1
|
Hauner A, Rogé S, Vanlerberghe V, Lepore L, Ndayisenga F, Shyaka A, Van Esbroeck M, Situma S, Nasimiyu C, Ahuka-Mundeke S, Njenga MK, Breiman RF, Masumu J, Mukadi-Bamuleka D, Ariën KK. Technical validation of a multiplex real-time PCR for combined detection of Rift Valley fever, chikungunya, Zika and dengue viruses. J Virol Methods 2025; 337:115174. [PMID: 40345597 DOI: 10.1016/j.jviromet.2025.115174] [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: 02/24/2025] [Revised: 05/02/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Several arthropod-borne (arbo)-viruses have overlapping symptoms, insect vectors and geographical occurrence. With little known about the importance of arboviruses as cause of acute undifferentiated fever (AUF) in East and Central Africa (ECA), there is a clear need for a multiplex-PCR allowing for multi-pathogen surveillance. A multiplex real-time RT-PCR (RDCZ-multiplex) was developed and validated for the simultaneous detection of Rift Valley fever virus (RVFV), dengue virus 1-4 (DENV), chikungunya virus (CHIKV) and Zika virus (ZIKV). Phocine distemper virus (PDV) was added to the PCR as sample extraction control. Validation was conducted following the MIQE-guidelines using a panel of retrospective clinical samples and Quality Control for Molecular Diagnostics (QCMD, https://www.qcmd.org/en/) samples with the simplex-PCR as reference. These included samples from RVFV in animals (n = 19), DENV (n = 15), CHIKV (n = 11), ZIKV (n = 2) and YFV (n = 1, QCMD), and 14 negative endemic controls. Extractions and PCRs were done with commercially available kits. Some loss of sensitivity was observed at low target concentrations for RVFV, DENV1 and DENV4, when comparing the standard curves of simplex-PCRs with the multiplex-PCR. The limit of detection of the multiplex-PCR was 2064 copies/ml for CHIKV, 3587 copies/ml for DENV1, 30,249 copies/ml for ZIKV and 73 PFU/ml for RVFV. Specificity of the multiplex-PCRs was 100 %. For 12 out of 48 positive samples with high Cq values, RVFV (n = 7), CHIKV (n = 2), DENV1 (n = 2), YFV (n = 1), the multiplex-PCRs were negative. Although PCR sensitivity of the RDCZ-multiplex is slightly lower with low target concentrations, it offers a useful tool for molecular surveillance and clinical diagnosis for arboviruses for the ECA-region.
Collapse
Affiliation(s)
- Anne Hauner
- Department of Biomedical Sciences, Unit of Virology, Institute of Tropical Medicine, Antwerp, Belgium.
| | - Stijn Rogé
- DRC office, Institute of Tropical Medicine, Antwerp, Belgium
| | - Veerle Vanlerberghe
- Department of Public Health, Unit of Emerging Infectious Diseases, Institute of Tropical Medicine, Antwerp, Belgium
| | - Luciana Lepore
- Department of Public Health, Unit of Emerging Infectious Diseases, Institute of Tropical Medicine, Antwerp, Belgium
| | - Fabrice Ndayisenga
- Rwanda Agriculture and Animal Resources Development Board, Kigali, Rwanda
| | - Anselme Shyaka
- Center for One Health, University of Global Health Equity, Butaro, Rwanda
| | - Marjan Van Esbroeck
- Department of Clinical Science, Institute of Tropical Medicine, Antwerp, Belgium
| | - Silvia Situma
- Washington State University Global Health-Kenya, Nairobi, Kenya
| | | | | | | | | | - Justin Masumu
- National Institute for Biomedical Research INRB, Kinshasa, Congo
| | - Daniel Mukadi-Bamuleka
- Rodolphe Mérieux INRB-Goma Laboratory, Goma, North Kivu, Congo; Service of Microbiology, Department of Medical Biology, Faculty of Medicine, University of Kinshasa, Kinshasa, Congo; Department of Virology, Institut National de Recherche Biomédicale, INRB, Kinshasa, Congo
| | - Kevin K Ariën
- Department of Biomedical Sciences, Unit of Virology, Institute of Tropical Medicine, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
2
|
Ateutchia-Ngouanet S, Nanfack-Minkeu F, Mavridis K, Wanji S, Demanou M, Vontas J, Djouaka R. Monitoring Aedes populations for arboviruses, Wolbachia, insecticide resistance and its mechanisms in various agroecosystems in Benin. Acta Trop 2024; 253:107178. [PMID: 38461924 DOI: 10.1016/j.actatropica.2024.107178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
Aedes mosquitoes are the main vectors of arboviruses in Benin. Cases of dengue have been reported in Benin with all four serotypes of the virus actively circulating in this region. Some agricultural settings are known to harbor Aedes vectors responsible for the transmission of arboviruses. The massive use of certain insecticides in agricultural settings has probably contributed to insecticide resistance in these vectors. In Benin, the susceptibility of arbovirus vectors to insecticides is poorly studied. In addition, the distribution of Wolbachia spp., which is used against some arboviruses is unknown. Moreover, there is limited information regarding the vectors responsible for the transmission of arboviruses in Benin. This present study monitored the species composition, arboviruses, and Wolbachia symbiont status, as well as the phenotypic and molecular insecticide resistance profile of Aedes populations from three agroecosystems in Benin. Aedes species identification was performed morphologically and confirmed using qPCR. (RT)-qPCR assay was applied for monitoring the presence of DENV, CHIKV, ZIKV, and WNV pathogens as well as for naturally occurring Wolbachia symbionts. Insecticide resistance was assessed phenotypically, by permethrin (0.75%) exposure of Adults (F0) using World Health Organization (WHO) bioassay protocols, and at the molecular level, using TaqMan (RT)-qPCR assays for assessing knock-down resistance (kdr) mutations (F1534C, V1016G/I, and S989P) and the expression levels of eight detoxification genes (P450s from the CYP9 and CYP6 families, carboxylesterases and glutathione-S-transferases). Aedes aegypti (Ae. aegypti) mosquitoes were the most abundant (93.9%) in the three agroecosystems studied, followed by Aedes albopictus (Ae. albopictus) mosquitoes (6.1%). No arboviruses were detected in the study's mosquito populations. Naturally occurring Wolbachia symbionts were present in 7 pools out of 15 pools tested. This could influence the effectiveness of vector control strategies based on exogenously introduced Wolbachia, all present in the three agroecosystems. Full susceptibility to permethrin was observed in all tested populations of Ae. albopictus. On the contrary, Ae. aegypti were found to be resistant in all three agroecosystem sites except for banana plantation sites, where full susceptibility was observed. Molecular analysis revealed that individual target site resistance kdr mutations F1534C and V1016G/I were detected in most Ae. aegypti populations. Additionally, double mutant (F1534C + V1016G/I) mosquitoes were found in some populations, and in one case, triple mutant (F1534C + V1016G/I + S989P) mosquitoes were detected. Metabolic resistance, as reflected by overexpression of three P450 genes (CYP6BB2, CYP9J26, and CYP9J32), was also detected in Ae. aegypti mosquitoes. Our study provides information that could be used to strategize future vector control strategies and highlights the importance of continuing vector surveillance. Future studies should assess the effect of piperonyl butoxide (PBO) on metabolic resistance and identify the different strains of Wolbachia spp., to choose the best vector control strategies in Benin.
Collapse
Affiliation(s)
- S Ateutchia-Ngouanet
- International Institute of Tropical Agriculture (IITA), 08 Tri-Postal, P.O. Box 0932, Cotonou, Benin; Department Microbiology and Parasitology, Faculty of Science, University of Buea, P.O. BOX 63, Buea, Cameroon.
| | - F Nanfack-Minkeu
- International Institute of Tropical Agriculture (IITA), 08 Tri-Postal, P.O. Box 0932, Cotonou, Benin; Department of Biology, The College of Wooster, OH, USA
| | - K Mavridis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 70013, Greece
| | - S Wanji
- Department Microbiology and Parasitology, Faculty of Science, University of Buea, P.O. BOX 63, Buea, Cameroon
| | - M Demanou
- Regional Yellow Fever Laboratory Coordinator World Health Organization, Inter-Country Support Team West Africa, 03 PO BOX 7019 Ouagadougou 03, Burkina Faso
| | - J Vontas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion 70013, Greece; Department of Crop Science, Pesticide Science Laboratory, Agricultural University of Athens, Athens 11855, Greece
| | - R Djouaka
- International Institute of Tropical Agriculture (IITA), 08 Tri-Postal, P.O. Box 0932, Cotonou, Benin
| |
Collapse
|
3
|
Zhu K, Hill C, Muirhead A, Basu M, Brown J, Brinton MA, Hayat MJ, Venegas-Vargas C, Reis MG, Casanovas-Massana A, Meschke JS, Ko AI, Costa F, Stauber CE. Zika virus RNA persistence and recovery in water and wastewater: An approach for Zika virus surveillance in resource-constrained settings. WATER RESEARCH 2023; 241:120116. [PMID: 37270953 PMCID: PMC10330535 DOI: 10.1016/j.watres.2023.120116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/14/2023] [Accepted: 05/22/2023] [Indexed: 06/06/2023]
Abstract
During the 2015-2016 Zika virus (ZIKV) epidemic in the Americas, serological cross-reactivity with other flaviviruses and relatively high costs of nucleic acid testing in the region hindered the capacity for widespread diagnostic testing. In such cases where individual testing is not feasible, wastewater monitoring approaches may offer a means of community-level public health surveillance. To inform such approaches, we characterized the persistence and recovery of ZIKV RNA in experiments where we spiked cultured ZIKV into surface water, wastewater, and a combination of both to examine the potential for detection in open sewers serving communities most affected by the ZIKV outbreak, such as those in Salvador, Bahia, Brazil. We used reverse transcription droplet digital PCR to quantify ZIKV RNA. In our persistence experiments, we found that the persistence of ZIKV RNA decreased with increasing temperature, significantly decreased in surface water versus wastewater, and significantly decreased when the initial concentration of virus was lowered by one order of magnitude. In our recovery experiments, we found higher percent recovery of ZIKV RNA in pellets versus supernatants from the same sample, higher recoveries in pellets using skimmed milk flocculation, lower recoveries of ZIKV RNA in surface water versus wastewater, and lower recoveries from a freeze thaw. We also analyzed samples collected from Salvador, Brazil during the ZIKV outbreak (2015-2016) that consisted of archived samples obtained from open sewers or environmental waters thought to be contaminated by sewage. Although we did not detect any ZIKV RNA in the archived Brazil samples, results from these persistence and recovery experiments serve to inform future wastewater monitoring efforts in open sewers, an understudied and important application of wastewater monitoring.
Collapse
Affiliation(s)
- Kevin Zhu
- Department of Civil and Environmental Engineering, College of Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Cailee Hill
- Department of Population Health Sciences, School of Public Health, Georgia State University, Atlanta, GA 30303, USA
| | - Aaron Muirhead
- Department of Population Health Sciences, School of Public Health, Georgia State University, Atlanta, GA 30303, USA
| | - Mausumi Basu
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 303034, USA
| | - Joe Brown
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Margo A Brinton
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA 303034, USA
| | - Matthew J Hayat
- Department of Population Health Sciences, School of Public Health, Georgia State University, Atlanta, GA 30303, USA
| | - Cristina Venegas-Vargas
- Department of Large Animal Clinical Sciences, College Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Mitermayer G Reis
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Rua Waldemar Falcão, 121, Salvador Bahia, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06511, USA
| | - Arnau Casanovas-Massana
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06511, USA
| | - J Scott Meschke
- Department of Environmental and Occupational Health, School of Public Health, University of Washington, Seattle, WA, USA
| | - Albert I Ko
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Rua Waldemar Falcão, 121, Salvador Bahia, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06511, USA
| | - Federico Costa
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz, Ministério da Saúde, Rua Waldemar Falcão, 121, Salvador Bahia, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06511, USA; Institute of Collective Health, Federal University of Bahia, Canela, Salvador 40110-040, Brazil
| | - Christine E Stauber
- Department of Population Health Sciences, School of Public Health, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
4
|
Franco EJ, Cella E, Tao X, Hanrahan KC, Azarian T, Brown AN. Favipiravir Suppresses Zika Virus (ZIKV) through Activity as a Mutagen. Microorganisms 2023; 11:1342. [PMID: 37317316 PMCID: PMC10224157 DOI: 10.3390/microorganisms11051342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/09/2023] [Accepted: 05/17/2023] [Indexed: 06/16/2023] Open
Abstract
In a companion paper, we demonstrated that the nucleoside analogue favipiravir (FAV) suppressed Zika virus (ZIKV) replication in three human-derived cell lines-HeLa, SK-N-MC, and HUH-7. Our results revealed that FAV's effect was most pronounced in HeLa cells. In this work, we aimed to explain variation in FAV activity, investigating its mechanism of action and characterizing host cell factors relevant to tissue-specific differences in drug effect. Using viral genome sequencing, we show that FAV therapy was associated with an increase in the number of mutations and promoted the production of defective viral particles in all three cell lines. Our findings demonstrate that defective viral particles made up a larger portion of the viral population released from HeLa cells both at increasing FAV concentrations and at increasing exposure times. Taken together, our companion papers show that FAV acts via lethal mutagenesis against ZIKV and highlight the host cell's influence on the activation and antiviral activity of nucleoside analogues. Furthermore, the information gleaned from these companion papers can be applied to gain a more comprehensive understanding of the activity of nucleoside analogues and the impact of host cell factors against other viral infections for which we currently have no approved antiviral therapies.
Collapse
Affiliation(s)
- Evelyn J. Franco
- Institute for Therapeutic Innovation, Department of Medicine, College of Medicine, University of Florida, Orlando, FL 32827, USA; (E.J.F.); (K.C.H.)
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA;
| | - Eleonora Cella
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, USA; (E.C.); (T.A.)
| | - Xun Tao
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA;
| | - Kaley C. Hanrahan
- Institute for Therapeutic Innovation, Department of Medicine, College of Medicine, University of Florida, Orlando, FL 32827, USA; (E.J.F.); (K.C.H.)
| | - Taj Azarian
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL 32827, USA; (E.C.); (T.A.)
| | - Ashley N. Brown
- Institute for Therapeutic Innovation, Department of Medicine, College of Medicine, University of Florida, Orlando, FL 32827, USA; (E.J.F.); (K.C.H.)
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA;
| |
Collapse
|
5
|
Fraiture MA, Coucke W, Pol M, Rousset D, Gourinat AC, Biron A, Broeders S, Vandermassen E, Dupont-Rouzeyrol M, Roosens NHC. Non-Invasive versus Invasive Samples for Zika Virus Surveillance: A Comparative Study in New Caledonia and French Guiana in 2015-2016. Microorganisms 2021; 9:microorganisms9061312. [PMID: 34208593 PMCID: PMC8235784 DOI: 10.3390/microorganisms9061312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/18/2022] Open
Abstract
Zika virus, an arbovirus responsible for major outbreaks, can cause serious health issues, such as neurological diseases. In the present study, different types of samples (serum, saliva, and urine), collected in 2015–2016 in New Caledonia and French Guiana from 53 patients presenting symptoms and clinical signs triggered by arbovirus infections, were analyzed using a recently developed, and in-house validated, 4-plex RT-qPCR TaqMan method for simultaneous detection and discrimination of the Zika and Chikungunya viruses. Subsequently, statistical analyses were performed in order to potentially establish recommendations regarding the choice of samples type to use for an efficient and early stage Zika infection diagnosis. On this basis, the use of only urine samples presented the highest probability to detect viral RNA from Zika virus. Moreover, such a probability was improved using both urine and saliva samples. Consequently, the added value of non-invasive samples, associated with a higher acceptance level for collection among patients, instead of serum samples, for the detection of Zika infections was illustrated.
Collapse
Affiliation(s)
- Marie-Alice Fraiture
- Transversal & Applied Genomics (TAG), Sciensano, rue Juliette Wytsman 14, 1050 Brussels, Belgium; (M.-A.F.); (S.B.); (E.V.)
| | - Wim Coucke
- Quality of Laboratories, Sciensano, rue Juliette Wytsman 14, 1050 Brussels, Belgium;
| | - Morgane Pol
- URE Dengue et Arboviroses, Institut Pasteur of New Caledonia, 11 avenue Paul Doumer, BP 61, CEDEX, 98845 Noumea, New Caledonia; (M.P.); (A.-C.G.); (A.B.); (M.D.-R.)
| | - Dominique Rousset
- Laboratoire de Virologie CNR Arbovirus, Institut Pasteur de la Guyane, 23 avenue Pasteur, BP 6010, CEDEX, 97306 Cayenne, French Guiana;
| | - Ann-Claire Gourinat
- URE Dengue et Arboviroses, Institut Pasteur of New Caledonia, 11 avenue Paul Doumer, BP 61, CEDEX, 98845 Noumea, New Caledonia; (M.P.); (A.-C.G.); (A.B.); (M.D.-R.)
| | - Antoine Biron
- URE Dengue et Arboviroses, Institut Pasteur of New Caledonia, 11 avenue Paul Doumer, BP 61, CEDEX, 98845 Noumea, New Caledonia; (M.P.); (A.-C.G.); (A.B.); (M.D.-R.)
| | - Sylvia Broeders
- Transversal & Applied Genomics (TAG), Sciensano, rue Juliette Wytsman 14, 1050 Brussels, Belgium; (M.-A.F.); (S.B.); (E.V.)
- Quality of Laboratories, Sciensano, rue Juliette Wytsman 14, 1050 Brussels, Belgium;
| | - Els Vandermassen
- Transversal & Applied Genomics (TAG), Sciensano, rue Juliette Wytsman 14, 1050 Brussels, Belgium; (M.-A.F.); (S.B.); (E.V.)
| | - Myrielle Dupont-Rouzeyrol
- URE Dengue et Arboviroses, Institut Pasteur of New Caledonia, 11 avenue Paul Doumer, BP 61, CEDEX, 98845 Noumea, New Caledonia; (M.P.); (A.-C.G.); (A.B.); (M.D.-R.)
| | - Nancy H. C. Roosens
- Transversal & Applied Genomics (TAG), Sciensano, rue Juliette Wytsman 14, 1050 Brussels, Belgium; (M.-A.F.); (S.B.); (E.V.)
- Correspondence: ; Tel.: +32-(0)-2-642-52-58
| |
Collapse
|
6
|
Yang D, Chu H, Lu G, Shuai H, Wang Y, Hou Y, Zhang X, Huang X, Hu B, Chai Y, Yuen TTT, Zhao X, Lee ACY, Ye Z, Li C, Chik KKH, Zhang AJ, Zhou J, Yuan S, Chan JFW. STAT2-dependent restriction of Zika virus by human macrophages but not dendritic cells. Emerg Microbes Infect 2021; 10:1024-1037. [PMID: 33979266 PMCID: PMC8205058 DOI: 10.1080/22221751.2021.1929503] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Zika virus (ZIKV) is an emerging mosquito-borne flavivirus that poses significant threats to global public health. Macrophages and dendritic cells are both key sentinel cells in the host immune response and play critical roles in the pathogenesis of flavivirus infections. Recent studies showed that ZIKV could productively infect monocyte-derived dendritic cells (moDCs), but the role of macrophages in ZIKV infection remains incompletely understood. In this study, we first compared ZIKV infection in monocyte-derived macrophages (MDMs) and moDCs derived from the same donors. We demonstrated that while both MDMs and moDCs were susceptible to epidemic (Puerto Rico) and pre-epidemic (Uganda) strains of ZIKV, virus replication was largely restricted in MDMs but not in moDCs. ZIKV induced significant apoptosis in moDCs but not MDMs. The restricted virus replication in MDMs was not due to inefficient virus entry but was related to post-entry events in the viral replication cycle. In stark contrast with moDCs, ZIKV failed to inhibit STAT1 and STAT2 phosphorylation in MDMs. This resulted in the lack of efficient antagonism of the host type I interferon-mediated antiviral responses. Importantly, depletion of STAT2 but not STAT1 in MDMs significantly rescued the replication of ZIKV and the prototype flavivirus yellow fever virus. Overall, our findings revealed a differential interplay between macrophages and dendritic cells with ZIKV. While dendritic cells may be exploited by ZIKV to facilitate virus replication, macrophages restricted ZIKV infection.
Collapse
Affiliation(s)
- Dong Yang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Gang Lu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, People's Republic of China.,Hainan-Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou, Hainan, People's Republic of China, and the The University of Hong Kong, Pokfulam, People's Republic of China.,Department of Pathogen Biology, Hainan Medical University, Haikou, Hainan, People's Republic of China
| | - Huiping Shuai
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Yixin Wang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Yuxin Hou
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Xi Zhang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Xiner Huang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Bingjie Hu
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Yue Chai
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Terrence Tsz-Tai Yuen
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Xiaoyu Zhao
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Andrew Chak-Yiu Lee
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Ziwei Ye
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Cun Li
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Kenn Ka-Heng Chik
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Anna Jinxia Zhang
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Jie Zhou
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Pokfulam, People's Republic of China.,Hainan-Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou, Hainan, People's Republic of China, and the The University of Hong Kong, Pokfulam, People's Republic of China.,Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, People's Republic of China.,Department of Microbiology, Queen Mary Hospital, Pokfulam, People's Republic of China.,Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People's Republic of China
| |
Collapse
|
7
|
Zhang X, Li G, Chen G, Zhu N, Wu D, Wu Y, James TD. Recent progresses and remaining challenges for the detection of Zika virus. Med Res Rev 2021; 41:2039-2108. [PMID: 33559917 DOI: 10.1002/med.21786] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 12/26/2022]
Abstract
Zika virus (ZIKV) has emerged as a particularly notorious mosquito-borne flavivirus, which can lead to a devastating congenital syndrome in the fetuses of pregnant mothers (e.g., microcephaly, spasticity, craniofacial disproportion, miscarriage, and ocular abnormalities) and cause the autoimmune disorder Guillain-Barre' syndrome of adults. Due to its severity and rapid dispersal over several continents, ZIKV has been acknowledged to be a global health concern by the World Health Organization. Unfortunately, the ZIKV has recently resurged in India with the potential for devastating effects. Researchers from all around the world have worked tirelessly to develop effective detection strategies and vaccines for the prevention and control of ZIKV infection. In this review, we comprehensively summarize the most recent research into ZIKV, including the structural biology and evolution, historical overview, pathogenesis, symptoms, and transmission. We then focus on the detection strategies for ZIKV, including viral isolation, serological assays, molecular assays, sensing methods, reverse transcription loop mediated isothermal amplification, transcription-mediated amplification technology, reverse transcription strand invasion based amplification, bioplasmonic paper-based device, and reverse transcription isothermal recombinase polymerase amplification. To conclude, we examine the limitations of currently available strategies for the detection of ZIKV, and outline future opportunities and research challenges.
Collapse
Affiliation(s)
- Xianlong Zhang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Guoliang Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Guang Chen
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Niu Zhu
- Department of Public Health, Xi'an Medical University, Xi'an, China
| | - Di Wu
- Institute for Global Food Security, Queen's University Belfast, Belfast, UK
| | - Yongning Wu
- NHC Key Laboratory of Food Safety Risk Assessment, Food Safety Research Unit (2019RU014) of Chinese Academy of Medical Science, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Tony D James
- Department of Chemistry, University of Bath, Bath, UK.,School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| |
Collapse
|
8
|
VectorDisk: A Microfluidic Platform Integrating Diagnostic Markers for Evidence-Based Mosquito Control. Processes (Basel) 2020. [DOI: 10.3390/pr8121677] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Effective mosquito monitoring relies on the accurate identification and characterization of the target population. Since this process requires specialist knowledge and equipment that is not widely available, automated field-deployable systems are highly desirable. We present a centrifugal microfluidic cartridge, the VectorDisk, which integrates TaqMan PCR assays in two feasibility studies, aiming to assess multiplexing capability, specificity, and reproducibility in detecting disk-integrated vector-related assays. In the first study, pools of 10 mosquitoes were used as samples. We tested 18 disks with 27 DNA and RNA assays each, using a combination of multiple microfluidic chambers and detection wavelengths (geometric and color multiplexing) to identify mosquito and malaria parasite species as well as insecticide resistance mechanisms. In the second study, purified nucleic acids served as samples to test arboviral and malaria infective mosquito assays. Nine disks were tested with 14 assays each. No false positive results were detected on any of the disks. The coefficient of variation in reproducibility tests was <10%. The modular nature of the platform, the easy adaptation of the primer/probe panels, the cold chain independence, the rapid (2–3 h) analysis, and the assay multiplexing capacity are key features, rendering the VectorDisk a potential candidate for automated vector analysis.
Collapse
|
9
|
Giovannoni F, Bosch I, Polonio CM, Torti MF, Wheeler MA, Li Z, Romorini L, Rodriguez Varela MS, Rothhammer V, Barroso A, Tjon EC, Sanmarco LM, Takenaka MC, Modaresi SMS, Gutiérrez-Vázquez C, Zanluqui NG, Dos Santos NB, Munhoz CD, Wang Z, Damonte EB, Sherr D, Gehrke L, Peron JPS, Garcia CC, Quintana FJ. AHR is a Zika virus host factor and a candidate target for antiviral therapy. Nat Neurosci 2020; 23:939-951. [PMID: 32690969 DOI: 10.1038/s41593-020-0664-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 06/03/2020] [Indexed: 02/07/2023]
Abstract
Zika virus (ZIKV) is a flavivirus linked to multiple birth defects including microcephaly, known as congenital ZIKV syndrome. The identification of host factors involved in ZIKV replication may guide efficacious therapeutic interventions. In genome-wide transcriptional studies, we found that ZIKV infection triggers aryl hydrocarbon receptor (AHR) activation. Specifically, ZIKV infection induces kynurenine (Kyn) production, which activates AHR, limiting the production of type I interferons (IFN-I) involved in antiviral immunity. Moreover, ZIKV-triggered AHR activation suppresses intrinsic immunity driven by the promyelocytic leukemia (PML) protein, which limits ZIKV replication. AHR inhibition suppressed the replication of multiple ZIKV strains in vitro and also suppressed replication of the related flavivirus dengue. Finally, AHR inhibition with a nanoparticle-delivered AHR antagonist or an inhibitor developed for human use limited ZIKV replication and ameliorated newborn microcephaly in a murine model. In summary, we identified AHR as a host factor for ZIKV replication and PML protein as a driver of anti-ZIKV intrinsic immunity.
Collapse
Affiliation(s)
- Federico Giovannoni
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Laboratorio de Estrategias Antivirales, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. CONICET-Instituto de Química Biológica, Buenos Aires, Argentina
| | - Irene Bosch
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | - Carolina Manganeli Polonio
- Neuroimmune Interactions Laboratory, Immunology Department-ICB IV, University of São Paulo, São Paulo, Brazil.,Scientific Platform Pasteur-USP, University of São Paulo, São Paulo, Brazil
| | - María F Torti
- Laboratorio de Estrategias Antivirales, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. CONICET-Instituto de Química Biológica, Buenos Aires, Argentina
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhaorong Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Leonardo Romorini
- Laboratorio de Investigación aplicada a Neurociencias, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia, Buenos Aires, Argentina
| | - María S Rodriguez Varela
- Laboratorio de Investigación aplicada a Neurociencias, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia, Buenos Aires, Argentina
| | - Veit Rothhammer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andreia Barroso
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily C Tjon
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Liliana M Sanmarco
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maisa C Takenaka
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Cristina Gutiérrez-Vázquez
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nágela Ghabdan Zanluqui
- Scientific Platform Pasteur-USP, University of São Paulo, São Paulo, Brazil.,Immunopathology and Allergy Post Graduate Program, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Nilton Barreto Dos Santos
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Carolina Demarchi Munhoz
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Zhongyan Wang
- Dept. of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Elsa B Damonte
- Laboratorio de Estrategias Antivirales, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. CONICET-Instituto de Química Biológica, Buenos Aires, Argentina
| | - David Sherr
- Dept. of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Lee Gehrke
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Program in Virology, Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - Jean Pierre Schatzmann Peron
- Neuroimmune Interactions Laboratory, Immunology Department-ICB IV, University of São Paulo, São Paulo, Brazil. .,Scientific Platform Pasteur-USP, University of São Paulo, São Paulo, Brazil. .,Immunopathology and Allergy Post Graduate Program, School of Medicine, University of São Paulo, São Paulo, Brazil.
| | - Cybele C Garcia
- Laboratorio de Estrategias Antivirales, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires. CONICET-Instituto de Química Biológica, Buenos Aires, Argentina.
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
10
|
Chu H, Yuen TTT, Chik KKH, Yuan S, Shuai H, Zou Z, Wang Y, Zhu Z, Yang D, Poon VKM, Chan CCS, Zhou J, Yin F, Kok KH, Yuen KY, Chan JFW. Targeting the Inositol-Requiring Enzyme-1 Pathway Efficiently Reverts Zika Virus-Induced Neurogenesis and Spermatogenesis Marker Perturbations. ACS Infect Dis 2020; 6:1745-1758. [PMID: 32329611 DOI: 10.1021/acsinfecdis.9b00526] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Zika virus (ZIKV) is an emerging flavivirus that may be associated with congenital anomalies in infected fetuses and severe neurological and genital tract complications in infected adults. Currently, antiviral treatments to revert these ZIKV-induced complications are lacking. ZIKV infection has recently been suggested to upregulate the host unfolded protein response, which may contribute to the congenital neurological anomalies. As an extension from these findings, we thoroughly investigated the ZIKV-induced unfolded protein response using a combination of the neuronal cell line, induced pluripotent stem cell-derived human neuronal stem and progenitor cells, and an interferon receptor-deficient A129 mouse model. Our results revealed a critical contribution of the inositol-requiring enzyme-1 (IRE1) arm of the unfolded protein response to ZIKV-induced neurological and testicular complications. Importantly, the inhibition of the IRE1 signaling pathway activation with KIRA6 (kinase-inhibiting RNAse attenuator 6), a selective small molecule IRE1 inhibitor that promotes cell survival, potently reverted the ZIKV-induced perturbations of the key gene expressions associated with neurogenesis and spermatogenesis in vitro and in vivo, highlighting the potential of IRE1 inhibition as a novel host-targeting antiviral strategy in combating against ZIKV-induced neurological and testicular pathologies.
Collapse
Affiliation(s)
- Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Terrence T. T. Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Kenn K. H. Chik
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Huiping Shuai
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Zijiao Zou
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Yixin Wang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Zheng Zhu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Dong Yang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Vincent K. M. Poon
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Chris C. S. Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Jie Zhou
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Feifei Yin
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University and The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, Hainan 571199, China
- Department of Pathogen Biology, Hainan Medical University, Haikou, Hainan 571199, China
| | - Kin-Hang Kok
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University and The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, Hainan 571199, China
- Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| | - Jasper F. W. Chan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University and The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, Haikou, Hainan 571199, China
- Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region 999077, China
| |
Collapse
|
11
|
Ou TP, Yun C, Auerswald H, In S, Leang R, Huy R, Choeung R, Dussart P, Duong V. Improved detection of dengue and Zika viruses using multiplex RT-qPCR assays. J Virol Methods 2020; 282:113862. [PMID: 32417207 DOI: 10.1016/j.jviromet.2020.113862] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 11/30/2022]
Abstract
Dengue virus (DENV) and Zika virus (ZIKV) are important viral pathogens, known to cause human infections with similar symptoms, are transmitted by common vectors and co-circulate in intertropical regions. Moreover, dengue fever results from infection with one of four different serotypes of dengue virus. Considering the recent ZIKV emergence, multiplex and up-to-date assays are more preferable for detection of both viruses in a single reaction. This study aimed to develop: (i) an one-step duplex real-time reverse transcription polymerase chain reaction (RT-qPCR) assay to efficiently and simultaneously detect and quantify DENV and ZIKV; (ii) a fourplex RT-qPCR to differentiate and quantify the four DENV serotypes. The detection limit of the duplex assay was 0.028 and 0.065 FFU (focus forming unit)/ml for DENV and ZIKV respectively. The lower limit of analytical sensitivity of fourplex assay was 0.01 FFU/ml for DENV-1 and 0.1 FFU/ml for DENV-2,-3 and -4. The assessment of specificity indicated both assays were highly specific to targeted viruses with negative results for other Flaviviridae such as Japanese encephalitis, West Nile, Yellow fever or Hepatitis C viruses. The newly developed RT-qPCRs were shown to be more sensitive than a previously described assay in detecting DENV in clinical samples and are suitable for the routine diagnosis.
Collapse
Affiliation(s)
- Tey Putita Ou
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia.
| | - Chanvannak Yun
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia.
| | - Heidi Auerswald
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia.
| | - Saraden In
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia.
| | - Rithea Leang
- National Center for Parasitology, Entomology & Malaria Control, Phnom Penh, Cambodia.
| | - Rekol Huy
- National Center for Parasitology, Entomology & Malaria Control, Phnom Penh, Cambodia.
| | - Rithy Choeung
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia.
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia.
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia.
| |
Collapse
|
12
|
Improved Molecular Diagnosis of COVID-19 by the Novel, Highly Sensitive and Specific COVID-19-RdRp/Hel Real-Time Reverse Transcription-PCR Assay Validated In Vitro and with Clinical Specimens. J Clin Microbiol 2020; 58:JCM.00310-20. [PMID: 32132196 PMCID: PMC7180250 DOI: 10.1128/jcm.00310-20] [Citation(s) in RCA: 654] [Impact Index Per Article: 130.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022] Open
Abstract
On 31 December 2019, the World Health Organization was informed of a cluster of cases of pneumonia of unknown etiology in Wuhan, China. Subsequent investigations identified a novel coronavirus, now named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), from the affected patients. Highly sensitive and specific laboratory diagnostics are important for controlling the rapidly evolving SARS-CoV-2-associated coronavirus disease 2019 (COVID-19) epidemic. In this study, we developed and compared the performance of three novel real-time reverse transcription-PCR (RT-PCR) assays targeting the RNA-dependent RNA polymerase (RdRp)/helicase (Hel), spike (S), and nucleocapsid (N) genes of SARS-CoV-2 with that of the reported RdRp-P2 assay, which is used in >30 European laboratories. On 31 December 2019, the World Health Organization was informed of a cluster of cases of pneumonia of unknown etiology in Wuhan, China. Subsequent investigations identified a novel coronavirus, now named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), from the affected patients. Highly sensitive and specific laboratory diagnostics are important for controlling the rapidly evolving SARS-CoV-2-associated coronavirus disease 2019 (COVID-19) epidemic. In this study, we developed and compared the performance of three novel real-time reverse transcription-PCR (RT-PCR) assays targeting the RNA-dependent RNA polymerase (RdRp)/helicase (Hel), spike (S), and nucleocapsid (N) genes of SARS-CoV-2 with that of the reported RdRp-P2 assay, which is used in >30 European laboratories. Among the three novel assays, the COVID-19-RdRp/Hel assay had the lowest limit of detection in vitro (1.8 50% tissue culture infective doses [TCID50]/ml with genomic RNA and 11.2 RNA copies/reaction with in vitro RNA transcripts). Among 273 specimens from 15 patients with laboratory-confirmed COVID-19 in Hong Kong, 77 (28.2%) were positive by both the COVID-19-RdRp/Hel and RdRp-P2 assays. The COVID-19-RdRp/Hel assay was positive for an additional 42 RdRp-P2-negative specimens (119/273 [43.6%] versus 77/273 [28.2%]; P < 0.001), including 29/120 (24.2%) respiratory tract specimens and 13/153 (8.5%) non-respiratory tract specimens. The mean viral load of these specimens was 3.21 × 104 RNA copies/ml (range, 2.21 × 102 to 4.71 × 105 RNA copies/ml). The COVID-19-RdRp/Hel assay did not cross-react with other human-pathogenic coronaviruses and respiratory pathogens in cell culture and clinical specimens, whereas the RdRp-P2 assay cross-reacted with SARS-CoV in cell culture. The highly sensitive and specific COVID-19-RdRp/Hel assay may help to improve the laboratory diagnosis of COVID-19.
Collapse
|
13
|
Yip CCY, Ho CC, Chan JFW, To KKW, Chan HSY, Wong SCY, Leung KH, Fung AYF, Ng ACK, Zou Z, Tam AR, Chung TWH, Chan KH, Hung IFN, Cheng VCC, Tsang OTY, Tsui SKW, Yuen KY. Development of a Novel, Genome Subtraction-Derived, SARS-CoV-2-Specific COVID-19-nsp2 Real-Time RT-PCR Assay and Its Evaluation Using Clinical Specimens. Int J Mol Sci 2020; 21:E2574. [PMID: 32276333 PMCID: PMC7177594 DOI: 10.3390/ijms21072574] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 01/02/2023] Open
Abstract
The pandemic novel coronavirus infection, Coronavirus Disease 2019 (COVID-19), has affected at least 190 countries or territories, with 465,915 confirmed cases and 21,031 deaths. In a containment-based strategy, rapid, sensitive and specific testing is important in epidemiological control and clinical management. Using 96 SARS-CoV-2 and 104 non-SARS-CoV-2 coronavirus genomes and our in-house program, GolayMetaMiner, four specific regions longer than 50 nucleotides in the SARS-CoV-2 genome were identified. Primers were designed to target the longest and previously untargeted nsp2 region and optimized as a probe-free real-time reverse transcription-polymerase chain reaction (RT-PCR) assay. The new COVID-19-nsp2 assay had a limit of detection (LOD) of 1.8 TCID50/mL and did not amplify other human-pathogenic coronaviruses and respiratory viruses. Assay reproducibility in terms of cycle threshold (Cp) values was satisfactory, with the total imprecision (% CV) values well below 5%. Evaluation of the new assay using 59 clinical specimens from 14 confirmed cases showed 100% concordance with our previously developed COVID-19-RdRp/Hel reference assay. A rapid, sensitive, SARS-CoV-2-specific real-time RT-PCR assay, COVID-19-nsp2, was developed.
Collapse
Affiliation(s)
- Cyril Chik-Yan Yip
- Department of Microbiology, Queen Mary Hospital, HKSAR, Hong Kong, China; (C.C.-Y.Y.); (T.W.-H.C.); (V.C.-C.C.)
| | - Chi-Chun Ho
- Genomics and Bioinformatics Programme, The Chinese University of Hong Kong, HKSAR, Hong Kong, China;
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, HKSAR, Hong Kong, China; (J.F.-W.C.); (K.K.-W.T.); (K.-H.C.)
- Department of Microbiology, The University of Hong Kong, HKSAR, Hong Kong, China; (K.-H.L.); (A.Y.-F.F.); (A.C.-K.N.); (Z.Z.)
- Department of Clinical Microbiology and Infection, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, HKSAR, Hong Kong, China
| | - Kelvin Kai-Wang To
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, HKSAR, Hong Kong, China; (J.F.-W.C.); (K.K.-W.T.); (K.-H.C.)
- Department of Microbiology, The University of Hong Kong, HKSAR, Hong Kong, China; (K.-H.L.); (A.Y.-F.F.); (A.C.-K.N.); (Z.Z.)
- Department of Clinical Microbiology and Infection, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, HKSAR, Hong Kong, China
| | | | | | - Kit-Hang Leung
- Department of Microbiology, The University of Hong Kong, HKSAR, Hong Kong, China; (K.-H.L.); (A.Y.-F.F.); (A.C.-K.N.); (Z.Z.)
| | - Agnes Yim-Fong Fung
- Department of Microbiology, The University of Hong Kong, HKSAR, Hong Kong, China; (K.-H.L.); (A.Y.-F.F.); (A.C.-K.N.); (Z.Z.)
| | - Anthony Chin-Ki Ng
- Department of Microbiology, The University of Hong Kong, HKSAR, Hong Kong, China; (K.-H.L.); (A.Y.-F.F.); (A.C.-K.N.); (Z.Z.)
| | - Zijiao Zou
- Department of Microbiology, The University of Hong Kong, HKSAR, Hong Kong, China; (K.-H.L.); (A.Y.-F.F.); (A.C.-K.N.); (Z.Z.)
| | | | - Tom Wai-Hin Chung
- Department of Microbiology, Queen Mary Hospital, HKSAR, Hong Kong, China; (C.C.-Y.Y.); (T.W.-H.C.); (V.C.-C.C.)
| | - Kwok-Hung Chan
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, HKSAR, Hong Kong, China; (J.F.-W.C.); (K.K.-W.T.); (K.-H.C.)
- Department of Microbiology, The University of Hong Kong, HKSAR, Hong Kong, China; (K.-H.L.); (A.Y.-F.F.); (A.C.-K.N.); (Z.Z.)
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, HKSAR, Hong Kong, China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, The University of Hong Kong, HKSAR, Hong Kong, China;
| | - Vincent Chi-Chung Cheng
- Department of Microbiology, Queen Mary Hospital, HKSAR, Hong Kong, China; (C.C.-Y.Y.); (T.W.-H.C.); (V.C.-C.C.)
| | - Owen Tak-Yin Tsang
- Department of Medicine and Geriatrics, Princess Margaret Hospital, HKSAR, Hong Kong, China;
| | - Stephen Kwok Wing Tsui
- School of Biomedical Sciences, The Chinese University of Hong Kong, HKSAR, Hong Kong, China;
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, HKSAR, Hong Kong, China; (J.F.-W.C.); (K.K.-W.T.); (K.-H.C.)
- Department of Microbiology, The University of Hong Kong, HKSAR, Hong Kong, China; (K.-H.L.); (A.Y.-F.F.); (A.C.-K.N.); (Z.Z.)
- Department of Clinical Microbiology and Infection, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
- Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, HKSAR, Hong Kong, China
| |
Collapse
|
14
|
Kim J, Jung S, Kim MY, Kim BK, Kwon SH, Kim SK. Thermo-Responsive Polymer Capsules in Real-Time One-Step RT-PCR for Highly Multiplex RNA Analysis. Adv Healthc Mater 2020; 9:e1900790. [PMID: 32134572 DOI: 10.1002/adhm.201900790] [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: 06/19/2019] [Revised: 12/13/2019] [Indexed: 11/09/2022]
Abstract
Rapid and simple detection of RNA targets is in high demand due to the growing threat of pandemic viruses. One-step real-time, reverse transcription-polymerase chain reaction (One-step RT-qPCR) using a controlled release system of thermo-responsive materials is developed in this paper to enable high-fidelity RNA analysis as suppressing by-products. The nanocapsules, consisting of upper critical solution temperature (UCST) material and PCR primers, carry or release the primers depending upon the temperature. The UCST nanocapsules are introduced into hydrogel microparticles incorporated with RT primers and then the target RNA is selectively amplified in the microparticle through one-step RT-qPCR. Severe side products are sharply subdued by separating the PCR primers from the RT process by means of the microparticles with nanocapsules. Because the one-step assay is now implemented in a single microparticle, multiple target RNAs can be analyzed in a simple RT-qPCR of multiple particles. Reliable 18-plex one-step RT-qPCR is successfully conducted within 30 min using single-color fluorescent optics. This work also explains the facile fabrication processes used for the thermo-responsive nanocapsules and hydrogel microparticles by the blending polymerization method. Extensible multiplex analysis of influenza virus demonstrates the versatile uses of this one-step RT-qPCR platform.
Collapse
Affiliation(s)
- Junsun Kim
- Center for Molecular Recognition ResearchMaterials and Life Science Research DivisionKorea Institute of Science and Technology Seoul 02792 Republic of Korea
- Chemical and Biological EngineeringKorea University Seoul 02841 Republic of Korea
| | - Seungwon Jung
- Center for Molecular Recognition ResearchMaterials and Life Science Research DivisionKorea Institute of Science and Technology Seoul 02792 Republic of Korea
- Applied ChemistryKyung Hee University Yongin 17104 Republic of Korea
| | - Mi Yeon Kim
- Center for Molecular Recognition ResearchMaterials and Life Science Research DivisionKorea Institute of Science and Technology Seoul 02792 Republic of Korea
- Chemical and Biological EngineeringKorea University Seoul 02841 Republic of Korea
| | - Bong Kyun Kim
- Center for Molecular Recognition ResearchMaterials and Life Science Research DivisionKorea Institute of Science and Technology Seoul 02792 Republic of Korea
- Biomedical EngineeringUniversity of Science and Technology (UST) Daejeon 34113 Republic of Korea
| | - Soon Hwan Kwon
- Armed Forces Medical Research Institute Daejeon 34059 Republic of Korea
| | - Sang Kyung Kim
- Center for Molecular Recognition ResearchMaterials and Life Science Research DivisionKorea Institute of Science and Technology Seoul 02792 Republic of Korea
- Biomedical EngineeringUniversity of Science and Technology (UST) Daejeon 34113 Republic of Korea
| |
Collapse
|
15
|
Jorge FA, Thomazella MV, Castro Moreira D, Lopes LDG, Teixeira JJV, Bertolini DA. Evolutions and upcoming on Zika virus diagnosis through an outbreak: A systematic review. Rev Med Virol 2020; 30:e2105. [DOI: 10.1002/rmv.2105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/12/2020] [Accepted: 03/15/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Fernando A. Jorge
- Department of Clinical Analysis and BiomedicineState University of Maringá Maringá Brazil
| | - Mateus V. Thomazella
- Medical Research Laboratory, School of MedicineUniversity of São Paulo São Paulo Brazil
| | - Deborah Castro Moreira
- Department of Clinical Analysis and BiomedicineState University of Maringá Maringá Brazil
| | - Luciana D. G. Lopes
- Department of Clinical Analysis and BiomedicineState University of Maringá Maringá Brazil
| | - Jorge J. V. Teixeira
- Department of Clinical Analysis and BiomedicineState University of Maringá Maringá Brazil
| | - Dennis A. Bertolini
- Department of Clinical Analysis and BiomedicineState University of Maringá Maringá Brazil
| |
Collapse
|
16
|
Guo Q, Chan JFW, Poon VKM, Wu S, Chan CCS, Hou L, Yip CCY, Ren C, Cai JP, Zhao M, Zhang AJ, Song X, Chan KH, Wang B, Kok KH, Wen Y, Yuen KY, Chen W. Immunization With a Novel Human Type 5 Adenovirus-Vectored Vaccine Expressing the Premembrane and Envelope Proteins of Zika Virus Provides Consistent and Sterilizing Protection in Multiple Immunocompetent and Immunocompromised Animal Models. J Infect Dis 2019; 218:365-377. [PMID: 29617816 DOI: 10.1093/infdis/jiy187] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/28/2018] [Indexed: 12/30/2022] Open
Abstract
Background Zika virus (ZIKV) infection may be associated with severe complications and disseminated via both vector-borne and nonvector-borne routes. Adenovirus-vectored vaccines represent a favorable controlling measure for the ZIKV epidemic because they have been shown to be safe, immunogenic, and rapidly generable for other emerging viral infections. Evaluations of 2 previously reported adenovirus-vectored ZIKV vaccines were performed using nonlethal animal models and/or nonepidemic ZIKV strain. Methods We constructed 2 novel human adenovirus 5 (Ad5)-vectored vaccines containing the ZIKV premembrane-envelope (Ad5-Sig-prM-Env) and envelope (Ad5-Env) proteins, respectively, and evaluated them in multiple nonlethal and lethal animal models using epidemic ZIKV strains. Results Both vaccines elicited robust humoral and cellular immune responses in immunocompetent BALB/c mice. Dexamethasone-immunosuppressed mice vaccinated with either vaccine demonstrated robust and durable antibody responses and significantly lower blood and tissue viral loads than controls (P < .05). Similar findings were also observed in interferon-α/β receptor-deficient A129 mice. In both of these immunocompromised animal models, Ad5-Sig-prM-Env-vaccinated mice had significantly (P < .05) higher titers of anti-ZIKV-specific neutralizing antibody titers and lower (undetectable) viral loads than Ad5-Env-vaccinated mice. The close correlation between the neutralizing antibody titer and viral load helped to explain the better protective effect of Ad5-Sig-prM-Env than Ad5-Env. Anamnestic response was absent in Ad5-Sig-prM-Env-vaccinated A129 mice. Conclusions Ad5-Sig-prM-Env provided sterilizing protection against ZIKV infection in mice.
Collapse
Affiliation(s)
- Qiang Guo
- Beijing Institute of Biotechnology, China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Vincent Kwok-Man Poon
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, China
| | - Chris Chung-Sing Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, China
| | - Cyril Chik-Yan Yip
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | | | - Jian-Piao Cai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | | | - Anna Jinxia Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | | | - Kwok-Hung Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Busen Wang
- Beijing Institute of Biotechnology, China
| | - Kin-Hang Kok
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yanbo Wen
- Beijing Institute of Biotechnology, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Wei Chen
- Beijing Institute of Biotechnology, China
| |
Collapse
|
17
|
Valiant WG, Mattapallil MJ, Higgs S, Huang YJS, Vanlandingham DL, Lewis MG, Mattapallil JJ. Simultaneous Coinfection of Macaques with Zika and Dengue Viruses Does not Enhance Acute Plasma Viremia but Leads to Activation of Monocyte Subsets and Biphasic Release of Pro-inflammatory Cytokines. Sci Rep 2019; 9:7877. [PMID: 31133721 PMCID: PMC6536518 DOI: 10.1038/s41598-019-44323-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 05/15/2019] [Indexed: 01/06/2023] Open
Abstract
The consequences of simultaneous infection with Zika (ZIKV) and Dengue (DENV) viruses are poorly understood. Here we show that rhesus macaques experimentally coinfected simultaneously with ZIKV and DENV-2 demonstrated ZIKV or DENV replication without an enhancement of either infection. Coinfection was accompanied by an increase in the proportions of CD14+CD16+ pro-inflammatory subsets of monocytes and release of pro-inflammatory cytokines in the plasma. Numerous cytokines such as I-TAC, Eotaxin, RANTES, MCP-1, IFNγ and MIG demonstrated a biphasic peak that coincided with the differences in kinetics of ZIKV and DENV replication suggesting that viral replication likely differentially modulated the release of these cytokines. Red blood cell indices significantly declined during acute infection suggesting transient anemia, and was accompanied by elevated levels of muscle, liver and renal injury markers. These findings have implications for understanding the pathogenesis of coinfection in ZIKV and DENV endemic regions, and is the 1st report of an experimental coinfection using the rhesus macaque model for ZIKV and DENV infections.
Collapse
Affiliation(s)
- William G Valiant
- F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, 20814, USA
| | - Mary J Mattapallil
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephen Higgs
- Biosecurity Research Institute, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Yan-Jang S Huang
- Biosecurity Research Institute, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Dana L Vanlandingham
- Biosecurity Research Institute, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | | | - Joseph J Mattapallil
- F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD, 20814, USA.
| |
Collapse
|
18
|
Mazzon M, Ortega-Prieto AM, Imrie D, Luft C, Hess L, Czieso S, Grove J, Skelton JK, Farleigh L, Bugert JJ, Wright E, Temperton N, Angell R, Oxenford S, Jacobs M, Ketteler R, Dorner M, Marsh M. Identification of Broad-Spectrum Antiviral Compounds by Targeting Viral Entry. Viruses 2019; 11:E176. [PMID: 30791609 PMCID: PMC6410080 DOI: 10.3390/v11020176] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/13/2019] [Accepted: 02/18/2019] [Indexed: 12/22/2022] Open
Abstract
Viruses are a major threat to human health and economic well-being. In recent years Ebola, Zika, influenza, and chikungunya virus epidemics have raised awareness that infections can spread rapidly before vaccines or specific antagonists can be made available. Broad-spectrum antivirals are drugs with the potential to inhibit infection by viruses from different groups or families, which may be deployed during outbreaks when specific diagnostics, vaccines or directly acting antivirals are not available. While pathogen-directed approaches are generally effective against a few closely related viruses, targeting cellular pathways used by multiple viral agents can have broad-spectrum efficacy. Virus entry, particularly clathrin-mediated endocytosis, constitutes an attractive target as it is used by many viruses. Using a phenotypic screening strategy where the inhibitory activity of small molecules was sequentially tested against different viruses, we identified 12 compounds with broad-spectrum activity, and found a subset blocking viral internalisation and/or fusion. Importantly, we show that compounds identified with this approach can reduce viral replication in a mouse model of Zika infection. This work provides proof of concept that it is possible to identify broad-spectrum inhibitors by iterative phenotypic screenings, and that inhibition of host-pathways critical for viral life cycles can be an effective antiviral strategy.
Collapse
Affiliation(s)
- Michela Mazzon
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Ana Maria Ortega-Prieto
- Section of Virology, Department of Medicine, School of Medicine, Imperial College London, London W2 1PG, UK.
| | - Douglas Imrie
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Christin Luft
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Lena Hess
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Stephanie Czieso
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Joe Grove
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London NW3 2QG, UK.
| | - Jessica Katy Skelton
- Section of Virology, Department of Medicine, School of Medicine, Imperial College London, London W2 1PG, UK.
| | - Laura Farleigh
- Medical Microbiology, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - Joachim J Bugert
- Medical Microbiology, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany.
| | | | - Nigel Temperton
- Medway School of Pharmacy, University of Kent, Chatham ME4 4TB, UK.
| | - Richard Angell
- School of Pharmacy, University College London, London WC1N 1AX, UK.
| | - Sally Oxenford
- School of Pharmacy, University College London, London WC1N 1AX, UK.
| | - Michael Jacobs
- Faculty of Medical Sciences, UCL Medical School, London NW3 2QG, UK.
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Marcus Dorner
- Section of Virology, Department of Medicine, School of Medicine, Imperial College London, London W2 1PG, UK.
| | - Mark Marsh
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
19
|
In-stem molecular beacon targeted to a 5'-region of tRNA inclusive of the D arm that detects mature tRNA with high sensitivity. PLoS One 2019; 14:e0211505. [PMID: 30695081 PMCID: PMC6351059 DOI: 10.1371/journal.pone.0211505] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/15/2019] [Indexed: 01/12/2023] Open
Abstract
Cellular functions are regulated by the up- and down-regulation and localization of RNA molecules. Therefore, many RNA detection methods have been developed to analyze RNA levels and localization. Molecular beacon (MB) is one of the major methods for quantitative RNA detection and analysis of RNA localization. Most oligonucleotide-based probes, including MB, are designed to target a long flexible region on the target RNA molecule, e.g., a single-stranded region. Recently, analyses of tRNA localization and levels became important, as it has been shown that environmental stresses and chemical reagents induce nuclear accumulation of tRNA and tRNA degradation in mammalian cells. However, tRNA is highly structured and does not harbor any long flexible regions. Hence, only a few methods are currently available for detecting tRNA. In the present study, we attempted to detect elongator tRNAMet (eMet) and initiator tRNAMet (iMet) by using an in-stem molecular beacon (ISMB), characterized by more effective quenching and significantly higher sensitivity than those of conventional MB. We found that ISMB1 targeted a 5′- region that includes the D arm of tRNA and that it detected eMet and iMet transcripts as well as mature eMet with high sensitivity. Moreover, the analysis revealed that the formation of the ISMB/tRNA transcript complex required more time than the formation of an ISMB/unstructured short RNA complex. These results suggest that ISMB-based tRNA detection can be a useful tool for various biological and medical studies.
Collapse
|
20
|
Targeting SUMO Modification of the Non-Structural Protein 5 of Zika Virus as a Host-Targeting Antiviral Strategy. Int J Mol Sci 2019; 20:ijms20020392. [PMID: 30658479 PMCID: PMC6359730 DOI: 10.3390/ijms20020392] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 01/28/2023] Open
Abstract
Post-translational modifications of host or viral proteins are key strategies exploited by viruses to support virus replication and counteract host immune response. SUMOylation is a post-translational modification process mediated by a family of ubiquitin-like proteins called small ubiquitin-like modifier (SUMO) proteins. Multiple sequence alignment of 78 representative flaviviruses showed that most (72/78, 92.3%) have a putative SUMO-interacting motif (SIM) at their non-structural 5 (NS5) protein’s N-terminal domain. The putative SIM was highly conserved among 414 pre-epidemic and epidemic Zika virus (ZIKV) strains, with all of them having a putative SIM core amino acid sequence of VIDL (327/414, 79.0%) or VVDL (87/414, 21.0%). Molecular docking predicted that the hydrophobic SIM core residues bind to the β2 strand of the SUMO-1 protein, and the acidic residues flanking the core strengthen the binding through interactions with the basic surface of the SUMO protein. The SUMO inhibitor 2-D08 significantly reduced replication of flaviviruses and protected cells against ZIKV-induced cytopathic effects in vitro. A SIM-mutated ZIKV NS5 failed to efficiently suppress type I interferon signaling. Overall, these findings may suggest SUMO modification of the viral NS5 protein to be an evolutionarily conserved post-translational modification process among flaviviruses to enhance virus replication and suppress host antiviral response.
Collapse
|
21
|
The celecoxib derivative kinase inhibitor AR-12 (OSU-03012) inhibits Zika virus via down-regulation of the PI3K/Akt pathway and protects Zika virus-infected A129 mice: A host-targeting treatment strategy. Antiviral Res 2018; 160:38-47. [PMID: 30326204 PMCID: PMC7113887 DOI: 10.1016/j.antiviral.2018.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 09/22/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022]
Abstract
Zika virus (ZIKV) is a human-pathogenic flavivirus that has recently emerged as a global public health threat. ZIKV infection may be associated with congenital malformations in infected fetuses and severe neurological and systemic complications in infected adults. There are currently limited treatment options for ZIKV infection. AR-12 (OSU-03012) is a celecoxib derivative cellular kinase inhibitor that has broad-spectrum antiviral activities. In this study, we investigated the antiviral activity and mechanism of AR-12 against ZIKV. We evaluated the in vitro anti-ZIKV activity of AR-12, using cell protection and virus yield reduction assays, in multiple clinically relevant cell lines, and the in vivo treatment effects of AR-12 in a lethal mouse model using type I interferon receptor-deficient A129 mice. AR-12 inhibited ZIKV strains belonging to both the African and Asian/American lineages in Huh-7 and/or neuronal cells. AR12's IC50 against ZIKV was consistently <2 μM in these cells. ZIKV-infected A129 mice treated with intraperitoneally or orally administered AR-12 had significantly higher survival rate (50.0%–83.3% vs 0%, P < 0.05), less body weight loss, and lower blood and tissue ZIKV RNA loads than untreated control A129 mice. These anti-ZIKV effects were likely the results of down-regulation of the PI3K/Akt pathway by AR-12. Clinical trials using the clinically available and broad-spectrum AR-12 as an empirical treatment should be considered especially for patients residing in or returning from areas endemic of ZIKV and other arboviral infections who present with an acute undifferentiated febrile illness. AR-12 (OSU-03012) inhibited the replication of Zika virus strains belonging to both the Asian/American and African lineages. AR-12 inhibited Zika virus replication in multiple cell types in vitro. AR-12 treatment improved clinical and virological outcome of Zika virus-infected type I interferon receptor-deficient mice. AR-12 inhibited Zika virus replication via down-regulation of protein kinase B (Akt).
Collapse
|
22
|
Romette JL, Prat CM, Gould EA, de Lamballerie X, Charrel R, Coutard B, Fooks AR, Bardsley M, Carroll M, Drosten C, Drexler JF, Günther S, Klempa B, Pinschewer D, Klimkait T, Avsic-Zupanc T, Capobianchi MR, Dicaro A, Ippolito G, Nitsche A, Koopmans M, Reusken C, Gorbalenya A, Raoul H, Bourhy H, Mettenleiter T, Reiche S, Batten C, Sabeta C, Paweska JT, Eropkin M, Zverev V, Hu Z, Mac Cullough S, Mirazimi A, Pradel F, Lieutaud P. The European Virus Archive goes global: A growing resource for research. Antiviral Res 2018; 158:127-134. [PMID: 30059721 PMCID: PMC7127435 DOI: 10.1016/j.antiviral.2018.07.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/21/2018] [Accepted: 07/23/2018] [Indexed: 11/28/2022]
Abstract
The European Virus Archive (EVA) was created in 2008 with funding from the FP7-EU Infrastructure Programme, in response to the need for a coordinated and readily accessible collection of viruses that could be made available to academia, public health organisations and industry. Within three years, it developed from a consortium of nine European laboratories to encompass associated partners in Africa, Russia, China, Turkey, Germany and Italy. In 2014, the H2020 Research and Innovation Framework Programme (INFRAS projects) provided support for the transformation of the EVA from a European to a global organization (EVAg). The EVAg now operates as a non-profit consortium, with 26 partners and 20 associated partners from 21 EU and non-EU countries. In this paper, we outline the structure, management and goals of the EVAg, to bring to the attention of researchers the wealth of products it can provide and to illustrate how end-users can gain access to these resources. Organisations or individuals who would like to be considered as contributors are invited to contact the EVAg coordinator, Jean-Louis Romette, at jean-louis.romette@univmed.fr.
Collapse
Affiliation(s)
- J L Romette
- Unite des Virus Emergents (UVE: Aix Marseille Univ, IRD 190, INSERM 1207, IHU Méditerranée Infection), Marseille, France.
| | - C M Prat
- Unite des Virus Emergents (UVE: Aix Marseille Univ, IRD 190, INSERM 1207, IHU Méditerranée Infection), Marseille, France
| | - E A Gould
- Unite des Virus Emergents (UVE: Aix Marseille Univ, IRD 190, INSERM 1207, IHU Méditerranée Infection), Marseille, France
| | - X de Lamballerie
- Unite des Virus Emergents (UVE: Aix Marseille Univ, IRD 190, INSERM 1207, IHU Méditerranée Infection), Marseille, France
| | - R Charrel
- Unite des Virus Emergents (UVE: Aix Marseille Univ, IRD 190, INSERM 1207, IHU Méditerranée Infection), Marseille, France
| | - B Coutard
- Architectures et Fonctions, des Macromolécules, Biologiques, Marseille, France
| | - A R Fooks
- Animal and Plant Health Agency, Weybridge, United Kingdom
| | - M Bardsley
- Animal and Plant Health Agency, Weybridge, United Kingdom
| | - M Carroll
- Department of Health-Special Pathogens Laboratory, Porton Down, United Kingdom
| | - C Drosten
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, 10117 Berlin, Germany
| | - J F Drexler
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, 10117 Berlin, Germany
| | - S Günther
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - B Klempa
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - D Pinschewer
- Department of Pathology and Immunology, University of Bales, Switzerland
| | - T Klimkait
- Department of Pathology and Immunology, University of Bales, Switzerland
| | - T Avsic-Zupanc
- Institute of Microbiology and Immunology, Lubljana, Slovenia
| | | | - A Dicaro
- UOC, Istituto Nazionale Malattie Infettive Roma, Italy
| | - G Ippolito
- UOC, Istituto Nazionale Malattie Infettive Roma, Italy
| | - A Nitsche
- Robert Koch Institut, Berlin, Germany
| | - M Koopmans
- ERASMUS Medical Center, Rotterdam, The Netherlands
| | - C Reusken
- ERASMUS Medical Center, Rotterdam, The Netherlands
| | - A Gorbalenya
- Leiden University Medical Center, Leiden, The Netherlands
| | - H Raoul
- Laboratoire Merieux, INSERM, Lyon, France
| | | | - T Mettenleiter
- Friedrich Loeffler Institut, Greifswald-Insel Riems, Germany
| | - S Reiche
- Friedrich Loeffler Institut, Greifswald-Insel Riems, Germany
| | - C Batten
- The Pirbright Institute, Pirbright, United Kingdom
| | - C Sabeta
- Onderstepoort Veterinary Institute, Praetoria, South Africa
| | - J T Paweska
- National Institute for Communicable Diseases, Johannesburg, South Africa
| | - M Eropkin
- Research Institute of Influenza, St. Petersburg, Russia
| | - V Zverev
- Mechnikov Scientific Research Institute for Vaccines and Sera, Moscow, Russia
| | - Z Hu
- Wuhan Institute of Virology, Wuhan, China
| | - S Mac Cullough
- Australian Animal Health Laboratory, Geelong, Australia Disease, Johannesburg, South Africa
| | | | - F Pradel
- Fondation Mérieux, réseau GABRIEL, Lyon, France
| | - P Lieutaud
- Unite des Virus Emergents (UVE: Aix Marseille Univ, IRD 190, INSERM 1207, IHU Méditerranée Infection), Marseille, France
| |
Collapse
|
23
|
Jiang D, Hao M, Ding F, Fu J, Li M. Mapping the transmission risk of Zika virus using machine learning models. Acta Trop 2018; 185:391-399. [PMID: 29932934 DOI: 10.1016/j.actatropica.2018.06.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 06/11/2018] [Accepted: 06/18/2018] [Indexed: 11/18/2022]
Abstract
Zika virus, which has been linked to severe congenital abnormalities, is exacerbating global public health problems with its rapid transnational expansion fueled by increased global travel and trade. Suitability mapping of the transmission risk of Zika virus is essential for drafting public health plans and disease control strategies, which are especially important in areas where medical resources are relatively scarce. Predicting the risk of Zika virus outbreak has been studied in recent years, but the published literature rarely includes multiple model comparisons or predictive uncertainty analysis. Here, three relatively popular machine learning models including backward propagation neural network (BPNN), gradient boosting machine (GBM) and random forest (RF) were adopted to map the probability of Zika epidemic outbreak at the global level, pairing high-dimensional multidisciplinary covariate layers with comprehensive location data on recorded Zika virus infection in humans. The results show that the predicted high-risk areas for Zika transmission are concentrated in four regions: Southeastern North America, Eastern South America, Central Africa and Eastern Asia. To evaluate the performance of machine learning models, the 50 modeling processes were conducted based on a training dataset. The BPNN model obtained the highest predictive accuracy with a 10-fold cross-validation area under the curve (AUC) of 0.966 [95% confidence interval (CI) 0.965-0.967], followed by the GBM model (10-fold cross-validation AUC = 0.964[0.963-0.965]) and the RF model (10-fold cross-validation AUC = 0.963[0.962-0.964]). Based on training samples, compared with the BPNN-based model, we find that significant differences (p = 0.0258* and p = 0.0001***, respectively) are observed for prediction accuracies achieved by the GBM and RF models. Importantly, the prediction uncertainty introduced by the selection of absence data was quantified and could provide more accurate fundamental and scientific information for further study on disease transmission prediction and risk assessment.
Collapse
Affiliation(s)
- Dong Jiang
- State Key Laboratory of Resources and Environmental Information System, Institute of Geographical Sciences and Natural Resources Research, Chinese Academy of Sciences, Beijing, 100101, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Mengmeng Hao
- State Key Laboratory of Resources and Environmental Information System, Institute of Geographical Sciences and Natural Resources Research, Chinese Academy of Sciences, Beijing, 100101, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Fangyu Ding
- State Key Laboratory of Resources and Environmental Information System, Institute of Geographical Sciences and Natural Resources Research, Chinese Academy of Sciences, Beijing, 100101, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jingying Fu
- State Key Laboratory of Resources and Environmental Information System, Institute of Geographical Sciences and Natural Resources Research, Chinese Academy of Sciences, Beijing, 100101, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Meng Li
- State Key Laboratory of Resources and Environmental Information System, Institute of Geographical Sciences and Natural Resources Research, Chinese Academy of Sciences, Beijing, 100101, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
24
|
Alcendor DJ. Zika virus infection and implications for kidney disease. J Mol Med (Berl) 2018; 96:1145-1151. [PMID: 30171265 PMCID: PMC6208949 DOI: 10.1007/s00109-018-1692-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 12/22/2022]
Abstract
High-level and persistent viruria observed in patients infected by Zika virus (ZIKV) has been well documented. However, renal pathology in acutely infected, immunocompetent patients remains subclinical. Moreover, the long-term impact of ZIKV infection, replication, and persistence in the renal compartment of adults and infants as well as immunosuppressed patients and solid organ transplant (SOT) recipients is unknown. Mechanisms involving host and viral factors that limit or control ZIKV pathogenesis in the renal compartment are important yet unexplored. The observation that long-term viral shedding occurs in the renal compartment in the absence of clinical disease requires further investigation. In this review, I explore Zika virus-induced renal pathology in animal models, the dynamics of virus shedding in urine, virus replication in glomerular cells, ZIKV infection in human renal transplantation, and the potential impact of long-term persistent ZIKV infection in the renal compartment.
Collapse
Affiliation(s)
- Donald J Alcendor
- Center for AIDS Health Disparities Research, Department of Microbiology, Immunology, and Physiology, School of Medicine, Meharry Medical College, 1005 Dr. D.B. Todd Jr. Blvd., Hubbard Hospital, 5th Floor, Rm. 5025, Nashville, TN, 37208, USA.
| |
Collapse
|
25
|
Draz MS, Venkataramani M, Lakshminarayanan H, Saygili E, Moazeni M, Vasan A, Li Y, Sun X, Hua S, Yu XG, Shafiee H. Nanoparticle-enhanced electrical detection of Zika virus on paper microchips. NANOSCALE 2018; 10:11841-11849. [PMID: 29881853 PMCID: PMC6033661 DOI: 10.1039/c8nr01646a] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Zika virus (ZIKV) is a reemerging flavivirus causing an ongoing pandemic and public health emergency worldwide. There are currently no effective vaccines or specific therapy for Zika infection. Rapid, low-cost diagnostics for mass screening and early detection are of paramount importance in timely management of the infection at the point-of-care (POC). The current Zika diagnostics are laboratory-based and cannot be implemented at the POC particularly in resource-limited settings. Here, we develop a nanoparticle-enhanced viral lysate electrical sensing assay for Zika virus detection on paper microchips with printed electrodes. The virus is isolated from biological samples using antibodies and labeled with platinum nanoparticles (PtNPs) to enhance the electrical signal. The captured ZIKV-PtNP complexes are lysed using a detergent to release the electrically charged molecules associated with the intact virus and the PtNPs on the captured viruses. The released charged molecules and PtNPs change the electrical conductivity of the solution, which can be measured on a cellulose paper microchip with screen-printed microelectrodes. The results confirmed a highly specific detection of ZIKV in the presence of other non-targeted viruses, including closely related flaviviruses such as dengue virus-1 and dengue virus-2 with a detection limit down to 101 virus particles per μl. The developed assay is simple, rapid, and cost-effective and has the potential for POC diagnosis of viral infections and treatment monitoring.
Collapse
Affiliation(s)
- Mohamed Shehata Draz
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
da Silva LRC. Zika Virus Trafficking and Interactions in the Human Male Reproductive Tract. Pathogens 2018; 7:E51. [PMID: 29751638 PMCID: PMC6027493 DOI: 10.3390/pathogens7020051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 12/28/2022] Open
Abstract
Sexual transmission of Zika virus (ZIKV) is a matter of great concern. Infectious viral particles can be shed in semen for as long as six months after infection and can be transferred to male and female sexual partners during unprotected sexual intercourse. The virus can be found inside spermatozoa and could be directly transferred to the oocyte during fertilization. Sexual transmission of ZIKV can contribute to the rise in number of infected individuals in endemic areas as well as in countries where the mosquito vector does not thrive. There is also the possibility, as has been demonstrated in mouse models, that the vaginal deposition of ZIKV particles present in semen could lead to congenital syndrome. In this paper, we review the current literature to understand ZIKV trafficking from the bloodstream to the human male reproductive tract and viral interactions with host cells in interstitial spaces, tubule walls, annexed glands and semen. We hope to highlight gaps to be filled by future research and potential routes for vaccine and antiviral development.
Collapse
|
27
|
Singh RK, Dhama K, Karthik K, Tiwari R, Khandia R, Munjal A, Iqbal HMN, Malik YS, Bueno-Marí R. Advances in Diagnosis, Surveillance, and Monitoring of Zika Virus: An Update. Front Microbiol 2018; 8:2677. [PMID: 29403448 PMCID: PMC5780406 DOI: 10.3389/fmicb.2017.02677] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/22/2017] [Indexed: 02/05/2023] Open
Abstract
Zika virus (ZIKV) is associated with numerous human health-related disorders, including fetal microcephaly, neurological signs, and autoimmune disorders such as Guillain-Barré syndrome (GBS). Perceiving the ZIKA associated losses, in 2016, the World Health Organization (WHO) declared it as a global public health emergency. In consequence, an upsurge in the research on ZIKV was seen around the globe, with significant attainments over developing several effective diagnostics, drugs, therapies, and vaccines countering this life-threatening virus at an early step. State-of-art tools developed led the researchers to explore virus at the molecular level, and in-depth epidemiological investigations to understand the reason for increased pathogenicity and different clinical manifestations. These days, ZIKV infection is diagnosed based on clinical manifestations, along with serological and molecular detection tools. As, isolation of ZIKV is a tedious task; molecular assays such as reverse transcription-polymerase chain reaction (RT-PCR), real-time qRT-PCR, loop-mediated isothermal amplification (LAMP), lateral flow assays (LFAs), biosensors, nucleic acid sequence-based amplification (NASBA) tests, strand invasion-based amplification tests and immune assays like enzyme-linked immunosorbent assay (ELISA) are in-use to ascertain the ZIKV infection or Zika fever. Herein, this review highlights the recent advances in the diagnosis, surveillance, and monitoring of ZIKV. These new insights gained from the recent advances can aid in the rapid and definitive detection of this virus and/or Zika fever. The summarized information will aid the strategies to design and adopt effective prevention and control strategies to counter this viral pathogen of great public health concern.
Collapse
Affiliation(s)
- Raj K. Singh
- ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, 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 Sansthan, Mathura, India
| | - Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | - Ashok Munjal
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | - Hafiz M. N. Iqbal
- School of Engineering and Science, Tecnologico de Monterrey, Monterrey, Mexico
| | - Yashpal S. Malik
- Division of Biological Standardization, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Rubén Bueno-Marí
- Laboratorios Lokímica, Departamento de Investigación y Desarrollo (I+D), Valencia, Spain
| |
Collapse
|
28
|
T cell immunity to Zika virus targets immunodominant epitopes that show cross-reactivity with other Flaviviruses. Sci Rep 2018; 8:672. [PMID: 29330423 PMCID: PMC5766511 DOI: 10.1038/s41598-017-18781-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 12/18/2017] [Indexed: 11/09/2022] Open
Abstract
Zika virus (ZIKV) Infection has several outcomes from asymptomatic exposure to rash, conjunctivitis, Guillain-Barré syndrome or congenital Zika syndrome. Analysis of ZIKV immunity is confounded by the fact that several related Flaviviruses infect humans, including Dengue virus 1-4, West Nile virus and Yellow Fever virus. HLA class II restricted T cell cross-reactivity between ZIKV and other Flaviviruses infection(s) or vaccination may contribute to protection or to enhanced immunopathology. We mapped immunodominant, HLA class II restricted, CD4 epitopes from ZIKV Envelope (Env), and Non-structural (NS) NS1, NS3 and NS5 antigens in HLA class II transgenic mice. In several cases, ZIKV primed CD4 cells responded to homologous sequences from other viruses, including DENV1-4, WNV or YFV. However, cross-reactive responses could confer immune deviation - the response to the Env DENV4 p1 epitope in HLA-DR1 resulted in IL-17A immunity, often associated with exacerbated immunopathogenesis. This conservation of recognition across Flaviviruses, may encompass protective and/or pathogenic components and poses challenges to characterization of ZIKV protective immunity.
Collapse
|
29
|
Sridhar S, Yip CCY, Chan JFW, To KKW, Cheng VCC, Yuen KY. Impact of inter-genotypic recombination and probe cross-reactivity on the performance of the Abbott RealTime HCV Genotype II assay for hepatitis C genotyping. Diagn Microbiol Infect Dis 2018; 91:34-37. [PMID: 29434000 DOI: 10.1016/j.diagmicrobio.2017.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 12/16/2017] [Accepted: 12/29/2017] [Indexed: 01/21/2023]
Abstract
The Abbott RealTime HCV Genotype II assay (Abbott-RT-HCV assay) is a real-time PCR based genotyping method for hepatitis C virus (HCV). This study measured the impact of inter-genotypic recombination and probe cross-reactivity on the performance of the Abbott-RT-HCV assay. 517 samples were genotyped using the Abbott-RT-HCV assay over a one-year period, 34 (6.6%) were identified as HCV genotype 1 without further subtype designation raising the possibility of inaccurate genotyping. These samples were subjected to confirmatory sequencing. 27 of these 34 (79%) samples were genotype 1b while five (15%) were genotype 6. One HCV isolate was an inter-genotypic 1a/4o recombinant. This is a novel natural HCV recombinant that has never been reported. Inter-genotypic recombination and probe cross-reactivity can affect the accuracy of the Abbott-RT-HCV assay, both of which have significant implications on antiviral regimen choice. Confirmatory sequencing of ambiguous results is crucial for accurate genotyping.
Collapse
Affiliation(s)
- Siddharth Sridhar
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cyril C Y Yip
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jasper F W Chan
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kelvin K W To
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Vincent C C Cheng
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Infection and Immunology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
30
|
Husstedt IW, Maschke M, Eggers C, Neuen-Jacob E, Arendt G. [Zika virus infection and the nervous system]. DER NERVENARZT 2018; 89:136-143. [PMID: 29318332 DOI: 10.1007/s00115-017-0472-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Zika virus is an arbovirus from the family of flaviviruses, which is transmitted by the mosquito Aedes aegyptii and also by the Asian mosquito Aedes albopticus. The largest observed Zika virus epidemic is currently taking place in North and South America, in the Caribbean, southern USA and Southeast Asia. In most cases the infection is an unspecific, acute, febrile disease. Neurological manifestations consist mainly of microcephaly in newborns and Guillain-Barré syndrome but other rare manifestations have also become known in the meantime, such as meningoencephalitis and myelitis. Therefore, the Zika virus, similar to other flaviviruses, has neuropathogenic properties. In particular, the drastic increase in microcephaly cases in Brazil has induced great research activities. The virus is transmitted perinatally and can be detected in the amniotic fluid, placenta and brain tissue of the newborn. Vaccination or a causal therapy does not yet exist. The significant increase in Guillain-Barré syndrome induced by the Zika virus was observed during earlier outbreaks. In the meantime, scientifically clear connections between a Zika virus infection and these neurological manifestations have been shown. Long-term studies and animal models should be used for a better understanding of the pathomechanisms of this disease.
Collapse
Affiliation(s)
- I W Husstedt
- Klinik Maria Frieden und Medizinische Fakultät, Westfälische Wilhelms-Universität Münster, Am Krankenhaus 1, 48291, Telgte, Deutschland.
| | - M Maschke
- Klinik für Neurologie, Krankenhaus der Barmherzigen Brüder, Trier, Deutschland
| | - C Eggers
- Krankenhaus der Barmherzigen Brüder, Linz, Österreich
| | - E Neuen-Jacob
- Institut für Neuropathologie, Universitätsklinikum Düsseldorf, Düsseldorf, Deutschland
| | - G Arendt
- Klinik für Neurologie, Universitätsklinikum Düsseldorf, Düsseldorf, Deutschland
| |
Collapse
|
31
|
|
32
|
Chen JH, Lam HY, Yip CC, Cheng VC, Chan JF, Leung TH, Sridhar S, Chan KH, Tang BS, Yuen KY. Evaluation of the molecular Xpert Xpress Flu/RSV assay vs. Alere i Influenza A & B assay for rapid detection of influenza viruses. Diagn Microbiol Infect Dis 2017; 90:177-180. [PMID: 29262988 DOI: 10.1016/j.diagmicrobio.2017.11.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/08/2017] [Accepted: 11/17/2017] [Indexed: 11/30/2022]
Abstract
A new FDA-approved Xpert Xpress Flu/RSV assay has been released for rapid influenza virus detection. We collected 134 nasopharyngeal specimens to compare the diagnostic performance of the Xpert assay and the Alere i Influenza A & B assay for influenza A and B virus detection. The Xpert assay demonstrated 100% and 96.3% sensitivity to influenza A and influenza B virus respectively. Its specificity was 100% for both viruses. The Alere i assay demonstrated slightly lower sensitivity but similar specificity to the Xpert Xpress assay. Although the Xpert assay (30 min) required longer processing time than the Alere assay (15 min), the handling procedure of the Alere assay was more complicated than the Xpert assay. As the GenXpert system has higher throughput than the Alere system, it is more suitable for hospital clinical laboratories. Overall, the new Xpert Xpress Flu/RSV assay is a reliable and useful tool for rapid influenza detection.
Collapse
Affiliation(s)
- J H Chen
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - H Y Lam
- Department of Pathology, Hong Kong Sanatorium and Hospital, Hong Kong Special Administrative Region, China
| | - C C Yip
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - V C Cheng
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China; Infection Control Team, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - J F Chan
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - T H Leung
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - S Sridhar
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - K H Chan
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - B S Tang
- Department of Pathology, Hong Kong Sanatorium and Hospital, Hong Kong Special Administrative Region, China
| | - K Y Yuen
- Department of Microbiology, Queen Mary Hospital, Hong Kong Special Administrative Region, China; Carol Yu Centre for Infection, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
33
|
Yuan S, Chan JFW, den-Haan H, Chik KKH, Zhang AJ, Chan CCS, Poon VKM, Yip CCY, Mak WWN, Zhu Z, Zou Z, Tee KM, Cai JP, Chan KH, de la Peña J, Pérez-Sánchez H, Cerón-Carrasco JP, Yuen KY. Structure-based discovery of clinically approved drugs as Zika virus NS2B-NS3 protease inhibitors that potently inhibit Zika virus infection in vitro and in vivo. Antiviral Res 2017; 145:33-43. [PMID: 28712942 DOI: 10.1016/j.antiviral.2017.07.007] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 06/01/2017] [Accepted: 07/11/2017] [Indexed: 10/19/2022]
Abstract
Zika virus (ZIKV) infection may be associated with severe complications in fetuses and adults, but treatment options are limited. We performed an in silico structure-based screening of a large chemical library to identify potential ZIKV NS2B-NS3 protease inhibitors. Clinically approved drugs belonging to different drug classes were selected among the 100 primary hit compounds with the highest predicted binding affinities to ZIKV NS2B-NS3-protease for validation studies. ZIKV NS2B-NS3 protease inhibitory activity was validated in most of the selected drugs and in vitro anti-ZIKV activity was identified in two of them (novobiocin and lopinavir-ritonavir). Molecular docking and molecular dynamics simulations predicted that novobiocin bound to ZIKV NS2B-NS3-protease with high stability. Dexamethasone-immunosuppressed mice with disseminated ZIKV infection and novobiocin treatment had significantly (P < 0.05) higher survival rate (100% vs 0%), lower mean blood and tissue viral loads, and less severe histopathological changes than untreated controls. This structure-based drug discovery platform should facilitate the identification of additional enzyme inhibitors of ZIKV.
Collapse
Affiliation(s)
- Shuofeng Yuan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Jasper Fuk-Woo Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region; Research Centre of Infection and Immunology, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region; Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region.
| | - Helena den-Haan
- Bioinformatics and High Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica San Antonio de Murcia (UCAM), Spain; Villapharma Research S.L., Parque Tecnológico de Fuente Álamo, Ctra. El Estrecho-Lobosillo, Km. 2.5, Av. Azul, Fuente álamo de Murcia, Murcia, Spain
| | - Kenn Ka-Heng Chik
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Anna Jinxia Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Chris Chung-Sing Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Vincent Kwok-Man Poon
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Cyril Chik-Yan Yip
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Winger Wing-Nga Mak
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Zheng Zhu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Zijiao Zou
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Kah-Meng Tee
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Jian-Piao Cai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Kwok-Hung Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Jorge de la Peña
- Bioinformatics and High Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica San Antonio de Murcia (UCAM), Spain
| | - Horacio Pérez-Sánchez
- Bioinformatics and High Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica San Antonio de Murcia (UCAM), Spain.
| | - José Pedro Cerón-Carrasco
- Bioinformatics and High Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica San Antonio de Murcia (UCAM), Spain.
| | - Kwok-Yung Yuen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region; State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region; Research Centre of Infection and Immunology, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region; Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region; The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region.
| |
Collapse
|