Sie sind auf Seite 1von 10

Review

pubs.acs.org/journal/aidcbc

Zika Virus: Emergence, Phylogenetics, Challenges, and Opportunities


Maaran M. Rajah,† Ryan D. Pardy,† Stephanie A. Condotta,† Martin J. Richer,*,†,‡
and Selena M. Sagan*,†,‡

Department of Microbiology and Immunology and ‡Microbiome and Disease Tolerance Centre (MDTC), McGill University,
Montréal, Québec, Canada H3A 2B4

ABSTRACT: Zika virus (ZIKV) is an emerging arthropod-


borne pathogen that has recently gained notoriety due to its
rapid and ongoing geographic expansion and its novel
association with neurological complications. Reports of
ZIKV-associated Guillain−Barré syndrome as well as fetal
microcephaly place emphasis on the need to develop
preventative measures and therapeutics to combat ZIKV
infection. Thus, it is imperative that models to study ZIKV
replication and pathogenesis and the immune response are
developed in conjunction with integrated vector control
strategies to mount an efficient response to the pandemic.
This paper summarizes the current state of knowledge on
ZIKV, including the clinical features, phylogenetic analyses, pathogenesis, and the immune response to infection. Potential
challenges in developing diagnostic tools, treatment, and prevention strategies are also discussed.
KEYWORDS: Zika virus, Guillain−Barré syndrome, fetal microcephaly

■ INTRODUCTION
Zika virus (ZIKV) is an emerging mosquito-borne pathogen
■ TRANSMISSION AND CLINICAL FEATURES OF
ZIKV INFECTION
that has recently become a significant global health concern due ZIKV is an arbovirus that belongs to the Flavivirus genus of the
to its rapid geographical expansion and pathogenesis associated Flaviviridae family. Viral transmission occurs through both
with infection. The virus was initially described in 1947 in the sylvatic and urban cycles involving a variety of Aedes species
Zika forest region of Uganda, where it was isolated from the mosquitos and either nonhuman primates or humans as the
blood of sentinel rhesus macaques.1 Serological surveys amplifying reservoir, respectively. It is not yet clear whether
conducted in West Africa demonstrated the presence of other mammals or mosquito species can serve as an amplifying
reservoir, but viral replication in cell culture has been
ZIKV antibodies in human populations; however, the first
demonstrated in a variety of animal cell lines.9 Recent papers
human infection was not reported until 1964.2,3 For the latter
have also described ZIKV replication in testicular tissue and
half of the 20th century the virus remained in relative obscurity excretion in semen, suggesting that sexual transmission is also a
with only isolated human cases reported until the first serious route of ZIKV infection.7,10−12 Although transmission of ZIKV
outbreak in 2007.4 During this outbreak, over 73% of the through breast milk has not been documented, it has been
population of Yap Island, Federated States of Micronesia, demonstrated to contain high viral loads, suggesting this may
became infected with ZIKV in a period of 4 months.5,6 This was represent another potential route of transmission.13,14 A deeper
followed by a major outbreak in 2013 in French Polynesia; understanding of the amplifying reservoirs for ZIKV and routes
together, these outbreaks represented the first significant of transmission will be critical to contain the ongoing epidemic
transmission of ZIKV outside its original endemic regions.6 and prevent further spread.
Since then, ZIKV has been introduced into the Western ZIKV replication is first thought to occur in human primary
Hemisphere, causing an ongoing epidemic in South America, dermal fibroblasts, epidermal keratinocytes, and immature
with localized epidemics in Argentina, Colombia, Brazil, El dendritic cells at the site of inoculation.15 From the site of
Salvador, Guatemala, Paraguay, and Venezuela, as well as recent the mosquito bite, ZIKV spreads to the draining lymph node,
outbreaks in the southern United States and Singapore.6−8 where it is amplified and disseminated through the bloodstream
to peripheral tissues and visceral organs. ZIKV is first detectable
Although ZIKV typically causes a mild, self-limiting febrile
in the blood within the first 10 days of infection, with peak viral
illness, its explosive spread across South America and its recent
association with more severe pathogenesis make ZIKV
infection a serious public health concern that must be rapidly Special Issue: Host-Pathogen Interactions
addressed through a concerted collaborative effort between Received: September 14, 2016
researchers, clinicians, and public health officials alike. Published: October 5, 2016

© 2016 American Chemical Society 763 DOI: 10.1021/acsinfecdis.6b00161


ACS Infect. Dis. 2016, 2, 763−772
ACS Infectious Diseases Review

Figure 1. Phylogenetic analysis of ZIKV. Translated amino acid sequences of 94 ZIKV polyproteins were aligned using ClustalW. Trees were
constructed by neighbor joining of pairwise amino acid distances with the program MEGA7 (according to the distance scale provided). Bootstrap
resampling was used to determine robustness of branches; values of ≥50% (from 1000 replicates) are shown. Human (black circles), monkey (gray
circles), and mosquito (open circles) isolates are indicated. Isolate name, country of origin, and year of isolation, as well as the unique accession
numbers for each sequence, are indicated.

loads coinciding with the onset of symptoms, which typically infection has been reported to cause a self-limiting illness that is
present between 2 and 12 days postinfection.16−18 ZIKV mostly asymptomatic, but can present with mild symptoms in
764 DOI: 10.1021/acsinfecdis.6b00161
ACS Infect. Dis. 2016, 2, 763−772
ACS Infectious Diseases Review

up to 20% of cases.19 These symptoms typically include fever, to microcephaly, a number of other malformations have been
maculopapular rash, headache, joint and muscle pain, fatigue, reported in fetuses and newborns with congenital ZIKV
and conjunctivitis.5,6,16 During the Yap Island outbreak, other infection, including intrauterine growth restriction, brain
symptoms including myalgia, headaches, retro-orbital pain, atrophy, cerebral and placental calcifications, arthrogryposis,
edema, and vomiting were also reported.5 The presentation of a and retinal and optic nerve abnormalities.37−40 Although recent
mild febrile illness can be misdiagnosed as Dengue or research has started to decipher some of the mechanisms
Chikungunya virus infection due to their similar clinical through which ZIKV can lead to fetal abnormalities, a
presentation.20 In areas of co-circulation of these pathogens, concerted effort is required to further understand all of the
multiple infections are likely very common;21,22 however, it factors associated with ZIKV-induced pathogenesis. Success will
remains to be seen whether there are synergistic effects due to be dependent on a collaborative multidisciplinary research to
co-infection, vaccination, or past infections.23−25 In the recent assemble epidemiological data along with the establishment of
outbreaks, beginning with the 2013 French Polynesia outbreak, models to study host−pathogen interactions that ultimately
ZIKV has been linked to an increase in more severe shape ZIKV pathogenesis.
neurological complications including Guillain−Barré syndrome
and fetal microcephaly. The factors contributing to this sudden
rise in ZIKV-associated neurological symptoms currently
■ ZIKV GENOME AND PHYLOGENETIC ANALYSES
Similar to other clinically relevant human pathogens belonging
remain unexplained, and research efforts are directed at to this family (e.g., yellow fever, West Nile, and Dengue
identifying host, pathogen, and environmental factors linked viruses), ZIKV is a single-stranded, positive-sense RNA virus
to these complications. with a single open reading frame flanked by highly structured 5′
Guillain−Barré Syndrome. Guillain−Barré syndrome is a and 3′ untranslated regions (UTRs). The genome encodes a
severe neurological autoimmune disease causing acute or single polyprotein that is cleaved by host and viral proteases
subacute flaccid paralysis that is attributable to peripheral into the structural [capsid (C), premembrane protein (prM),
nerve damage.26,27 During the French Polynesia outbreak, there and envelope (E)] and nonstructural proteins (NS1, NS2A,
were 48 reported cases of Guillain−Barré syndrome, and 88% NS2B, NS3, NS4A, 2K, NS4B, and NS5).19,41,42 The structural
of the affected patients reported a symptomatic ZIKV infection proteins form the virion particle, whereas the nonstructural
prior to the onset of neurological symptoms; this was proteins participate in polyprotein processing, viral RNA
comparatively higher than the 5 cases per annum reported replication, virion assembly, and evasion of the host immune
over the previous 4 years.27 A case-controlled study on 42 of response.
these patients demonstrated that all of them had neutralizing Phylogenetic analyses have established that there are two
antibodies directed against ZIKV.27 Although none of these main ZIKV lineages, African and Asian (Figure 1).41−43
patients died, 50% of them were still unable to walk without Notably, all of the contemporary human strains have greater
assistance 3 months after discharge. A similar pattern was sequence homology to the mosquito strain P6-740 (Malaysia,
observed in the current epidemic in Latin America and the 1966) than the African strains, suggesting that the currently
Caribbean, where the incidence of Guillain−Barré syndrome in circulating strains evolved from the Asian lineage, anchored by
seven different countries was reported to be 2−9.8-fold higher P6-740.42,43 In addition, all of the strains identified in the
than baseline.28 Although fatalities among Guillain−Barré 2015−2016 epidemic are more closely related to the H/PF/
syndrome patients are rare, the severity and burden of the 2013 French Polynesia strain than the FSM/2007 Micronesia
disease on the health care system as well as to the affected strain, suggesting that these sublineages may have evolved
patients and their families suggest that research is needed to independently from a common ancestor (Figure 1). Interest-
better understand the underlying pathology and improve ingly, the current ZIKV outbreak in Singapore is of the Asian
diagnosis and treatment.26 lineage, but sequence analyses suggest that it evolved
Fetal Microcephaly. One of the most alarming aspects of independently from a strain that was already circulating in
the recent outbreaks has been the association of ZIKV infection Southeast Asia, rather than being derived from an imported
with an increase in congenital malformations including fetal case from the ongoing South American outbreak. Comparative
microcephaly (a neurodevelopmental disorder that results in analyses of historical ZIKV strains in both in vitro and in vivo
malformation of the brain and head), which can result in severe models is likely to answer important questions regarding the
life-long limitations for the child and family.29 Reports from the emergence and pathogenesis of ZIKV during the current
Brazilian Ministry of Health suggest that there is a 20-fold outbreaks.
increase in the incidence of microcephaly that coincides with Comparative genetic analyses suggest that there are several
the current ZIKV epidemic.30 Recent studies have strongly amino acid polymorphisms that are common to the Asian
linked ZIKV infection during pregnancy to microcephaly, lineage isolates with known clinical outcomes when compared
including the detection of viral RNA in amniotic fluid, ZIKV- with the African (MR766, CDC reference strain) and P6-740
specific IgM antibodies in the cerebrospinal fluid of micro- strains.42,43 The recent acquisition of these amino acid
cephalic neonates (indicative of active central nervous system polymorphisms could potentially contribute to the increased
infection), and reports that ZIKV attenuates growth of human pathogenesis and dissemination seen in the current outbreaks.
neural progenitor cells.6,31−33 Although questions remain about On the basis of their locations within the ZIKV polyprotein,
the type of exposure and whether symptomatic or asympto- these mutations are predicted to contribute to ZIKV infection,
matic infection poses the greatest risk to the fetus, the first replication, pathogenesis, and the immune response to
trimester of pregnancy has been identified as the gestational infection.42,43 Amino acid polymorphisms in the prM, E, and
period at major risk for microcephaly.34,35 This is further NS1 proteins may contribute to infectivity and the immune
supported by a recent study that demonstrated ZIKV-induced response, whereas those in NS2A and NS3 are likely to affect
apoptosis in first trimester human trophoblasts as well as polyprotein processing and replication. Interestingly, the NS4B
detrimental effects on trophoblast differentiation.36 In addition and NS5 proteins contain large clusters of amino acid
765 DOI: 10.1021/acsinfecdis.6b00161
ACS Infect. Dis. 2016, 2, 763−772
ACS Infectious Diseases Review

polymorphisms.42,43 This may be of particular interest because ment) generated from human induced pluripotent stem
NS4B is implicated in replication, pathogenesis, and antiviral cells.33,48−50 These studies indicate that ZIKV can infect
signaling, whereas the NS5 protein is the viral RNA-dependent hNPCs with high efficiency, resulting in increased cell death
RNA polymerase responsible for viral RNA synthesis.42,43 and dysregulation of the cell cycle, as well as impaired growth
Although it is clear that ZIKV has undergone significant and morphogenesis of healthy neurospheres.33 Furthermore,
evolution since its discovery, little is known about how these infected hNPCs release infectious viral particles, presenting a
recently acquired nucleotide and amino acid polymorphisms challenge for the development of therapeutics to halt or block
have contributed to the increased pathogenesis and rapid the impact of infection.33,48−50 Importantly, these findings
dissemination of ZIKV in the current outbreaks.42,43 These suggest a potential mechanism of ZIKV-induced microcephaly
studies have been hampered by the lack of ZIKV reverse as hNPCs are essential for the development of the cortex and
genetics systems and suitable small animal models (discussed in brain. Whereas these cell culture approaches are of great
more detail below). importance in understanding ZIKV infectivity, viral fitness,

■ UNDERSTANDING ZIKV PATHOGENESIS AND THE


IMMUNE RESPONSE TO INFECTION
replication, and antiviral responses, these approaches do not
take into account the complexity of host−pathogen interactions
and need to be complemented by animal models of infection.
Most of our knowledge of the ZIKV life cycle has been Animal Models. The establishment of animal models is
extrapolated from our knowledge of related flaviviruses. As extremely important for understanding ZIKV pathogenesis and
such, much remains to be determined regarding ZIKV the immune response to infection, as well as for the design and
replication, cell tropism, transplacental transmission, patho- testing of effective vaccine strategies and therapeutics. Nonhu-
genesis and the immune response to infection. Furthermore, it man primates are similar to humans in terms of gestation and
is unclear what factors have contributed to the explosive spread fetal development and therefore may provide important
and increased virulence of ZIKV in the recent and ongoing translational insights. To date, the majority of nonhuman
epidemics. The development of genetic tools and experimental primate ZIKV studies have focused on viral dissemination and
systems, including mosquito transmission models, reverse vaccine protection;51,52 however, a recent study demonstrated
genetics systems, and animal models, will be crucial to improve that subcutaneous ZIKV infection of a pregnant pigtail
our understanding of ZIKV transmission, replication and macaque results in arrested fetal brain development and
pathogenesis and the immune response to infection. neuro-invasion.53 Future studies in these models are likely to
Furthermore, these experimental systems will be critical to further our understanding of ZIKV fetal pathogenesis.
facilitate the development of novel vaccine and therapeutic However, nonhuman primates present a number of logistical
strategies. challenges that make them less suitable for certain research
Cell Culture Systems. In vitro and ex vivo models are avenues than other small animal models.
useful for dissecting the molecular mechanisms of viral The use of the wild-type (WT) C57BL/6 or 129 Sv/Ev
infection, fitness, replication, and host−virus interactions. To mouse strains to create a model of ZIKV pathogenesis has been
date, in vitro studies have been focused on determining suitable hampered by the apparent lack of infectivity of ZIKV in these
cell types for ZIKV infection and replication as well as those mice. Indeed, it has been shown in vitro that although the ZIKV
involved in transplacental transmission and neural damage to NS5 protein is capable of impeding IFN signaling in human
investigate ZIKV pathogenesis. The link between ZIKV cells by binding to the STAT2 protein, it is unable to do so in
infection and fetal microcephaly has been demonstrated by murine cells.54 To address this, the majority of mouse models
detection of viral RNA in the placenta, amniotic fluid, and have used mice lacking the IFN-α/β receptor (IFNAR;
brain, as well as ZIKV IgM-specific antibodies in the IFNAR−/− or A129 mice), both IFNAR and the IFN-γ receptor
cerebrospinal fluid of microcephalic neonates.6,29,31−33,44 (IFNGR; AG129 mice), or Irf3−/−Irf5−/−Irf7−/− triple-knock-
Recent studies suggest that ZIKV is able to infect and replicate out mice that produce limited type I IFN (Table 1).10,55−57
in primary human placental cells (cytotrophoblasts, endothelial The ability of ZIKV to cause severe pathogenesis and even fatal
cells, fibroblasts, and Hofbauer cells) from mid to late gestation infection in many of these models emphasizes the importance
as well as villus explants from first-trimester human placentas, of a robust type I IFN response in the innate immune response
suggesting two possible routes of ZIKV transmission to the to ZIKV.10,55−57 Analyses of viral dissemination and patho-
fetus (placental and paraplacental).45,46 Furthermore, ZIKV genesis within these mice have also highlighted ZIKV’s capacity
replication in Hofbauer cells (placental macrophages) was to infect and replicate within immune privileged sites such as
associated with induction of type I interferons (IFN), pro- the reproductive organs, eyes, and brain, similar to reports in
inflammatory cytokines, and antiviral gene expression, but human patients.6,12,58−61 Mice deficient in IFNAR signaling
minimal cytopathic effects, suggesting that these cells may allow have also been used to address the effects of ZIKV infection on
ZIKV to gain access to the fetal compartment and could play a fetal development. After subcutaneous infection of pregnant
role in viral dissemination.46 In contrast, primary human IFNAR−/− mice, ZIKV was detectable in both the placenta and
trophoblasts (the barrier cells of the placenta) from full-term fetal brain, which led to intrauterine growth restriction and fetal
placentas are refractory to ZIKV infection and produce type III demise.58 Similarly, a recent paper demonstrated that intra-
IFN that are postulated to protect trophoblast and non- vaginal infection of IFNAR−/− dams mated with WT males led
trophoblast cells from infection during later stages of to total fetal resorption when infection occurred early in
pregnancy.47 pregnancy.59 Although structural differences between the
Several groups have investigated the link between ZIKV human and mouse placenta must be considered, these studies
infection and neural pathogenesis in vitro using human neural provide an important first step in understanding the ability of
progenitor cells (hNPCs), neurons, and cerebral organoids ZIKV to transverse the placenta, as well as its tropism for neural
(three-dimensional, self-organized, stem-cell-derived models progenitor cells in the developing brain.62 Intriguingly,
that recapitulate the first trimester of human neurodevelop- intravaginal infection of pregnant WT mice still caused
766 DOI: 10.1021/acsinfecdis.6b00161
ACS Infect. Dis. 2016, 2, 763−772
ACS Infectious Diseases Review

Table 1. Murine Models of ZIKV Infection: Opportunities introduce specific mutations as well as generate reporter viruses
and Challenges that will facilitate antiviral drug discovery efforts. Together,
these advances will significantly accelerate the capacity of the
research community to understand ZIKV molecular biology,
pathogenesis, and the immune response and will aid in the
development of tools to counter the growing threat of ZIKV
infection.

■ CHALLENGES FOR ZIKV RESEARCH


Vector Control. The concept of mosquito vector control
has been suggested for many years as an approach to control
vector-borne infections. However, many challenges arise that
can hamper the implementation of effective control strategies.68
For example, the mosquito life cycle is a major determinant for
executing successful vector control initiatives. The primary
mosquito vector for ZIKV transmission is Aedes aegypti, and
recently Aedes albopictus has also been implicated in trans-
mission.69−72 Although both of these species belong to the
Aedes genus, they have different habitat and behavioral
characteristics. For instance, Aedes aegypti species bite during
the day, typically rest indoors, and lay their eggs in artificial
containers in and around homes,68 whereas Aedes albopictus
species bite in the early morning and late afternoon, rest
intrauterine growth restriction in the absence of systemic outdoors, and prefer to lay their eggs in natural containers such
viremia.59 Together, these models provide excellent tools for as tree holes and ground pools.73 These individualistic features
interrogating ZIKV pathogenesis and will be extremely useful need to be considered when vector control approaches are
for testing potential therapeutics. employed; as such, entomological surveillance is imperative for
Despite the challenges associated with infection of WT mice, efficacious vector control initiatives.
there is still an urgent need for immunocompetent mouse Prevention targeted at different mosquito life stages is one
models for ZIKV infection. The lack of an intact type I IFN control method that can be easily implemented by individuals
response in the models described above precludes in-depth at home and through public health community awareness.
analyses of the immune response to ZIKV infection. In addition Mosquitoes go through four distinct and separate life stages:
to a role in innate immune responses, type I IFN plays a critical egg, larvae, pupae, and adult. Monitoring areas of mosquito
role in the induction of an optimal adaptive immune response, populations and eliminating breeding grounds is the first step.
including CD8+ T cells, which are required for clearance of Individuals at home can remove standing water in receptacles in
intracellular pathogens.63 To date, only one immunocompetent and around the home, which will eliminate mosquito eggs,
mouse model has been proposed, which used WT SJL mice larvae, and pupae. Spraying with insecticides will kill adult
(Table 1).50 In this strain, pregnant WT mice infected with a mosquitoes, and because of their unique resting preferences,
Brazilian ZIKV isolate gave birth to pups with severe insecticides should be targeted to the appropriate areas to kill
intrauterine growth restriction, ocular deformities, and reduced adult Aedes aegypti and Aedes albopictus, thus ultimately
cell number and cortical thickness in the brain (symptoms reducing virus transmission. Another aspect to consider is the
associated with microcephaly in humans).50 No such defects possibility of overwintering and vertical transmission.74
were observed in pregnant WT C57BL/6 mice, suggesting Mosquito eggs can survive dry seasons (desiccation) and cold
there may be a genetic influence to ZIKV-induced micro- periods (diapause), facilitating long-term mosquito survival.75
cephaly.50 Analyses of the ZIKV immune response in both During the change of seasons (shorter days and colder
C57BL/6 and SJL mice will be of interest to determine specific temperatures), Aedes albopictus is able to lay diapausing eggs,
differences that dictate the apparent increased susceptibility of ensuring survival until the next hatching period.75 A
SJL mice to ZIKV infection. In addition, immunocompetent consequence of this behavior is that there is a possibility that
mouse models will be critical to understand whether newly ZIKV-infected females could vertically transmit ZIKV to
acquired viral polymorphisms contribute to the capacity of progeny. In fact, vertical transmission of ZIKV has already
ZIKV to counter host immune defenses. been documented in Aedes aegypti, suggesting a potential
Until recently, both in vitro and in vivo studies of ZIKV have mechanism for virus persistence during adverse conditions.76
relied on serial passage of ZIKV isolates in suckling mice or cell Thus, although vector control is an attractive option to reduce
culture. However, these systems are not suitable for the ZIKV transmission, there are a large number of variables that
dissection of viral polymorphisms (limited to those mutations need to be considered to implement effective control protocols.
present in viral isolates) and may result in the selection of viral Biological control and genetic manipulation represent
mutants (e.g., cell culture adaptations). Understanding the another set of tools that can be used for vector control. The
contribution of viral polymorphisms to the explosive trans- bacterium Wolbachia has been demonstrated to reduce the
mission and increased pathogenesis of currently circulating ability of mosquitoes to transmit viruses to humans.77,78 When
ZIKV strains has been hampered by the lack of ZIKV reverse introduced into Aedes aegypti populations, a symbiotic strain of
genetics models; however, several groups have now reported Wolbachia reduces ZIKV replication within the mosquito,
the development of full-length infectious cDNA clones.64−67 delaying virus dissemination to the mosquito salivary glands
These ZIKV reverse genetic systems will allow researchers to and therefore reducing vector competence.77,78 Genetic
767 DOI: 10.1021/acsinfecdis.6b00161
ACS Infect. Dis. 2016, 2, 763−772
ACS Infectious Diseases Review

modification of mosquitos is another strategy that has been inhibition of several flaviviruses, including ZIKV, suggesting
proposed. The introduction of the OX513A gene into the that these compounds might represent lead candidates for the
mosquito genome results in normal development of larvae; development of specific ZIKV antivirals.90−92 An alternative
however, these genetically modified mosquitos die before pharmacologic strategy is to perform drug-repurposing screens
adulthood, potentially reducing the risk of virus trans- of existing clinical compounds for their potential anti-ZIKV
mission.79−82 The possibility of environmental damage and a activity. This approach has been undertaken for other emerging
general public distrust of genetically modified organisms are viruses, and two such screens have now been reported for
two of the current limitations hindering the widespread ZIKV.93,94 These studies each screened hundreds of clinically
implementation of vector control strategies in ZIKV-affected approved drugs, clinical trial drug candidates, and pharmaco-
areas.83 However, the effectiveness of such strategies has been logically active compounds that exhibited anti-ZIKV activity or
demonstrated in recent field studies, where the sustained were neuroprotective in nature.93,94 Several active compounds
release of OX513A Aedes aegypti males successfully reduced the were identified in both studies that were further validated in
local Aedes aegypti population by 95% in Bahia, Brazil.84 The ZIKV-infected human neural stem cells and primary amnion
future of vector control will thus require an integrated approach cells, demonstrating the efficacy of such screening strategies to
where basic strategies such as eliminating breeding grounds and identify lead compounds for anti-ZIKV drug development.
larval source management will need to be implemented in As an alternative to small-molecule-based inhibitors,
conjunction with more sophisticated strategies, including therapeutic antibodies could also be developed to prevent or
biocontrol and genetic manipulation.83 treat ZIKV infections. Neutralizing antibodies have already
Diagnostics. Definitive diagnosis of ZIKV infection is been studied in the context of Dengue virus infection and have
challenging due to the similar symptomology and broad cross- demonstrated reduction in viral loads in a variety of
reactivity of Flavivirus antibodies induced during infection. As experimental systems.95 In addition to reducing viremia in
such, reliable diagnosis can be achieved only by laboratory the mother, the ability of IgG antibodies to cross the placenta
assays. At present, laboratory diagnosis of ZIKV infection relies suggests that ZIKV-neutralizing antibodies could reach the
on isolation of the virus in culture, detection of viral RNA in fetus, protecting against infection and fetal microcephaly.
blood or other biological samples, or serologic testing. Thus, a However, the use of anti-ZIKV antibodies should be carefully
major challenge in ZIKV research is the development of selected to minimize side effects and potential disease
diagnostic assays that can rapidly and definitively diagnose enhancement.96 No matter the strategy, the major challenge
ZIKV infection. in the development of ZIKV inhibitors will be the time and
Although the gold standard to diagnose viral infections is caution required to critically evaluate the efficacy and toxicity
isolation of the virus in culture, this requires sufficient viral load, for use in pregnant women, who represent the highest risk
appropriate timing of sample collection, and significant population.
resources to perform. This renders this approach difficult to Vaccines. A variety of different vaccine platforms are
implement in the field. Detection of viral RNA using RT-PCR currently being explored for ZIKV. These include subunit and
is the most widespread diagnostic assay used to detect ZIKV in DNA-based vaccines as well as the inactivated or live attenuated
blood, urine, saliva, or semen specimens.85−87 RT-PCR-based vaccines. A recent study reported efficacy of both plasmid
assays have the best sensitivity during the first week of DNA- and rhesus adenovirus vector-based vaccines, both of
symptom onset while patients are still viremic;87 however, viral which expressed the ZIKV prM and E proteins, which were able
RNA can still be detected in samples several months after the to successfully confer full protection against ZIKV challenge.51
onset of symptoms.11,87−89 Although the time to development for a subunit or DNA-based
Both in-house and commercial assays have been developed vaccine is likely to be shorter than inactivated or live attenuated
for serologic testing for ZIKV IgM and IgG antibodies in vaccine strategies, this approach is likely to require multiple
serum.85 However, many ZIKV immunoassays have significant doses to achieve protective immunity. In addition, this
cross-reactivity with heterologous flaviviruses from previous approach is likely to induce only an antibody response, thus
infections or vaccination.18,85 Positive results from enzyme- making it more susceptible to being countered by viral
linked immunosorbent assays (ELISA) or immunofluorescence evolution. In contrast, live attenuated approaches typically
assays should be confirmed by neutralization assays; however, elicit both antibody and cell-mediated immunity and are thus
in patients with secondary Flavivirus infections, neutralization likely to provide more robust protection. In line with this, a
assays may not be conclusive due to the induction of broadly recent study demonstrated that a purified inactivated virus
neutralizing antibodies from previous infections.18,85 This is a vaccine was indeed able to confer full protection against the
particular concern in regions of South America, where other Brazilian and Puerto Rican strains of ZIKV in rhesus
flaviviruses, such as Dengue virus, are endemic and have macaques.51 Although each of these approaches has merit,
infected the large majority of the population. Thus, the given that there are already licensed inactivated or live
development of more specific antigens is likely required to attenuated vaccines for several flaviviruses, it is likely that one
improve serologic diagnosis of ZIKV infection in the future. of these approaches will have the greatest likelihood of
Therapeutics. To date, there are no clinically approved success.97
antiviral therapeutics for flaviviruses. However, some of the The major complications surrounding successful vaccine
experience gained from drug discovery efforts for Dengue virus development arise from distinct features of ZIKV infection and
and related viruses can be applied to ZIKV. Both viral infection include the low incidence of clinical presentation, short period
models and viral enzyme assays will be useful in identifying of viremia, the risk of neurological symptoms, and the risk of
ZIKV-specific inhibitors from small-molecule compound sexual transmission.98 Further complications for vaccine
libraries. In fact, nucleotide/nucleoside-based inhibitors, such development could result from the close phylogenetic relation-
as 7-deaza-2′-C methyladenosine, T-705 (favipiravir), and 2′-C- ship between ZIKV and other arthropod-borne flaviviruses.99
methylated nucleosides, have already shown promise for Pre-existing antibodies to these related viruses could attenuate
768 DOI: 10.1021/acsinfecdis.6b00161
ACS Infect. Dis. 2016, 2, 763−772
ACS Infectious Diseases Review

optimal adaptive immune responses and hamper the efficacy of *(M.J.R.) Mail: McGill University, 3775 University Street,
vaccines in Flavivirus endemic populations and may result in Room 406A, Montréal, QC, Canada H3A 2B4. E-mail: martin.j.
antibody-dependent enhancement (where cross-reactive anti- richer@mcgill.ca.
bodies from previous Flavivirus infection or vaccination can Notes
enhance subsequent infections).96,99 Two recent studies The authors declare no competing financial interest.


suggest that cross-reactive antibodies produced in response to
prior Dengue virus infection can indeed exacerbate ZIKV
ACKNOWLEDGMENTS
infection.96,100 As such, vaccination efforts must carefully
consider the risk of antibody-dependent enhancement and This work was supported by start-up funds from McGill
assess Flavivirus serostatus in test populations.98 University (S.M.S. and M.J.R.) as well as operating funds from


the Fonds de Recherche du Québec Nature et Technologies
(S.M.S.). S.M.S. is a Tier II Canada Research Chair in RNA
SUMMARY AND CONCLUSIONS
Biology and Viral Infections. M.J.R. received salary support
From its initial isolation in the mid-20th century in Uganda, from the Fonds de Recherche du Québec Santé − Chercheurs-
ZIKV remained in relative obscurity until recent outbreaks in Boursiers Junior 1. M.M.R. thanks the McGill University
Micronesia (2007), French Polynesia (2013), and Latin Faculty of Medicine Max E. Binz Fellowship for graduate
America (ongoing) brought it to the forefront of global public training. R.D.P. thanks the Natural Sciences and Engineering
health consciousness. Whereas ZIKV infection typically Research Council of Canada (NSERC) Canada Graduate
presents as a mild febrile illness, recent epidemics have been Scholarship − Masters (CGS-M) for graduate support.


associated with novel pathogenesis including Guillain−Barré
syndrome and fetal microcephaly. The rapid mobilization of the REFERENCES
scientific community has already led to several advances in (1) Dick, G. W., Kitchen, S. F., and Haddow, A. J. (1952) Zika virus.
understanding the pathogenesis of ZIKV and how this may be I. Isolations and serological specificity. Trans. R. Soc. Trop. Med. Hyg.
linked to these novel neurological complications. Furthermore, 46 (5), 509−520.
phylogenetic analyses suggest that viral evolution may have (2) Dick, G. W. (1952) Zika virus. II. Pathogenicity and physical
played a role in the rapid emergence and pathogenesis observed properties. Trans. R. Soc. Trop. Med. Hyg. 46 (5), 521−534.
in the recent outbreaks. Comparative genetic analyses and (3) Simpson, D. I. (1964) Zika Virus Infection in Man. Trans. R. Soc.
reverse genetic models, as well as in vitro and in vivo models, Trop. Med. Hyg. 58, 335−337.
are likely to provide more insight into how host−pathogen (4) Hayes, E. B. (2009) Zika virus outside Africa. Emerging Infect. Dis.
interactions and viral evolution have shaped ZIKV transmission 15 (9), 1347−1350.
(5) Duffy, M. R., Chen, T. H., Hancock, W. T., Powers, A. M., Kool,
and the pathogenesis associated with the current outbreaks. In J. L., Lanciotti, R. S., Pretrick, M., Marfel, M., Holzbauer, S., Dubray,
addition, this outbreak represents a unique opportunity to gain C., Guillaumot, L., Griggs, A., Bel, M., Lambert, A. J., Laven, J., Kosoy,
further insight into the factors that can influence the capacity of O., Panella, A., Biggerstaff, B. J., Fischer, M., and Hayes, E. B. (2009)
emerging viruses to establish epidemics. Zika virus outbreak on Yap Island, Federated States of Micronesia. N.
Despite the rapid mobilization of the scientific community, Engl. J. Med. 360 (24), 2536−2543.
there remain a number of outstanding questions that need to be (6) Chang, C., Ortiz, K., Ansari, A., and Gershwin, M. E. (2016) The
addressed: Did viral evolution lead to changes that resulted in Zika outbreak of the 21st century. J. Autoimmun. 68, 1−13.
increased infectivity and pathogenesis of ZIKV? Are there host (7) Nicastri, E., Castilletti, C., Liuzzi, G., Iannetta, M., Capobianchi,
M. R., and Ippolito, G. Persistent detection of Zika virus RNA in
genetic factors associated with the increase in neurological symptoms
semen for six months after symptom onset in a traveller returning from
such as Guillain−Barré syndrome? Has the prevalence of infection Haiti to Italy, February 2016. Euro Surveill. 2016, 21 (32), DOI:
with heterologous f laviviruses in endemic regions contributed to the 10.2807/1560-7917.ES.2016.21.32.30314.
sudden rise in pathogenesis? What are the correlates of immune (8) Centers for Disease Control and Prevention. (2016) Zika virus:
protection and are these somehow compromised in pregnant case counts in the US; http://www.cdc.gov/zika/geo/united-states.
women? The answers to these questions will be critical for the html.
development of novel therapeutic and vaccine strategies aimed (9) Chan, J. F., Yip, C. C., Tsang, J. O., Tee, K. M., Cai, J. P., Chik, K.
at preventing ZIKV infection. K., Zhu, Z., Chan, C. C., Choi, G. K., Sridhar, S., Zhang, A. J., Lu, G.,
Significant research efforts will be required to curb the Chiu, K., Lo, A. C., Tsao, S. W., Kok, K. H., Jin, D. Y., Chan, K. H., and
Yuen, K. Y. (2016) Differential cell line susceptibility to the emerging
growing global threat posed by ZIKV infection. The rapid Zika virus: implications for disease pathogenesis, non-vector-borne
response from the research community has already provided human transmission and animal reservoirs. Emerging Microbes Infect. 5,
several in vitro and in vivo models that will help provide a e93.
greater understanding of viral transmission, replication, patho- (10) Lazear, H. M., Govero, J., Smith, A. M., Platt, D. J., Fernandez,
genesis, and the immune response to infection. Future research E., Miner, J. J., and Diamond, M. S. (2016) A Mouse Model of Zika
efforts are thus likely to be focused on implementation of Virus Pathogenesis. Cell Host Microbe 19, 720.
appropriate vector control strategies and development of novel (11) Musso, D., Roche, C., Robin, E., Nhan, T., Teissier, A., and Cao-
diagnostic tools and antivirals, as well as design and testing of Lormeau, V. M. (2015) Potential sexual transmission of Zika virus.
ZIKV vaccine strategies. Emerging Infect. Dis. 21 (2), 359−361.


(12) Mansuy, J. M., Pasquier, C., Daudin, M., Chapuy-Regaud, S.,
Moinard, N., Chevreau, C., Izopet, J., Mengelle, C., and Bujan, L.
AUTHOR INFORMATION (2016) Zika virus in semen of a patient returning from a non-epidemic
area. Lancet Infect. Dis. 16 (8), 894−895.
Corresponding Authors (13) Besnard, M., Lastere, S., Teissier, A., Cao-Lormeau, V., and
*(S.M.S.) Mail: McGill University, 3775 University Street, Musso, D., Evidence of perinatal transmission of Zika virus, French
Room 608, Montréal, QC, Canada H3A 2B4. E-mail: selena. Polynesia, December 2013 and February 2014. Euro Surveill. 2014, 19
sagan@mcgill.ca. (13), 2075110.2807/1560-7917.ES2014.19.13.20751

769 DOI: 10.1021/acsinfecdis.6b00161


ACS Infect. Dis. 2016, 2, 763−772
ACS Infectious Diseases Review

(14) Dupont-Rouzeyrol, M., Biron, A., O’Connor, O., Huguon, E., (31) Calvet, G., Aguiar, R. S., Melo, A. S., Sampaio, S. A., de Filippis,
and Descloux, E. (2016) Infectious Zika viral particles in breastmilk. I., Fabri, A., Araujo, E. S., de Sequeira, P. C., de Mendonca, M. C., de
Lancet 387 (10023), 1051. Oliveira, L., Tschoeke, D. A., Schrago, C. G., Thompson, F. L., Brasil,
(15) Hamel, R., Dejarnac, O., Wichit, S., Ekchariyawat, P., Neyret, A., P., Dos Santos, F. B., Nogueira, R. M., Tanuri, A., and de Filippis, A.
Luplertlop, N., Perera-Lecoin, M., Surasombatpattana, P., Talignani, L., M. (2016) Detection and sequencing of Zika virus from amniotic fluid
Thomas, F., Cao-Lormeau, V. M., Choumet, V., Briant, L., Despres, P., of fetuses with microcephaly in Brazil: a case study. Lancet Infect. Dis.
Amara, A., Yssel, H., and Misse, D. (2015) Biology of Zika Virus 16, 653.
Infection in Human Skin Cells. J. Virol. 89 (17), 8880−8896. (32) Cordeiro, M. T., Pena, L. J., Brito, C. A., Gil, L. H., and
(16) Campos, G. S., Bandeira, A. C., and Sardi, S. I. (2015) Zika Marques, E. T. (2016) Positive IgM for Zika virus in the cerebrospinal
Virus Outbreak, Bahia, Brazil. Emerging Infect. Dis. 21 (10), 1885− fluid of 30 neonates with microcephaly in Brazil. Lancet 387, 1811.
1886. (33) Tang, H., Hammack, C., Ogden, S. C., Wen, Z., Qian, X., Li, Y.,
(17) Gourinat, A. C., O’Connor, O., Calvez, E., Goarant, C., and Yao, B., Shin, J., Zhang, F., Lee, E. M., Christian, K. M., Didier, R. A.,
Dupont-Rouzeyrol, M. (2015) Detection of Zika virus in urine. Jin, P., Song, H., and Ming, G. L. (2016) Zika Virus Infects Human
Emerging Infect. Dis. 21 (1), 84−86. Cortical Neural Progenitors and Attenuates Their Growth. Cell Stem
(18) Lanciotti, R. S., Kosoy, O. L., Laven, J. J., Velez, J. O., Lambert, Cell 18 (5), 587−590.
A. J., Johnson, A. J., Stanfield, S. M., and Duffy, M. R. (2008) Genetic (34) Cauchemez, S., Besnard, M., Bompard, P., Dub, T., Guillemette-
and serologic properties of Zika virus associated with an epidemic, Yap Artur, P., Eyrolle-Guignot, D., Salje, H., Van Kerkhove, M. D., Abadie,
State, Micronesia, 2007. Emerging Infect. Dis. 14 (8), 1232−1239. V., Garel, C., Fontanet, A., and Mallet, H. P. (2016) Association
(19) Lazear, H. M., and Diamond, M. S. (2016) Zika Virus: New between Zika virus and microcephaly in French Polynesia, 2013−15: a
Clinical Syndromes and Its Emergence in the Western Hemisphere. J. retrospective study. Lancet 387 (10033), 2125−2132.
Virol. 90 (10), 4864−4875. (35) Pacheco, O., Beltran, M., Nelson, C. A., Valencia, D., Tolosa, N.,
(20) Pabbaraju, K., Wong, S., Gill, K., Fonseca, K., Tipples, G. A., and Farr, S. L., Padilla, A. V., Tong, V. T., Cuevas, E. L., Espinosa-Bode, A.,
Tellier, R. (2016) Simultaneous detection of Zika, Chikungunya and Pardo, L., Rico, A., Reefhuis, J., Gonzalez, M., Mercado, M., Chaparro,
Dengue viruses by a multiplex real-time RT-PCR assay. J. Clin. Virol. P., Martinez Duran, M., Rao, C. Y., Munoz, M. M., Powers, A. M.,
83, 66−71. Cuellar, C., Helfand, R., Huguett, C., Jamieson, D. J., Honein, M. A.,
(21) Cardoso, C. W., Paploski, I. A., Kikuti, M., Rodrigues, M. S., and Ospina Martinez, M. L. (2016) Zika Virus Disease in Colombia −
Silva, M. M., Campos, G. S., Sardi, S. I., Kitron, U., Reis, M. G., and Preliminary Report. N. Engl. J. Med., DOI: 10.1056/NEJMoa1604037.
Ribeiro, G. S. (2015) Outbreak of Exanthematous Illness Associated (36) Aldo, P., You, Y., Szigeti, K., Horvath, T. L., Lindenbach, B., and
with Zika, Chikungunya, and Dengue Viruses, Salvador, Brazil. Mor, G. (2016) HSV-2 enhances ZIKV infection of the placenta and
Emerging Infect. Dis. 21 (12), 2274−2276. induces apoptosis in first-trimester trophoblast cells. Am. J. Reprod.
(22) Villamil-Gomez, W. E., Gonzalez-Camargo, O., Rodriguez-
Immunol., DOI: 10.1111/aji.12578.
Ayubi, J., Zapata-Serpa, D., and Rodriguez-Morales, A. J. (2016)
(37) Brasil, P., Pereira, J. P., Jr., Raja Gabaglia, C., Damasceno, L.,
Dengue, chikungunya and Zika co-infection in a patient from
Wakimoto, M., Ribeiro Nogueira, R. M., Carvalho de Sequeira, P.,
Colombia. J. Infect Public Health 9 (5), 684−686.
Machado Siqueira, A., Abreu de Carvalho, L. M., Cotrim da Cunha, D.,
(23) Butler, D. (2016) Brazil asks whether Zika acts alone to cause
Calvet, G. A., Neves, E. S., Moreira, M. E., Rodrigues Baiao, A. E.,
birth defects. Nature 535 (7613), 475−476.
Nassar de Carvalho, P. R., Janzen, C., Valderramos, S. G., Cherry, J. D.,
(24) Dejnirattisai, W., Supasa, P., Wongwiwat, W., Rouvinski, A.,
Barba-Spaeth, G., Duangchinda, T., Sakuntabhai, A., Cao-Lormeau, V. Bispo de Filippis, A. M., and Nielsen-Saines, K. (2016) Zika Virus
M., Malasit, P., Rey, F. A., Mongkolsapaya, J., and Screaton, G. R. Infection in Pregnant Women in Rio de Janeiro − Preliminary Report.
(2016) Dengue virus sero-cross-reactivity drives antibody-dependent New Engl. J. Med., DOI: 10.1056/NEJMoa1602412.
enhancement of infection with zika virus. Nat. Immunol. 17 (9), 1102− (38) Meaney-Delman, D., Oduyebo, T., Polen, K. N., White, J. L.,
1108. Bingham, A. M., Slavinski, S. A., Heberlein-Larson, L., St, George, K.,
(25) Kawiecki, A. B., and Christofferson, R. C. (2016) Zika-induced Rakeman, J. L., Hills, S., Olson, C. K., Adamski, A., Culver Barlow, L.,
antibody response enhances dengue serotype 2 replication in vitro. J. Lee, E. H., Likos, A. M., Munoz, J. L., Petersen, E. E., Dufort, E. M.,
Infect. Dis., jiw377. Dean, A. B., Cortese, M. M., Santiago, G. A., Bhatnagar, J., Powers, A.
(26) Yuki, N., and Hartung, H. P. (2012) Guillain-Barre syndrome. M., Zaki, S., Petersen, L. R., Jamieson, D. J., Honein, M. A., and U.S.
N. Engl. J. Med. 366 (24), 2294−2304. Zika Pregnancy Registry Prolonged Viremia Working Group. (2016)
(27) Cao-Lormeau, V. M., Blake, A., Mons, S., Lastere, S., Roche, C., Prolonged Detection of Zika Virus RNA in Pregnant Women. Obstet.
Vanhomwegen, J., Dub, T., Baudouin, L., Teissier, A., Larre, P., Vial, A. Gynecol. 128, 724.
L., Decam, C., Choumet, V., Halstead, S. K., Willison, H. J., Musset, L., (39) Franca, G. V., Schuler-Faccini, L., Oliveira, W. K., Henriques, C.
Manuguerra, J. C., Despres, P., Fournier, E., Mallet, H. P., Musso, D., M., Carmo, E. H., Pedi, V. D., Nunes, M. L., Castro, M. C., Serruya, S.,
Fontanet, A., Neil, J., and Ghawche, F. (2016) Guillain-Barre Silveira, M. F., Barros, F. C., and Victora, C. G. (2016) Congenital
Syndrome outbreak associated with Zika virus infection in French Zika virus syndrome in Brazil: a case series of the first 1501 livebirths
Polynesia: a case-control study. Lancet 387 (10027), 1531−1539. with complete investigation. Lancet 388, 891.
(28) Dos Santos, T., Rodriguez, A., Almiron, M., Sanhueza, A., (40) de Paula Freitas, B., de Oliveira Dias, J. R., Prazeres, J.,
Ramon, P., de Oliveira, W. K., Coelho, G. E., Badaro, R., Cortez, J., Sacramento, G. A., Ko, A. I., Maia, M., and Belfort, R., Jr. (2016)
Ospina, M., Pimentel, R., Masis, R., Hernandez, F., Lara, B., Montoya, Ocular Findings in Infants With Microcephaly Associated With
R., Jubithana, B., Melchor, A., Alvarez, A., Aldighieri, S., Dye, C., and Presumed Zika Virus Congenital Infection in Salvador, Brazil. JAMA
Espinal, M. A. (2016) Zika Virus and the Guillain-Barre Syndrome − Ophthalmol 134, 529.
Case Series from Seven Countries. N. Engl. J. Med., DOI: 10.1056/ (41) Kuno, G., and Chang, G. J. (2007) Full-length sequencing and
NEJMc1609015. genomic characterization of Bagaza, Kedougou, and Zika viruses. Arch.
(29) Mlakar, J., Korva, M., Tul, N., Popovic, M., Poljsak-Prijatelj, M., Virol. 152 (4), 687−696.
Mraz, J., Kolenc, M., Resman Rus, K., Vesnaver Vipotnik, T., Fabjan (42) Wang, L., Valderramos, S. G., Wu, A., Ouyang, S., Li, C., Brasil,
Vodusek, V., Vizjak, A., Pizem, J., Petrovec, M., and Avsic Zupanc, T. P., Bonaldo, M., Coates, T., Nielsen-Saines, K., Jiang, T., Aliyari, R.,
(2016) Zika Virus Associated with Microcephaly. N. Engl. J. Med. 374 and Cheng, G. (2016) From Mosquitos to Humans: Genetic
(10), 951−958. Evolution of Zika Virus. Cell Host Microbe 19, 561.
(30) Fauci, A. S., and Morens, D. M. (2016) Zika Virus in the (43) Ye, Q., Liu, Z. Y., Han, J. F., Jiang, T., Li, X. F., and Qin, C. F.
Americas − Yet Another Arbovirus Threat. N. Engl. J. Med. 374 (7), (2016) Genomic characterization and phylogenetic analysis of Zika
601−604. virus circulating in the Americas. Infect., Genet. Evol. 43, 43−49.

770 DOI: 10.1021/acsinfecdis.6b00161


ACS Infect. Dis. 2016, 2, 763−772
ACS Infectious Diseases Review

(44) Driggers, R. W., Ho, C. Y., Korhonen, E. M., Kuivanen, S., (54) Grant, A., Ponia, S. S., Tripathi, S., Balasubramaniam, V., Miorin,
Jaaskelainen, A. J., Smura, T., Rosenberg, A., Hill, D. A., DeBiasi, R. L., L., Sourisseau, M., Schwarz, M. C., Sanchez-Seco, M. P., Evans, M. J.,
Vezina, G., Timofeev, J., Rodriguez, F. J., Levanov, L., Razak, J., Best, S. M., and Garcia-Sastre, A. (2016) Zika Virus Targets Human
Iyengar, P., Hennenfent, A., Kennedy, R., Lanciotti, R., du Plessis, A., STAT2 to Inhibit Type I Interferon Signaling. Cell Host Microbe 19,
and Vapalahti, O. (2016) Zika Virus Infection with Prolonged 882.
Maternal Viremia and Fetal Brain Abnormalities. N. Engl. J. Med. (55) Aliota, M. T., Caine, E. A., Walker, E. C., Larkin, K. E.,
374 (22), 2142−2151. Camacho, E., and Osorio, J. E. (2016) Characterization of Lethal Zika
(45) Tabata, T., Petitt, M., Puerta-Guardo, H., Michlmayr, D., Wang, Virus Infection in AG129 Mice. PLoS Neglected Trop. Dis. 10 (4),
C., Fang-Hoover, J., Harris, E., and Pereira, L. (2016) Zika Virus e0004682.
Targets Different Primary Human Placental Cells, Suggesting Two (56) Dowall, S. D., Graham, V. A., Rayner, E., Atkinson, B., Hall, G.,
Routes for Vertical Transmission. Cell Host Microbe 20 (2), 155−166. Watson, R. J., Bosworth, A., Bonney, L. C., Kitchen, S., and Hewson,
(46) Quicke, K. M., Bowen, J. R., Johnson, E. L., McDonald, C. E., R. (2016) A Susceptible Mouse Model for Zika Virus Infection. PLoS
Ma, H., O’Neal, J. T., Rajakumar, A., Wrammert, J., Rimawi, B. H., Neglected Trop. Dis. 10 (5), e0004658.
Pulendran, B., Schinazi, R. F., Chakraborty, R., and Suthar, M. S. (57) Rossi, S. L., Tesh, R. B., Azar, S. R., Muruato, A. E., Hanley, K.
(2016) Zika Virus Infects Human Placental Macrophages. Cell Host A., Auguste, A. J., Langsjoen, R. M., Paessler, S., Vasilakis, N., and
Microbe 20 (1), 83−90. Weaver, S. C. (2016) Characterization of a Novel Murine Model to
(47) Bayer, A., Lennemann, N. J., Ouyang, Y., Bramley, J. C., Study Zika Virus. Am. J. Trop. Med. Hyg. 94 (6), 1362−1369.
Morosky, S., Marques, E. T., Jr., Cherry, S., Sadovsky, Y., and Coyne, (58) Miner, J. J., Cao, B., Govero, J., Smith, A. M., Fernandez, E.,
C. B. (2016) Type III Interferons Produced by Human Placental Cabrera, O. H., Garber, C., Noll, M., Klein, R. S., Noguchi, K. K.,
Trophoblasts Confer Protection against Zika Virus Infection. Cell Host Mysorekar, I. U., and Diamond, M. S. (2016) Zika Virus Infection
Microbe 19 (5), 705−712. during Pregnancy in Mice Causes Placental Damage and Fetal Demise.
(48) Garcez, P. P., Loiola, E. C., Madeiro da Costa, R., Higa, L. M., Cell 165 (5), 1081−1091.
Trindade, P., Delvecchio, R., Nascimento, J. M., Brindeiro, R., Tanuri, (59) Yockey, L. J., Varela, L., Rakib, T., Khoury-Hanold, W., Fink, S.
A., and Rehen, S. K. (2016) Zika virus impairs growth in human L., Stutz, B., Szigeti-Buck, K., Van den Pol, A., Lindenbach, B. D.,
neurospheres and brain organoids. Science 352 (6287), 816−818. Horvath, T. L., and Iwasaki, A. (2016) Vaginal Exposure to Zika Virus
(49) Qian, X., Nguyen, H. N., Song, M. M., Hadiono, C., Ogden, S. during Pregnancy Leads to Fetal Brain Infection. Cell 166 (5), 1247−
C., Hammack, C., Yao, B., Hamersky, G. R., Jacob, F., Zhong, C., 1256-e4.
Yoon, K. J., Jeang, W., Lin, L., Li, Y., Thakor, J., Berg, D. A., Zhang, C., (60) Furtado, J. M., Esposito, D. L., Klein, T. M., Teixeira-Pinto, T.,
Kang, E., Chickering, M., Nauen, D., Ho, C. Y., Wen, Z., Christian, K. and da Fonseca, B. A. (2016) Uveitis Associated with Zika Virus
M., Shi, P. Y., Maher, B. J., Wu, H., Jin, P., Tang, H., Song, H., and Infection. N. Engl. J. Med. 375 (4), 394−396.
Ming, G. L. (2016) Brain-Region-Specific Organoids Using Mini- (61) Miner, J. J., Sene, A., Richner, J. M., Smith, A. M., Santeford, A.,
bioreactors for Modeling ZIKV Exposure. Cell 165 (5), 1238−1254. Ban, N., Weger-Lucarelli, J., Manzella, F., Ruckert, C., Govero, J.,
(50) Cugola, F. R., Fernandes, I. R., Russo, F. B., Freitas, B. C., Dias, Noguchi, K. K., Ebel, G. D., Diamond, M. S., and Apte, R. S. (2016)
J. L., Guimaraes, K. P., Benazzato, C., Almeida, N., Pignatari, G. C., Zika Virus Infection in Mice Causes Panuveitis with Shedding of Virus
Romero, S., Polonio, C. M., Cunha, I., Freitas, C. L., Brandao, W. N., in Tears. Cell Rep. 16, 3208.
Rossato, C., Andrade, D. G., Faria Dde, P., Garcez, A. T., Buchpigel, C. (62) Coyne, C. B., and Lazear, H. M. (2016) Zika virus − reigniting
A., Braconi, C. T., Mendes, E., Sall, A. A., Zanotto, P. M., Peron, J. P., the TORCH. Nat. Rev. Microbiol., DOI: 10.1038/nrmicro.2016.125.
Muotri, A. R., and Beltrao-Braga, P. C. (2016) The Brazilian Zika virus (63) Kolumam, G. A., Thomas, S., Thompson, L. J., Sprent, J., and
strain causes birth defects in experimental models. Nature 534 (7606), Murali-Krishna, K. (2005) Type I interferons act directly on CD8 T
267−271. cells to allow clonal expansion and memory formation in response to
(51) Abbink, P., Larocca, R. A., De La Barrera, R. A., Bricault, C. A., viral infection. J. Exp. Med. 202 (5), 637−650.
Moseley, E. T., Boyd, M., Kirilova, M., Li, Z., Ng’ang’a, D., (64) Shan, C., Xie, X., Muruato, A. E., Rossi, S. L., Roundy, C. M.,
Nanayakkara, O., Nityanandam, R., Mercado, N. B., Borducchi, E. Azar, S. R., Yang, Y., Tesh, R. B., Bourne, N., Barrett, A. D., Vasilakis,
N., Agarwal, A., Brinkman, A. L., Cabral, C., Chandrashekar, A., Giglio, N., Weaver, S. C., and Shi, P. Y. (2016) An Infectious cDNA Clone of
P. B., Jetton, D., Jimenez, J., Lee, B. C., Mojta, S., Molloy, K., Shetty, Zika Virus to Study Viral Virulence, Mosquito Transmission, and
M., Neubauer, G. H., Stephenson, K. E., Peron, J. P., Zanotto, P. M., Antiviral Inhibitors. Cell Host Microbe 19 (6), 891−900.
Misamore, J., Finneyfrock, B., Lewis, M. G., Alter, G., Modjarrad, K., (65) Tsetsarkin, K. A., Kenney, H., Chen, R., Liu, G., Manukyan, H.,
Jarman, R. G., Eckels, K. H., Michael, N. L., Thomas, S. J., and Whitehead, S. S., Laassri, M., Chumakov, K., and Pletnev, A. G., A Full-
Barouch, D. H. (2016) Protective efficacy of multiple vaccine Length Infectious cDNA Clone of Zika Virus from the 2015 Epidemic
platforms against Zika virus challenge in rhesus monkeys. Science in Brazil as a Genetic Platform for Studies of Virus-Host Interactions
353, 1129. and Vaccine Development. mBio 2016, 7 (4), e01114-1610.1128/
(52) Dudley, D. M., Aliota, M. T., Mohr, E. L., Weiler, A. M., Lehrer- mBio.01114-16.
Brey, G., Weisgrau, K. L., Mohns, M. S., Breitbach, M. E., Rasheed, M. (66) Gadea, G., Bos, S., Krejbich-Trotot, P., Clain, E., Viranaicken,
N., Newman, C. M., Gellerup, D. D., Moncla, L. H., Post, J., Schultz- W., El-Kalamouni, C., Mavingui, P., and Despres, P. (2016) A robust
Darken, N., Schotzko, M. L., Hayes, J. M., Eudailey, J. A., Moody, M. method for the rapid generation of recombinant Zika virus expressing
A., Permar, S. R., O’Connor, S. L., Rakasz, E. G., Simmons, H. A., the GFP reporter gene. Virology 497, 157−162.
Capuano, S., Golos, T. G., Osorio, J. E., Friedrich, T. C., and (67) Schwarz, M. C., Sourisseau, M., Espinosa, M. M., Gray, E. S.,
O’Connor, D. H. (2016) A rhesus macaque model of Asian-lineage and Chambers, M. T. (2016) Rescue of the 1947 Zika Virus Prototype
Zika virus infection. Nat. Commun. 7, 12204. Strain with a Cytomegalovirus Promoter-Driven cDNA Clone.
(53) Adams Waldorf, K. M., Stencel-Baerenwald, J. E., Kapur, R. P., mSphere 1 (5), e00246-16.
Studholme, C., Boldenow, E., Vornhagen, J., Baldessari, A., Dighe, M. (68) Tolle, M. A. (2009) Mosquito-borne diseases. Curr. Probl
K., Thiel, J., Merillat, S., Armistead, B., Tisoncik-Go, J., Green, R. R., Pediatr Adolesc Health Care 39 (4), 97−140.
Davis, M. A., Dewey, E. C., Fairgrieve, M. R., Gatenby, J. C., Richards, (69) Marchette, N. J., Garcia, R., and Rudnick, A. (1969) Isolation of
T., Garden, G. A., Diamond, M. S., Juul, S. E., Grant, R. F., Kuller, L., Zika virus from Aedes aegypti mosquitoes in Malaysia. Am. J. Trop. Med.
Shaw, D. W., Ogle, J., Gough, G. M., Lee, W., English, C., Hevner, R. Hyg. 18 (3), 411−415.
F., Dobyns, W. B., Gale, M., Jr., and Rajagopal, L. (2016) Fetal brain (70) Vorou, R. (2016) Zika virus, vectors, reservoirs, amplifying
lesions after subcutaneous inoculation of Zika virus in a pregnant hosts, and their potential to spread worldwide: what we know and
nonhuman primate. Nat. Med., DOI: 10.1038/nm.4193. what we should investigate urgently. Int. J. Infect. Dis. 48, 85−90.

771 DOI: 10.1021/acsinfecdis.6b00161


ACS Infect. Dis. 2016, 2, 763−772
ACS Infectious Diseases Review

(71) Grard, G., Caron, M., Mombo, I. M., Nkoghe, D., Mboui Ondo, (90) Schul, W., Liu, W., Xu, H. Y., Flamand, M., and Vasudevan, S. G.
S., Jiolle, D., Fontenille, D., Paupy, C., and Leroy, E. M. (2014) Zika (2007) A dengue fever viremia model in mice shows reduction in viral
virus in Gabon (Central Africa) − 2007: a new threat from Aedes replication and suppression of the inflammatory response after
albopictus? PLoS Neglected Trop. Dis. 8 (2), e2681. treatment with antiviral drugs. J. Infect. Dis. 195 (5), 665−674.
(72) Ayres, C. F. (2016) Identification of Zika virus vectors and (91) Julander, J. G., Shafer, K., Smee, D. F., Morrey, J. D., and Furuta,
implications for control. Lancet Infect. Dis. 16 (3), 278−279. Y. (2009) Activity of T-705 in a hamster model of yellow fever virus
(73) Bonizzoni, M., Gasperi, G., Chen, X., and James, A. A. (2013) infection in comparison with that of a chemically related compound,
The invasive mosquito species Aedes albopictus: current knowledge and T-1106. Antimicrob. Agents Chemother. 53 (1), 202−209.
future perspectives. Trends Parasitol. 29 (9), 460−468. (92) Eyer, L., Nencka, R., Huvarova, I., Palus, M., Joao Alves, M.,
(74) Saiz, J. C., Vazquez-Calvo, A., Blazquez, A. B., Merino-Ramos, Gould, E. A., De Clercq, E., and Ruzek, D. (2016) Nucleoside
T., Escribano-Romero, E., and Martin-Acebes, M. A. (2016) Zika Inhibitors of Zika Virus. J. Infect. Dis. 214 (5), 707−711.
Virus: the Latest Newcomer. Front. Microbiol. 7, 496. (93) Barrows, N. J., Campos, R. K., Powell, S. T., Prasanth, K. R.,
(75) Waldock, J., Chandra, N. L., Lelieveld, J., Proestos, Y., Michael, Schott-Lerner, G., Soto-Acosta, R., Galarza-Munoz, G., McGrath, E. L.,
E., Christophides, G., and Parham, P. E. (2013) The role of Urrabaz-Garza, R., Gao, J., Wu, P., Menon, R., Saade, G., Fernandez-
environmental variables on Aedes albopictus biology and chikungunya Salas, I., Rossi, S. L., Vasilakis, N., Routh, A., Bradrick, S. S., and
epidemiology. Pathog. Global Health 107 (5), 224−241. Garcia-Blanco, M. A. (2016) A Screen of FDA-Approved Drugs for
(76) Thangamani, S., Huang, J., Hart, C. E., Guzman, H., and Tesh, Inhibitors of Zika Virus Infection. Cell Host Microbe 20 (2), 259−270.
R. B. (2016) Vertical Transmission of Zika Virus in Aedes aegypti (94) Xu, M., Lee, E. M., Wen, Z., Cheng, Y., Huang, W. K., Qian, X.,
Mosquitoes. Am. J. Trop. Med. Hyg., DOI: 10.4269/ajtmh.16-0448. Tcw, J., Kouznetsova, J., Ogden, S. C., Hammack, C., Jacob, F.,
(77) Aliota, M. T., Peinado, S. A., Velez, I. D., and Osorio, J. E. Nguyen, H. N., Itkin, M., Hanna, C., Shinn, P., Allen, C., Michael, S.
(2016) The wMel strain of Wolbachia Reduces Transmission of Zika G., Simeonov, A., Huang, W., Christian, K. M., Goate, A., Brennand, K.
virus by Aedes aegypti. Sci. Rep. 6, 28792. J., Huang, R., Xia, M., Ming, G. L., Zheng, W., Song, H., and Tang, H.
(78) Dutra, H. L., Rocha, M. N., Dias, F. B., Mansur, S. B., Caragata, (2016) Identification of small-molecule inhibitors of Zika virus
E. P., and Moreira, L. A. (2016) Wolbachia Blocks Currently infection and induced neural cell death via a drug repurposing screen.
Circulating Zika Virus Isolates in Brazilian Aedes aegypti Mosquitoes. Nat. Med. 22, 1101.
Cell Host Microbe 19 (6), 771−774. (95) Dejnirattisai, W., Wongwiwat, W., Supasa, S., Zhang, X., Dai, X.,
(79) Bargielowski, I., Nimmo, D., Alphey, L., and Koella, J. C. (2011) Rouvinski, A., Jumnainsong, A., Edwards, C., Quyen, N. T.,
Comparison of life history characteristics of the genetically modified Duangchinda, T., Grimes, J. M., Tsai, W. Y., Lai, C. Y., Wang, W.
OX513A line and a wild type strain of Aedes aegypti. PLoS One 6 (6), K., Malasit, P., Farrar, J., Simmons, C. P., Zhou, Z. H., Rey, F. A.,
e20699. Mongkolsapaya, J., and Screaton, G. R. (2015) A new class of highly
(80) Curtis, Z., Matzen, K., Neira Oviedo, M., Nimmo, D., Gray, P., potent, broadly neutralizing antibodies isolated from viremic patients
Winskill, P., Locatelli, M. A., Jardim, W. F., Warner, S., Alphey, L., and infected with dengue virus. Nat. Immunol. 16 (2), 170−177.
Beech, C. (2015) Assessment of the Impact of Potential Tetracycline (96) Stettler, K., Beltramello, M., Espinosa, D. A., Graham, V.,
Exposure on the Phenotype of Aedes aegypti OX513A: Implications for Cassotta, A., Bianchi, S., Vanzetta, F., Minola, A., Jaconi, S., Mele, F.,
Field Use. PLoS Neglected Trop. Dis. 9 (8), e0003999. Foglierini, M., Pedotti, M., Simonelli, L., Dowall, S., Atkinson, B.,
(81) Subbaraman, N. (2011) Science snipes at Oxitec transgenic- Percivalle, E., Simmons, C. P., Varani, L., Blum, J., Baldanti, F.,
mosquito trial. Nat. Biotechnol. 29 (1), 9−11. Cameroni, E., Hewson, R., Harris, E., Lanzavecchia, A., Sallusto, F.,
(82) Waltz, E. (2016) GM mosquitoes fire first salvo against Zika and Corti, D. (2016) Specificity, cross-reactivity, and function of
virus. Nat. Biotechnol. 34 (3), 221−222. antibodies elicited by Zika virus infection. Science 353 (6301), 823−
(83) Hunter, P. (2016) Challenges and options for disease vector 826.
control: The outbreak of Zika virus in South America and increasing (97) Guy, B., Briand, O., Lang, J., Saville, M., and Jackson, N. (2015)
insecticide resistance among mosquitoes have rekindled efforts for Development of the Sanofi Pasteur tetravalent dengue vaccine: One
controlling disease vectors. EMBO Rep. 17, 1370. more step forward. Vaccine 33 (50), 7100−7111.
(84) Carvalho, D. O., McKemey, A. R., Garziera, L., Lacroix, R., (98) Durbin, A. P. (2016) Vaccine Development for Zika Virus-
Donnelly, C. A., Alphey, L., Malavasi, A., and Capurro, M. L. (2015) Timelines and Strategies. Semin. Reprod. Med., DOI: 10.1055/s-0036-
Suppression of a Field Population of Aedes aegypti in Brazil by 1592070.
Sustained Release of Transgenic Male Mosquitoes. PLoS Neglected (99) Martins, K. A., Dye, J. M., and Bavari, S. (2016) Considerations
Trop. Dis. 9 (7), e0003864. for the development of Zika virus vaccines. Vaccine 34 (33), 3711−
(85) Rabe, I. B., Staples, J. E., Villanueva, J., Hummel, K. B., Johnson, 3712.
J. A., Rose, L., Mts, Hills, S., Wasley, A., Fischer, M., and Powers, A. M. (100) Priyamvada, L., Quicke, K. M., Hudson, W. H., Onlamoon, N.,
(2016) Interim Guidance for Interpretation of Zika Virus Antibody Sewatanon, J., Edupuganti, S., Pattanapanyasat, K., Chokephaibulkit,
Test Results. MMWR Morb. Mortal. Wkly. Rep. 65 (21), 543−546. K., Mulligan, M. J., Wilson, P. C., Ahmed, R., Suthar, M. S., and
(86) Waggoner, J. J., and Pinsky, B. A. (2016) Zika Virus: Diagnostics Wrammert, J. (2016) Human antibody responses after dengue virus
infection are highly cross-reactive to Zika virus. Proc. Natl. Acad. Sci. U.
for an Emerging Pandemic Threat. J. Clin. Microbiol. 54 (4), 860−867.
S. A. 113 (28), 7852−7857.
(87) Bingham, A. M., Cone, M., Mock, V., Heberlein-Larson, L.,
Stanek, D., Blackmore, C., and Likos, A. (2016) Comparison of Test
Results for Zika Virus RNA in Urine, Serum, and Saliva Specimens
from Persons with Travel-Associated Zika Virus Disease - Florida,
2016. MMWR Morb. Mortal. Wkly. Rep. 65 (18), 475−478.
(88) Barzon, L., Pacenti, M., Berto, A., Sinigaglia, A., Franchin, E.,
Lavezzo, E., Brugnaro, P., and Palu, G. Isolation of infectious Zika virus
from saliva and prolonged viral RNA shedding in a traveller returning
from the Dominican Republic to Italy, January 2016. Euro Surveill.
2016, 21 (10), DOI: 10.2807/1560-7917.ES.2016.21.10.30159.
(89) Lustig, Y., Mendelson, E., Paran, N., Melamed, S., and Schwartz,
E. Detection of Zika virus RNA in whole blood of imported Zika virus
disease cases up to 2 months after symptom onset, Israel, December
2015 to April 2016. Euro Surveill. 2016, 21 (26), DOI: 10.2807/1560-
7917.ES.2016.21.26.30269.

772 DOI: 10.1021/acsinfecdis.6b00161


ACS Infect. Dis. 2016, 2, 763−772

Das könnte Ihnen auch gefallen