1
|
Pastor AF, Mahaney SM, Garcia J, Morales M, Quintanilla O, Arriaga MA, Thomas JM, VandeBerg JL. The Laboratory Opossum ( Monodelphis domestica) Is a Unique Model for Research on Zika Virus: Robust Immune Response, Widespread Dissemination, and Long-Term Persistence. Viruses 2024; 16:1847. [PMID: 39772157 PMCID: PMC11680235 DOI: 10.3390/v16121847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
The Zika virus (ZIKV) epidemic elicited a rapid commitment to the development of animal models for ZIKV research. Non-human primates (NHPs) and mice have made significant contributions to this research, but NHPs are expensive, have a long gestation period, and are available only in small numbers; non-genetically modified mice are resistant to infection. To address these deficiencies, we have established the laboratory opossum, Monodelphis domestica, as a small animal model that complements the mouse and monkey models. We developed and validated an indirect ELISA for measuring antibodies to ZIKV in opossums, as well as an immunohistochemistry (IHC) method to detect ZIKV NS1 protein in tissue samples. Opossum pups inoculated intracerebrally as embryos, juveniles inoculated by several routes, and mothers that cannibalized inoculated pups became persistently infected with ZIKV. The virus spread to multiple organs and persisted for up to 38 weeks (the latest endpoint of the experiments). A robust humoral immune response was mounted, and high titers of antibodies also persisted for 38 weeks. The results establish M. domestica as a natural, non-genetically modified animal model in which ZIKV persists long-term after experimental exposure and as a unique animal model for research on the immune response to ZIKV.
Collapse
Affiliation(s)
- André Filipe Pastor
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA; (S.M.M.); (M.A.A.); (J.M.T.III)
- Center for Vector-Borne Disease, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
- Sertão Pernambucano Federal Institute of Education, Science, and Technology, Floresta 56400000, Pernambuco, Brazil
| | - Susan M. Mahaney
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA; (S.M.M.); (M.A.A.); (J.M.T.III)
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
| | - Juan Garcia
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA (O.Q.)
| | - Marisol Morales
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA (O.Q.)
| | - Oscar Quintanilla
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA (O.Q.)
| | - Marco A. Arriaga
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA; (S.M.M.); (M.A.A.); (J.M.T.III)
| | - John M. Thomas
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA; (S.M.M.); (M.A.A.); (J.M.T.III)
- Center for Vector-Borne Disease, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA (O.Q.)
| | - John L. VandeBerg
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA; (S.M.M.); (M.A.A.); (J.M.T.III)
- Center for Vector-Borne Disease, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
| |
Collapse
|
2
|
Kim N, Choi H, Kim U, Kim S, Kim YB, Shin HY. Sustained Microglial Activation Promotes Synaptic Loss and Neuronal Dysfunction after Recovery from ZIKV Infection. Int J Mol Sci 2024; 25:9451. [PMID: 39273400 PMCID: PMC11394746 DOI: 10.3390/ijms25179451] [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: 07/18/2024] [Revised: 08/19/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Zika virus (ZIKV), transmitted by Aedes mosquitoes, has been a global health concern since 2007. It primarily causes fetal microcephaly and neuronal defects through maternal transmission and induces neurological complications in adults. Recent studies report elevated proinflammatory cytokines and persistent neurological alterations post recovery, but the in vivo mechanisms remain unclear. In our study, viral RNA loads in the brains of mice infected with ZIKV peaked at 7 days post infection and returned to baseline by day 21, indicating recovery. RNA sequencing of the cerebral cortex at 7 and 21 days revealed upregulated genes related to neuroinflammation and microglial activation. Histological analyses indicated neuronal cell death and altered neurite morphology owing to severe neuroinflammation. Additionally, sustained microglial activation was associated with increased phospho-Tau levels, constituting a marker of neurodegeneration. These findings highlight how persistent microglial activation leads to neuronal dysfunction post ZIKV recovery, providing insights into the molecular pathogenesis of ZIKV-induced brain abnormalities.
Collapse
Affiliation(s)
- Nahyun Kim
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Hanul Choi
- Department of Bio-Industrial Technologies, Konkuk University, Seoul 05029, Republic of Korea
| | - Uijin Kim
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Suyeon Kim
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Young Bong Kim
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea
- Department of Bio-Industrial Technologies, Konkuk University, Seoul 05029, Republic of Korea
| | - Ha Youn Shin
- Department of Biomedical Science & Engineering, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
3
|
Zhang Q, Zhang F, Chang X, Hu J, Zhang Z, Cui X, Zheng X, Wang X. A Neonatal Murine Model for Caprine Enterovirus Infection and the Viral Tissue Tropism. Viruses 2023; 15:v15020475. [PMID: 36851688 PMCID: PMC9962493 DOI: 10.3390/v15020475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
As the first caprine enterovirus identified from goat herds characterized by severe diarrhea with a high morbidity and mortality rate, the underlying pathogenesis and tissue tropism for CEV-JL14 remains largely unknown. Here, we reported the establishment of a neonatal murine model for caprine enterovirus and the unveiling of the tissue tropism and underlying pathogenesis for CEV-JL14 enterovirus. Susceptible murine strains, the infective dose, the infective routes, viral loads, and tissue tropism for CEV-JL14 infection were determined. The findings showed that ICR mice were susceptible to CEV-JL14 infection via all infection routes. Tissue viral load analysis showed that CEV-JL14 was detected in almost all tissues including the heart, liver, spleen, lung, kidney, intestine, brain, and muscle, with significantly higher viral loads in the heart, liver, lung, kidney, and intestine. These results revealed the pattern of viral load and tropism for CEV-JL14 and provided a model system for elucidating the pathogenesis of CEV-JL14 viruses.
Collapse
|
4
|
Negi V, Kuhn RJ, Fekete DM. Exploring the Expression and Function of cTyro3, a Candidate Zika Virus Receptor, in the Embryonic Chicken Brain and Inner Ear. Viruses 2023; 15:247. [PMID: 36680287 PMCID: PMC9867072 DOI: 10.3390/v15010247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
The transmembrane protein Axl was proposed as an entry receptor for Zika virus (ZIKV) infection in vitro, but conflicting results from in vivo studies have made it difficult to establish Axl as a physiologically relevant ZIKV receptor. Both the functional redundancy of receptors and the experimental model used can lead to variable results. Therefore, it can be informative to explore alternative animal models to analyze ZIKV receptor candidates as an aid in discovering antivirals. This study used chicken embryos to examine the role of chicken Tyro3 (cTyro3), the equivalent of human Axl. Results show that endogenous cTyro3 mRNA expression overlaps with previously described hot spots of ZIKV infectivity in the brain and inner ear. We asked if ectopic expression or knockdown of cTyro3 influenced ZIKV infection in embryos. Tol2 vectors or replication-competent avian retroviruses were used in ovo to introduce full-length or truncated (presumed dominant-negative) cTyro3, respectively, into the neural tube on embryonic day two (E2). ZIKV was delivered to the brain 24 h later. cTyro3 manipulations did not alter ZIKV infection or cell death in the E5/E6 brain. Moreover, delivery of truncated cTyro3 variants to the E3 otocyst had no effect on inner ear formation on E6 or E10.
Collapse
Affiliation(s)
| | | | - Donna M. Fekete
- Department of Biological Sciences, Purdue Institute for Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47906, USA
| |
Collapse
|
5
|
Gomes JA, Wachholz GE, Boquett JA, Vianna FSL, Schuler-Faccini L, Fraga LR. Molecular Mechanisms of ZIKV-Induced Teratogenesis: A Systematic Review of Studies in Animal Models. Mol Neurobiol 2022; 60:68-83. [PMID: 36215025 PMCID: PMC9549063 DOI: 10.1007/s12035-022-03046-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 09/21/2022] [Indexed: 12/09/2022]
Abstract
Zika virus (ZIKV) is a teratogen that causes congenital anomalies, being linked to microcephaly in children exposed during pregnancy. Animal studies have been conducted to investigate the molecular mechanisms related to ZIKV teratogenesis. Although animal models can mimic the effects of ZIKV in human embryo development, few in vivo studies have addressed molecular changes following ZIKV infection in embryos. Moreover, few literature reviews have been conducted with these studies. The aim of this systematic review is to evaluate the molecular mechanisms of ZIKV teratogenesis determined from studies in animal models. PubMed/MEDLINE, EMBASE, Web of Science, and Scopus as well as grey literature were searched for studies that evaluated molecular alterations related to ZIKV teratogenesis which occurred during embryonic development. Nine studies were included: six with mice, one with mice and guinea pigs, one with pigs and one with chickens. In general, studies presented an unclear or high risk of bias for methodological criteria. Most of studies reported embryos exposed to ZIKV presenting microcephaly, reduced cortex thickness, and growth restriction. Different techniques were used to evaluated molecular changes in the animals following ZIKV infection: RNA sequencing, RT-qPCR, and in situ hybridization. It was found that common pathways are changed in most studies, being pathways related to immune response upregulated and those involved to neurodevelopment downregulated.
Collapse
Affiliation(s)
- Julia A Gomes
- Teratology Information Service, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil
- Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil
| | - Gabriela E Wachholz
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Juliano A Boquett
- Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Postgraduate Program in Child and Adolescent Health, Faculty of Medicine, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Fernanda S L Vianna
- Teratology Information Service, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil
- Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil
- Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Postgraduate Program in Medicine: Medical Sciences, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, 90035-003, Brazil
| | - Lavínia Schuler-Faccini
- Teratology Information Service, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil
- Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
- Postgraduate Program in Medicine: Medical Sciences, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, 90035-003, Brazil
| | - Lucas R Fraga
- Teratology Information Service, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil.
- Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil.
- Postgraduate Program in Medicine: Medical Sciences, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, 90035-003, Brazil.
- Department of Morphological Sciences, Institute of Health Sciences, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, 90050-170, Brazil.
| |
Collapse
|
6
|
Mask E, Hodara VL, Callery JE, Parodi LM, Obregon-Perko V, Yagi S, Glenn J, Frost P, Clemmons E, Patterson JL, Cox LA, Giavedoni LD. Molecular Approaches for the Validation of the Baboon as a Nonhuman Primate Model for the Study of Zika Virus Infection. Front Cell Infect Microbiol 2022; 12:880860. [PMID: 35493734 PMCID: PMC9046911 DOI: 10.3389/fcimb.2022.880860] [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: 02/22/2022] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Nonhuman primates (NHP) are particularly important for modeling infections with viruses that do not naturally replicate in rodent cells. Zika virus (ZIKV) has been responsible for sporadic epidemics, but in 2015 a disseminated outbreak of ZIKV resulted in the World Health Organization declaring it a global health emergency. Since the advent of this last epidemic, several NHP species, including the baboon, have been utilized for modeling and understanding the complications of ZIKV infection in humans; several health issues related to the outcome of infection have not been resolved yet and require further investigation. This study was designed to validate, in baboons, the molecular signatures that have previously been identified in ZIKV-infected humans and macaque models. We performed a comprehensive molecular analysis of baboons during acute ZIKV infection, including flow cytometry, cytokine, immunological, and transcriptomic analyses. We show here that, similar to most human cases, ZIKV infection of male baboons tends to be subclinical, but is associated with a rapid and transient antiviral interferon-based response signature that induces a detectable humoral and cell-mediated immune response. This immunity against the virus protects animals from challenge with a divergent ZIKV strain, as evidenced by undetectable viremia but clear anamnestic responses. These results provide additional support for the use of baboons as an alternative animal model to macaques and validate omic techniques that could help identify the molecular basis of complications associated with ZIKV infections in humans.
Collapse
Affiliation(s)
- Emma Mask
- Department of Biology, Trinity University, San Antonio, TX, United States
| | - Vida L. Hodara
- Southwest National Primate Research Center, San Antonio, TX, United States,Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jessica E. Callery
- Department of Biology, Trinity University, San Antonio, TX, United States
| | - Laura M. Parodi
- Southwest National Primate Research Center, San Antonio, TX, United States,Texas Biomedical Research Institute, San Antonio, TX, United States
| | | | - Shigeo Yagi
- California Department of Public Health, Richmond, CA, United States
| | - Jeremy Glenn
- Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Patrice Frost
- Southwest National Primate Research Center, San Antonio, TX, United States
| | - Elizabeth Clemmons
- Southwest National Primate Research Center, San Antonio, TX, United States
| | | | - Laura A. Cox
- Southwest National Primate Research Center, San Antonio, TX, United States,Center for Precision Medicine, Wake Forest Health Sciences University, Winston Salem, NC, United States
| | - Luis D. Giavedoni
- Department of Biology, Trinity University, San Antonio, TX, United States,Southwest National Primate Research Center, San Antonio, TX, United States,*Correspondence: Luis D. Giavedoni,
| |
Collapse
|
7
|
Nazerai L, Buus S, Stryhn A, Thomsen AR, Christensen JP. Efficient Control of Zika Virus Infection Induced by a Non-Replicating Adenovector Encoding Zika Virus NS1/NS2 Antigens Fused to the MHC Class II-Associated Invariant Chain. Viruses 2021; 13:2215. [PMID: 34835021 PMCID: PMC8625593 DOI: 10.3390/v13112215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022] Open
Abstract
It is generally believed that a successful Zika virus (ZIKV) vaccine should induce neutralizing antibodies against the ZIKV envelope (E) protein to efficiently halt viral infection. However, E-specific neutralizing antibodies have been implicated in a phenomenon called antibody-dependent enhancement, which represents an ongoing concern in the flavivirus-vaccinology field. In this report, we investigated the vaccination potential of replication-deficient adenoviral vectors encoding the ZIKV non-structural proteins 1 and 2 (NS1/NS2) and employed the strategy of linking the antigens to the MHC-II associated invariant chain (li) to improve immunogenicity and by inference, the level of protection. We demonstrated that li-linkage enhanced the production of anti-NS1 antibodies and induced an accelerated and prolonged polyfunctional CD8 T cell response in mice, which ultimately resulted in a high degree of protection against ZIKV infection of the CNS.
Collapse
Affiliation(s)
| | | | | | | | - Jan Pravsgaard Christensen
- Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; (L.N.); (S.B.); (A.S.); (A.R.T.)
| |
Collapse
|
8
|
Are the Organoid Models an Invaluable Contribution to ZIKA Virus Research? Pathogens 2021; 10:pathogens10101233. [PMID: 34684182 PMCID: PMC8537471 DOI: 10.3390/pathogens10101233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 12/16/2022] Open
Abstract
In order to prevent new pathogen outbreaks and avoid possible new global health threats, it is important to study the mechanisms of microbial pathogenesis, screen new antiviral agents and test new vaccines using the best methods. In the last decade, organoids have provided a groundbreaking opportunity for modeling pathogen infections in human brains, including Zika virus (ZIKV) infection. ZIKV is a member of the Flavivirus genus, and it is recognized as an emerging infectious agent and a serious threat to global health. Organoids are 3D complex cellular models that offer an in-scale organ that is physiologically alike to the original one, useful for exploring the mechanisms behind pathogens infection; additionally, organoids integrate data generated in vitro with traditional tools and often support those obtained in vivo with animal model. In this mini-review the value of organoids for ZIKV research is examined and sustained by the most recent literature. Within a 3D viewpoint, tissue engineered models are proposed as future biological systems to help in deciphering pathogenic processes and evaluate preventive and therapeutic strategies against ZIKV. The next steps in this field constitute a challenge that may protect people and future generations from severe brain defects.
Collapse
|
9
|
Miller MR, Fagre AC, Clarkson TC, Markle ED, Foy BD. Three Immunocompetent Small Animal Models That Do Not Support Zika Virus Infection. Pathogens 2021; 10:pathogens10080971. [PMID: 34451435 PMCID: PMC8401401 DOI: 10.3390/pathogens10080971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that is primarily transmitted to humans through the bite of an infected mosquito. ZIKV causes disease in infected humans with added complications of Guillain-Barré syndrome and birth defects in infants born to mothers infected during pregnancy. There are several large immunocompetent animal models for ZIKV including non-human primates (NHPs). NHP models closely reflect human infection; however, due to sample size restrictions, investigations into the effects of transmission route and the impacts on disease dynamics have been understudied. Mice have been widely used for modeling ZIKV infection, yet there are few ZIKV-susceptible immunocompetent mouse models and none of these have been used to investigate sexual transmission. In an effort to identify a small immunocompetent animal model to characterize sexual transmission of ZIKV, we attempt experimental infection of multimammate mice, New Zealand white rabbits, and Hartley guinea pigs. The multimammate mouse is the natural reservoir of Lassa fever virus and has been identified to harbor other human pathogens. Likewise, while NZW rabbits are susceptible to West Nile virus, they have not yet been examined for their susceptibility to infection with ZIKV. Guinea pigs have been successfully used as models for ZIKV infection, but only in immunocompromised life stages (young or pregnant). Here, it was found that the multimammate mouse and New Zealand White (NZW) rabbits are not susceptible ZIKV infection as determined by a lack viral RNA in tissues and fluids collected. Sexually mature male Hartley guinea pigs were inoculated subcutaneously and by mosquito bite, but found to be refractory to ZIKV infection, contrary to findings of other studies in young and pregnant guinea pigs. Interestingly, here it is shown that adult male guinea pigs are not susceptible to ZIKV infection, even when infected by natural route (e.g., mosquito bite). Although a new small animal model for the sexual transmission for ZIKV was not established through this study, these findings provide information on outbred animal species that are not permissive to infection (NZW rabbits and multimammate mice) and new information surrounding limitations of a previously established animal model (guinea pigs).
Collapse
|
10
|
Wachholz GE, do Amaral Gomes J, Boquett JA, Vianna FSL, Schuler-Faccini L, Fraga LR. Molecular mechanisms of Zika virus teratogenesis from animal studies: a systematic review protocol. Syst Rev 2021; 10:160. [PMID: 34051864 PMCID: PMC8164069 DOI: 10.1186/s13643-021-01713-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 05/18/2021] [Indexed: 02/21/2023] Open
Abstract
BACKGROUND Due to the diversity of studies in animal models reporting that molecular mechanisms are involved in the teratogenic effect of the Zika virus (ZIKV), the objective of the present study is to evaluate the methodological quality of these studies, as well as to demonstrate which genes and which molecular pathways are affected by ZIKV in different animal models. METHODS This search will be performed in four databases: PubMed/MEDLINE, EMBASE, Web of Science, and Scopus, as well as in the grey literature. The studies selection process will be reported through the PRISMA Statement diagram model. All studies describing the molecular mechanisms possibly involved in the development of malformations caused by embryonic/fetal ZIKV exposure in animal models with an appropriate control group and methodology will be included (including, for instance, randomized and non-randomized studies). All animals used as experimental models for ZIKV teratogenesis may be included as long as exposure to the virus occurred during the embryonic/fetal period. From the selected studies, data will be extracted using a previously prepared standard form. Bias risk evaluation will be conducted following the SYRCLE's Risk of Bias tool. All data obtained will be tabulated and organized by outcomes (morphological and molecular). DISCUSSION With the proposed systematic review, we expect to present results about the methodological quality of the published studies with animal models that investigated the molecular mechanisms involved in the teratogenic effect of ZIKV, as well as to show the studies with greater reliability. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42019157316.
Collapse
Affiliation(s)
- Gabriela Elis Wachholz
- Graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, 91501-970, Brazil.,Teratology Information Service, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil.,Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil
| | - Julia do Amaral Gomes
- Graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, 91501-970, Brazil.,Teratology Information Service, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil.,Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil
| | - Juliano André Boquett
- Graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, 91501-970, Brazil.,Graduate Program in Child and Adolescent Health, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Sales Luiz Vianna
- Graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, 91501-970, Brazil.,Teratology Information Service, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil.,Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil.,Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90035-003, Brazil
| | - Lavínia Schuler-Faccini
- Graduate Program in Genetics and Molecular Biology, Department of Genetics, Biosciences Institute, Universidade Federal do Rio Grande do Sul, Porto Alegre, 91501-970, Brazil.,Teratology Information Service, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil.,Graduate Program in Child and Adolescent Health, Faculty of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lucas Rosa Fraga
- Teratology Information Service, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil. .,Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, 90035-903, Brazil. .,Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90035-003, Brazil. .,Department of Morphological Sciences, Institute Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90050-170, Brazil.
| |
Collapse
|
11
|
Wachholz GE, Varela APM, Teixeira TF, de Matos SMS, Rigon da Luz Soster P, Vianna FSL, de Souza DOG, Roehe PM, Schuler-Faccini L, Fraga LR. Zika virus-induced brain malformations in chicken embryos. Birth Defects Res 2020; 113:22-31. [PMID: 33009728 DOI: 10.1002/bdr2.1813] [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] [Received: 07/06/2020] [Revised: 09/07/2020] [Accepted: 09/18/2020] [Indexed: 11/12/2022]
Abstract
BACKGROUND Zika virus (ZIKV) was confirmed to be related to microcephaly in 2016. However, there is still a need for understanding the embryonic morphological changes induced by ZIKV and when they occur. Here, chicken embryos were chosen as experimental model of ZIKV to evaluate virus-associated morphological alterations that might take place during embryonic development. METHODS A screening with different viral doses was conducted in embryos at HH Stage 10-12 (E1.5) as well as a follow up of the first 5 days postinfection (dpi) was performed to observe the main morphologic changes post ZIKV infection. RESULTS ZIKV exposed embryos presented a higher prevalence of mortality and defects such as brain malformation when compared to controls. Moreover, we observed that the phenotypes become more evident at 4dpi, when the viral load quantification reaches a peak. CONCLUSIONS We found that ZIKV exposed embryos presented a high prevalence of mortality and central nervous system (CNS) abnormalities in a dose-dependent manner. The phenotype was more evident 4 days postinfection, when the viral load quantification reached a peak.
Collapse
Affiliation(s)
- Gabriela Elis Wachholz
- Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Teratogen Information Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Ana Paula Muterle Varela
- Postgraduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Thais Fumaco Teixeira
- Department of Microbiology, Immunology and Parasitology, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Sophia Martins Simon de Matos
- Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Paula Rigon da Luz Soster
- Department of Morphological Sciences, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Sales Luiz Vianna
- Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Teratogen Information Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Diogo Onofre Gomes de Souza
- Postgraduate Program in Biochemistry, Departamento f Biochemistry, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Paulo Michel Roehe
- Department of Microbiology, Immunology and Parasitology, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Lavínia Schuler-Faccini
- Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Teratogen Information Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Lucas Rosa Fraga
- Teratogen Information Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Laboratory of Genomic Medicine, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Department of Morphological Sciences, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
12
|
Nazerai L, Schøller AS, Bassi MR, Buus S, Stryhn A, Christensen JP, Thomsen AR. Effector CD8 T Cell-Dependent Zika Virus Control in the CNS: A Matter of Time and Numbers. Front Immunol 2020; 11:1977. [PMID: 32973802 PMCID: PMC7461798 DOI: 10.3389/fimmu.2020.01977] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/21/2020] [Indexed: 12/29/2022] Open
Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus, came into the spotlight in 2016 when it was found to be associated with an increased rate of microcephalic newborns in Brazil. The virus has further been recognized to cause neurologic complications in children and adults in the form of myelitis, encephalitis, acute disseminated encephalomyelitis (ADEM) and Guillain Barre Syndrome in a fraction of infected individuals. With the ultimate goal of identifying correlates of protection to guide the design of an effective vaccine, the study of the immune response to ZIKV infection has become the focus of research worldwide. Both innate and adaptive immune responses seem to be essential for controlling the infection. Induction of sufficient levels of neutralizing antibodies has been strongly correlated with protection against reinfection in various models, while the role of CD8 T cells as antiviral effectors in the CNS has been controversial. In an attempt to improve our understanding regarding the role of ZIKV-induced CD8 T cells in protective immunity inside the CNS, we have expanded on previous studies in intracranially infected mice. In a recent study, we have demonstrated that, peripheral ZIKV infection in adult C57BL/6 mice induces a robust CD8 T cell response that peaks within a week. In the present study, we used B cell deficient as well as wild-type mice to show that there is a race between CXCR3-dependent recruitment of the effector CD8 T cells and local ZIKV replication, and that CD8 T cells are capable of local viral control if they arrive in the brain early after viral invasion, in appropriate numbers and differentiation state. Our data highlight the benefits of considering this subset when designing vaccines against Zika virus.
Collapse
Affiliation(s)
- Loulieta Nazerai
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Amalie Skak Schøller
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Maria Rosaria Bassi
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Søren Buus
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Anette Stryhn
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | - Allan Randrup Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Young G, Bohning KJ, Zahralban-Steele M, Hather G, Tadepalli S, Mickey K, Godin CS, Sanisetty S, Sonnberg S, Patel HK, Dean HJ. Complete Protection in Macaques Conferred by Purified Inactivated Zika Vaccine: Defining a Correlate of Protection. Sci Rep 2020; 10:3488. [PMID: 32103097 PMCID: PMC7044319 DOI: 10.1038/s41598-020-60415-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/12/2020] [Indexed: 01/01/2023] Open
Abstract
A critical global health need exists for a Zika vaccine capable of mitigating the effects of future Zika epidemics. In this study we evaluated the antibody responses and efficacy of an aluminum hydroxide adjuvanted purified inactivated Zika vaccine (PIZV) against challenge with Zika virus (ZIKV) strain PRVABC59. Indian rhesus macaques received two doses of PIZV at varying concentrations ranging from 0.016 µg - 10 µg and were subsequently challenged with ZIKV six weeks or one year following the second immunization. PIZV induced a dose-dependent immune response that was boosted by a second immunization. Complete protection against ZIKV infection was achieved with the higher PIZV doses of 0.4 µg, 2 µg, and 10 µg at 6 weeks and with 10 ug PIZV at 1 year following vaccination. Partial protection was achieved with the lower PIZV doses of 0.016 µg and 0.08 µg. Based on these data, a neutralizing antibody response above 3.02 log10 EC50 was determined as a correlate of protection in macaques. PIZV elicited a dose-dependent neutralizing antibody response which is protective for at least 1 year following vaccination.
Collapse
Affiliation(s)
| | | | | | - Greg Hather
- Takeda Pharmaceuticals, Inc., Cambridge, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Modelling Neurotropic Flavivirus Infection in Human Induced Pluripotent Stem Cell-Derived Systems. Int J Mol Sci 2019; 20:ijms20215404. [PMID: 31671583 PMCID: PMC6862117 DOI: 10.3390/ijms20215404] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/24/2019] [Accepted: 10/29/2019] [Indexed: 02/06/2023] Open
Abstract
Generation of human induced pluripotent stem cells (hiPSCs) and their differentiation into a variety of cells and organoids have allowed setting up versatile, non-invasive, ethically sustainable, and patient-specific models for the investigation of the mechanisms of human diseases, including viral infections and host–pathogen interactions. In this study, we investigated and compared the infectivity and replication kinetics in hiPSCs, hiPSC-derived neural stem cells (NSCs) and undifferentiated neurons, and the effect of viral infection on host innate antiviral responses of representative flaviviruses associated with diverse neurological diseases, i.e., Zika virus (ZIKV), West Nile virus (WNV), and dengue virus (DENV). In addition, we exploited hiPSCs to model ZIKV infection in the embryo and during neurogenesis. The results of this study confirmed the tropism of ZIKV for NSCs, but showed that WNV replicated in these cells with much higher efficiency than ZIKV and DENV, inducing massive cell death. Although with lower efficiency, all flaviviruses could also infect pluripotent stem cells and neurons, inducing similar patterns of antiviral innate immune response gene expression. While showing the usefulness of hiPSC-based infection models, these findings suggest that additional virus-specific mechanisms, beyond neural tropism, are responsible for the peculiarities of disease phenotype in humans.
Collapse
|
15
|
Therapeutic Advances Against ZIKV: A Quick Response, a Long Way to Go. Pharmaceuticals (Basel) 2019; 12:ph12030127. [PMID: 31480297 PMCID: PMC6789873 DOI: 10.3390/ph12030127] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that spread throughout the American continent in 2015 causing considerable worldwide social and health alarm due to its association with ocular lesions and microcephaly in newborns, and Guillain-Barré syndrome (GBS) cases in adults. Nowadays, no licensed vaccines or antivirals are available against ZIKV, and thus, in this very short time, the scientific community has conducted enormous efforts to develop vaccines and antivirals. So that, different platforms (purified inactivated and live attenuated viruses, DNA and RNA nucleic acid based candidates, virus-like particles, subunit elements, and recombinant viruses) have been evaluated as vaccine candidates. Overall, these vaccines have shown the induction of vigorous humoral and cellular responses, the decrease of viremia and viral RNA levels in natural target organs, the prevention of vertical and sexual transmission, as well as that of ZIKV-associated malformations, and the protection of experimental animal models. Some of these vaccine candidates have already been assayed in clinical trials. Likewise, the search for antivirals have also been the focus of recent investigations, with dozens of compounds tested in cell culture and a few in animal models. Both direct acting antivirals (DAAs), directed to viral structural proteins and enzymes, and host acting antivirals (HAAs), directed to cellular factors affecting all steps of the viral life cycle (binding, entry, fusion, transcription, translation, replication, maturation, and egress), have been evaluated. It is expected that this huge collaborative effort will produce affordable and effective therapeutic and prophylactic tools to combat ZIKV and other related still unknown or nowadays neglected flaviviruses. Here, a comprehensive overview of the advances made in the development of therapeutic measures against ZIKV and the questions that still have to be faced are summarized.
Collapse
|
16
|
Park JG, Ávila-Pérez G, Madere F, Hilimire TA, Nogales A, Almazán F, Martínez-Sobrido L. Potent Inhibition of Zika Virus Replication by Aurintricarboxylic Acid. Front Microbiol 2019; 10:718. [PMID: 31031722 PMCID: PMC6473159 DOI: 10.3389/fmicb.2019.00718] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/21/2019] [Indexed: 11/14/2022] Open
Abstract
Zika virus (ZIKV) is one of the recently emerging vector-borne viruses in humans and is responsible for severe congenital abnormalities such as microcephaly in the Western Hemisphere. Currently, only a few vaccine candidates and therapeutic drugs are being developed for the treatment of ZIKV infections, and as of yet none are commercially available. The polyanionic aromatic compound aurintricarboxylic acid (ATA) has been shown to have a broad-spectrum antimicrobial and antiviral activity. In this study, we evaluated ATA as a potential antiviral drug against ZIKV replication. The antiviral activity of ATA against ZIKV replication in vitro showed median inhibitory concentrations (IC50) of 13.87 ± 1.09 μM and 33.33 ± 1.13 μM in Vero and A549 cells, respectively; without showing any cytotoxic effect in both cell lines (median cytotoxic concentration (CC50) > 1,000 μM). Moreover, ATA protected both cell types from ZIKV-induced cytopathic effect (CPE) and apoptosis in a time- and concentration-dependent manner. In addition, pre-treatment of Vero cells with ATA for up to 72 h also resulted in effective suppression of ZIKV replication with similar IC50. Importantly, the inhibitory effect of ATA on ZIKV infection was effective against strains of the African and Asian/American lineages, indicating that this inhibitory effect was not strain dependent. Overall, these results demonstrate that ATA has potent inhibitory activity against ZIKV replication and may be considered as a potential anti-ZIKV therapy for future clinical evaluation.
Collapse
Affiliation(s)
- Jun-Gyu Park
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Ginés Ávila-Pérez
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Ferralita Madere
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Thomas A Hilimire
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Aitor Nogales
- Center for Animal Health Research, INIA-CISA, Madrid, Spain
| | - Fernando Almazán
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
17
|
Pre-Clinical Pregnancy Models for Evaluating Zika Vaccines. Trop Med Infect Dis 2019; 4:tropicalmed4020058. [PMID: 30959955 PMCID: PMC6630727 DOI: 10.3390/tropicalmed4020058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/26/2019] [Accepted: 04/04/2019] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) infection during pregnancy can result in a variety of developmental abnormalities in the fetus, referred to as Congenital Zika Syndrome (CZS). The effects of CZS can range from the loss of the viable fetus to a variety of neurological defects in full-term infants, including microcephaly. The clinical importance of ZIKV-induced CZS has driven an intense effort to develop effective vaccines. Consequently, there are approximately 45 different ZIKV vaccine candidates at various stages of development with several undergoing phase I and II clinical trials. These vaccine candidates have been shown to effectively prevent infection in adult animal models, however, there has been less extensive testing for their ability to block vertical transmission to the fetus during pregnancy or prevent the development of CZS. In addition, it is becoming increasingly difficult to test vaccines in the field as the intensity of the ZIKV epidemic has declined precipitously, making clinical endpoint studies difficult. These ethical and practical challenges in determining efficacy of ZIKV vaccine candidates in preventing CZS have led to increased emphasis on pre-clinical testing in animal pregnancy models. Here we review the current status of pre-clinical pregnancy models for testing the ability of ZIKV vaccines to prevent CZS.
Collapse
|
18
|
Martín-Acebes MA, Saiz JC. The Scientific Response to Zika Virus. J Clin Med 2019; 8:jcm8030369. [PMID: 30884762 PMCID: PMC6463034 DOI: 10.3390/jcm8030369] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/04/2019] [Accepted: 03/15/2019] [Indexed: 01/08/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne pathogen that became renowned in 2015 in Brazil mainly due to its association with microcephaly in newborns. Although most infections in adults are asymptomatic or cause mild illnesses, in a reduced number of cases, ZIKV can also produce severe complications that include neurological disorders (Guillain–Barré syndrome), ocular lesions, or reproductive alterations. From 2015 the efforts of a significant part of the scientific community were placed on ZIKV research, which has resulted in an unpredicted escalation of the knowledge of the biology and pathology of this virus. The rapid response of the scientific community against ZIKV highlights its enormous potential to counter attack a viral threat within a short time period. It is expected that this huge collaborative effort will produce affordable and effective prophylactic and therapeutic tools against ZIKV. Nevertheless, there are still other arboviral threats different from ZIKV that should not be forgotten.
Collapse
Affiliation(s)
- Miguel A Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, 28040, Spain.
| | - Juan-Carlos Saiz
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, 28040, Spain.
| |
Collapse
|