Zhuofei Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Road, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Road, Wuhan, 430070, Hubei, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, No.1 Shizishan Road, Wuhan, 430070, Hubei, China.
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China.
| |
Collapse
|
7
|
Structure-based design of a novel inhibitor of the ZIKA virus NS2B/NS3 protease. Bioorg Chem 2022; 128:106109. [DOI: 10.1016/j.bioorg.2022.106109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/02/2022] [Accepted: 08/19/2022] [Indexed: 11/24/2022]
|
8
|
Zika Virus Infection and Development of Drug Therapeutics. Appl Microbiol 2022. [DOI: 10.3390/applmicrobiol2040059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Zika virus (ZIKV) is an emerging flavivirus that is associated with neurological complications, such as neuroinflammatory Guillain Barré Syndrome in adults and microcephaly in newborns, and remains a potentially significant and international public health concern. The World Health Organization is urging the development of novel antiviral therapeutic strategies against ZIKV, as there are no clinically approved vaccines or drugs against this virus. Given the public health crisis that is related to ZIKV cases in the last decade, efficient strategies should be identified rapidly to combat or treat ZIKV infection. Several promising strategies have been reported through drug repurposing studies, de novo design, and the high-throughput screening of compound libraries in only a few years. This review summarizes the genome and structure of ZIKV, viral life cycle, transmission cycle, clinical manifestations, cellular and animal models, and antiviral drug developments, with the goal of increasing our understanding of ZIKV and ultimately defeating it.
Collapse
|
9
|
Chakrabartty I, Khan M, Mahanta S, Chopra H, Dhawan M, Choudhary OP, Bibi S, Mohanta YK, Emran TB. Comparative overview of emerging RNA viruses: Epidemiology, pathogenesis, diagnosis and current treatment. Ann Med Surg (Lond) 2022; 79:103985. [PMID: 35721786 PMCID: PMC9188442 DOI: 10.1016/j.amsu.2022.103985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023] Open
Abstract
From many decades, emerging infections have threatened humanity. The pandemics caused by different CoVs have already claimed and will continue to claim millions of lives. The SARS, Ebola, MERS epidemics and the most recent emergence of COVID-19 pandemic have threatened populations across borders. Since a highly pathogenic CoV has been evolved into the human population in the twenty-first century known as SARS, scientific advancements and innovative methods to tackle these viruses have increased in order to improve response preparedness towards the unpredictable threat posed by these rapidly emerging pathogens. Recently published review articles on SARS-CoV-2 have mainly focused on its pathogenesis, epidemiology and available treatments. However, in this review, we have done a systematic comparison of all three CoVs i.e., SARS, MERS and SARS-CoV-2 along with Ebola and Zika in terms of their epidemiology, virology, clinical features and current treatment strategies. This review focuses on important emerging RNA viruses starting from Zika, Ebola and the CoVs which include SARS, MERS and SARS-CoV-2. Each of these viruses has been elaborated on the basis of their epidemiology, virulence, transmission and treatment. However, special attention has been given to SARS-CoV-2 and the disease caused by it i.e., COVID-19 due to current havoc caused worldwide. At the end, insights into the current understanding of the lessons learned from previous epidemics to combat emerging CoVs have been described. The travel-related viral spread, the unprecedented nosocomial outbreaks and the high case-fatality rates associated with these highly transmissible and pathogenic viruses highlight the need for new prophylactic and therapeutic actions which include but are not limited to clinical indicators, contact tracing, and laboratory investigations as important factors that need to be taken into account in order to arrive at the final conclusion. Recently published review articles on SARS-CoV-2 have mainly focused on its pathogenesis, epidemiology and available treatments. The pandemics caused by different CoVs have already claimed and will continue to claim millions of lives. This review focuses on important emerging RNA viruses starting from Zika, Ebola and the CoVs which include SARS, MERS and SARS-CoV-2. Globally, numerous studies and researchers have recently started fighting this virus.
Collapse
Affiliation(s)
- Ishani Chakrabartty
- Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya (USTM), 9th Mile, Techno City, Baridua, Ri-Bhoi 793101, Meghalaya, India
| | - Maryam Khan
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, U.P, India
| | - Saurov Mahanta
- National Institute of Electronics and Information Technology (NIELIT), Guwahati Centre Guwahati, 781008, Assam, India
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, 141004, Punjab, India.,Trafford College, Altrincham, Manchester, WA14 5PQ, UK
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy and Histology, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Selesih, Aizawl, India
| | - Shabana Bibi
- Department of Biosciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan.,Yunnan Herbal Laboratory, College of Ecology and Environmental Sciences, Yunnan University, Kunming, 650091, China
| | - Yugal Kishore Mohanta
- Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya (USTM), 9th Mile, Techno City, Baridua, Ri-Bhoi 793101, Meghalaya, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh.,Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| |
Collapse
|
10
|
Naqvi AAT, Anjum F, Shafie A, Badar S, Elasbali AM, Yadav DK, Hassan MI. Investigating host-virus interaction mechanism and phylogenetic analysis of viral proteins involved in the pathogenesis. PLoS One 2021; 16:e0261497. [PMID: 34914801 PMCID: PMC8675761 DOI: 10.1371/journal.pone.0261497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023] Open
Abstract
Since the emergence of yellow fever in the Americas and the devastating 1918 influenza pandemic, biologists and clinicians have been drawn to human infecting viruses to understand their mechanisms of infection better and develop effective therapeutics against them. However, the complex molecular and cellular processes that these viruses use to infect and multiply in human cells have been a source of great concern for the scientific community since the discovery of the first human infecting virus. Viral disease outbreaks, such as the recent COVID-19 pandemic caused by a novel coronavirus, have claimed millions of lives and caused significant economic damage worldwide. In this study, we investigated the mechanisms of host-virus interaction and the molecular machinery involved in the pathogenesis of some common human viruses. We also performed a phylogenetic analysis of viral proteins involved in host-virus interaction to understand the changes in the sequence organization of these proteins during evolution for various strains of viruses to gain insights into the viral origin's evolutionary perspectives.
Collapse
Affiliation(s)
| | - Farah Anjum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Sufian Badar
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Abdelbaset Mohamed Elasbali
- Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Sakakah, Saudi Arabia
| | - Dharmendra Kumar Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, Yeonsu-gu, Incheon City, South Korea
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| |
Collapse
|
11
|
Cavazzoni CB, Bozza VB, Lucas TC, Conde L, Maia B, Mesin L, Schiepers A, Ersching J, Neris RL, Conde JN, Coelho DR, Lima TM, Alvim RG, Castilho LR, de Paula Neto HA, Mohana-Borges R, Assunção-Miranda I, Nobrega A, Victora GD, Vale AM. The immunodominant antibody response to Zika virus NS1 protein is characterized by cross-reactivity to self. J Exp Med 2021; 218:e20210580. [PMID: 34292314 PMCID: PMC8302445 DOI: 10.1084/jem.20210580] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/24/2021] [Accepted: 06/30/2021] [Indexed: 12/20/2022] Open
Abstract
Besides antigen-specific responses to viral antigens, humoral immune response in virus infection can generate polyreactive and autoreactive antibodies. Dengue and Zika virus infections have been linked to antibody-mediated autoimmune disorders, including Guillain-Barré syndrome. A unique feature of flaviviruses is the secretion of nonstructural protein 1 (NS1) by infected cells. NS1 is highly immunogenic, and antibodies targeting NS1 can have both protective and pathogenic roles. In the present study, we investigated the humoral immune response to Zika virus NS1 and found NS1 to be an immunodominant viral antigen associated with the presence of autoreactive antibodies. Through single B cell cultures, we coupled binding assays and BCR sequencing, confirming the immunodominance of NS1. We demonstrate the presence of self-reactive clones in germinal centers after both infection and immunization, some of which present cross-reactivity with NS1. Sequence analysis of anti-NS1 B cell clones showed sequence features associated with pathogenic autoreactive antibodies. Our findings demonstrate NS1 immunodominance at the cellular level as well as a potential role for NS1 in ZIKV-associated autoimmune manifestations.
Collapse
Affiliation(s)
- Cecilia B. Cavazzoni
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Vicente B.T. Bozza
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tostes C.V. Lucas
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Conde
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Maia
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Ariën Schiepers
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Jonatan Ersching
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Romulo L.S. Neris
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jonas N. Conde
- Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diego R. Coelho
- Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tulio M. Lima
- Programa de Engenharia Química, Laboratório de Engenharia de Cultivos Celulares, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata G.F. Alvim
- Programa de Engenharia Química, Laboratório de Engenharia de Cultivos Celulares, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leda R. Castilho
- Programa de Engenharia Química, Laboratório de Engenharia de Cultivos Celulares, Instituto Alberto Luiz Coimbra de Pós-Graduação e Pesquisa de Engenharia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heitor A. de Paula Neto
- Laboratório de Alvos Moleculares, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ronaldo Mohana-Borges
- Laboratório de Genômica Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Iranaia Assunção-Miranda
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alberto Nobrega
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriel D. Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY
| | - Andre M. Vale
- Laboratório de Biologia de Linfócitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Rother M, Naumann M. Signal peptidase complex subunit 1 is an essential Zika virus host factor in placental trophoblasts. Virus Res 2021; 296:198338. [PMID: 33577859 DOI: 10.1016/j.virusres.2021.198338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 01/12/2021] [Accepted: 02/02/2021] [Indexed: 10/22/2022]
Abstract
Zika is a major teratogenic virus that can be transmitted from pregnant women to the fetus via the transplacental route. At present, no specific vaccines or treatments are available. Large-scale functional genomics approaches for the analysis of host cell function in infection greatly improve the understanding of molecular infection processes and advance the discovery of antiviral targets. We conducted a pooled CRISPR/Cas9 screen to explore trophoblast function upon Zika infection. The identified Zika virus host factors enrich in the ER membrane complex and the signal peptide processing pathway. Finally, we demonstrate that signal peptidase complex subunit 1 (SPCS1) is crucial for virus replication in trophoblasts.
Collapse
Affiliation(s)
- Marion Rother
- Max Planck Institute for Infection Biology, Department of Molecular Biology, 10117, Berlin, Germany; Institute of Experimental Internal Medicine, Otto Von Guericke University Magdeburg, 39120, Magdeburg, Germany.
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto Von Guericke University Magdeburg, 39120, Magdeburg, Germany
| |
Collapse
|
13
|
Paul D, Sharif IH, Sayem A, Ahmed H, Saleh MA, Mahmud S. In silico prediction of a highly immunogenic and conserved epitope against Zika Virus. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
14
|
Hossein F. An overview of the current medical literature on Zika virus. Biophys Rev 2020; 12:1133-1138. [PMID: 32880054 DOI: 10.1007/s12551-020-00748-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/28/2020] [Indexed: 01/07/2023] Open
Abstract
Zika virus is a member of the family of Flaviviridae, which is primarily spread to humans by mosquito bites. It has been linked to microcephaly in neonates, and as such, it poses a significant risk to human pregnancy. Zika virus infection is also implicated in other severe neurological disorders such as Guillain-Barre syndrome. There is currently no vaccine available to treat Zika virus disease, and as such, it represents a serious challenge to public health. Antigenic similarities between Zika and dengue can suggest artificially high infection rates of Zika within specific population groups. Here, we review recent literature and provide an update on the status of the Zika outbreak, including a description of available medical countermeasure options and current diagnosis methodology.
Collapse
Affiliation(s)
- Fria Hossein
- School of Engineering, Nanotechnology and Integrated Bio-Engineering Centre (NIBEC), Ulster University, Shore Road, BT37 0QB, Newtownabbey, United Kingdom.
| |
Collapse
|
15
|
Haddad JG, Grauzdytė D, Koishi AC, Viranaicken W, Venskutonis PR, Nunes Duarte dos Santos C, Desprès P, Diotel N, El Kalamouni C. The Geraniin-Rich Extract from Reunion Island Endemic Medicinal Plant Phyllanthus phillyreifolius Inhibits Zika and Dengue Virus Infection at Non-Toxic Effect Doses in Zebrafish. Molecules 2020; 25:molecules25102316. [PMID: 32429073 PMCID: PMC7287739 DOI: 10.3390/molecules25102316] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
The mosquito-borne viruses dengue (DENV) and Zika (ZIKV) viruses are two medically important pathogens in tropical and subtropical regions of the world. There is an urgent need of therapeutics against DENV and ZIKV, and medicinal plants are considered as a promising source of antiviral bioactive metabolites. In the present study, we evaluated the ability of Phyllanthus phillyreifolius, an endemic medicinal plant from Reunion Island, to prevent DENV and ZIKV infection in human cells. At non-cytotoxic concentration in vitro, incubation of infected A549 cells with a P. phillyreifolius extract or its major active phytochemical geraniin resulted in a dramatic reduction of virus progeny production for ZIKV as well as four serotypes of DENV. Virological assays showed that P. phillyreifolius extract-mediated virus inhibition relates to a blockade in internalization of virus particles into the host cell. Infectivity studies on ZIKV showed that both P. phillyreifolius and geraniin cause a loss of infectivity of the viral particles. Using a zebrafish model, we demonstrated that administration of P. phillyreifolius and geraniin has no effect on zebrafish locomotor activity while no morbidity nor mortality was observed up to 5 days post-inoculation. Thus, P. phillyreifolius could act as an important source of plant metabolite geraniin which is a promising antiviral compound in the fight against DENV and ZIKV.
Collapse
Affiliation(s)
- Juliano G. Haddad
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (J.G.H.); (W.V.); (P.D.)
| | - Dovilė Grauzdytė
- Department of Food Science and Technology, Kaunas University of Technology, Radvilėnų pl. 19, Kaunas LT-50254, Lithuania; (D.G.); (P.R.V.)
| | - Andrea Cristine Koishi
- Laboratorio de Virologia Molecular, Instituto Carlos Chagas, ICC/FIOCRUZ/PR, Curitiba 81350-010, Brazil; (A.C.K.); (C.N.D.d.S.)
| | - Wildriss Viranaicken
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (J.G.H.); (W.V.); (P.D.)
| | - Petras Rimantas Venskutonis
- Department of Food Science and Technology, Kaunas University of Technology, Radvilėnų pl. 19, Kaunas LT-50254, Lithuania; (D.G.); (P.R.V.)
| | - Claudia Nunes Duarte dos Santos
- Laboratorio de Virologia Molecular, Instituto Carlos Chagas, ICC/FIOCRUZ/PR, Curitiba 81350-010, Brazil; (A.C.K.); (C.N.D.d.S.)
| | - Philippe Desprès
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (J.G.H.); (W.V.); (P.D.)
| | - Nicolas Diotel
- Université de La Réunion, INSERM, UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), 97490 Saint-Denis de La Réunion, France;
| | - Chaker El Kalamouni
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, France; (J.G.H.); (W.V.); (P.D.)
- Correspondence: ; Tel.: +33-262-938822
| |
Collapse
|
16
|
Hussain W, Rasool N, Khan YD. A Sequence-Based Predictor of Zika Virus Proteins Developed by Integration of PseAAC and Statistical Moments. Comb Chem High Throughput Screen 2020; 23:797-804. [PMID: 32342804 DOI: 10.2174/1386207323666200428115449] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND ZIKV has been a well-known global threat, which hits almost all of the American countries and posed a serious threat to the entire globe in 2016. The first outbreak of ZIKV was reported in 2007 in the Pacific area, followed by another severe outbreak, which occurred in 2013/2014 and subsequently, ZIKV spread to all other Pacific islands. A broad spectrum of ZIKV associated neurological malformations in neonates and adults has driven this deadly virus into the limelight. Though tremendous efforts have been focused on understanding the molecular basis of ZIKV, the viral proteins of ZIKV have still not been studied extensively. OBJECTIVES Herein, we report the first and the novel predictor for the identification of ZIKV proteins. METHODS We have employed Chou's pseudo amino acid composition (PseAAC), statistical moments and various position-based features. RESULTS The predictor is validated through 10-fold cross-validation and Jackknife testing. In 10- fold cross-validation, 94.09% accuracy, 93.48% specificity, 94.20% sensitivity and 0.80 MCC were achieved while in Jackknife testing, 96.62% accuracy, 94.57% specificity, 97.00% sensitivity and 0.88 MCC were achieved. CONCLUSION Thus, ZIKVPred-PseAAC can help in predicting the ZIKV proteins efficiently and accurately and can provide baseline data for the discovery of new drugs and biomarkers against ZIKV.
Collapse
Affiliation(s)
- Waqar Hussain
- National Center of Artificial Intelligence, Punjab University College of Information Technology, University of the
Punjab, Lahore, Pakistan,Center for Professional Studies, Lahore, Pakistan
| | | | - Yaser D Khan
- Department of Computer Science, University of Management and Technology, Lahore, Pakistan
| |
Collapse
|
17
|
Davidson RB, Hendrix J, Geiss BJ, McCullagh M. RNA-Dependent Structures of the RNA-Binding Loop in the Flavivirus NS3 Helicase. J Phys Chem B 2020; 124:2371-2381. [PMID: 32105483 DOI: 10.1021/acs.jpcb.0c00457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The flavivirus NS3 protein is a helicase that has pivotal functions during the viral genome replication process, where it unwinds double-stranded RNA and translocates along the nucleic acid polymer in a nucleoside triphosphate hydrolysis-dependent mechanism. Crystallographic and computational studies of the flavivirus NS3 helicase have identified the RNA-binding loop as an interesting structural element that may function as a component of the RNA-enhanced NTPase activity observed for this family of helicases. Microsecond-long unbiased molecular dynamics and extensive replica exchange umbrella sampling simulations of the Zika NS3 helicase have been performed to investigate the RNA dependence of this loop's structural conformations. Specifically, the effect of the bound single-stranded RNA (ssRNA) oligomer on the putative "open" and "closed" conformations of this loop is studied. In the Apo substrate state, the two loop conformations are nearly isoergonic (ΔAO→C = -0.22 kcal mol-1), explaining the structural ambiguity observed in Apo NS3h crystal structures. The bound ssRNA is seen to stabilize the "open" conformation (ΔAO→C = 1.97 kcal mol-1) through direct protein-RNA interactions at the top of the loop. Interestingly, a small ssRNA oligomer bound over 13 Å away from the loop is seen to affect the free energy surface to favor the "open" structure, while minimizing barriers between the two states. Both the mechanism of the "open" to "closed" transition and important residues of the RNA-binding loop structures are characterized. From these results, point mutations that are hypothesized to stabilize the "closed" RNA-binding loop and negatively impact RNA-binding and the RNA-enhanced NTPase activity are posited.
Collapse
Affiliation(s)
- Russell B Davidson
- Department of Chemistry, Colorado State University, Fort Collins 80523, Colorado, United States
| | - Josie Hendrix
- Department of Chemistry, Colorado State University, Fort Collins 80523, Colorado, United States
| | - Brian J Geiss
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins 80523, Colorado, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins 80523, Colorado, United States
| | - Martin McCullagh
- Department of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74074, United States
| |
Collapse
|
18
|
Abstract
Zika virus (ZIKV) belongs to the Flavivirus genus of the Flaviviridae family. It is an arbovirus that can cause congenital abnormalities and is sexually transmissible. A series of outbreaks accompanied by unexpected severe clinical complications have captured medical attention to further characterize the clinical features of congenital ZIKV syndrome and its underlying pathophysiological mechanisms. Endoplasmic reticulum (ER) and ER-related proteins are essential in ZIKV genome replication. This review highlights the subcellular localization of ZIKV to the ER and ZIKV modulation on the architecture of the ER. This review also discusses ZIKV interaction with ER proteins such as signal peptidase complex subunit 1 (SPCS1), ER membrane complex (EMC) subunits, and ER translocon for viral replication. Furthermore, the review covers several important resulting effects of ZIKV infection to the ER and cellular processes including ER stress, reticulophagy, and paraptosis-like death. Pharmacological targeting of ZIKV-affected ER-resident proteins and ER-associated components demonstrate promising signs of combating ZIKV infection and rescuing host organisms from severe neurologic sequelae.
Collapse
|
19
|
Sharma V, Sharma M, Dhull D, Sharma Y, Kaushik S, Kaushik S. Zika virus: an emerging challenge to public health worldwide. Can J Microbiol 2019; 66:87-98. [PMID: 31682478 DOI: 10.1139/cjm-2019-0331] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Zika virus (ZIKV) is a mosquito-borne virus that was first isolated from Zika forest, Uganda, in 1947. Since its inception, major and minor outbreaks have been documented from several parts of world. Aedes spp. mosquitoes are the primary vectors of ZIKV, but the virus can also be transmitted through sexual practices, materno-fetal transmission, and blood transfusion. The clinical presentations of symptomatic ZIKV infections are similar to dengue and chikungunya, including fever, headache, arthralgia, retro-orbital pain, conjunctivitis, and rash. ZIKV often causes mild illness in the majority of cases, but in some instances, it is linked with congenital microcephaly and autoimmune disorders like Guillain-Barré syndrome. The recent Indian ZIKV outbreak suggests that the virus is circulating in the South East Asian region and may cause new outbreaks in future. At present, no specific vaccines or antivirals are available to treat ZIKV, so management and control of ZIKV infections rely mostly on preventive measures.
Collapse
Affiliation(s)
- Vikrant Sharma
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Manisha Sharma
- Department of Biotechnology, Himachal Pradesh University, Shimla-171005, Himachal Pradesh, India
| | - Divya Dhull
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Yashika Sharma
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Sulochana Kaushik
- Department of Genetics, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Samander Kaushik
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| |
Collapse
|
20
|
Liu X, Zhao Y, Zhang JZ. Molecular mechanism of ligand bindings to Zika virus at SAM site. Chem Phys Lett 2019. [DOI: 10.1016/j.cplett.2019.136771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
21
|
Zandi K, Bassit L, Amblard F, Cox BD, Hassandarvish P, Moghaddam E, Yueh A, Libanio Rodrigues GO, Passos I, Costa VV, AbuBakar S, Zhou L, Kohler J, Teixeira MM, Schinazi RF. Nucleoside Analogs with Selective Antiviral Activity against Dengue Fever and Japanese Encephalitis Viruses. Antimicrob Agents Chemother 2019; 63:e00397-19. [PMID: 31061163 PMCID: PMC6591611 DOI: 10.1128/aac.00397-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/03/2019] [Indexed: 02/07/2023] Open
Abstract
Dengue virus (DENV) and Japanese encephalitis virus (JEV) are important arthropod-borne viruses from the Flaviviridae family. DENV is a global public health problem with significant social and economic impacts, especially in tropical and subtropical areas. JEV is a neurotropic arbovirus endemic to east and southeast Asia. There are no U.S. FDA-approved antiviral drugs available to treat or to prevent DENV and JEV infections, leaving nearly one-third of the world's population at risk for infection. Therefore, it is crucial to discover potent antiviral agents against these viruses. Nucleoside analogs, as a class, are widely used for the treatment of viral infections. In this study, we discovered nucleoside analogs that possess potent and selective anti-JEV and anti-DENV activities across all serotypes in cell-based assay systems. Both viruses were susceptible to sugar-substituted 2'-C-methyl analogs with either cytosine or 7-deaza-7-fluoro-adenine nucleobases. Mouse studies confirmed the anti-DENV activity of these nucleoside analogs. Molecular models were assembled for DENV serotype 2 (DENV-2) and JEV RNA-dependent RNA polymerase replication complexes bound to nucleotide inhibitors. These models show similarities between JEV and DENV-2, which recognize the same nucleotide inhibitors. Collectively, our findings provide promising compounds and a structural rationale for the development of direct-acting antiviral agents with dual activity against JEV and DENV infections.
Collapse
Affiliation(s)
- Keivan Zandi
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Tropical Infectious Disease Research and Education Center, Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Leda Bassit
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Franck Amblard
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Bryan D Cox
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Pouya Hassandarvish
- Tropical Infectious Disease Research and Education Center, Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ehsan Moghaddam
- Tropical Infectious Disease Research and Education Center, Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Andrew Yueh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Taiwan, Republic of China
| | - Gisele Olinto Libanio Rodrigues
- Center for Research and Drug Development, Instituto de Ciencias Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ingredy Passos
- Center for Research and Drug Development, Instituto de Ciencias Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian V Costa
- Center for Research and Drug Development, Instituto de Ciencias Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sazaly AbuBakar
- Tropical Infectious Disease Research and Education Center, Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Longhu Zhou
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - James Kohler
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mauro M Teixeira
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Research and Drug Development, Instituto de Ciencias Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Raymond F Schinazi
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
22
|
Identification of potential Zika virus NS2B-NS3 protease inhibitors via docking, molecular dynamics and consensus scoring-based virtual screening. J Mol Model 2019; 25:194. [PMID: 31209577 DOI: 10.1007/s00894-019-4076-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 05/27/2019] [Indexed: 02/08/2023]
Abstract
The Zika virus has recently become a subject of acute interest after the discovery of the link between viral infection and microcephaly in infants. Though a number of treatments are under active investigation, there are currently no approved treatments for the disease. To address this critical need, we screened more than 7 million compounds targeting the NS2B-NS3 protease in an attempt to identify promising inhibitor candidates. Starting with commercially and freely available compounds, we identified six hits utilizing an exhaustive consensus screening protocol, followed by molecular dynamics simulation and binding energy estimation using MM/GBSA and MM/PBSA methods. These compounds feature a variety of cores and functionalities, and all are predicted to have good pharmacokinetic profiles, making them promising candidates for screening assays. Graphical abstract Virtual screen of potential Zika virus NS2B-NS3 protease inhibitors.
Collapse
|
23
|
Shaily S, Upadhya A. Zika virus: Molecular responses and tissue tropism in the mammalian host. Rev Med Virol 2019; 29:e2050. [PMID: 31095819 DOI: 10.1002/rmv.2050] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 12/15/2022]
Abstract
Zika virus (ZIKV) outbreaks have raised alarm because of reports of congenital Zika virus syndrome in infants. The virus is also known to cause the debilitating Guillain-Barré syndrome in adults. As a result, extensive research has been carried out on the virus over the past few years. To study the molecular responses of viral infectivity in mammals, in vitro two-dimensional and three-dimensional cellular models have been employed. The in vivo models of mouse, pig, chicken, and nonhuman primates are primarily used to investigate the teratogenicity of the virus, to study effects of the virus on specific tissues, and to study the systemic effects of a proposed antiviral agent. The virus exhibits wide tissue tropism in the mammalian host. The major host tissues of viral persistence and propagation are neural tissue, ocular tissue, testicular tissue and placental tissue. An understanding of the function of viral components, viral replication cycle, and the molecular responses elicited in the host tissues is imperative for designing antiviral treatment strategies and for development of vaccines. This review provides an update on ZIKV research models and mammalian host responses with respect to ZIKV tissue infection.
Collapse
Affiliation(s)
- Sangya Shaily
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, Mumbai, India
| | - Archana Upadhya
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, Mumbai, India
| |
Collapse
|
24
|
Muñoz LS, Parra B, Pardo CA. Neurological Implications of Zika Virus Infection in Adults. J Infect Dis 2019; 216:S897-S905. [PMID: 29267923 DOI: 10.1093/infdis/jix511] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The 2015-2016 epidemic of Zika virus (ZIKV) in the Americas and the Caribbean was associated with an unprecedented burden of neurological disease among adults. Clinically, Guillain-Barre syndrome (GBS) predominated among regions affected by the ZIKV epidemic, but the spectrum of neurological disease in the adults appears broader as cases of encephalopathy, encephalitis, meningitis, myelitis, and seizures have also been reported. A para-infectious temporal profile of ZIKV-associated GBS (ZIKV-GBS) has been described in clinical studies, which may suggest a direct viral neuropathic effect. However, ZIKV neuropathogenesis has not yet been fully understood. Mechanisms for ZIKV-GBS and other neurological syndromes have been hypothesized, such as adaptive viral genetic changes, immunological interactions with other circulating flaviviruses, and host and factors. This review summarizes the current evidence on ZIKV-associated neurological complications in the adults.
Collapse
Affiliation(s)
- Laura S Muñoz
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Beatriz Parra
- Department of Microbiology, Universidad del Valle School of Medicine, Cali, Colombia
| | - Carlos A Pardo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
25
|
Poveda-Cuevas S, Etchebest C, Barroso da Silva FL. Insights into the ZIKV NS1 Virology from Different Strains through a Fine Analysis of Physicochemical Properties. ACS OMEGA 2018; 3:16212-16229. [PMID: 31458257 PMCID: PMC6643396 DOI: 10.1021/acsomega.8b02081] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 11/14/2018] [Indexed: 05/02/2023]
Abstract
The flavivirus genus has several organisms responsible for generating various diseases in humans. Recently, especially in tropical regions, Zika virus (ZIKV) has raised great health concerns due to the high number of cases affecting the area during the last years that has been accompanied by a rise in the cases of the Guillain-Barré syndrome and fetal and neonatal microcephaly. Diagnosis is still difficult since the clinical symptoms between ZIKV and other flaviviruses (e.g., dengue and yellow fever) are highly similar. The understanding of their common physicochemical properties that are pH-dependent and biomolecular interaction features and their differences sheds light on the relation strain-virulence and might suggest alternative strategies toward differential serological diagnostics and therapeutic intervention. Due to their immunogenicity, the primary focus of this study was on the ZIKV nonstructural proteins 1 (NS1). By means of computational studies and semiquantitative theoretical analyses, we calculated the main physicochemical properties of this protein from different strains that are directly responsible for the biomolecular interactions and, therefore, can be related to the differential infectivity of the strains. We also mapped the electrostatic differences at both the sequence and structural levels for the strains from Uganda to Brazil, which could suggest possible molecular mechanisms for the increase of the virulence of ZIKV in Brazil. Exploring the interfaces used by NS1 to self-associate in some different oligomeric states and interact with membranes and the antibody, we could map the strategy used by the ZIKV during its evolutionary process. This indicates possible molecular mechanisms that can be correlated with the different immunological responses. By comparing with the known antibody structure available for the West Nile virus, we demonstrated that this antibody would have difficulties to neutralize the NS1 from the Brazilian strain. The present study also opens up perspectives to computationally design high-specificity antibodies.
Collapse
Affiliation(s)
- Sergio
A. Poveda-Cuevas
- Programa
Interunidades em Bioinformática, Universidade de São Paulo, São Paulo 05508-090, Brazil
- Departamento
de Física e Química, Faculdade de Ciências Farmacêuticas
de Ribeirão Preto, Universidade de
São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil
- University
of São Paulo and Université Sorbonne Paris Cité
Joint International Laboratory in Structural Bioinformatics
| | - Catherine Etchebest
- Institut
National de la Transfusion Sanguine, Paris 75015, France
- Biologie
Intégrée du Globule Rouge, Equipe 2, Dynamique des Structures
et des Interactions Moléculaires, Institut National de la Santé et de la Recherche Médicale,
UMR_S 1134, Paris 75015, France
- Université
Sorbonne Paris Cité and Université Paris Diderot, 75013 Paris, France
- University
of São Paulo and Université Sorbonne Paris Cité
Joint International Laboratory in Structural Bioinformatics
| | - Fernando L. Barroso da Silva
- Programa
Interunidades em Bioinformática, Universidade de São Paulo, São Paulo 05508-090, Brazil
- Departamento
de Física e Química, Faculdade de Ciências Farmacêuticas
de Ribeirão Preto, Universidade de
São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil
- University
of São Paulo and Université Sorbonne Paris Cité
Joint International Laboratory in Structural Bioinformatics
- Department
of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27606, United States
- E-mail: and
| |
Collapse
|
26
|
Sinigaglia A, Riccetti S, Trevisan M, Barzon L. In silico approaches to Zika virus drug discovery. Expert Opin Drug Discov 2018; 13:825-835. [PMID: 30160181 DOI: 10.1080/17460441.2018.1515909] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION After the WHO declared Zika virus (ZIKV) as a public health emergency of international concern, intense research for the development of vaccines and drugs has been undertaken, leading to the development of several candidates. Areas covered: This review discusses the developments achieved so far by computational methods in the discovery of candidate compounds targeting ZIKV proteins, i.e. the envelope and capsid structural proteins, the NS3 helicase/protease, and the NS5 methyltransferase/RNA-dependent RNA polymerase. Expert opinion: Research for effective drugs against ZIKV is still in a very early discovery phase. Notwithstanding the intense efforts for the development of new drugs and the identification of several promising candidates by using different approaches, including computational methods, so far only a few candidates have been experimentally tested. An important caveat of anti-flavivirus drug development is represented by the difficult of reproducing the in vivo microenvironment of the replication complex, which may lead to discrepancies between in vitro results and experimental evaluation in vivo. Moreover, anti-ZIKV drugs have the additional requirement of an excellent safety profile in pregnancy and ability to diffuse to different tissues, including the central nervous system, the testis, and the placenta.
Collapse
Affiliation(s)
| | - Silvia Riccetti
- a Department of Molecular Medicine , University of Padova , Padova , Italy
| | - Marta Trevisan
- a Department of Molecular Medicine , University of Padova , Padova , Italy
| | - Luisa Barzon
- a Department of Molecular Medicine , University of Padova , Padova , Italy
| |
Collapse
|
27
|
Abstract
Background Synthetic virology is an important multidisciplinary scientific field, with emerging applications in biotechnology and medicine, aiming at developing methods to generate and engineer synthetic viruses. In particular, many of the RNA viruses, including among others the Dengue and Zika, are widespread pathogens of significant importance to human health. The ability to design and synthesize such viruses may contribute to exploring novel approaches for developing vaccines and virus based therapies. Results Here we develop a full multidisciplinary pipeline for generation and analysis of synthetic RNA viruses and specifically apply it to Dengue virus serotype 2 (DENV-2). The major steps of the pipeline include comparative genomics of endogenous and synthetic viral strains. Specifically, we show that although the synthetic DENV-2 viruses were found to have lower nucleotide variability, their phenotype, as reflected in the study of the AG129 mouse model morbidity, RNA levels, and neutralization antibodies, is similar or even more pathogenic in comparison to the wildtype master strain. Additionally, the highly variable positions, identified in the analyzed DENV-2 population, were found to overlap with less conserved homologous positions in Zika virus and other Dengue serotypes. These results may suggest that synthetic DENV-2 could enhance virulence if the correct sequence is selected. Conclusions The approach reported in this study can be used to generate and analyze synthetic RNA viruses both on genotypic and on phenotypic level. It could be applied for understanding the functionality and the fitness effects of any set of mutations in viral RNA and for editing RNA viruses for various target applications. Electronic supplementary material The online version of this article (10.1186/s12859-018-2132-3) contains supplementary material, which is available to authorized users.
Collapse
|
28
|
Lee I, Bos S, Li G, Wang S, Gadea G, Desprès P, Zhao RY. Probing Molecular Insights into Zika Virus⁻Host Interactions. Viruses 2018; 10:v10050233. [PMID: 29724036 PMCID: PMC5977226 DOI: 10.3390/v10050233] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 04/26/2018] [Accepted: 04/28/2018] [Indexed: 12/13/2022] Open
Abstract
The recent Zika virus (ZIKV) outbreak in the Americas surprised all of us because of its rapid spread and association with neurologic disorders including fetal microcephaly, brain and ocular anomalies, and Guillain–Barré syndrome. In response to this global health crisis, unprecedented and world-wide efforts are taking place to study the ZIKV-related human diseases. Much has been learned about this virus in the areas of epidemiology, genetic diversity, protein structures, and clinical manifestations, such as consequences of ZIKV infection on fetal brain development. However, progress on understanding the molecular mechanism underlying ZIKV-associated neurologic disorders remains elusive. To date, we still lack a good understanding of; (1) what virologic factors are involved in the ZIKV-associated human diseases; (2) which ZIKV protein(s) contributes to the enhanced viral pathogenicity; and (3) how do the newly adapted and pandemic ZIKV strains alter their interactions with the host cells leading to neurologic defects? The goal of this review is to explore the molecular insights into the ZIKV–host interactions with an emphasis on host cell receptor usage for viral entry, cell innate immunity to ZIKV, and the ability of ZIKV to subvert antiviral responses and to cause cytopathic effects. We hope this literature review will inspire additional molecular studies focusing on ZIKV–host Interactions.
Collapse
Affiliation(s)
- Ina Lee
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Sandra Bos
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France.
| | - Ge Li
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Shusheng Wang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Gilles Gadea
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France.
| | - Philippe Desprès
- Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 94791 Sainte Clotilde, La Réunion, France.
| | - Richard Y Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
29
|
Panwar U, Singh SK. An Overview on Zika Virus and the Importance of Computational Drug Discovery. JOURNAL OF EXPLORATORY RESEARCH IN PHARMACOLOGY 2018; 3:43-51. [DOI: 10.14218/jerp.2017.00025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
30
|
Mittal R, Nguyen D, Debs LH, Patel AP, Liu G, Jhaveri VM, S. Kay SI, Mittal J, Bandstra ES, Younis RT, Chapagain P, Jayaweera DT, Liu XZ. Zika Virus: An Emerging Global Health Threat. Front Cell Infect Microbiol 2017; 7:486. [PMID: 29276699 PMCID: PMC5727043 DOI: 10.3389/fcimb.2017.00486] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/13/2017] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is an emerging healthcare threat. The presence of the mosquito Aedes species across South and Central America in combination with complementary climates have incited an epidemic of locally transmitted cases of ZIKV infection in Brazil. As one of the most significant current public health concerns in the Americas, ZIKV epidemic has been a cause of alarm due to its known and unknown complications. At this point, there has been a clear association between ZIKV infection and severe clinical manifestations in both adults and neonates, including but not limited to neurological deficits such as Guillain-Barré syndrome (GBS) and microcephaly, respectively. The gravity of the fetal anomalies linked to ZIKV vertical transmission from the mother has prompted a discussion on whether to include ZIKV as a formal member of the TORCH [Toxoplasma gondii, other, rubella virus, cytomegalovirus (CMV), and herpes] family of pathogens known to breach placental barriers and cause congenital disease in the fetus. The mechanisms of these complex phenotypes have yet to be fully described. As such, diagnostic tools are limited and no effective modalities are available to treat ZIKV. This article will review the recent advancements in understanding the pathogenesis of ZIKV infection as well as diagnostic tests available to detect the infection. Due to the increase in incidence of ZIKV infections, there is an immediate need to develop new diagnostic tools and novel preventive as well as therapeutic modalities based on understanding the molecular mechanisms underlying the disease.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States,*Correspondence: Rahul Mittal
| | - Desiree Nguyen
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Luca H. Debs
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Amit P. Patel
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - George Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Vasanti M. Jhaveri
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sae-In S. Kay
- Department of Surgery, Division of Otorhinolaryngology, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Emmalee S. Bandstra
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ramzi T. Younis
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States,Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States,Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Prem Chapagain
- Department of Physics and Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Dushyantha T. Jayaweera
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States,Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States,Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States,Xue Zhong Liu
| |
Collapse
|
31
|
Structures and Functions of the Envelope Glycoprotein in Flavivirus Infections. Viruses 2017; 9:v9110338. [PMID: 29137162 PMCID: PMC5707545 DOI: 10.3390/v9110338] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 10/28/2017] [Accepted: 11/10/2017] [Indexed: 12/26/2022] Open
Abstract
Flaviviruses are enveloped, single-stranded RNA viruses that widely infect many animal species. The envelope protein, a structural protein of flavivirus, plays an important role in host cell viral infections. It is composed of three separate structural envelope domains I, II, and III (EDI, EDII, and EDIII). EDI is a structurally central domain of the envelope protein which stabilizes the overall orientation of the protein, and the glycosylation sites in EDI are related to virus production, pH sensitivity, and neuroinvasiveness. EDII plays an important role in membrane fusion because of the immunodominance of the fusion loop epitope and the envelope dimer epitope. Additionally, EDIII is the major target of neutralization antibodies. The envelope protein is an important target for research to develop vaccine candidates and antiviral therapeutics. This review summarizes the structures and functions of ED I/II/III, and provides practical applications for the three domains, with the ultimate goal of implementing strategies to utilize the envelope protein against flavivirus infections, thus achieving better diagnostics and developing potential flavivirus therapeutics and vaccines.
Collapse
|
32
|
Mottin M, Braga RC, da Silva RA, Silva JHMD, Perryman AL, Ekins S, Andrade CH. Molecular dynamics simulations of Zika virus NS3 helicase: Insights into RNA binding site activity. Biochem Biophys Res Commun 2017; 492:643-651. [DOI: 10.1016/j.bbrc.2017.03.070] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 02/23/2017] [Accepted: 03/15/2017] [Indexed: 12/14/2022]
|
33
|
Shankar A, Patil AA, Skariyachan S. Recent Perspectives on Genome, Transmission, Clinical Manifestation, Diagnosis, Therapeutic Strategies, Vaccine Developments, and Challenges of Zika Virus Research. Front Microbiol 2017; 8:1761. [PMID: 28959246 PMCID: PMC5603822 DOI: 10.3389/fmicb.2017.01761] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/30/2017] [Indexed: 12/13/2022] Open
Abstract
One of the potential threats to public health microbiology in 21st century is the increased mortality rate caused by Zika virus (ZIKV), a mosquito-borne flavivirus. The severity of ZIKV infection urged World Health Organization (WHO) to declare this virus as a global concern. The limited knowledge on the structure, virulent factors, and replication mechanism of the virus posed as hindrance for vaccine development. Several vector and non-vector-borne mode of transmission are observed for spreading the disease. The similarities of the virus with other flaviviruses such as dengue and West Nile virus are worrisome; hence, there is high scope to undertake ZIKV research that probably provide insight for novel therapeutic intervention. Thus, this review focuses on the recent aspect of ZIKV research which includes the outbreak, genome structure, multiplication and propagation of the virus, current animal models, clinical manifestations, available treatment options (probable vaccines and therapeutics), and the recent advancements in computational drug discovery pipelines, challenges and limitation to undertake ZIKV research. The review suggests that the infection due to ZIKV became one of the universal concerns and an interdisciplinary environment of in vitro cellular assays, genomics, proteomics, and computational biology approaches probably contribute insights for screening of novel molecular targets for drug design. The review tried to provide cutting edge knowledge in ZIKV research with future insights required for the development of novel therapeutic remedies to curtail ZIKV infection.
Collapse
Affiliation(s)
- Apoorva Shankar
- R&D Centre, Department of Biotechnology Engineering, Dayananda Sagar InstitutionsBengaluru, India
| | - Amulya A Patil
- R&D Centre, Department of Biotechnology Engineering, Dayananda Sagar InstitutionsBengaluru, India
| | - Sinosh Skariyachan
- R&D Centre, Department of Biotechnology Engineering, Dayananda Sagar InstitutionsBengaluru, India.,Visvesvaraya Technological UniversityBelagavi, India
| |
Collapse
|
34
|
Aliota MT, Bassit L, Bradrick SS, Cox B, Garcia-Blanco MA, Gavegnano C, Friedrich TC, Golos TG, Griffin DE, Haddow AD, Kallas EG, Kitron U, Lecuit M, Magnani DM, Marrs C, Mercer N, McSweegan E, Ng LFP, O'Connor DH, Osorio JE, Ribeiro GS, Ricciardi M, Rossi SL, Saade G, Schinazi RF, Schott-Lerner GO, Shan C, Shi PY, Watkins DI, Vasilakis N, Weaver SC. Zika in the Americas, year 2: What have we learned? What gaps remain? A report from the Global Virus Network. Antiviral Res 2017; 144:223-246. [PMID: 28595824 PMCID: PMC5920658 DOI: 10.1016/j.antiviral.2017.06.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/01/2017] [Indexed: 12/25/2022]
Abstract
In response to the outbreak of Zika virus (ZIKV) infection in the Western Hemisphere and the recognition of a causal association with fetal malformations, the Global Virus Network (GVN) assembled an international taskforce of virologists to promote basic research, recommend public health measures and encourage the rapid development of vaccines, antiviral therapies and new diagnostic tests. In this article, taskforce members and other experts review what has been learned about ZIKV-induced disease in humans, its modes of transmission and the cause and nature of associated congenital manifestations. After describing the make-up of the taskforce, we summarize the emergence of ZIKV in the Americas, Africa and Asia, its spread by mosquitoes, and current control measures. We then review the spectrum of primary ZIKV-induced disease in adults and children, sites of persistent infection and sexual transmission, then examine what has been learned about maternal-fetal transmission and the congenital Zika syndrome, including knowledge obtained from studies in laboratory animals. Subsequent sections focus on vaccine development, antiviral therapeutics and new diagnostic tests. After reviewing current understanding of the mechanisms of emergence of Zika virus, we consider the likely future of the pandemic.
Collapse
Affiliation(s)
- Matthew T Aliota
- Department of Pathobiological Sciences, University of Wisconsin-Madison, USA
| | - Leda Bassit
- Center for AIDS Research, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Shelton S Bradrick
- Department of Biochemistry and Molecular Biology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Bryan Cox
- Center for AIDS Research, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Mariano A Garcia-Blanco
- Department of Biochemistry and Molecular Biology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Christina Gavegnano
- Center for AIDS Research, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Thomas C Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison, USA; Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA; Department of Comparative Biosciences, University of Wisconsin-Madison, USA; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, USA
| | - Diane E Griffin
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Andrew D Haddow
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Virology Division, United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, 21702, USA
| | - Esper G Kallas
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, Brazil
| | - Uriel Kitron
- Department of Environmental Sciences, Emory University, Atlanta, GA, USA
| | - Marc Lecuit
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Institut Pasteur, Biology of Infection Unit and INSERM Unit 1117, France; Paris Descartes University, Sorbonne Paris Cité, Division of Infectious Diseases and Tropical Medicine, Necker- Enfants Malades University Hospital, Institut Imagine, Paris, France
| | - Diogo M Magnani
- Department of Pathology, University of Miami, Miami, FL, USA
| | - Caroline Marrs
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Natalia Mercer
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA
| | | | - Lisa F P Ng
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - David H O'Connor
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, USA
| | - Jorge E Osorio
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Department of Pathobiological Sciences, University of Wisconsin-Madison, USA
| | - Guilherme S Ribeiro
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz and Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | | | - Shannan L Rossi
- Department of Microbiology & Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - George Saade
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Raymond F Schinazi
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Center for AIDS Research, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Geraldine O Schott-Lerner
- Department of Biochemistry and Molecular Biology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Chao Shan
- Department of Biochemistry and Molecular Biology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - David I Watkins
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Department of Pathology, University of Miami, Miami, FL, USA
| | - Nikos Vasilakis
- Department of Pathology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Scott C Weaver
- Global Virus Network, 725 West Lombard St., Baltimore, MD, USA; Department of Microbiology & Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
35
|
Baez CF, Barel VA, de Souza AMT, Rodrigues CR, Varella RB, Cirauqui N. Analysis of worldwide sequence mutations in Zika virus proteins E, NS1, NS3 and NS5 from a structural point of view. MOLECULAR BIOSYSTEMS 2017; 13:122-131. [PMID: 27805221 DOI: 10.1039/c6mb00645k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Zika virus (ZIKV) is an emergent arbovirus that has attracted attention in the last year as a possible causative agent of congenital malformation; it shows a remarkably increased microcephaly risk during otherwise healthy pregnancies. We present here an analysis of all ZIKV sequences available in Genbank up to April 2016, studying the mutations in the whole polyprotein and their possible structural implications for the proteins E, NS1, NS3 and NS5. This study suggests that microcephaly is not a consequence of any particular amino acid substitution but, conceivably, is a feature of ZIKV itself. Moreover, the structural analysis of ZIKV proteins, together with the mutational landscape of ZIKV and a structure-sequence comparison with other flaviviruses, allows the suggestion of regions that could be exploited as anti-ZIKV targets, including some allosteric sites found in the NS3 and NS5 proteins of DENV.
Collapse
Affiliation(s)
- C F Baez
- Preventive Medicine Department, Rio de Janeiro Federal University Hospital, Brazil
| | - V A Barel
- ModMolQSAR Laboratory, Faculty of Pharmaceutical Sciences, Rio de Janeiro Federal University, Brazil.
| | - A M T de Souza
- ModMolQSAR Laboratory, Faculty of Pharmaceutical Sciences, Rio de Janeiro Federal University, Brazil.
| | - C R Rodrigues
- ModMolQSAR Laboratory, Faculty of Pharmaceutical Sciences, Rio de Janeiro Federal University, Brazil.
| | - R B Varella
- Microbiology and Parasitology Department, Biomedical Institute, Fluminense Federal University, Brazil
| | - N Cirauqui
- ModMolQSAR Laboratory, Faculty of Pharmaceutical Sciences, Rio de Janeiro Federal University, Brazil.
| |
Collapse
|
36
|
Roy A, Lim L, Srivastava S, Lu Y, Song J. Solution conformations of Zika NS2B-NS3pro and its inhibition by natural products from edible plants. PLoS One 2017; 12:e0180632. [PMID: 28700665 PMCID: PMC5503262 DOI: 10.1371/journal.pone.0180632] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 06/19/2017] [Indexed: 12/20/2022] Open
Abstract
The recent Zika viral (ZIKV) epidemic has been associated with severe neurological pathologies such as neonatal microcephaly and Guillain-Barre syndrome but unfortunately no vaccine or medication is effectively available yet. Zika NS2B-NS3pro is essential for the proteolysis of the viral polyprotein and thereby viral replication. Thus NS2B-NS3pro represents an attractive target for anti-Zika drug discovery/design. Here, we have characterized the solution conformations and catalytic parameters of both linked and unlinked Zika NS2B-NS3pro complexes and found that the unlinked complex manifested well-dispersed NMR spectra. Subsequently with selective isotope-labeling using NMR spectroscopy, we demonstrated that C-terminal residues (R73-K100) of NS2B is highly disordered without any stable tertiary and secondary structures in the Zika NS2B-NS3pro complex in the free state. Upon binding to the well-characterized serine protease inhibitor, bovine pancreatic trypsin inhibitor (BPTI), only the extreme C-terminal residues (L86-K100) remain disordered. Additionally, we have identified five flavonoids and one natural phenol rich in edible plants including fruits and vegetables, which inhibit Zika NS2B-NS3pro in a non-competitive mode, with Ki ranging from 770 nM for Myricetin to 34.02 μM for Apigenin. Molecular docking showed that they all bind to a pocket on the back of the active site and their structure-activity relationship was elucidated. Our study provides valuable insights into the solution conformation of Zika NS2B-NS3pro and further deciphers its susceptibility towards allosteric inhibition by natural products. As these natural product inhibitors fundamentally differ from the currently-known active site inhibitors in terms of both inhibitory mode and chemical scaffold, our finding might open a new avenue for development of better allosteric inhibitors to fight ZIKV infection.
Collapse
Affiliation(s)
- Amrita Roy
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Liangzhong Lim
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Shagun Srivastava
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yimei Lu
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Jianxing Song
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
37
|
Song BH, Yun SI, Woolley M, Lee YM. Zika virus: History, epidemiology, transmission, and clinical presentation. J Neuroimmunol 2017; 308:50-64. [DOI: 10.1016/j.jneuroim.2017.03.001] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/01/2017] [Accepted: 03/01/2017] [Indexed: 10/20/2022]
|
38
|
Cai L, Sun Y, Song Y, Xu L, Bei Z, Zhang D, Dou Y, Wang H. Viral polymerase inhibitors T-705 and T-1105 are potential inhibitors of Zika virus replication. Arch Virol 2017; 162:2847-2853. [PMID: 28597088 PMCID: PMC5563514 DOI: 10.1007/s00705-017-3436-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/19/2017] [Indexed: 02/01/2023]
Abstract
Since 2015, 69 countries and territories have reported evidence of vector-borne Zika virus (ZIKV) transmission. Currently, there are no effective licensed vaccines or drugs available for the treatment or prevention of ZIKV infection. We tested a series of compounds for their ability to inhibit ZIKV replication in cell culture. The compounds in T-705 (favipiravir) and T-1105 were found to have antiviral activity, suggesting that these compounds are promising candidates for further development as specific antiviral drugs against ZIKV.
Collapse
Affiliation(s)
- Lei Cai
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Science, Beijing, 100071, China
| | - Yajie Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Science, Beijing, 100071, China
| | - Yabin Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Science, Beijing, 100071, China
| | - Likun Xu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Science, Beijing, 100071, China
| | - Zhuchun Bei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Science, Beijing, 100071, China
| | - Dongna Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Science, Beijing, 100071, China
| | - Yuanyuan Dou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Science, Beijing, 100071, China
| | - Hongquan Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Science, Beijing, 100071, China.
| |
Collapse
|
39
|
Gavegnano C, Bassit LC, Cox BD, Hsiao HM, Johnson EL, Suthar M, Chakraborty R, Schinazi RF. Jak Inhibitors Modulate Production of Replication-Competent Zika Virus in Human Hofbauer, Trophoblasts, and Neuroblastoma cells. Pathog Immun 2017; 2:199-218. [PMID: 28776046 PMCID: PMC5538373 DOI: 10.20411/pai.v2i2.190] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Zika Virus (ZIKV) is a flavivirus that has been implicated in causing brain deformations, birth defects, and microcephaly in fetuses, and associated with Guillain-Barre syndrome. Mechanisms responsible for transmission of ZIKV across the placenta to the fetus are incompletely understood. Herein, we define key events modulating infection in clinically relevant cells, including primary placental macrophages (human Hofbauer cells; HC), trophoblasts, and neuroblastoma cells. Consistent with previous findings, HC and trophoblasts are permissive to ZIKV infection. Decrease of interferon signaling by Jak ½ inhibition (using ruxolitinib) significantly increased ZIKV replication in HC, trophoblasts, and neuroblasts. Enhanced ZIKV production in ruxolitinib-treated HC was associated with increased expression of HLA-DR and DC-SIGN. Nucleoside analogs blocked ruxolitinib-mediated production of extracellular virus. Although low-level ZIKV infection occurred in untreated HC and trophoblasts, replicating virions were incapable of infecting naive Vero cells. These deficient virions from untreated HC have “thin-coats” suggesting an immature structure. Blocking Jak ½ signaling (with ruxolitinib) restored replication competence as virions produced under these conditions confer cytopathic effects to naive Vero cells. These data demonstrate that Jak-STAT signaling directly impacts the ability of primary placental cells to produce replication-competent virus and is a key determinant in the production of mature virions in clinically relevant cells, including HC and trophoblasts. Design of targeted agents to prevent ZIKV replication in the placenta should consider Jak ½ signaling, the impact of its block on ZIKV infection, and subsequent transmission to the fetus.
Collapse
Affiliation(s)
- Christina Gavegnano
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Leda C Bassit
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Bryan D Cox
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Hui-Mien Hsiao
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Erica L Johnson
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Mehul Suthar
- Emory Vaccine Center, Yerkes National Primate Center, Emory University, Atlanta, Georgia
| | - Rana Chakraborty
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Raymond F Schinazi
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| |
Collapse
|
40
|
Liang D, Leung RKK, Lee SS, Kam KM. Insights into intercontinental spread of Zika virus. PLoS One 2017; 12:e0176710. [PMID: 28448611 PMCID: PMC5407806 DOI: 10.1371/journal.pone.0176710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 04/16/2017] [Indexed: 12/02/2022] Open
Abstract
The epidemic of Zika virus (ZIKV) infection in South America has led to World Health Organization's declaration of a Public Health Emergency of International Concern. To further inform effective public health policy, an understanding of ZIKV's transmission mechanisms is crucial. To characterize the intercontinental transmission of ZIKV, we compiled and analyzed more than 250 gene sequences together with their sequence-related geographic and temporal information, sampled across 27 countries spanning from 1947 to 2016. After filtering and selecting appropriate sequences, extensive phylogenetic analyses were performed. Although phylogeographic reconstruction supported the transmission route of the virus in Africa, South-eastern Asia, Oceania and Latin America, we discovered that the Eastern Africa origin of ZIKV was disputable. On a molecular level, purifying selection was found to be largely responsible for the evolution of non-structural protein 5 and envelope protein E. Our dataset and ancestral sequences reconstruction analysis captured previously unidentified amino acid changes during evolution. Finally, based on the estimation of the time to the most recent common ancestors for the non-structural protein 5 gene, we hypothesized potential specific historic events that occurred in the 1940s and might have facilitated the spread of Zika virus from Africa to South-eastern Asia. Our findings provide new insights into the transmission characteristics of ZIKV, while further genetic and serologic studies are warranted to support the design of tailored prevention strategies.
Collapse
Affiliation(s)
- Dachao Liang
- Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, China
| | - Ross Ka Kit Leung
- Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, China
| | - Shui Shan Lee
- Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, China
| | - Kai Man Kam
- Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
41
|
Advances in research on Zika virus. ASIAN PAC J TROP MED 2017; 10:321-331. [PMID: 28552102 DOI: 10.1016/j.apjtm.2017.03.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 02/14/2017] [Accepted: 03/13/2017] [Indexed: 11/22/2022] Open
Abstract
Zika virus (ZIKV) is rapidly spreading across the America and its devastating outcomes for pregnant women and infants have driven this previously ignored pathogen into the limelight. Clinical manifestations are fever, joint pain or rash and conjunctivitis. Emergence of ZIKV started with a first outbreak in the Pacific area in 2007, a second large outbreak occurred in the Pacific in 2013/2014 and subsequently the virus spread in other Pacific islands. Threat of explosive global pandemic and severe clinical complications linked with the more immediate and recurrent epidemics necessitate the development of an effective vaccine. Several vaccine platforms such as DNA vaccine, recombinant subunit vaccine, ZIKV purified inactivated vaccine, and chimeric vaccines have shown potent efficacy in vitro and in vivo trials. Moreover, number of drugs such as Sofosbuvir, BCX4450, NITD008 and 7-DMA are ready to enter phase I clinical trial because of proven anti-ZIKV activity. Monoclonal based antibodies offer promise as an intervention effective for use in pregnant women. In this review, we describe the advances in research on ZIKV such as research strategies for the development of antiviral drugs & vaccines, molecular evolution, epidemiology emergence, neurological complications and other teratogenic outcomes as well as pathogenesis.
Collapse
|
42
|
Yun SI, Lee YM. Zika virus: An emerging flavivirus. J Microbiol 2017; 55:204-219. [PMID: 28243937 DOI: 10.1007/s12275-017-7063-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 02/15/2017] [Indexed: 01/18/2023]
Abstract
Zika virus (ZIKV) is a previously little-known flavivirus closely related to Japanese encephalitis, West Nile, dengue, and yellow fever viruses, all of which are primarily transmitted by blood-sucking mosquitoes. Since its discovery in Uganda in 1947, ZIKV has continued to expand its geographic range, from equatorial Africa and Asia to the Pacific Islands, then further afield to South and Central America and the Caribbean. Currently, ZIKV is actively circulating not only in much of Latin America and its neighbors but also in parts of the Pacific Islands and Southeast Asia. Although ZIKV infection generally causes only mild symptoms in some infected individuals, it is associated with a range of neuroimmunological disorders, including Guillain-Barré syndrome, meningoencephalitis, and myelitis. Recently, maternal ZIKV infection during pregnancy has been linked to neonatal malformations, resulting in various degrees of congenital abnormalities, microcephaly, and even abortion. Despite its emergence as an important public health problem, however, little is known about ZIKV biology, and neither vaccine nor drug is available to control ZIKV infection. This article provides a brief introduction to ZIKV with a major emphasis on its molecular virology, in order to help facilitate the development of diagnostics, therapeutics, and vaccines.
Collapse
Affiliation(s)
- Sang-Im Yun
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, 84322-4815, USA
| | - Young-Min Lee
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, 84322-4815, USA. .,Utah Science Technology and Research, College of Agriculture and Applied Sciences, Utah State University, Logan, UT, 84322-4815, USA.
| |
Collapse
|
43
|
Characterization of cytopathic factors through genome-wide analysis of the Zika viral proteins in fission yeast. Proc Natl Acad Sci U S A 2017; 114:E376-E385. [PMID: 28049830 DOI: 10.1073/pnas.1619735114] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The Zika virus (ZIKV) causes microcephaly and the Guillain-Barré syndrome. Little is known about how ZIKV causes these conditions or which ZIKV viral protein(s) is responsible for the associated ZIKV-induced cytopathic effects, including cell hypertrophy, growth restriction, cell-cycle dysregulation, and cell death. We used fission yeast for the rapid, global functional analysis of the ZIKV genome. All 14 proteins or small peptides were produced under an inducible promoter, and we measured the intracellular localization and the specific effects on ZIKV-associated cytopathic activities of each protein. The subcellular localization of each ZIKV protein was in overall agreement with its predicted protein structure. Five structural and two nonstructural ZIKV proteins showed various levels of cytopathic effects. The expression of these ZIKV proteins restricted cell proliferation, induced hypertrophy, or triggered cellular oxidative stress leading to cell death. The expression of premembrane protein (prM) resulted in cell-cycle G1 accumulation, whereas membrane-anchored capsid (anaC), membrane protein (M), envelope protein (E), and nonstructural protein 4A (NS4A) caused cell-cycle G2/M accumulation. A mechanistic study revealed that NS4A-induced cellular hypertrophy and growth restriction were mediated specifically through the target of rapamycin (TOR) cellular stress pathway involving Tor1 and type 2A phosphatase activator Tip41. These findings should provide a reference for future research on the prevention and treatment of ZIKV diseases.
Collapse
|
44
|
Ramharack P, Oguntade S, Soliman MES. Delving into Zika virus structural dynamics – a closer look at NS3 helicase loop flexibility and its role in drug discovery. RSC Adv 2017. [DOI: 10.1039/c7ra01376k] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
This study demonstrates the structural alterations in the P-loop of ZIKV helicase subsequent to binding of potent inhibitor, NITD008.
Collapse
Affiliation(s)
- Pritika Ramharack
- Molecular Modeling and Drug Design Research Group
- School of Health Sciences
- University of KwaZulu-Natal
- Durban 4001
- South Africa
| | - Sofiat Oguntade
- Molecular Modeling and Drug Design Research Group
- School of Health Sciences
- University of KwaZulu-Natal
- Durban 4001
- South Africa
| | - Mahmoud E. S. Soliman
- Molecular Modeling and Drug Design Research Group
- School of Health Sciences
- University of KwaZulu-Natal
- Durban 4001
- South Africa
| |
Collapse
|
45
|
Munjal A, Khandia R, Dhama K, Sachan S, Karthik K, Tiwari R, Malik YS, Kumar D, Singh RK, Iqbal HMN, Joshi SK. Advances in Developing Therapies to Combat Zika Virus: Current Knowledge and Future Perspectives. Front Microbiol 2017; 8:1469. [PMID: 28824594 PMCID: PMC5541032 DOI: 10.3389/fmicb.2017.01469] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/20/2017] [Indexed: 02/05/2023] Open
Abstract
Zika virus (ZIKV) remained largely quiescent for nearly six decades after its first appearance in 1947. ZIKV reappeared after 2007, resulting in a declaration of an international "public health emergency" in 2016 by the World Health Organization (WHO). Until this time, ZIKV was considered to induce only mild illness, but it has now been established as the cause of severe clinical manifestations, including fetal anomalies, neurological problems, and autoimmune disorders. Infection during pregnancy can cause congenital brain abnormalities, including microcephaly and neurological degeneration, and in other cases, Guillain-Barré syndrome, making infections with ZIKV a substantial public health concern. Genomic and molecular investigations are underway to investigate ZIKV pathology and its recent enhanced pathogenicity, as well as to design safe and potent vaccines, drugs, and therapeutics. This review describes progress in the design and development of various anti-ZIKV therapeutics, including drugs targeting virus entry into cells and the helicase protein, nucleosides, inhibitors of NS3 protein, small molecules, methyltransferase inhibitors, interferons, repurposed drugs, drugs designed with the aid of computers, neutralizing antibodies, convalescent serum, antibodies that limit antibody-dependent enhancement, and herbal medicines. Additionally, covalent inhibitors of viral protein expression and anti-Toll-like receptor molecules are discussed. To counter ZIKV-associated disease, we need to make rapid progress in developing novel therapies that work effectually to inhibit ZIKV.
Collapse
Affiliation(s)
- Ashok Munjal
- Department of Biochemistry and Genetics, Barkatullah UniversityBhopal, India
| | - Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah UniversityBhopal, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research InstituteBareilly, India
- *Correspondence: Kuldeep Dhama,
| | - Swati Sachan
- Immunology Section, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences UniversityChennai, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, UP Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan SansthanMathura, India
| | - Yashpal S. Malik
- Division of Biological Standardization, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Deepak Kumar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Raj K. Singh
- ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Hafiz M. N. Iqbal
- School of Engineering and Science, Tecnologico de Monterrey, Campus MonterreyMonterrey, Mexico
| | - Sunil K. Joshi
- Cellular Immunology Lab, Frank Reidy Research Center of Bioelectrics, Old Dominion University, NorfolkVA, United States
| |
Collapse
|
46
|
Sofosbuvir: an antiviral drug with potential efficacy against Zika infection. Int J Infect Dis 2016; 55:29-30. [PMID: 27988410 DOI: 10.1016/j.ijid.2016.12.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/20/2016] [Accepted: 12/08/2016] [Indexed: 11/23/2022] Open
|
47
|
Angeletti S, Lo Presti A, Giovanetti M, Grifoni A, Amicosante M, Ciotti M, Alcantara LCJ, Cella E, Ciccozzi M. Phylogenesys and homology modeling in Zika virus epidemic: food for thought. Pathog Glob Health 2016; 110:269-274. [PMID: 27670692 DOI: 10.1080/20477724.2016.1235337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Zika virus (ZIKV) is an emerging Flavivirus that have recently caused an outbreak in Brazil and rapid spread in several countries. In this study, the consequences of ZIKV evolution on protein recognition by the host immune system have been analyzed. Evolutionary analysis was combined with homology modeling and T-B cells epitope predictions. Two separate clades, the African one with the Uganda sequence, as the most probable ancestor, and the second one containing all the most recent sequences from the equatorial belt were identified. Brazilian strains clustered all together and closely related to the French Polynesia isolates. A strong presence of a negatively selected site in the envelope gene (Env) protein was evidenced, suggesting a probable purging of deleterious polymorphisms in functionally important genes. Our results show relative conservancy of ZIKV sequences when envelope and other non-structural proteins (NS3 and NS5) are analyzed by homology modeling. However, some regions within the consensus sequence of NS5 protein and to a lesser extent in the envelope protein, show localized high mutation frequency corresponding to a considerable alteration in protein stability. In terms of viral immune escape, envelope protein is under a higher selective pressure than NS5 and NS3 proteins for HLA class I and II molecules. Moreover, envelope mutations that are not strictly related to T-cell immune responses are mostly located on the surface of the protein in putative B-cell epitopes, suggesting an important contribution of B cells in the immune response as well.
Collapse
Affiliation(s)
- Silvia Angeletti
- a Unit of Clinical Pathology and Microbiology , University Campus Bio-Medico , Rome , Italy
| | - Alessandra Lo Presti
- b Department of Infectious, Parasitic, and Immune-Mediated Diseases, Epidemiology Unit, Reference Centre on Phylogeny, Molecular Epidemiology, and Microbial Evolution (FEMEM) , National Institute of Health , Rome , Italy
| | - Marta Giovanetti
- c Oswaldo Cruz Foundation (FIOCRUZ) , Salvador , Brazil.,d Department of Biology , University of Rome 'Tor Vergata' , Rome , Italy
| | | | - Massimo Amicosante
- e ProxAgen Ltd , Sofia , Bulgaria.,f Department of Biomedicine and Prevention , University of Rome "Tor Vergata" , Rome , Italy
| | - Marco Ciotti
- g Laboratory of Molecular Virology , Polyclinic Tor Vergata Foundation , Rome , Italy
| | | | - Eleonora Cella
- b Department of Infectious, Parasitic, and Immune-Mediated Diseases, Epidemiology Unit, Reference Centre on Phylogeny, Molecular Epidemiology, and Microbial Evolution (FEMEM) , National Institute of Health , Rome , Italy.,h Department of Public Health and Infectious Diseases , Sapienza University of Rome , Rome , Italy
| | - Massimo Ciccozzi
- a Unit of Clinical Pathology and Microbiology , University Campus Bio-Medico , Rome , Italy.,b Department of Infectious, Parasitic, and Immune-Mediated Diseases, Epidemiology Unit, Reference Centre on Phylogeny, Molecular Epidemiology, and Microbial Evolution (FEMEM) , National Institute of Health , Rome , Italy
| |
Collapse
|