Skip to main content
The Journal of Infectious Diseases logoLink to The Journal of Infectious Diseases
. 2017 Dec 16;216(Suppl 10):S919–S927. doi: 10.1093/infdis/jix465

Small-Animal Models of Zika Virus

Justin G Julander 1,, Venkatraman Siddharthan 1
PMCID: PMC5853237  PMID: 29267919

Abstract

Zika virus (ZIKV) infection can result in serious consequences, including severe congenital manifestations, persistent infection in the testes, and neurologic sequelae. After a pandemic emergence, the virus has spread to much of North and South America and has been introduced to many countries outside of ZIKV-endemic areas as infected travelers return to their home countries. Rodent models have been important in gaining a better understanding of the wide range of disease etiologies associated with ZIKV infection and for the initial phase of developing countermeasures to prevent or treat viral infections. We discuss herein the advantages and disadvantages of small-animal models that have been developed to replicate various aspects of disease associated with ZIKV infection.

Keywords: Zika virus, rodent, congenital, flavivirus

ACUTE VIRAL DISEASE

There are many advantages to modeling viral disease in mice and other small-animal species. Various advantages and disadvantages of rodent species that have been used to replicate disease manifestations of Zika virus (ZIKV) disease are included in Table 1. This article will summarize the major findings of work performed in various small-animal models to characterize disease outcomes after challenge with ZIKV.

Table 1.

Comparison of Advantages and Disadvantages of Small-Animal Species Used to Model Zika Virus Infection and Disease

Variable WT Mice Ab-Induced Immune Deficient Mice IFN Pathway KO Mice STAT2-/- Hamsters Guinea Pigs
Advantages Readily available, inexpensive, normal immunity, well characterized, diverse immune profiles Readily available, induced immune deficiency as needed, high virus replication Age-dependent disease, diverse disease manifestations, lethality, high virus replication Intermediate sensitivity, persistent infection of testes, lower mortality rate Readily available, similar placentation to human, naturally susceptible to infection, normal immunity
Disadvantages Naturally resilient, low virus replication, requires high virus inoculum, difficult infection routes Ab expense/availability, reversion to WT and clearance of virus, little overt disease Limited availability, may require in-house colony, susceptible to other pathogens, abnormally severe disease Lack of reagents, limited availability, underlying polyomavirus, variable disease manifestation Lack of reagents, large size, not well characterized, expensive to house, somewhat difficult to work with

Abbreviations: Ab, antibody; IFN, interferon; KO, knockout; WT, wild type.

Initial studies investigating ZIKV shortly after its discovery in the mid 1900s included challenge of various mouse strains, as well as a human challenge model [1–4]. These early studies demonstrated that mice were somewhat refractory to ZIKV infection [5], so work during the 20th century was limited to around 40 published reports. Emergence of ZIKV in the early 21st century resulted in an increased effort to learn more about this virus, with around 2800 papers being published (as of August 2017). One of the main goals was to develop small-animal models for use in delineating the infection cycle, identifying consequences of virus infection, and discovering antiviral countermeasures. A brief summary of the work to develop mouse models of acute ZIKV infection are included in Table 2.

Table 2.

Mouse Models of Acute Zika Virus (ZIKV) Infection and Disease

Mouse Strain(s) Age, wk ZIKV Strain, Dose, Route Pathological Finding Reference
Neurological Other
Porton 0, 4 MR 766, titration, intracerebral Not reported LD50 of 107.2 pfu in 1–2-d-old mice and 106.4 pfu in adult mice [5]
IFNAR-/-or AG129 5, 11 Cambodian, 105 pfu, intraperitoneal Brain virus 3, tremors 6 d after virus injection Viremia detected 2 d after virus injection, virus in spleen and testis [8]
AG129 3–4 or 8 H/PF/2013, 100–5 pfu, subcutaneous, 105.3 pfu, intraperitoneal Increased virus level in brain, neurodegeneration Viremia detected 2 d after virus injection, myofiber necrosis, inflammatory cell infiltration, nuclear rowing [6]
AG129 8 MR 766, 101–5 pfu, intraperitoneal Paralysis, conjunctivitis, neurodegeneration, viral antigen in brain and spinal cord, acute encephalitis IFN-γ and IL-18 levels increased in serum, viral RNA detected in brain, spleen, liver, kidney [9]
IFNAR-/- 5–6 MP1751, 106 pfu, subcutaneous Perivascular cuffing, polymorphonuclear cells in gray/white matter near blood vessels, neurodegeneration in hippocampus Viral RNA detected in brain and spleen 3 d after virus injection, apoptosis in spleen, hematopoiesis in liver [84]
AG129 8–10 P 6–740, 103.0 pfu, subcutaneous Paralysis, conjunctivitis, hyperexcitability, seizure, tremor, ZIKV Ag in neurons and astrocytes, encephalitis, and myelitis Viral RNA level peaked 5 d after virus injection; virus level in testis peaked 7 d after virus injection, including in Leydig cells; viral RNA shedding in urine [7]
IFNAR-/- or Irf3-/-Irf5-/-Ifr7-/- 5–6 H/PF/2013 or MR766, 102 ffu, subcutaneous (footpad) ZIKV RNA in brain and spinal cord, higher paralysis incidence after intravenous delivery Viral RNA detected 2–6 d after virus injection, 106 ffu virus in testes of IFNAR-/- mice [12]
C57BL/6 + Ifnar Ab 5 Dakar, 106.4 pfu, subcutaneous or intraperitoneal Paralysis 8 d after virus injection after intraperitoneal challenge, viral RNA by in situ hybridization, neurodegeneration in cerebellum and hippocampus Viral RNA detected in brain, kidney, skeletal muscle, and spleen 3 d after virus injection; increased glial fibrillary acidic protein level in cerebrum, brain titer 15 d after virus injection [17]
C57BL/6, IFNAR-/- 0–1 PRVABC-59, 103.3–4.3 pfu, subcutaneous Wide array of neurologic symptoms, virus in neurons Increased inflammatory gene expression, T-cell– driven immunity, viral load in brain and spleen [85]
STAT2-/-, IFNAR-/- 5–6 MR-766, 103 pfu, subcutaneous Viral RNA in brain, hind limb paralysis, elevated cytokine levels in brain Viral RNA detected in various tissues, upregulated IFN expression [13]
BALB/c + immunosuppression with dexamethasone 6–8 PRVABC-59, 106.5 pfu, intraperitoneal Neurons in hippocampus stain positive for ZIKV NS1 Orchitis [18]
Swiss 0 SPH 2015, dose not provided, intracerebral or subcutaneous Neurological disease and death of neurons, gliosis Atonic urinary bladder [19]
STAT2-/- 5–8 PRVABC-59, 103.7 pfu, intradermal Elevated ZIKV replication in spinal cord Enhanced disease severity after convalescent sera treatment [23]

Abbreviations: Ag, antigen; ffu, focus-forming units; IFN, interferon; IL-18, interleukin 18; LD50, median lethal dose; NS1, nonstructural protein 1; pfu, plaque-forming units.

Experiments conducted during the last several years often used immune-compromised mouse strains, which were permissive for virus infection and displayed various neurologic signs of disease, including some that replicated severe disease manifestations in humans. The AG129 mouse strain, which lacks receptors for interferon α/β (IFN-α/β) and IFN-γ were susceptible to infection after inoculation with various strains of ZIKV injected by various routes [6–9]. Virus replicates in a wide range of tissues, and viral RNA can be detected in the serum during the course of infection, with timing depending on the strain and route of virus infection. Relevant disease signs include lacrimation and neurologic involvement. The severity of disease observed in this mouse strain underscores the importance of the IFN response in controlling ZIKV infection.

Other IFN-pathway-knockout mice also show varying degrees of disease severity after infection with ZIKV and provide a suitable model for virus replication in tissues. Infection of IFNAR-/- mice, lacking IFN-α/β receptors, with ZIKV results in age-dependent severity, with mortality occurring in mice that are around 3 weeks of age or younger [8]. The presence of IFN-γ receptors provides intermediate protection in 5-week-old mice and complete survival in mice that are 11 weeks old. Another knockout strain, IFNGR-/- mice, lack IFN-γ receptors and are susceptible to ZIKV infection. These mice were used to compare the pathogenicity of various ZIKV infectious clones [10]. Mosquito transmission of ZIKV to IFNAR-/- mice has helped identify the vector competency for several virus strains [11]. STAT2-/- knockout mice and mice lacking IFN response factors 3, 5, and 7 are also susceptible to intravenous infection with ZIKV [12, 13].

Histologic analysis of AG129 mice infected with ZIKV identified neurons throughout the central nervous system that were positive for ZIKV antigen, with outcomes including neurodegenerative multifocal neutrophilic encephalitis and myelitis [7]. The observation of ZIKV in motor neurons in the ventral horn of the spinal cord was similar to disease observed after infection with West Nile virus [14]. Astrocytes were also heavily infected in various regions of the brain and spinal cord after ZIKV infection, which was similar to pathology observed after challenge of mice with Venezuelan equine encephalitis virus, an alphavirus [15]. Interestingly, infected AG129 mice displayed rear limb myofiber degeneration and necrosis with inflammatory cell infiltration in the absence of hind-limb paralysis, suggesting that this virus infects muscle cells [6]. Viral antigen positivity, however, was not directly observed in the muscle tissue. Direct intraocular inoculation with ZIKV results in infection of the cells lining the blood-retinal barrier and causes chorioretinal atrophy [16].

Since wild-type mice generally display only a very transient viral infection after ZIKV challenge, various methods are used to increase the susceptibility of wild-type, immunocompetent strains. Mice can be treated with function-blocking antibodies (MAR1-5A3) targeting the IFN-α/β receptors to increase the susceptibility of the mice to infection following challenge [17]. Viral RNA was detected in the cerebrum and hippocampus in mice treated with function-blocking antibody. Dexamethasone immunosuppression renders BALB/c mice susceptible to ZIKV infection, resulting in detectable virus titer in various tissues and lethality [18]. Infection of neonatal mice shortly after birth also results in morbidity and mortality, with more-severe disease and death observed in younger mouse pups [19]. Infected neonates developed neurologic complications, including tremors, seizure, hyperactivity, and limb collapse, with detectable virus in the brain as late as 15 days after infection.

Preexisting flavivirus antibodies are implicated in worsened disease during infection, a concept known as antibody enhancement of disease. This phenomenon is well-known for dengue virus (DENV), where antibodies to one strain bind at low levels to an incoming heterotypic strain, resulting in an increased uptake of virus by Fc receptor–bearing cells of the immune system. Infection in this manner effectively increases the host cells available for viral replication and thereby increases the antigen load, host response, and immunopathology of the infection [20]. This has been demonstrated in immunodeficient mouse models of DENV infection [21, 22]. Enhanced disease in ZIKV-infected mice after treatment with immune serum–containing antibodies to DENV or West Nile virus has been demonstrated [23]. Because of the overlap in sequence between DENV and ZIKV, it is possible that antibody enhancement of disease might also play a role in ZIKV pathogenesis, as has been demonstrated in rodent models. However, there is no clinical evidence to support this idea, and care should be taken not to overinterpret data from rodent models. Additionally, mounting data from epidemiological studies and studies in nonhuman primates suggest that antibodies to DENV play more of a protective role, rather than a role in disease enhancement [24, 25]. Previous exposure to DENV did not correlate with worsened outcome in ZIKV infection during pregnancy [26]. Further epidemiological data are needed to provide further insight into this subject.

Aside from mouse models, other small-animal species have been used to model ZIKV, including chickens and guinea pigs. Infection of chicken embryos with ZIKV results in virus infection of the developing nervous system, causing fetal demise at higher virus challenge doses and a microcephaly-like phenotype at lower doses, replicating some aspects of congenital infection [27]. Immunocompetent guinea pigs infected with ZIKV developed a transient viremia, had detectable viral RNA in whole blood and serum, had an increase in cytokine and chemokine levels, developed infection of various tissues, and developed neutralizing antibodies [28]. These additional models of ZIKV infection and disease may provide useful systems for the evaluation of countermeasures or in disease characterization.

CONGENITAL INFECTION

Intrauterine exposure of a developing fetus to ZIKV infection can result in debilitating manifestations in the fetus, the most severe and obvious of which is microcephaly. An increase in the incidence of microcephaly during the recent ZIKV outbreak in Brazil eventually led to the discovery of the causative role of the virus in developmental abnormalities of fetuses after exposure in utero. Aside from microcephaly, other disease manifestations have been reported, including smaller birth weight, brain abnormalities despite normal head size, hearing loss, optic nerve hypoplasia, joint and bone deformities (arthrogryposis), and many other less apparent effects of ZIKV congenital exposure [29]. A significantly higher fetal mortality rate was associated with ZIKV infection among pregnant women in Brazil, compared with that due to other etiologies (approximately 5.1% vs 1.4%; P < .0001) [30]. This section will focus on various consequences of intrauterine exposure to ZIKV that have been replicated in small-animal models, as well as those aspects of congenital infection that are dissimilar between rodents and humans.

In attempting to recapitulate disease associated with intrauterine infection in small-animal models, various laboratories have independently developed mouse models of congenital infection (Table 3). Despite the use of a diverse range of ZIKV strains, as well as differences in the dose, route, and timing of infection during gestation, some consistent consequences of infection have been delineated in rodents. Virus was generally detected in the placenta [31–34], while fetal infection was dependent on the timing of maternal challenge. Specifically, lower fetal infection rates were generally observed when the female was challenged at gestational stages after embryonic day 12 (as discussed below). Another commonly observed consequence was intrauterine growth restriction and spontaneous abortion of developing fetuses [31, 35, 36]. These findings are similar to outcomes in natural congenital infection [30] and can be used to further characterize congenital infection, as well as to identify countermeasures to inhibit or prevent fetal infection. The period between embryonic days 3 and 14, when pregnant mice have been challenged with ZIKV across several studies, corresponds to gestation days 4–48 during the first trimester in humans.

Table 3.

Rodent Models of Congenital Infection With Zika Virus (ZIKV)

Strain(s), Rodent Origin(s), Virus Strain(s) Infection Route(s), Dose(s) Embryonic Day(s) Major Findings Reference
IFNAR1-/- × C57BL/6, mouse French Polynesia, H/PF/2013 Subcutaneous (footpad), 103 ffu 6.5, 7.5 Fetal and placental infection, IUGR, fetal demise, placental and fetal brain apoptosis [31]
C57BL/6, mouse French Polynesia, H/PF/2013 (MAR1-5A3 Ab E5.5) subcutaneous (footpad), 103 ffu 6.5, 7.5 Placental and fetal infection, IUGR [31]
SJL, mouse Paraiba, Brazil, 2015 Intravenous/103, 1010.6, or 1012 pfu/mL 10–13 IUGR, upregulation of apoptosis genes, cortical malformations [35]
ICR, mouse SZ01 Lateral ventricle (injection of fetus), 105.8 pfu/mL (1 µL) 13.5 Brain replication in ventricular subventricular zones, cortical thinning, infection of neural progenitor or intermediate progenitor cells [45]
C57BL/6, mouse SZ01 Intrauterine (fetal brain injection), 105.5 pfu 13.5 ZIKV infection of placenta and fetal brain, reduction of cortex founder cells [33]
C57BL/6, IRF3/7-/-, and IFNAR-/-, mouse Cambodia FSS13025 Intravaginal, 104.4–105.7 pfu 4.5–8.5 IUGR, fetal demise, ZIKV infection of fetal brain (based on RNA detection and electron microscopy findings) [36]
CD-1, mouse 2010 Cambodia, 2015 Brazil, 2015 Puerto Rico, 1968 Nigeria Intrauterine, intraperitoneal /106 TCID50 10, 14 High aborted fetus rate (30%–45%), virus in fetus/ dam, and increased interferon expression 48 h after virus injection w/infection at embryonic d 10 (not embryonic d 14); neuroinflammation and cortical thinning in neonatal brain [43]
STAT2-/-, hamster Malaysia, P 6–740 Subcutaneous/102.7 CCID50 8.5 Virus in fetal brain, placental pathology, live births [32]
FVB/NJ, C57BL/6, mouse Bahia, Brazil, HS-2015-BA-01 Intravenous (jugular)/105 pfu 5.5, 7.5, 9.5 Infection of placenta/fetus, pathology of fetal brain, fetal demise, arthrogryposis [34]

Abbreviations: CCID50, 50% cell culture infectious dose; ffu, focus-forming units; IUGR, intrauterine growth restriction; pfu, plaque-forming units; TCID50, 50% tissue culture infectious dose.

The interaction between virus in maternal blood and fetal tissues occurs in the placenta. Although the placental structure of mice is quite distinct from that of humans, there are various similarities that are found between the two that make the mouse a reasonable model for various aspects of human placentation, including notably the lining of fetal structures with syncytiotrophoblast cells that contact maternal blood [37]. Trophoblast cells of the mouse conceptus will differentiate, forming a branched villous structure, which invades the placental wall decidua beginning around embryonic day 8.5. The placenta of mice is considered functional at approximately 10 days after coitus [38–40].

The timing of virus challenge during pregnancy is important in the context of congenital infection of the fetus in rodent models. Congenital infection of mice with other flaviviruses was demonstrated when virus challenge occurred between embryonic days 5 and 12, but detection of virus or viral antigen in the fetus was less likely as development proceeded past embryonic day 12 [41, 42]. Similar observations have been reported with ZIKV infection in mouse models [34, 43]. Although peripheral challenge of pregnant dams with ZIKV after embryonic day 12 results in virus infection of the placenta, infection at this time does not typically result in fetal infection in rodents. The timing of reduced congenital virus infection in rodents appears to be associated with placental development. This is somewhat dissimilar to congenital infection in humans, where ZIKV infection during the second and third trimesters can result in transmission to the fetus [44]. This observation could indicate partial control of the dissemination of virus to the fetus at the placental barrier, or it could suggest other mechanisms involved with transplacental infection.

A functional placenta is present in mice around embryonic day 10. Challenge of mice between embryonic days 4 and 14 results in productive placental infection in mice. The detection of virus in the placenta after challenge from embryonic days 4 to 14 suggests a high degree of susceptibility of this organ [43]. After the development of a functional placenta in mice, transplacental transmission of ZIKV and other flaviviruses to the fetus is reduced unless the fetuses are challenged directly [33, 43, 45]. The lack of infection at later times in mice may suggest the placental barrier is preventing transplacental movement of ZIKV in the mouse. However, this is dissimilar to congenital infection of human fetuses, where brain lesions have been observed in newborn babies after ZIKV infection during the second and third trimesters of pregnancy [46]. However, these second and third trimester infections of human placentas appear to be limited primarily to Hofbauer cells, and the inflammatory villitis that is seen during first trimester placental infections is reportedly absent at later infection times [47]. Indeed, Hofbauer cells isolated from human placentas have been the focus of several studies, which demonstrate susceptibility of these cells to ZIKV [47–49]. In contrast, infection of the placenta during the first trimester of human pregnancy results in infection in a wider array of placental cells. Mesenchymal cells and cytotrophoblasts support ZIKV replication, along with Hofbauer cells. This gestationally dependent infection of placental cells was further supported by a study that demonstrated expression of genes for various entry factors used by ZIKV in early stage–type trophoblast cells [50]. These cells were highly susceptible to infection in cell culture, while cells isolated from term placentas did not express these genes and were relatively resilient to ZIKV infection. While mice and humans may differ in some aspects of congenital infection, challenge of pregnant mice during early gestation (ie, before embryonic day 10) models many aspects of natural infection.

Most of the studies have terminated at some point during gestation, and few live births have been recorded. The consequences of intrauterine exposure to ZIKV on the development of neonates will be an important step in future research to further identify consequences of congenital infection. Depending on the strain of virus used, the immune state of the dam, and the timing of infection, the aborted fetus rate can be relatively high [43], which would result in fewer births. Disease manifestations in females just after birth may cause the dams to neglect or cannibalize pups [32], further complicating studies designed to characterize the effect of intrauterine exposure to ZIKV on the development of neonates. Future studies should include live births to determine the suitability of mice to model the effects of intrauterine infection on developing neonates.

SEXUAL TRANSMISSION

A somewhat unanticipated consequence of ZIKV infection is sexual transmission. Evidence of non–mosquito-borne transmission was available as early as 2008, when a returning scientist that was infected with ZIKV in Senegal transmitted the virus to his wife, which was suspected to be through sexual contact [51]. Closely related flaviviruses, such as DENV, are not known to be transmitted through sexual contact. It has been well documented that men who were exposed to ZIKV in areas of endemicity could further spread the virus to their sex partners, which may or may not include the manifestation of disease. Various clinical studies have provided evidence of sexual transmission of ZIKV from male to female partners, from female to male partners, and from male to male partners [52–56], although the great majority of transmission cases are from male to female partners. Transmission has been reported to occur several weeks after returning from areas of endemicity, with the majority of cases occurring within 3 weeks of return. Viral RNA was detected in the semen of infected males up to 6 months after disease onset [57], although very few studies have evaluated the duration of persistence of ZIKV in semen. While little is known about the localization of virus in the reproductive tract of infected men, mouse models have provided insight into which cell types are infected and potential mechanisms of sexual transmission (Table 4).

Table 4.

Rodent Models of Zika Virus (ZIKV) Sexual Transmission and Infection of the Male Reproductive Tract

Model Virus Origin, Strain Infection Route(s)/Dose(s) Major Findings Reference
C57BL/6, (MAR1-5A3), Rag1-/-, and Axl-/- mice Dakar 41519 (ma) H/PF/2013 Subcutaneous (footpad)/106.0 or 103.0 pfu Persistence of ZIKV in testicles/epididymides (21 d after virus injection), reduced testis size, low testosterone level, infected spermatogonia, seminiferous tubule degradation [59]
IFNAR-/- mice MEX2-81 Subcutaneous (footpad)/105 pfu Presence of viral RNA in testicles/epididymides 21 d after virus injection, testicular atrophy, reduced testosterone level [60]
IFNAR-/- mice ZIKA-SMGC-1 Intraperitoneal/103.0 pfu, intratesticular/102.6 pfu Atrophy of the reproductive tract, virus in testicles/epididymis but not seminal vesicle or prostate [61]
AG129 mice Puerto Rico, PRVABC59 Intraperitoneal/103 pfu ZIKV RNA/virus observed in semen, demonstration of sexual transmission of virus and fetal infection, virus/inflammation in testicles/ epididymides [58]
STAT2-/- hamsters P 6–740 Subcutaneous/102.7 CCID50 Infection of Sertoli cells and spermatogonia [32]
BALB/c mice (dexamethasone) Puerto Rico, PRVABC59 Intraperitoneal/106.5 pfu Pathology of seminiferous tubules, lymphocytic infiltration (12–14 d after virus injection), reduced viral load in testicles/epididymides and prostate after interferon treatment [86]
AG129 and LysMCre+ IFNAR mice Cambodia, FSS13025 Intravaginal/105 or 106 ffu Estrus cycle–dependent susceptibility of females [62]
IFNAR-/- mice Puerto Rico, PRVABC59 Rectal/106.5 pfu Nonlethal, virus in rectum, testis, brain, and spleen on d 21; inflammation; and splenomegaly Martinez et al, unpublished data

Abbreviations: CCID50, 50% cell culture infectious dose; ffu, focus-forming units; pfu, plaque-forming units.

Many published reports have demonstrated ZIKV infection of the male reproductive tract of rodents, primarily indicating detection of infectious virus or viral RNA in the testes of infected males [7–9, 12]. The testes support very high levels of virus, particularly in immunocompromised mice. Virus has been localized in various cells of the testis, including Leydig cells and spermatogenic precursors, and in epithelial cells of the epididymis [32, 58]. Interstitial inflammation and inflammatory cell infiltrates were observed. Necrotic cells that stained positively for viral antigen were observed in the lumen of the epididymides [59], representing a potential source of virus for transmission. Other severe disease manifestions, such as orchitis and testicular atrophy, have been observed in mouse models [60].

The implications of testis infection observed in mice may not be directly translatable to human infection. For example, orchitis, or inflammation of the testicle, is a very painful condition in people and is generally the consequence of bacterial infection, including sexually transmitted infections. This is commonly observed in immune-suppressed mice infected with ZIKV [18, 61]. If a common consequence of ZIKV in infected men was orchitis, the pain would be intense, and they would surely seek medical attention. Orchitis is likely not a commonly occurring consequence of ZIKV infection of the male reproductive tract.

Intravaginal infection of female mice with ZIKV can cause a systemic infection and may also be transmitted to offspring [58, 62]. The estrous cycle may influence the susceptibility of females, as AG129 mice that were inoculated intravaginally with ZIKV during a hormonally induced estrus-like phase did not die of viral infection and had a relative lack of virus replication in various tissues, while those that were in an induced diestrus-like phase displayed virus replication and mortality [62]. Transmission from infected males to naive females has been demonstrated in AG129 mice, including transmission from vasectomized mice, despite a lower virus load as compared to intact males [58]. Males also had detectable virus in the semen several weeks after virus challenge, modeling persistent virus present in men infected naturally with ZIKV. Rectal inoculation of male mice results in systemic spread of virus to various tissues, including the testes, and supports observations of male-to-male transmission (Martinez et al, unpublished data).

USE OF MODELS IN THE DEVELOPMENT OF COUNTERMEASURES

As there are currently no Food and Drug Administration (FDA)–approved drugs to treat acute flaviviral diseases, it is unknown how direct-acting antiviral compounds would affect infection and disease outcomes in people infected with ZIKV. There is potential for antiviral treatment to reduce disease burden and further spread of the virus if therapy is initiated soon after the onset of clinical disease or if prophylactic treatment, in the context of a widespread viral outbreak, is used. In regard to the treatment of Zika, antiviral agents could have use in preventing fetal transmission, clearing sequelae from the testes, or reducing viral load during acute infection to reduce or inhibit further transmission.

Various nucleoside analogs, including 7-deaza-2ʹ-C-methyladenosine, BCX4430, sofosbuvir, and NITD008, have been shown to be active against ZIKV in cell culture and in mouse models [7, 9, 63, 64]. These viral RNA-dependent RNA polymerase inhibitors delay or prevent mortality, reduce virus titer in relevant tissues, and improve disease severity in infected mice. Although these broad-spectrum antivirals did not completely eliminate disease, efficacy in knockout mouse models was nevertheless impressive because of the acute susceptibility of these mice to ZIKV infection and would likely fare better against natural infection in an immunocompetent host. Sofosbuvir is an FDA-approved drug used to treat chronic hepatitis C virus infection [65]. If this compound has clinical efficacy against ZIKV, the approval process would be truncated because the compound is well characterized for use in humans. BCX4430 has been shown to be effective against a wide range of viruses of human concern [66–68], and clinical trials have been initiated. NITD008 was initially identified as a potential antiviral to treat DENV but had toxicity after long-term treatment [69]. Short-term treatment, applicable to treatment of acute arboviral diseases, did not result in appreciable toxicity, and further clinical investigation may be warranted. Broad-spectrum activity of NITD008 has also been observed in various animal models [69, 70].

Indirect-acting antiviral agents have also been identified to have activity against ZIKV in mouse models. The compound 25-hydroxycholesterol is an enzymatic product of cholesterol-25-hydroxylase. Treatment with 25-hydroxycholesterol reduced the ZIKV RNA level in the serum by blocking viral entry in mice and macaques, including reduction of the incidence of microcephaly in a congenital model [71]. Azithromycin, a macrolide antibiotic that is FDA approved for use, including during pregnancy, prevented ZIKV production and viral-mediated cell death in primary human brain tissue by reducing viral proliferation and cytopathic effects in glial cells and astrocytes [72]. However, activity has not yet been demonstrated in a small-animal model.

Antibody therapy has been used to control virus infection and disease during outbreaks [73]. Antibodies targeting the envelope protein and, in particular, domain III have been shown to include potent neutralizers that would be suitable for use in therapy [74, 75]. Efficient neutralizing antibodies have been isolated from the serum of people who have been infected with ZIKV, revealing the importance of antibodies targeting the envelope protein in the context of clearance of ZIKV infection [76]. Some of these antibodies have shown activity in mouse models and are effective in preventing or reducing disease severity after ZIKV infection, including prevention of congenital infection after treatment of pregnant dams [76, 77]. This is consistent with previous studies with the related West Nile virus, where prevention of congenital infection was observed in rodent models treated with pooled human immune serum [78].

Care must be taken in regard to use of antibody therapy in the context of ZIKV infection, as treatment with convalescent plasma from donors possessing antibodies specific for flaviviruses, including DENV and West Nile virus, may enhance disease severity [23]. Antibodies containing mutations in the Fc receptor region maintain the specificity and neutralizing capabilities of the antibodies but remove the possibility for negative enhancement interactions of antibodies with immune cells. Antibodies with an inactivated Fc receptor have been shown to be effective in small-animal models [76].

Vaccines are very important in controlling acute arboviral diseases and have demonstrated efficacy in substantially reducing the disease burden of yellow fever virus, the archetypical flavivirus [79]. Various types of vaccines have been developed to protect against ZIKV (Table 5), and additional studies are being published at a rapid pace. Inactivated or attenuated viruses may also elicit long-term immunity, as is the case with the attenuated 17D yellow fever virus vaccine. Inactivated and modified live-attenuated viruses (which have a 10-nucleotide deletion in the 3ʹ untranslated region) have shown promise in preventing ZIKV infection in mouse models [80, 81]. Development of vaccines that target the envelope protein have been prominent and show promise in various animal models [82, 83]. As with antibody therapy, an important question in regard to vaccine development is whether the antibodies elicited by vaccination might serve to enhance infection with ZIKV. Some reports of enhancement in small-animal models have been reported [23].

Table 5.

Various Anti–Zika Virus (ZIKV) Countermeasures Tested in Small-Animal Models

Countermeasure, Mouse Strain (Age) Virus Strain Treatment Protocol(s) Outcome(s) Reference
Antiviral therapy
AG129 (8–14 wk) MR 766 7 DMA, oral, 50 mg/kg/d, once daily × 10 beginning -1 h Reduced viral RNA level by 0.5–1.3 log10, delayed mean time to death to 15–23 d after virus injection [9]
AG129 (8–10 wk) P 6–740 BCX4430, 300 mg/kg/d, intramuscular, twice daily × 7 d beginning 0–7 d after virus injection Reduced viral RNA level by approximately 2 log10 5 d after virus injection, delayed or prevented death, reduced disease severity, survival after rechallenge [7]
IFNAR-/- (4 wk) GZ01/2016 NITD008, 50 mg/kg, oral, 4, 24, 48, 72, and 96 h after virus injection Reduced viral RNA level 2.6-fold 2 d after virus injection, 50% survival, reduced disease signs [63]
C57BL/6 + Ifnar1 mAb DAKAR 41519 Sofosbuvir, 33 mg/kg/d, oral × 7 d, beginning 24 h after virus injection Improved survival, extended mean time to death [64]
BALB/c, IFNAR-/- (3–4 wk) GZ01/2016 25-Hydroxycholesterol, 50 mg/kg, intraperitoneal × 7 d, beginning -12 h Reduced viral RNA level in serum (BALB/c), improved survival and reduced viral RNA level in various tissues (IFNAR-/-) [71]
BALB/c (6–8 wk) + DEX PRVABC-59 Pegylated interferon alfa 2b, 103.3 IU, subcutaneous, every 96 h 1–9 d after virus injection; interferon beta 1b, 105.2 IU/dose, intraperitoneal, every 48 h 1–9 d after virus injection Reduced viral loads in various tissues, 100% survival, no prominent inflammation in any of the tissues tested [18]
Antibody therapy
Pregnant ICR mice E13.5 SZ01 Convalescent sera, 100 µL, intraperitoneal, 1 d (E14.5) and 2 d (E15.5) after virus injection) nAb titer of 161, reduced caspase 3 level in cortex, rescued cortical plate thinning [77]
IFNAR-/- H/PF/2013 Human mAb from DENV-infected patients, 10 µg, intraperitoneal, -1 and 9 d after virus injection mAb EDE1 C10 neutralized ZIKV, 100% survival [87]
C57BL/6 + Ifnar1 mAb ZIKV- Dakar mAb ZIKV-117 from human antisera, intraperitoneal, 6.7 mg/kg 1 d after virus injection or 16.7 mg/kg 5 d after virus injection 100% survival (1 d after virus injection) or 70%–75% survival (5 d after virus injection); reduced transmission, pathology, and mortality [76]
Ifnar1 -/- female × WT male ZIKV- Dakar (E6.5) mAb ZIKV-117, dams treated with 6.7 mg/kg 1 d after virus injection or 16.7 mg/kg 5 d after virus injection 4–5-log10 reduced virus level in fetal and maternal tissues, protection in pregnancy model due to neutralization [76]
Pregnant C57BL/6 mice ZIKV- Dakar (E5.5) mAb ZIKV-117, dams treated with 16.7 mg/kg 1 d after virus injection Reduced virus in dams, placenta, and fetus; improved fetal and placental disease severity; prevented vertical transmission [76]
Vaccine
AG129 (4–6 wk) FSS13025 or MR 766 FSS13025 was administered subcutaneously. Inactivated MR 766 virus, vaccine, 10 µg/dose, 0 and 21 d, intramuscular 100% survival in MR 766 and FSS13025 infection, absence of detectable viremia in serum up to 6 d after virus injection [81]
BALB/c, C57BL/6 or SJL (4 wk) ZIKV2015, PRVABC-59 DNA prM-Env or DNA Env vaccines, 50 µg, intramuscular, -28 d after virus injection Reduced viral RNA level, correlation of efficacy with nAb, Ab transfer improves [82]
IFNAR-/- FSS13025 10-del ZIKV, route, -4 wk High nAb titer, robust T-cell response, 100% survival, undetectable viremia [80]
CD-1 (1 d) FSS13025 10-del ZIKV, as above 100% survival
AG129 (6 wk) P6-740 prM-Env mRNA, -6 wk High nAb, 100% survival [83]
C57BL/6 + Ifnar1 mAb (8 or 18 wk) Dakar 41519 Modified prM-Env mRNA, -6 wk High nAb, 100% survival
C57BL/6, BALB/c MR-766 VSV-ZprME, VSV-ZENV, intravenous or intramuscular, -3 wk Offspring born to vaccinated females were protected from challenge on postnatal d 7 [88]

Abbreviations: conv, convalescent serum; Env, envelope protein; ffu, focus-forming units; mAb, monoclonal antibody; mRNA, messenger RNA; nAb, neutralizing antibody; pfu, plaque-forming units; RDRP, RNA-dependent RNA polymerase; VSV, vesicular stomatitis virus.

CONCLUSIONS

Small-animal models are important in delineating the consequences of ZIKV. The use of various rodent species and strains, including genetic knockouts, has provided useful information to help us better understand disease as a result of infection with this virus. Small-animal models are useful for the evaluation of antiviral agents and vaccines during preclinical studies. This includes the usefulness of these models for evaluating countermeasures against congenital and sexual transmission, which is very difficult in larger-animal models. Rodent models appear to be more susceptible to enhancement by antibodies to other flaviviruses, compared with primates, and may not be the best model system for humans. Robust infection of rodents typically requires knock down of the IFN response to cause disease, which has required the use of immunodeficient rodent strains and includes more-severe disease pathology. Continued efforts will provide information to aid in the development of countermeasures to reduce the disease burden of this emerging virus, and it is anticipated that improved small animals will be developed that better replicate the features of natural ZIKV infection and disease.

Notes

Supplement sponsorship. This work is part of a supplement sponsored by the National Institute of Allergy and Infectious Diseases (NIAID), part of the National Institutes of Health (NIH).

Potential conflicts of interest. Both authors: No reported conflicts of interest. Both authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

References

  • 1. Bearcroft WG. Zika virus infection experimentally induced in a human volunteer. Trans R Soc Trop Med Hyg 1956; 50:442–8. [PubMed] [Google Scholar]
  • 2. Dick GW. Zika virus. II. Pathogenicity and physical properties. Trans R Soc Trop Med Hyg 1952; 46:521–34. [DOI] [PubMed] [Google Scholar]
  • 3. Dick GW, Kitchen SF, Haddow AJ. Zika virus. I. Isolations and serological specificity. Trans R Soc Trop Med Hyg 1952; 46:509–20. [DOI] [PubMed] [Google Scholar]
  • 4. Reagan RL, Chang SC, Brueckner AL. Electron micrographs of erythrocytes from Swiss albino mice infected with Zika virus. Tex Rep Biol Med 1955; 13:934–8. [PubMed] [Google Scholar]
  • 5. Way JH, Bowen ET, Platt GS. Comparative studies of some African arboviruses in cell culture and in mice. J Gen Virol 1976; 30:123–30. [DOI] [PubMed] [Google Scholar]
  • 6. Aliota MT, Caine EA, Walker EC, Larkin KE, Camacho E, Osorio JE. Characterization of lethal Zika virus infection in AG129 mice. PLoS Negl Trop Dis 2016; 10:e0004682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Julander JG, Siddharthan V, Evans J et al. Efficacy of the broad-spectrum antiviral compound BCX4430 against Zika virus in cell culture and in a mouse model. Antiviral Res 2017; 137:14–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Rossi SL, Tesh RB, Azar SR et al. Characterization of a novel murine model to study Zika virus. Am J Trop Med Hyg 2016; 94:1362–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Zmurko J, Marques RE, Schols D, Verbeken E, Kaptein SJ, Neyts J. The viral polymerase inhibitor 7-Deaza-2’-C-Methyladenosine is a potent inhibitor of in vitro Zika virus replication and delays disease progression in a robust mouse infection model. PLoS Negl Trop Dis 2016; 10:e0004695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Widman DG, Young E, Yount BL et al. A reverse genetics platform that spans the Zika virus family tree. mBio 2017; 8:doi: 10.1128/mBio.02014-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Roundy CM, Azar SR, Rossi SL et al. Variation in Aedes aegypti mosquito competence for Zika virus transmission. Emerg Infect Dis 2017; 23:625–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Lazear HM, Govero J, Smith AM et al. A mouse model of Zika virus pathogenesis. Cell Host Microbe 2016; 19:720–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Tripathi S, Balasubramaniam VR, Brown JA et al. A novel Zika virus mouse model reveals strain specific differences in virus pathogenesis and host inflammatory immune responses. PLoS Pathog 2017; 13:e1006258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Siddharthan V, Wang H, Motter NE et al. Persistent West Nile virus associated with a neurological sequela in hamsters identified by motor unit number estimation. J Virol 2009; 83:4251–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Julander JG, Skirpstunas R, Siddharthan V et al. C3H/HeN mouse model for the evaluation of antiviral agents for the treatment of Venezuelan equine encephalitis virus infection. Antiviral Res 2008; 78:230–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Singh PK, Guest JM, Kanwar M et al. Zika virus infects cells lining the blood-retinal barrier and causes chorioretinal atrophy in mouse eyes. JCI Insight 2017; 2:e92340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Smith DR, Hollidge B, Daye S et al. Neuropathogenesis of Zika virus in a highly susceptible immunocompetent mouse model after antibody blockade of type I interferon. PLoS Negl Trop Dis 2017; 11:e0005296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Chan JF, Zhang AJ, Chan CC et al. Zika virus infection in dexamethasone-immunosuppressed mice demonstrating disseminated infection with multi-organ involvement including orchitis effectively treated by recombinant type I interferons. EBioMedicine 2016; 14:112–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Fernandes NC, Nogueira JS, Réssio RA et al. Experimental Zika virus infection induces spinal cord injury and encephalitis in newborn Swiss mice. Exp Toxicol Pathol 2017; 69:63–71. [DOI] [PubMed] [Google Scholar]
  • 20. Halstead SB. Pathogenesis of dengue: dawn of a new era. F1000Res 2015; 4:doi: 10.12688/f1000research.7024.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Balsitis SJ, Williams KL, Lachica R et al. Lethal antibody enhancement of dengue disease in mice is prevented by Fc modification. PLoS Pathog 2010; 6:e1000790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Zellweger RM, Prestwood TR, Shresta S. Enhanced infection of liver sinusoidal endothelial cells in a mouse model of antibody-induced severe dengue disease. Cell Host Microbe 2010; 7:128–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Bardina SV, Bunduc P, Tripathi S et al. Enhancement of Zika virus pathogenesis by preexisting antiflavivirus immunity. Science 2017; 356:175–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Pantoja P, Pérez-Guzmán EX, Rodríguez IV et al. Zika virus pathogenesis in rhesus macaques is unaffected by pre-existing immunity to dengue virus. Nat Commun 2017; 8:15674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Priyamvada L, Hudson W, Ahmed R, Wrammert J. Humoral cross-reactivity between Zika and dengue viruses: implications for protection and pathology. Emerg Microbes Infect 2017; 6:e33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Halai UA, Nielsen-Saines K, Moreira ME et al. Maternal Zika virus disease severity, virus load, prior dengue antibodies and their relationship to birth outcomes. Clin Infect Dis 2017; doi: 10.1093/cid/cix472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Goodfellow FT, Tesla B, Simchick G et al. Zika virus induced mortality and microcephaly in chicken embryos. Stem Cells Dev 2016; 25:1691–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Kumar M, Krause KK, Azouz F, Nakano E, Nerurkar VR. A guinea pig model of Zika virus infection. Virol J 2017; 14:75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Moore CA, Staples JE, Dobyns WB et al. Characterizing the pattern of anomalies in congenital Zika syndrome for pediatric clinicians. JAMA Pediatr 2017; 171:288–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. França GV, Schuler-Faccini L, Oliveira WK et al. Congenital Zika virus syndrome in Brazil: a case series of the first 1501 livebirths with complete investigation. Lancet 2016; 388:891–7. [DOI] [PubMed] [Google Scholar]
  • 31. Miner JJ, Cao B, Govero J et al. Zika virus infection during pregnancy in mice causes placental damage and fetal demise. Cell 2016; 165:1081–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Siddharthan V, Van Wettere AJ, Li R et al. Zika virus infection of adult and fetal STAT2 knock-out hamsters. Virology 2017; 507:89–95. [DOI] [PubMed] [Google Scholar]
  • 33. Wu KY, Zuo GL, Li XF et al. Vertical transmission of Zika virus targeting the radial glial cells affects cortex development of offspring mice. Cell Res 2016; 26:645–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Xavier-Neto J, Carvalho M, Pascoalino BD et al. Hydrocephalus and arthrogryposis in an immunocompetent mouse model of ZIKA teratogeny: a developmental study. PLoS Negl Trop Dis 2017; 11:e0005363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Cugola FR, Fernandes IR, Russo FB et al. The Brazilian Zika virus strain causes birth defects in experimental models. Nature 2016; 534:267–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Yockey LJ, Varela L, Rakib T et al. Vaginal exposure to Zika virus during pregnancy leads to fetal brain infection. Cell 2016; 166:1247–56.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Rossant J, Cross JC. Placental development: lessons from mouse mutants. Nat Rev Genet 2001; 2:538–48. [DOI] [PubMed] [Google Scholar]
  • 38. Cross JC, Werb Z, Fisher SJ. Implantation and the placenta: key pieces of the development puzzle. Science 1994; 266:1508–18. [DOI] [PubMed] [Google Scholar]
  • 39. Enders AC. A comparative study of the fine structure of the trophoblast in several hemochorial placentas. Am J Anat 1965; 116:29–67. [DOI] [PubMed] [Google Scholar]
  • 40. Georgiades P, Ferguson-Smith AC, Burton GJ. Comparative developmental anatomy of the murine and human definitive placentae. Placenta 2002; 23:3–19. [DOI] [PubMed] [Google Scholar]
  • 41. Julander JG, Winger QA, Rickords LF et al. West Nile virus infection of the placenta. Virology 2006; 347:175–82. [DOI] [PubMed] [Google Scholar]
  • 42. Mathur A, Arora KL, Chaturvedi UC. Congenital infection of mice with Japanese encephalitis virus. Infect Immun 1981; 34:26–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Vermillion MS, Lei J, Shabi Y et al. Intrauterine Zika virus infection of pregnant immunocompetent mice models transplacental transmission and adverse perinatal outcomes. Nat Commun 2017; 8:14575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Brasil P, Pereira JP Jr, Moreira ME et al. Zika virus infection in pregnant women in Rio de Janeiro. N Engl J Med 2016; 375:2321–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Li C, Xu D, Ye Q et al. Zika virus disrupts neural progenitor development and leads to microcephaly in mice. Cell Stem Cell 2016; 19:672. [DOI] [PubMed] [Google Scholar]
  • 46. Victora CG, Castro MC, França GV, Schuler-Faccini L, Barros FC. Zika rash and increased risk of congenital brain abnormalities—Authors’ reply. Lancet 2017; 389:152. [DOI] [PubMed] [Google Scholar]
  • 47. Schwartz DA. Viral infection, proliferation, and hyperplasia of Hofbauer cells and absence of inflammation characterize the placental pathology of fetuses with congenital Zika virus infection. Arch Gynecol Obstet 2017; 295:1361–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Rosenberg AZ, Yu W, Hill DA, Reyes CA, Schwartz DA. Placental pathology of Zika virus: viral infection of the placenta induces villous stromal macrophage (Hofbauer Cell) proliferation and hyperplasia. Arch Pathol Lab Med 2017; 141:43–8. [DOI] [PubMed] [Google Scholar]
  • 49. Simoni MK, Jurado KA, Abrahams VM, Fikrig E, Guller S. Zika virus infection of hofbauer cells. Am J Reprod Immunol 2017; 77:doi: 10.1111/aji.12613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Sheridan MA, Yunusov D, Balaraman V et al. Vulnerability of primitive human placental trophoblast to Zika virus. Proc Natl Acad Sci U S A 2017; 114:E1587–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Foy BD, Kobylinski KC, Chilson Foy JL et al. Probable non-vector-borne transmission of Zika virus, Colorado, USA. Emerg Infect Dis 2011; 17:880–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. D’Ortenzio E, Matheron S, Yazdanpanah Y et al. Evidence of sexual transmission of Zika virus. N Engl J Med 2016; 374:2195–8. [DOI] [PubMed] [Google Scholar]
  • 53. Davidson A, Slavinski S, Komoto K, Rakeman J, Weiss D. Suspected female-to-male sexual transmission of Zika Virus—New York City, 2016. MMWR Morb Mortal Wkly Rep 2016; 65:716–7. [DOI] [PubMed] [Google Scholar]
  • 54. Deckard DT, Chung WM, Brooks JT et al. Male-to-male sexual transmission of Zika virus–Texas, January 2016. MMWR Morb Mortal Wkly Rep 2016; 65:372–4. [DOI] [PubMed] [Google Scholar]
  • 55. Hills SL, Russell K, Hennessey M et al. Transmission of Zika virus through sexual contact with travelers to areas of ongoing transmission—Continental United States, 2016. MMWR Morb Mortal Wkly Rep 2016; 65:215–6. [DOI] [PubMed] [Google Scholar]
  • 56. Musso D, Roche C, Robin E, Nhan T, Teissier A, Cao-Lormeau VM. Potential sexual transmission of Zika virus. Emerg Infect Dis 2015; 21:359–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Moreira J, Peixoto TM, Siqueira AM, Lamas CC. Sexually acquired Zika virus: a systematic review. Clin Microbiol Infect 2017; 23:296–305. [DOI] [PubMed] [Google Scholar]
  • 58. Duggal NK, Ritter JM, Pestorius SE et al. Frequent Zika virus sexual transmission and prolonged viral RNA shedding in an immunodeficient mouse model. Cell Rep 2017; 18:1751–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Govero J, Esakky P, Scheaffer SM et al. Zika virus infection damages the testes in mice. Nature 2016; 540:438–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Uraki R, Hwang J, Jurado KA et al. Zika virus causes testicular atrophy. Sci Adv 2017; 3:e1602899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Ma W, Li S, Ma S et al. Zika virus causes testis damage and leads to male infertility in mice. Cell 2016; 167:1511–24.e10. [DOI] [PubMed] [Google Scholar]
  • 62. Tang WW, Young MP, Mamidi A, Regla-Nava JA, Kim K, Shresta S. A mouse model of zika virus sexual transmission and vaginal viral replication. Cell Rep 2016; 17:3091–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Deng YQ, Zhang NN, Li CF et al. Adenosine analog NITD008 is a potent inhibitor of Zika virus. Open Forum Infect Dis 2016; 3:ofw175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Bullard-Feibelman KM, Govero J, Zhu Z et al. The FDA-approved drug sofosbuvir inhibits Zika virus infection. Antiviral Res 2017; 137:134–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Olsen DB, Eldrup AB, Bartholomew L et al. A 7-deaza-adenosine analog is a potent and selective inhibitor of hepatitis C virus replication with excellent pharmacokinetic properties. Antimicrob Agents Chemother 2004; 48:3944–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Julander JG, Bantia S, Taubenheim BR et al. BCX4430, a novel nucleoside analog, effectively treats yellow fever in a Hamster model. Antimicrob Agents Chemother 2014; 58:6607–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Taylor R, Kotian P, Warren T et al. BCX4430—a broad-spectrum antiviral adenosine nucleoside analog under development for the treatment of Ebola virus disease. J Infect Public Health 2016; 9:220–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Warren TK, Wells J, Panchal RG et al. Protection against filovirus diseases by a novel broad-spectrum nucleoside analogue BCX4430. Nature 2014; 508:402–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Yin Z, Chen YL, Schul W et al. An adenosine nucleoside inhibitor of dengue virus. Proc Natl Acad Sci U S A 2009; 106:20435–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Nelson J, Roe K, Orillo B, Shi PY, Verma S. Combined treatment of adenosine nucleoside inhibitor NITD008 and histone deacetylase inhibitor vorinostat represents an immunotherapy strategy to ameliorate West Nile virus infection. Antiviral Res 2015; 122:39–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Li C, Deng YQ, Wang S et al. 25-Hydroxycholesterol protects host against Zika virus infection and its associated microcephaly in a mouse model. Immunity 2017; 46:446–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Retallack H, Di Lullo E, Arias C et al. Zika virus cell tropism in the developing human brain and inhibition by azithromycin. Proc Natl Acad Sci U S A 2016; 113:14408–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Winkler AM, Koepsell SA. The use of convalescent plasma to treat emerging infectious diseases: focus on Ebola virus disease. Curr Opin Hematol 2015; 22:521–6. [DOI] [PubMed] [Google Scholar]
  • 74. Diamond MS, Pierson TC, Fremont DH. The structural immunology of antibody protection against West Nile virus. Immunol Rev 2008; 225:212–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Sukupolvi-Petty S, Austin SK, Purtha WE et al. Type- and subcomplex-specific neutralizing antibodies against domain III of dengue virus type 2 envelope protein recognize adjacent epitopes. J Virol 2007; 81:12816–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Sapparapu G, Fernandez E, Kose N et al. Neutralizing human antibodies prevent Zika virus replication and fetal disease in mice. Nature 2016; 540:443–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Wang Q, Yang H, Liu X et al. Molecular determinants of human neutralizing antibodies isolated from a patient infected with Zika virus. Sci Transl Med 2016; 8:369ra179. [DOI] [PubMed] [Google Scholar]
  • 78. Julander JG, Winger QA, Olsen AL, Day CW, Sidwell RW, Morrey JD. Treatment of West Nile virus-infected mice with reactive immunoglobulin reduces fetal titers and increases dam survival. Antiviral Res 2005; 65:79–85. [DOI] [PubMed] [Google Scholar]
  • 79. Monath TP. Review of the risks and benefits of yellow fever vaccination including some new analyses. Expert Rev Vaccines 2012; 11:427–48. [DOI] [PubMed] [Google Scholar]
  • 80. Shan C, Muruato AE, Nunes BTD et al. A live-attenuated Zika virus vaccine candidate induces sterilizing immunity in mouse models. Nat Med 2017; 23:763–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Sumathy K, Kulkarni B, Gondu RK et al. Protective efficacy of Zika vaccine in AG129 mouse model. Sci Rep 2017; 7:46375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Larocca RA, Abbink P, Peron JP et al. Vaccine protection against Zika virus from Brazil. Nature 2016; 536:474–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Richner JM, Himansu S, Dowd KA et al. Modified mRNA vaccines protect against Zika virus infection. Cell 2017; 168:1114–25.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Dowall SD, Graham VA, Rayner E et al. A susceptible mouse model for Zika virus infection. PLoS Negl Trop Dis 2016; 10:e0004658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Manangeeswaran M, Ireland DD, Verthelyi D. Zika (PRVABC59) infection is associated with T cell Infiltration and neurodegeneration in CNS of immunocompetent neonatal C57Bl/6 mice. PLoS Pathog 2016; 12:e1006004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Costa F, Sarno M, Khouri R et al. Emergence of congenital Zika syndrome: viewpoint from the front lines. Ann Intern Med 2016; 164:689–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Swanstrom JA, Plante JA, Plante KS et al. Dengue virus envelope dimer epitope monoclonal antibodies isolated from dengue patients are protective against Zika virus. mBio 2016; 7:doi: 10.1128/mBio.01123-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Betancourt D, de Queiroz NM, Xia T, Ahn J, Barber GN. Cutting edge: innate immune augmenting vesicular stomatitis virus expressing Zika virus proteins confers protective immunity. J Immunol 2017; 198:3023–8. [DOI] [PubMed] [Google Scholar]

Articles from The Journal of Infectious Diseases are provided here courtesy of Oxford University Press

RESOURCES