Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Journal of Neuroimmunology 308 (2017) 50–64

Contents lists available at ScienceDirect

Journal of Neuroimmunology

journal homepage: www.elsevier.com/locate/jneuroim

Review article

Zika virus: History, epidemiology, transmission, and clinical presentation


Byung-Hak Song, Sang-Im Yun, Michael Woolley, Young-Min Lee ⁎
Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA

a r t i c l e i n f o a b s t r a c t

Article history: Zika virus (ZIKV), a mosquito-borne positive-stranded RNA virus of the family Flaviviridae (genus Flavivirus), is
Received 22 December 2016 now causing an unprecedented large-scale outbreak in the Americas. Historically, ZIKV spread eastward from
Received in revised form 1 March 2017 equatorial Africa and Asia to the Pacific Islands during the late 2000s to early 2010s, invaded the Caribbean
Accepted 1 March 2017
and Central and South America in 2015, and reached North America in 2016. Although ZIKV infection generally
causes no symptoms or only a mild self-limiting illness, it has recently been linked to a rising number of severe
Keywords:
Zika virus
neurological diseases, including microcephaly and Guillain-Barré syndrome. Because of the continuous geo-
Flavivirus graphic expansion of both the virus and its mosquito vectors, ZIKV poses a serious threat to public health around
Arbovirus the globe. However, there are no vaccines or antiviral therapies available against this pathogen. This review sum-
Mosquito-borne pathogen marizes a fast-growing body of literature on the history, epidemiology, transmission, and clinical presentation of
Zika outbreak ZIKV and highlights the urgent need for the development of efficient control strategies for this emerging
Zika pandemic pathogen.
Microcephaly © 2017 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
Guillain-Barré syndrome
(http://creativecommons.org/licenses/by-nc-nd/4.0/).

Contents

1. Classification and nomenclature . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51


2. History and epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
2.1. Virus discovery and seroprevalence in Africa and Asia prior to the year 2000 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
2.2. The Yap outbreak and sporadic cases in Southeast Asia during the late 2000s to mid-2010s . . . . . . . . . . . . . . . . . . . . . . . . 52
2.3. The French Polynesia outbreak and spread in the Pacific Islands in the early 2010s . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
2.4. The Brazil outbreak and spread in the Americas in 2015–2016 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
2.5. Current status and future prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
3. Genetic diversity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
4. Transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
4.1. Vector-borne transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
4.2. Non-vector-borne transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
5. Virology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
6. Clinical presentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
6.1. Common signs and symptoms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
6.2. Guillain-Barré syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
6.3. Microcephaly . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
6.4. Other neurological complications. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
7. Tissue and cell tropism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
8. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59

⁎ Corresponding author at: Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, 9830 Old Main Hill, Logan, UT
84322, USA.
E-mail address: youngmin.lee@usu.edu (Y.-M. Lee).

http://dx.doi.org/10.1016/j.jneuroim.2017.03.001
0165-5728/© 2017 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64 51

1. Classification and nomenclature zone, however, it first emerged in Yap Island in 2007, spread eastward
to French Polynesia and other Pacific Islands in 2013–2014, reached
Zika virus (ZIKV) belongs to the genus Flavivirus in the family Latin America in 2015, and disseminated further to North America in
Flaviviridae (Lindenbach et al., 2007). Currently, the genus comprises 2016. Now, ZIKV is circulating in the Americas, Southeast Asia, and the
53 virus species (Simmonds et al., 2012), which are transmitted by the Pacific Islands.
bite of mosquitoes (27 species), ticks (12 species), or no known arthro-
pod vector (14 species). Within the Flavivirus genus, ZIKV is a mosquito- 2.1. Virus discovery and seroprevalence in Africa and Asia prior to the year
borne virus that is phylogenetically closely related to other medically 2000
important mosquito-borne flaviviruses of global public health signifi-
cance (Fig. 1), such as Japanese encephalitis (JEV), West Nile (WNV), ZIKV was first isolated in 1947 from the serum of a sentinel rhesus
dengue (DENV), and yellow fever (YFV) viruses (Gubler et al., 2007). macaque monkey that was placed in the Zika forest on the Entebbe
These mosquito-borne flaviviruses can be divided into two major clas- peninsula, Uganda, during the course of surveillance for YFV (Dick
ses based on their clinical presentation in humans (Gaunt et al., 2001; et al., 1952). This isolate, named MR-766, is the African prototype
Kramer and Ebel, 2003): (1) encephalitic flaviviruses (represented by strain of ZIKV. Shortly thereafter, it was also recovered on multiple oc-
JEV and WNV), which cause invasive neurological diseases, with birds casions from A. africanus mosquitoes caught in the same area (Dick,
serving as their natural vertebrate hosts and Culex species mosquitoes 1952; Dick et al., 1952; Haddow et al., 1964; Weinbren and
as their principal vectors (Brinton, 2013; Yun and Lee, 2014); and Williams, 1958). Although there was no indication that ZIKV caused
(2) non-encephalitic or viscerotropic flaviviruses (exemplified by disease in the residents of Uganda, the prevalence of antibodies
DENV and YFV), which cause lethal hemorrhagic fever, with non- against the virus in their serum was approximately 10–20% (Dick,
human primates acting as their vertebrate hosts and Aedes species mos- 1953; Dick et al., 1952). Despite the need for caution because of anti-
quitoes as their primary vectors (Monath and Vasconcelos, 2015; body cross-reactivity with other flaviviruses, a large number of sero-
Weaver and Barrett, 2004). Of note, DENV has fully adapted to humans logical studies in the half century since the discovery of ZIKV have
and no longer needs animal hosts for viral transmission (Clyde et al., revealed a broad but confined geographic distribution of human infec-
2006; Mackenzie et al., 2004). tion with the virus, across a relatively narrow equatorial belt running
from Africa to Asia: Senegal (Monlun et al., 1993), Sierra Leone (Robin
2. History and epidemiology and Mouchet, 1975), Nigeria (Adekolu-John and Fagbami, 1983;
Fagbami, 1977; Macnamara, 1954), Gabon (Jan et al., 1978; Saluzzo
Discovered in Uganda in 1947, ZIKV was confined for the first et al., 1982), Central African Republic (Saluzzo et al., 1981), Egypt
60 years to an equatorial zone across Africa and Asia. Outside this (Smithburn et al., 1954b), Uganda (Smithburn, 1952), Tanzania

Fig. 1. A rooted phylogenetic tree based on the nucleotide sequence of complete or near-complete genomes of all 46 available flaviviruses (as of November 2016). Multiple sequence
alignments were produced by ClustalX (Thompson et al., 1997), and the rooted phylogenetic tree was generated by the neighbor-joining method (Saitou and Nei, 1987). The scale bar
represents the genetic distance in nucleotide substitutions per site. Bootstrap values (1000 replications) are shown at each node. The complete genome sequence of the bovine viral
diarrhea virus, a member of the genus Pestivirus in the family Flaviviridae, was used as an outgroup. The virus abbreviations assigned by the International Committee on Taxonomy of
Viruses are listed, and the strain names and their GenBank accession numbers are provided in parenthesis. The known or probable vector (mosquito or tick) is indicated on the right
side of the tree.
52 B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64

(Smithburn, 1952), Kenya (Geser et al., 1970), Pakistan (Darwish (Heang et al., 2012), Malaysia (Tappe et al., 2015), Indonesia (Kwong
et al., 1983), India (Smithburn et al., 1954a), Thailand (Pond, 1963), et al., 2013; Perkasa et al., 2016), and the Philippines (Alera et al., 2015).
Malaysia (Pond, 1963; Smithburn, 1954), Vietnam (Pond, 1963),
Indonesia (Olson et al., 1983), and the Philippines (Hammon et al.,
1958). In 1966, the first non-African ZIKV strain, designated P6-740, 2.3. The French Polynesia outbreak and spread in the Pacific Islands in the
was isolated from a pool of A. aegypti mosquitoes collected in early 2010s
Malaysia (Marchette et al., 1969).
In 2013–2014, a major epidemic of ZIKV occurred in French Polyne-
sia (Fig. 2), a French overseas territory located in the middle of the
2.2. The Yap outbreak and sporadic cases in Southeast Asia during the late southern Pacific Ocean, with its ~ 270,000 people living on 67 islands
2000s to mid-2010s distributed among five archipelagoes (Cao-Lormeau et al., 2014). Dur-
ing this outbreak, ~11% of the total population was estimated to have
Human illness caused by ZIKV infection was first reported in 1954 sought medical treatment for suspected ZIKV infection (ECDC, 2014;
during an outbreak of jaundice in Nigeria, when infection was con- Mallet et al., 2015; Musso et al., 2014b). The magnitude of the outbreak
firmed in three patients by isolation of the virus or a rise in serum anti- was presumably the result of a combination of the low level of pre-
body titer, with a correlation observed between the development of existing immunity to ZIKV and the high density of competent mosquito
ZIKV neutralizing antibodies and jaundice (Macnamara, 1954). From vectors in that area (Aubry et al., 2015). Although a vast majority of the
that time until the early 2000s, only about a dozen cases of benign clinical cases seen in this outbreak were similar to those observed in the
human ZIKV-associated illness were documented in countries in Africa 2007 Yap outbreak, a small fraction of severe cases were associated with
and Asia (Hayes, 2009), such as Uganda (Simpson, 1964), Nigeria neurological complications, such as Guillain-Barré syndrome, in the
(Fagbami, 1979; Moore et al., 1975), and Indonesia (Olson et al., context of co-circulating DENV and chikungunya (ECDC, 2014; Oehler
1981). In 2007, however, ZIKV caused the first large outbreak outside et al., 2014). Retrospectively, the incidence of Guillain-Barré syndrome
of Africa and Asia on Yap Island, a part of the Federated States of was estimated to be increased by ~20-fold in French Polynesia (Oehler
Micronesia (Fig. 2) in the northwestern Pacific Ocean, with a relatively et al., 2014). Although the origin of the ZIKV in French Polynesia remains
mild disease characterized by fever, rash, arthralgia, and conjunctivitis unknown, it is genetically related to the strains isolated from Yap Island
(Duffy et al., 2009; Lanciotti et al., 2008). During this outbreak, ~ 73% in 2007 and from Cambodia in 2010 (Cao-Lormeau et al., 2014). During
of the 6892 Yap residents aged ≥3 years were estimated to be infected or shortly after the French Polynesia outbreak, ZIKV spread further to
with ZIKV, and ~ 18% of the infected people had a clinical illness that other neighboring islands in the South Pacific Ocean (Cao-Lormeau
was probably attributable to ZIKV infection (Duffy et al., 2009). Se- and Musso, 2014; Musso et al., 2015a; Musso et al., 2014b), including
quence analysis suggested that ZIKV was introduced to Yap Island New Caledonia (Dupont-Rouzeyrol et al., 2015), the Cook Islands
from Southeast Asia (Duffy et al., 2009; Haddow et al., 2012; Lanciotti (Roth et al., 2014), and Easter Island (Tognarelli et al., 2016). Also, it
et al., 2008). In the early to mid-2010s, a handful of sporadic cases of was imported to other faraway countries such as Australia (Pyke et al.,
ZIKV infection were also reported in Southeast Asian countries, such 2014), Italy (Zammarchi et al., 2015a), Japan (Kutsuna et al., 2014),
as Thailand (Buathong et al., 2015; Tappe et al., 2014), Cambodia and Norway (Waehre et al., 2014).

Fig. 2. Countries and territories that have reported autochthonous mosquito-borne transmission of ZIKV in the past 3 months. The term “widespread transmission” (red) indicates one of
the following three situations: (1) more than 10 locally transmitted ZIKV cases reported in a single area, (2) at least two separate areas reporting locally transmitted ZIKV cases, and
(3) ZIKV transmission ongoing in an area for more than 3 months. The term “sporadic transmission” (orange) indicates that no more than 10 locally transmitted ZIKV cases have been
reported in a single area in the past 3 months. The term “past transmission” (blue) indicates that local ZIKV transmission has been reported since 2007, but not in the past 3 months.
Source: European Centre for Disease Prevention and Control, current status of ZIKV transmission in the world as of November 23, 2016 (http://ecdc.europa.eu/en/healthtopics/zika_virus_
infection/zika-outbreak/pages/zika-countries-with-transmission.aspx).
B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64 53

2.4. The Brazil outbreak and spread in the Americas in 2015–2016 Islands, New Caledonia, Palau, Papua New Guinea, Samoa, and Tonga),
Southeast Asia (Singapore, Thailand, the Philippines, and Vietnam),
At the beginning of 2015, the first autochthonous transmission of and the islands of Cape Verde off the coast of West Africa (CDC,
ZIKV was detected in the northeastern part of Brazil (Campos et al., 2016a; ECDC, 2016a; WHO, 2016b). In addition, since early 2015,
2015; Zammarchi et al., 2015b; Zanluca et al., 2015), in association there have been an increasing number of travel-related imported ZIKV
with an outbreak of an acute exanthematous illness (Cardoso et al., cases in non-endemic countries: Australia (AGDH, 2016; Pyke et al.,
2015). Toward the end of 2015, ZIKV activity expanded into at least 14 2016), Belgium (De Smet et al., 2016), Canada (GC, 2016), China (Yin
Brazilian states (WHO, 2015), with an estimated 440,000–1,300,000 et al., 2016; Zhang et al., 2016b), France (Maria et al., 2016), the US
suspected cases (Hennessey et al., 2016). To our surprise, it was noted (Hawaii) (CDC, 2016b), Italy (Nicastri et al., 2016b), Portugal (Ze-Ze
in Brazil that the number of newborn infants with microcephaly had in- et al., 2016), Spain (Diaz-Menendez et al., 2016), Switzerland
creased in the ZIKV-affected areas by September 2015 (Schuler-Faccini (Gyurech et al., 2016), and the Netherlands (Duijster et al., 2016).
et al., 2016), and N4000 cases of suspected microcephaly were reported Using species distribution modeling techniques, a recent study has pre-
by February 2016, although these cases may have been misdiagnosed in dicted that a large portion of the tropical and sub-tropical regions of the
some cases or over-reported (Victora et al., 2016). In agreement with globe has suitable environmental conditions but has not yet reported
this finding, retrospective studies in French Polynesia indicated an in- symptomatic cases of ZIKV infection (Fig. 3), with over two billion peo-
creased number of instances of microcephaly and other fetal abnormal- ple living in these areas (Messina et al., 2016). Thus, the presence of a
ities after the 2013–2014 ZIKV outbreak (Cauchemez et al., 2016; much larger susceptible population in suitable environments for ZIKV,
Jouannic et al., 2016). In October 2015, Colombia reported the local combined with its travel-mediated rapid dissemination, has raised con-
transmission of ZIKV infection outside Brazil (WHO, 2015), and by cerns about the high risk of introducing and establishing new autoch-
March 2016, a total of 51,473 suspected ZIKV infections were recorded thonous transmission in these areas (Bogoch et al., 2016; Fauci and
in that country, with 2090 laboratory-confirmed cases (WHO, 2016a). Morens, 2016; Gatherer and Kohl, 2016; Musso and Gubler, 2016).
Since its emergence in Brazil, ZIKV has spread at an alarming rate The situation is particularly challenging in the US, because ZIKV is likely
throughout much of Central and South America and the Caribbean, to spread further, from Florida to other southern states (Fig. 3, inset),
and the possibility that microcephaly is linked to ZIKV has increased, given the estimated range of the two major competent mosquito
prompting the World Health Organization to declare a “public health vectors, A. aegypti and A. albopictus (CDC, 2016e). For the same reason,
emergency of international concern” from February to November it is very likely that ZIKV will become endemic in the Americas.
2016 (WHO, 2016c, 2016d). ZIKV is still causing an unprecedented on-
going epidemic in Latin America and threatening North America and po- 3. Genetic diversity
tentially the rest of the world (Fauci and Morens, 2016; Lessler et al.,
2016). During the seven decades since its accidental discovery in 1947,
As of November 17, 2016, 48 countries and territories in the N500 ZIKV isolates have been obtained sporadically in Africa, Asia, the
Americas had reported the autochthonous mosquito-borne transmis- Pacific Islands, and the Americas, but only a very limited number of
sion of ZIKV (PAHO/WHO, 2016a), with an accumulated number of these strains have been fully sequenced (Baronti et al., 2014; Barzon
171,553 confirmed cases (PAHO/WHO, 2016b): Anguilla; Antigua and et al., 2016a; Berthet et al., 2014; Cunha et al., 2016; Ellison et al.,
Barbuda; Argentina; Aruba; the Bahamas; Barbados; Belize; Bolivia; 2016; Giovanetti et al., 2016; Kuno and Chang, 2007; Ladner et al.,
Bonaire, Sint Eustatius and Saba; Brazil; the British Virgin Islands; 2016; Lanciotti et al., 2008; Lednicky et al., 2016; Liu et al., 2016; Zhu
Cayman Islands; Colombia; Costa Rica; Cuba; Curaçao; Dominica; the et al., 2016). As shown in Fig. 4, a phylogenetic analysis using the nucle-
Dominican Republic; Ecuador; El Salvador; French Guiana; Grenada; otide sequence of all 29 complete or near-complete ZIKV genomes re-
Guadeloupe; Guatemala; Guyana; Haiti; Honduras; Jamaica; trievable from GenBank (as of May 2016) revealed that the
Martinique; Mexico; Montserrat; Nicaragua; Panama; Paraguay; Peru; geographically and temporally distinct ZIKV strains can be grouped
Puerto Rico; Saint Barthélemy; Saint Kitts and Nevis; Saint Lucia; Saint into two major genetic lineages, African and Asian, with all eighteen
Martin; Saint Vincent and the Grenadines; Sint Maarten; Suriname; 2015–2016 American epidemic strains derived from a common ances-
Trinidad and Tobago; Turks and Caicos Islands; the United States of tor of the Asian lineage (Yun et al., 2016). This spatiotemporal relation-
America (US); the US Virgin Islands; and Venezuela (Fig. 2). Sexually ship is consistent with recent reports (Calvet et al., 2016; Enfissi et al.,
transmitted cases have also been reported in Argentina, Canada, Chile, 2016; Faria et al., 2016; Faye et al., 2014; Haddow et al., 2012;
Peru, and the US (PAHO/WHO, 2016a). In November 2016, a decreasing Lanciotti et al., 2016; Mlakar et al., 2016; Wang et al., 2016). Moreover,
trend in ZIKV cases had been noted in all the ZIKV-affected countries the most recent data from phylogenetic and molecular clock analyses
and territories in the Americas, except for Mexico, Panama, and the suggest that the 2015–2016 American epidemic began with a single in-
islands of Turks and Caicos (PAHO/WHO, 2016a). To date, a total of 20 troduction of ZIKV into the Americas during May–December 2013,
countries and territories in the Americas have documented 2311 con- along with the increase in air traffic from ZIKV-endemic areas to Brazil
firmed cases of ZIKV-associated congenital syndrome (PAHO/WHO, and the emergence of ZIKV in the Pacific Islands (Faria et al., 2016). De-
2016a, 2016b). In the US states and federal districts, there were 4444 spite its considerable degree of genetic variation, little is known about
confirmed ZIKV cases reported to ArboNET as of November 23, 2016; the impact of viral genetic variation on the pathogenicity of ZIKV of
of these, 36 were sexually transmitted cases throughout the country, the African and Asian lineages, as well as among different strains within
and 182 were locally acquired mosquito-borne cases in Florida, where the Asian lineage (Wang et al., 2016). Thus, more research is needed to
autochthonous transmission was first reported in July 2016 and is cur- extend our understanding of the evolution and diversity of ZIKV and to
rently ongoing in the area of Miami Beach and in the county of Miami- explore the biological effects of viral genetic variation on the outcome of
Dade (CDC, 2016b, 2016f). By November 17, 2016, a total of 33 newborn ZIKV infection.
infants and pregnancy losses with birth defects had been reported to the
US Zika Pregnancy Registry (CDC, 2016d). 4. Transmission

2.5. Current status and future prospects ZIKV is transmitted to humans primarily through the bite of infected
mosquitoes, but it can also be passed from mother to child during preg-
As illustrated in Fig. 2, ZIKV is a potential pandemic threat, currently nancy or spread through sexual contact, breastfeeding, or blood transfu-
circulating not only in the Americas but also in the Pacific Islands sion. The multiple modes of ZIKV transmission make it difficult to
(American Samoa, Federated States of Micronesia, Fiji, Marshall develop control strategies against the pathogen.
54 B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64

Fig. 3. Global environmental suitability for ZIKV. Highlighted are the countries that are highly environmentally suitable, having a suitable area of more than 10,000 km2, but not yet
reporting symptomatic ZIKV cases (Source:Messina et al., 2016). An inset shows an estimated range for A. aegypti and A. albopictus in the US.
Source: Centers for Disease Control and Prevention (https://www.cdc.gov/zika/vector/range.html).

Fig. 4. An unrooted phylogenetic tree based on the nucleotide sequence of complete or near-complete genomes of all 29 available ZIKVs (as of May 2016). Multiple sequence alignments
were performed, and the unrooted phylogenetic tree was reconstructed, as described in Fig. 1. Two major genetic clusters are indicated: African lineage (green circle disc) and Asian lineage
(orange circle disc). Also, the tree shows that all of the eighteen 2015–2016 American epidemic strains (red circle disc) belong to the Asian lineage. The ZIKV strains used in our phylo-
genetic analysis are listed at the bottom of the tree, with a description in parenthesis of the year, country, host of isolation, and their GenBank accession numbers. Note that the genome
of MR-766 has been fully sequenced by four independent groups, and their nucleotide sequences are not identical, likely because of variations in the cultivation history of the virus.
B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64 55

4.1. Vector-borne transmission wide geographic distribution in tropical and temperate regions, there
is a high potential for A. albopictus to become a major vector for the
ZIKV is an arthropod-borne virus (arbovirus) that is transmitted by urban transmission of ZIKV across the globe.
mosquito vectors, with two distinct transmission cycles (Weaver et al., Both A. aegypti and A. albopictus are usually active during daylight
2016): (i) a sylvatic cycle, involved in the maintenance of ZIKV between hours (Ponlawat and Harrington, 2005; Scott and Takken, 2012) and
non-human primates and arboreal mosquitoes in forests; and (ii) an are widely distributed throughout the tropical and subtropical regions
urban cycle, involved in the transmission of ZIKV between humans of the world, with the habitat of A. albopictus extending further into
and urban mosquitoes in towns (Fig. 5). Occasionally, the virus is pre- cool temperate regions (Kraemer et al., 2015; Paupy et al., 2009;
sumably transmitted by arboreal mosquitoes from non-human pri- Thomas et al., 2012). In the US, both A. aegypti and A. albopictus are pres-
mates to humans when they are in close proximity. ZIKV has been ent in practically the entire southern and southeastern half of the coun-
isolated in Africa and Asia from a number of different mosquito species try (Fig. 3, inset map), according to a recent estimate by the Centers for
in the genus Aedes (e.g., A. aegypti, A. africanus, A. albopictus, Disease Control and Prevention (CDC, 2016e), reaching their maximal
A. apicoargenteus, A. furcifer, A. luteocephalus, A. opok, and A. vittatus), abundance in June–October each year (Monaghan et al., 2016). A recent
which can potentially act as vectors for viral transmission in a given en- study has shown that although they are susceptible to ZIKV infection,
vironment of those endemic areas (Akoua-Koffi et al., 2001; Berthet American populations of both A. aegypti and A. albopictus collected
et al., 2014; Cornet et al., 1979b; Diallo et al., 2014; Dick et al., 1952; from Florida have unexpectedly low levels of vector competence (the
Fagbami, 1979; Grard et al., 2014; Haddow et al., 1964; Marchette mosquito's ability to biologically transmit ZIKV) (Chouin-Carneiro
et al., 1969; McCrae and Kirya, 1982; Weinbren and Williams, 1958). et al., 2016). However, A. aegypti (and to a lesser extent A. albopictus)
Also, A. hensilli and A. polynesiensis were suggested to be vectors in the is thought to have a high level of vectorial capacity (a measurement of
Yap (Ledermann et al., 2014) and French Polynesia (Cao-Lormeau the efficiency of ZIKV transmission) because it primarily bites humans,
et al., 2014; Musso et al., 2014b) outbreaks, respectively. In most infects several persons during a single blood meal, and lives in close as-
cases, however, their vector competence in ZIKV transmission has not sociation with humans (Gubler, 2002). Collectively, there is real poten-
been fully investigated (Diagne et al., 2015). Based on previous studies tial for the spread of ZIKV across portions of the southern US over the
with other mosquito-borne flaviviruses (DENV and YFV), the vector next few years.
competence is apparently influenced not only by the genetic variation Moreover, ZIKV has been isolated or detected occasionally in other
in the virus and vector (Gubler et al., 1979) but also by the population mosquitoes, such as Anopheles coustani, Culex perfuscus, and Mansonia
density of the vectors in a particular environment (Miller et al., 1989). uniformis (Diallo et al., 2014; Faye et al., 2013). These mosquito species
This observation is also likely true for ZIKV (Diagne et al., 2015). may contribute to the transmission of ZIKV in particular environments,
In an urban cycle, the transmission of ZIKV is believed to be mediat- but the mere isolation of the virus from mosquitoes or its detection in
ed predominantly by two Aedes species mosquitoes: A. aegypti, recog- the mosquitoes does not necessarily identify it as an actual vector. Fur-
nized by a bright lyre-shaped dorsal pattern with white bands on its ther field and experimental studies are needed to better define vector
legs, and A. albopictus, characterized by a single longitudinal dorsal competence for ZIKV and to determine whether there are any other
stripe with white bands on its legs (ECDC, 2016b). Of these two species, mosquito vectors or animal hosts.
A. aegypti is considered to be the primary vector associated with ZIKV
outbreaks, as supported by (1) isolating or detecting the virus in a 4.2. Non-vector-borne transmission
pool of A. aegypti mosquitoes (Akoua-Koffi et al., 2001; Diallo et al.,
2014; Marchette et al., 1969); (2) showing the susceptibility of Direct human-to-human transmission of ZIKV has been documented
A. aegypti mosquitoes to infection with the virus (Boorman and to occur perinatally, sexually, and through breastfeeding or blood trans-
Porterfield, 1956; Cornet et al., 1979a; Li et al., 2012); and (3) demon- fusion: (i) ZIKV can be passed from an infected mother to her fetus dur-
strating the transmission of the virus from artificially fed A. aegypti mos- ing pregnancy, as evidenced not only by the detection of viral RNA in the
quitoes to rhesus monkeys and mice (Boorman and Porterfield, 1956). amniotic fluid, urine, or serum of mothers whose fetuses had brain ab-
As a secondary vector for ZIKV, A. albopictus has been suggested to be in- normalities (Brasil et al., 2016; Calvet et al., 2016; Driggers et al.,
volved in the urban transmission in Gabon, where five human sera and 2016; Jouannic et al., 2016; Meaney-Delman et al., 2016; Oliveira Melo
two A. albopictus pools collected in an urban setting in 2007 were posi- et al., 2016; Sarno et al., 2016; Villamil-Gomez et al., 2016), but also
tive for ZIKV (Grard et al., 2014), and in Singapore, where high dissem- by probing of viral RNA/proteins/particles in the brain, placenta, or
ination and transmission rates of ZIKV in local A. albopictus have been serum of newborns with microcephaly and those aborted (Besnard
reported (Wong et al., 2013). Because of its high invasive ability and et al., 2014; Butler, 2016; Driggers et al., 2016; Faria et al., 2016;

Fig. 5. Vector-borne transmission of ZIKV. There are two mosquito-driven transmission cycles: (1) a sylvatic cycle, in which the virus cycles between non-human primates and arboreal
mosquitoes; and (2) an urban cycle, in which the virus cycles between humans and urban mosquitoes. Under certain circumstances, ZIKV can presumably be transmitted from non-human
primates to humans via arboreal mosquitoes.
56 B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64

Martines et al., 2016; Mlakar et al., 2016; Sarno et al., 2016). (ii) ZIKV 5. Virology
can be sexually transmitted from an infected person to his or her part-
ners, as shown by the detection of the virus in patient's semen, often ZIKV, a 50-nm enveloped particle, contains an inner nucleocapsid
in high titer, and transmission of the virus between both sexes, with composed of a linear plus-strand genomic RNA and multiple copies of
the male-to-female transmission occurring more frequently than the viral capsid (C) protein and the outer host cell-derived lipid bilayer
female-to-male or male-to-male transmission (Armstrong et al., 2016; bearing 180 copies each of two proteins: the viral membrane (M, a
Atkinson et al., 2016; Deckard et al., 2016; Foy et al., 2011; Frank et al., cleavage product of prM) protein and the envelope (E) protein
2016; Freour et al., 2016; Hills et al., 2016; Mansuy et al., 2016; Musso (Kostyuchenko et al., 2016; Sirohi et al., 2016). In the case of the
et al., 2015c; Venturi et al., 2016). In addition to semen, it is important American ZIKV strain PRVABC-59 (GenBank accession no. KX377337)
to note that ZIKV has also been detected in urine, saliva, and nasopha- isolated from a patient in Puerto Rico in 2015 (Lanciotti et al., 2016),
ryngeal swabs, potentially facilitating the non-vector-borne transmis- the genomic RNA is 10,807 nucleotides (nt) long and comprises a single
sion of ZIKV (Atkinson et al., 2016; Barzon et al., 2016a; Besnard et al., 10,272-nt open reading frame (ORF) flanked by a 107-nt 5′ noncoding
2014; Brasil et al., 2016; Fonseca et al., 2014; Gourinat et al., 2015; region (NCR) and a 428-nt 3′ NCR (Yun et al., 2016) (Fig. 6A). The ORF
Korhonen et al., 2016; Kutsuna et al., 2014; Leung et al., 2015; Maria encodes a large polyprotein of 3423 amino acids (aa), which is predicted
et al., 2016; Musso et al., 2015b; Musso et al., 2015c; Roze et al., 2016; to be cleaved by viral and cellular proteases into at least 10 individual
Shinohara et al., 2016). (iii) ZIKV RNA/particles have been detected in proteins, designated in an N- to C-terminal direction (Kim et al., 2015;
breast milk, suggesting a potential risk of viral transmission through Yun et al., 2016): C (122 aa), prM (168 aa), E (504 aa), NS1 (352 aa),
breastfeeding (Besnard et al., 2014; Dupont-Rouzeyrol et al., 2016). NS2A (226 aa), NS2B (130 aa), NS3 (617 aa), NS4A (150 aa), NS4B
(iv) ZIKV is likely to be transmitted through transfusions of blood (251 aa), and NS5 (903 aa). The three N-terminal structural proteins
from donors who have been infected with the virus (Aubry et al., are necessary for the formation of infectious virions (Kostyuchenko
2015; Aubry et al., 2016; Faria et al., 2016; Musso et al., 2014a). et al., 2016; Sirohi et al., 2016), and the seven C-terminal nonstructural
(v) Direct transmission, although uncommon, can take place through proteins are essential for the replication of the viral genomic RNA
the skin or mucous membranes, as exemplified by the index patient, (Brinton, 2013; Villordo and Gamarnik, 2009; Westaway et al., 2002;
who had a high viral load of 2 × 108 genome copies per ml in serum Yun and Lee, 2006). Viral RNA replication is catalyzed by the two viral
(Swaminathan et al., 2016). Although these non-vector-borne transmis- nonstructural proteins: (1) NS3 has an N-terminal serine protease do-
sions of ZIKV are believed to be uncommon, further studies are neces- main, which requires NS2B as a cofactor for its activity (Lei et al.,
sary to identify the risk factors involved in these routes of viral 2016; Phoo et al., 2016); it also has a C-terminal RNA helicase domain
transmission. that also possesses nucleoside triphosphatase and RNA 5′-

Fig. 6. Genome structure and replication cycle of ZIKV. (A) A schematic diagram of the ZIKV genomic RNA. The single open reading frame (ORF) encoded in the viral genome is shown as a
long gray barrel, with the three structural (green) and seven nonstructural (magenta) proteins defined below the ORF. NCR, non-coding region. Open arrowheads indicate the cleavage
sites for cellular signalases, and solid arrowheads mark the cleavage sites for the viral serine protease (NS2B-NS3). A gray arrowhead indicates the cleavage site for furin or a furin-like
protease. The cleavage at the NS1-NS2A junction is mediated by an unknown protease(s). (B) Proposed life cycle of ZIKV. The eight major steps of the viral life cycle are: attachment,
endocytosis, membrane fusion, translation, RNA replication, assembly, maturation, and release.
Source: Yun and Lee, 2014.
B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64 57

triphosphatase activities (Jain et al., 2016). (2) NS5 has an N-terminal spontaneously within a week, but arthralgia may persist for up to a
methyltransferase domain (Coloma et al., 2016; Zhang et al., 2016a) month (Foy et al., 2011).
and a C-terminal RNA-dependent RNA polymerase domain (Adiga,
2016; Cox et al., 2015). 6.2. Guillain-Barré syndrome
There are very few direct experimental data available concerning the
replication cycle of ZIKV, but the current understanding of the molecular Guillain-Barré syndrome is an autoimmune disease in which the im-
biology of other flaviviruses provides a clear path forward for ZIKV re- mune system attacks part of the peripheral nervous system, causing tin-
search (Lindenbach et al., 2013). As schematically illustrated in Fig. 6B, gling, muscle weakness, paralysis, and even death (Creange, 2016;
flaviviruses enter their host cells initially by binding nonspecifically to Goodfellow and Willison, 2016). Previously, this neuromuscular com-
the cell surface (Chen et al., 1997; Davis et al., 2006; Navarro-Sanchez plication had been associated with infection by other arboviruses,
et al., 2003; Pokidysheva et al., 2006; Tassaneetrithep et al., 2003), such as DENV (Carod-Artal et al., 2013; Ralapanawa et al., 2015;
followed by internalization specifically through clathrin-mediated en- Simon et al., 2016; Verma et al., 2014) and chikungunya virus
docytosis in a viral glycoprotein E-dependent manner (Perera-Lecoin (Wielanek et al., 2007). The temporal and geographic association of
et al., 2013; Pierson and Kielian, 2013). When the virions are delivered ZIKV with Guillain-Barré syndrome was initially observed during the
to endosomes, the E glycoprotein undergoes a series of low pH- 2013–2014 outbreak reported in French Polynesia (ECDC, 2014;
induced structural changes, enabling the fusion of the viral membrane Musso et al., 2014b; Oehler et al., 2014) and subsequently during the
with the endosomal membrane and the release of the viral genome 2015–2016 outbreak that is still ongoing in the Americas (Broutet
into the cytoplasm (Harrison, 2008; Kaufmann and Rossmann, 2011; et al., 2016; Roze et al., 2016; Thomas et al., 2016). During the previous
Smit et al., 2011; Stiasny et al., 2011). Viral RNA replication takes place French Polynesia outbreak, the incidence of Guillain-Barré syndrome
in close association with virus-induced intracellular membrane struc- was estimated to be ~ 20-fold higher than its basal incidence of 1–2
tures known as replication complexes, which contain the viral RNA cases per 100,000 population per year (Oehler et al., 2014; Sejvar
and proteins, as well as presumably a set of the host cell factors required et al., 2011). More definitively, a recent case-control study revealed
for viral RNA synthesis (Gillespie et al., 2010; Hsu et al., 2010; Welsch that anti-ZIKV IgM or IgG was detected in 41 (98%) of 42 patients with
et al., 2009). Assembly of immature virions is initiated by budding of a Guillain-Barré syndrome, and all had neutralizing antibodies against
viral genomic RNA-C protein complex into the endoplasmic reticulum ZIKV, as compared to 54 (55%) of 98 controls: age-, sex- and
(Zhang et al., 2003; Zhang et al., 2007), where it is enveloped by the residence-matched patients with a non-febrile illness (Cao-Lormeau
lipid bilayer with the viral prM and E proteins embedded (Lorenz et al., 2016). The same study also showed that patients with Guillain-
et al., 2002). These immature virions undergo a maturation process Barré syndrome had electrophysiological characteristics consistent
while moving through the host secretory pathway (Mackenzie and with the acute motor axonal neuropathy type of the disease (Cao-
Westaway, 2001) and particularly the trans-Golgi network, going Lormeau et al., 2016). Thus far, ZIKV-induced Guillain-Barré syndrome
through post-translational modifications that include the cleavage of has been transient, and most patients have recovered fully. Currently,
prM by furin or a furin-like protease to produce the mature M protein the mechanism by which ZIKV infection leads to Guillain-Barré syn-
(Stadler et al., 1997; Yu et al., 2008). Finally, the completely or partially drome is unknown but is under active investigation.
mature virions are released from the cell via exocytosis (Pierson and
Diamond, 2012). For future studies, it is important to understand the 6.3. Microcephaly
similarities and differences in molecular biology between ZIKV and
other relatively well-characterized flaviviruses (e.g., JEV, WNV, DENV, Microcephaly is a neurological condition in which the brain of a baby
and YFV), with respect to viral replication and pathogenesis. does not develop properly, causing the head to be smaller than normal
(Klase et al., 2016; Panchaud et al., 2016; Tang, 2016). It is divided into
two types: (1) primary or congenital microcephaly, which is present in
6. Clinical presentation
utero or at birth; and (2) secondary or postnatal microcephaly, which
develops after birth (Alcantara and O'Driscoll, 2014; Passemard et al.,
ZIKV can produce a wide variety of clinical symptoms in humans. A
2013). While primary microcephaly is likely caused by a decrease in
growing body of evidence suggests that in some severe cases, ZIKV
the number of neurons produced during neurogenesis, secondary mi-
causes neurological diseases, such as Guillain-Barré syndrome in ZIKV-
crocephaly is presumably caused by a reduction in the number of
infected adults and microcephaly in infants born to ZIKV-infected
dendritic processes and synaptic connections (Woods, 2004). Micro-
women.
cephaly can be caused by a variety of genetic mutations, peri- and
post-natal brain injuries, teratogenic agents, and congenital infections
6.1. Common signs and symptoms (Dahlgren and Wilson, 2001; von der Hagen et al., 2014). Earlier, con-
genital microcephaly had been described in association with rubella
ZIKV infections are symptomatic in only ~20–25% of the infected in- virus (De Santis et al., 2006; Webster, 1998) and cytomegalovirus
dividuals who develop a mild and self-limited illness, with an incuba- (Plosa et al., 2012; Swanson and Schleiss, 2013). For ZIKV, a causal rela-
tion period of 4–10 days (Bearcroft, 1956; CDC, 2016g; Cerbino-Neto tionship between prenatal infection and microcephaly emerged in
et al., 2016; Duffy et al., 2009; Lazear and Diamond, 2016; Musso Brazil, as the number of newborns with microcephaly began to rise in
et al., 2014a). In symptomatic cases, the common symptoms are non- September 2015 (Microcephaly Epidemic Research Group, 2016;
specific and resemble those of a flu-like syndrome, including transient Schuler-Faccini et al., 2016); thereafter, 8301 cases of microcephaly
low-grade fever, itchy maculopapular rash, arthritis or arthralgia, and were recorded from November 2015 to July 2016 in that country
nonpurulent conjunctivitis; at a lesser frequency, retro-orbital pain, (Magalhaes-Barbosa et al., 2016). Subsequently, the potential risk of mi-
headache, myalgia, edema, and vomiting are seen (Brasil et al., 2016; crocephaly associated with ZIKV infection has been suggested by two
Cao-Lormeau et al., 2014; Duffy et al., 2009; Macnamara, 1954; Musso retrospective studies from French Polynesia (Cauchemez et al., 2016;
et al., 2014a; Zammarchi et al., 2015a). Other clinical manifestations ob- Jouannic et al., 2016) and one prospective study from Brazil (Brasil
served with acute ZIKV infection include hematospermia (Foy et al., et al., 2016). In addition to this spatiotemporal association, ZIKV or its
2011; Musso et al., 2015c), hearing difficulties (Tappe et al., 2015), gene expression has been detected in the amniotic fluid (Calvet et al.,
thrombocytopenia, and subcutaneous bleeding (Chraibi et al., 2016; 2016; Jouannic et al., 2016; Meaney-Delman et al., 2016; Sarno et al.,
Duijster et al., 2016; Karimi et al., 2016; Sharp et al., 2016). The symp- 2016) and in various tissues of fetuses with microcephaly and those
toms generally appear along with the viremia and disappear who died after birth or following abortion (Besnard et al., 2014;
58 B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64

Butler, 2016; Driggers et al., 2016; Faria et al., 2016; Martines et al., mechanism for microcephaly, since hNPCs drive the development of
2016; Mlakar et al., 2016; Sarno et al., 2016). Moreover, ZIKV-specific the cortex of fetal brains. Consistent with these findings, ZIKV infection
IgM has been identified in the cerebrospinal fluid and serum of neonates of human brain organoids (three-dimensional cell culture systems reca-
with microcephaly (Cordeiro et al., 2016; de Araujo et al., 2016). pitulating early events in brain development) led to a reduction in pro-
According to the most recent guideline of the Centers for Disease liferative zones and disrupted cortical layers, thereby causing a decrease
Control and Prevention, congenital microcephaly is defined as: (i) for in overall organoid size reminiscent of microcephaly (Cugola et al.,
live births, having a head circumference (HC) at birth below the third 2016; Dang et al., 2016; Garcez et al., 2016; Qian et al., 2016). In mice
percentile for the age and sex of the baby or, if the HC at birth is unavail- (SJL, ICR, and C57 strains), ZIKV can replicate in the brains of embryos
able, an HC below the third percentile within the first 2 weeks after by targeting different neuronal lineages, causing cell cycle arrest, apo-
birth; and (ii) for stillbirths and elective abortions, having a HC at deliv- ptosis, and inhibition of NPC differentiation, all of which affect the corti-
ery below the third percentile for the age and sex of the baby (CDC, cal development of the embryos (Cugola et al., 2016; Li et al., 2016; Wu
2016c). In general, HC is a good indication of total brain volume that et al., 2016). Also, intracranial infection of ZIKV in early postnatal C57BL/
has been used to evaluate brain growth and size (Barbier et al., 2013; 6 mice has been shown to deplete proliferating cells in the ventricular
Bolduc and Shevell, 2005; Sullivan et al., 2014). A significance difference zone of the neural stem cell compartment and destroy corticospinal py-
in brain size is an important risk factor for motor and cognitive dysfunc- ramidal neurons, a cell type implicated in ZIKV-induced microcephaly
tion (von der Hagen et al., 2014; Watemberg et al., 2002). Accumulated (Huang et al., 2016). In addition to the direct disruption of NPCs, mature
data on pre- and post-natal clinical cases to date suggest that ZIKV infec- neurons, and other cells in the brain (Bayless et al., 2016; Dang et al.,
tion during pregnancy is linked to microcephaly and other congenital 2016), the differentiation and migration of neurons in the developing
abnormalities (e.g., facial disproportionality, cutis girata, hypertonia/ brain are also believed to be affected by ZIKV infection. Further studies
spasticity, and hyperreflexia), occasionally accompanied by neuroimag- are warranted to elucidate the pathobiology of ZIKV-induced brain
ing abnormalities such as calcifications, ventriculomegaly, and malformations and their underlying mechanisms. Furthermore,
lissencephaly (Brasil et al., 2016; Driggers et al., 2016; Hazin et al., in vitro infection studies of human placental cells have shown that mac-
2016; Meaney-Delman et al., 2016; Microcephaly Epidemic Research rophages, trophoblasts, and endothelial cells are susceptible to ZIKV in-
Group, 2016; Miranda-Filho Dde et al., 2016; Mlakar et al., 2016; fection to various degrees, presumably depending on gestational age
Oliveira Melo et al., 2016; Petersen et al., 2016; Sarno et al., 2016; and host genetic variation (Bayer et al., 2016; Quicke et al., 2016;
Schuler-Faccini et al., 2016). Recently, the ZIKV-microcephaly associa- Tabata et al., 2016).
tion has been supported by a preliminary report of the first case- In human and mouse studies, ZIKV is shown to be able to establish a
control study, showing that 13 of 32 newborns with microcephaly and productive infection not only in the fetus and placenta during pregnan-
none of 62 controls without microcephaly had laboratory-confirmed cy but also in the eyes and male and female reproductive tracts.
ZIKV infection (de Araujo et al., 2016). In addition, congenital ZIKV in- (1) Eyes: ZIKV has been detected and isolated from human conjunctival
fection has also been suggested to be associated with hearing loss swab samples (Sun et al., 2016). Consistent with this clinical finding,
(Leal et al., 2016a; Leal et al., 2016b; Miranda-Filho Dde et al., 2016) productive ZIKV infection has been observed in ocular tissues in
and ocular anomalies (de Paula Freitas et al., 2016; Miranda-Filho Dde Ifnar1−/− mice, as evidenced by the shedding of viral RNA in tears and
et al., 2016; Oliveira Melo et al., 2016; Ventura et al., 2016a; Ventura detection of viral RNA in the cornea, optic nerve, and ganglion and bipo-
et al., 2016b; Ventura et al., 2016c; Ventura et al., 2016d). lar cells in the retina (Miner et al., 2016). (2) Male reproductive tract: In
mice, a mouse-adapted ZIKV can infect spermatogonia, Sertoli cells, and
6.4. Other neurological complications Leydig cells in the testis and epididymis, causing testicular tissue dam-
age that is associated with reduced levels of the sex hormones (testos-
Case reports suggest that ZIKV infection may be associated with terone and inhibin B) and oligospermia (Chan et al., 2016; Govero
other neurological complications, including the case of an 81-year-old et al., 2016; Ma et al., 2016). Although persistent shedding of ZIKV
man who developed meningoencephalitis (Carteaux et al., 2016) and RNA in semen has been reported in humans for 6 months after the
the case of a 15-year-old girl who was diagnosed with acute myelitis onset of symptoms (Barzon et al., 2016b; Mansuy et al., 2016; Nicastri
(Mecharles et al., 2016). As research progresses, it is likely that other et al., 2016a), such severe injury on testicular tissue as seen in mice
neurological and non-neurological complications caused by ZIKV infec- has not yet been described in humans. (3) Female reproductive tract:
tion will be identified in the future. Case-control or cohort studies, to- A case report has shown that ZIKV RNA is detectable in human cervical
gether with well-characterized case reports, will be required in order mucus at 11 days after symptom onset, while it is undetectable in blood
for us to better understand the potential role of ZIKV infection in adults and urine samples (Prisant et al., 2016). Another case report has also
and newborns (Musso and Baud, 2016; Rasmussen et al., 2016). shown that ZIKV RNA is present in human vaginal mucosal specimens
for over 11 weeks (Murray et al., 2017). An in vitro infection experiment
7. Tissue and cell tropism has indicated that human uterine fibroblasts are susceptible to ZIKV in-
fection, potentially contributing to the heterosexual transmission of
ZIKV RNA has been detected both in the brains of infants with micro- ZIKV in women (Chen et al., 2016). Overall, ZIKV appears to have a
cephaly who died and in the placentas of women who had spontaneous broad tissue tropism, infecting a wide range of cell types. Therefore, an
abortion during the first or second trimester because of suspected ZIKV important area of ZIKV research in tissue/cell tropism is the identifica-
infection (Bhatnagar et al., 2017). After subcutaneous infection of ZIKV tion of host factors that promote or restrict viral replication.
in a pregnant nonhuman primate (pigtail macaque), viral RNA has
been identified in fetal and maternal tissues, specifically the fetal brain 8. Conclusion
and liver, the chorionic villi of the placenta, and the maternal brain,
eyes, spleen and liver; of these tissues, the placenta has the highest ZIKV, first isolated in Uganda in 1947, is transmitted among people
viral RNA load (Adams Waldorf et al., 2016). Over the last year, rapid primarily by the bite of infected Aedes species mosquitoes
progress has been made in the development of both human cell-based (e.g., A. aegypti and A. albopictus) or secondarily through non-vector
in vitro models and in vivo mouse models for the study of ZIKV modes (i.e., vertical transmission, sexual transmission, and blood trans-
neuropathogenesis. To date, it has been demonstrated that ZIKV, al- fusion). ZIKV was confined to Africa and Asia for over half a century after
though capable of infecting mature neurons, preferentially infects its discovery, before it emerged in the Pacific Islands by the late 2000s
human neural progenitor cells (hNPCs) and triggers apoptosis and early 2010s. Now it has gained a major foothold in the Americas
(Onorati et al., 2016; Tang et al., 2016), suggesting a potential and is poised to jump to the rest of the world. From a clinical standpoint,
B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64 59

ZIKV infection was considered to be associated only with mild nonspe- Bayless, N.L., Greenberg, R.S., Swigut, T., Wysocka, J., Blish, C.A., 2016. Zika virus infection
induces cranial neural crest cells to produce cytokines at levels detrimental for
cific flu-like illness prior to the 2013–2014 French Polynesian outbreak, neurogenesis. Cell Host Microbe 20, 423–428.
when serious neurological complications were reported, and the Bearcroft, W.G., 1956. Zika virus infection experimentally induced in a human volunteer.
2015–2016 American outbreak, when severe congenital malformations Trans. R. Soc. Trop. Med. Hyg. 50, 442–448.
Berthet, N., Nakoune, E., Kamgang, B., Selekon, B., Descorps-Declere, S., Gessain, A.,
were documented. The most worrying aspect of ZIKV infection is that Manuguerra, J.C., Kazanji, M., 2014. Molecular characterization of three Zika
we do not yet know the real scope of its complications, which may be flaviviruses obtained from sylvatic mosquitoes in the Central African Republic. Vector
more far-reaching than those initially anticipated. Much more research Borne Zoonotic Dis. 14, 862–865.
Besnard, M., Lastere, S., Teissier, A., Cao-Lormeau, V., Musso, D., 2014. Evidence of perina-
effort is required to fill the knowledge gaps concerning ZIKV pathogen- tal transmission of Zika virus, French Polynesia, December 2013 and February 2014.
esis and to develop medical countermeasures against this previously Euro Surveill. 19, 20751.
neglected but newly emerging human pathogen. Bhatnagar, J., Rabeneck, D.B., Martines, R.B., Reagan-Steiner, S., Ermias, Y., Estetter, L.B.,
Suzuki, T., Ritter, J., Keating, M.K., Hale, G., Gary, J., Muehlenbachs, A., Lambert, A.,
Lanciotti, R., Oduyebo, T., Meaney-Delman, D., Bolanos, F., Saad, E.A., Shieh, W.J.,
Zaki, S.R., 2017. Zika virus RNA replication and persistence in brain and placental tis-
Acknowledgments sue. Emerg. Infect. Dis. 23, 405–414.
Bogoch, I.I., Brady, O.J., Kraemer, M.U., German, M., Creatore, M.I., Kulkarni, M.A.,
Brownstein, J.S., Mekaru, S.R., Hay, S.I., Groot, E., Watts, A., Khan, K., 2016. Anticipating
This work was supported by the Utah Science Technology and the international spread of Zika virus from Brazil. Lancet 387, 335–336.
Research Initiative (USTAR A34637 and A36815) and by a grant Bolduc, F.V., Shevell, M.I., 2005. Corrected head circumference centiles as a possible pre-
(200784-00001) from the American Board of Obstetrics and Gynecolo- dictor of developmental performance in high-risk neonatal intensive care unit survi-
vors. Dev. Med. Child Neurol. 47, 766–770.
gy, the American College of Obstetricians and Gynecologists, the Boorman, J.P., Porterfield, J.S., 1956. A simple technique for infection of mosquitoes with
American Society for Reproductive Medicine, and the Society for Repro- viruses; transmission of Zika virus. Trans. R. Soc. Trop. Med. Hyg. 50, 238–242.
ductive Endocrinology and Infertility. We thank Dr. Deborah McClellan Brasil, P., Pereira Jr., J.P., Moreira, M.E., Ribeiro Nogueira, R.M., Damasceno, L., Wakimoto,
M., Rabello, R.S., Valderramos, S.G., Halai, U.A., Salles, T.S., Zin, A.A., Horovitz, D.,
for her careful and critical reading of our paper. There is no conflict of in- Daltro, P., Boechat, M., Raja Gabaglia, C., Carvalho de Sequeira, P., Pilotto, J.H.,
terest in this paper. Medialdea-Carrera, R., Cotrim da Cunha, D., Abreu de Carvalho, L.M., Pone, M.,
Machado Siqueira, A., Calvet, G.A., Rodrigues Baião, A.E., Neves, E.S., Nassar de
Carvalho, P.R., Hasue, R.H., Marschik, P.B., Einspieler, C., Janzen, C., Cherry, J.D., Bispo
References
de Filippis, A.M., Nielsen-Saines, K., 2016. Zika virus infection in pregnant women
in Rio de Janeiro. N. Engl. J. Med. 375, 2321–2334.
Adams Waldorf, K.M., Stencel-Baerenwald, J.E., Kapur, R.P., Studholme, C., Boldenow, E.,
Brinton, M.A., 2013. Replication cycle and molecular biology of the West Nile virus. Virus-
Vornhagen, J., Baldessari, A., Dighe, M.K., Thiel, J., Merillat, S., Armistead, B.,
es 6, 13–53.
Tisoncik-Go, J., Green, R.R., Davis, M.A., Dewey, E.C., Fairgrieve, M.R., Gatenby, J.C.,
Broutet, N., Krauer, F., Riesen, M., Khalakdina, A., Almiron, M., Aldighieri, S., Espinal, M.,
Richards, T., Garden, G.A., Diamond, M.S., Juul, S.E., Grant, R.F., Kuller, L., Shaw,
Low, N., Dye, C., 2016. Zika virus as a cause of neurologic disorders. N. Engl. J. Med.
D.W., Ogle, J., Gough, G.M., Lee, W., English, C., Hevner, R.F., Dobyns, W.B., Gale Jr.,
374, 1506–1509.
M., Rajagopal, L., 2016. Fetal brain lesions after subcutaneous inoculation of Zika
Buathong, R., Hermann, L., Thaisomboonsuk, B., Rutvisuttinunt, W., Klungthong, C.,
virus in a pregnant nonhuman primate. Nat. Med. 22, 1256–1259.
Chinnawirotpisan, P., Manasatienkij, W., Nisalak, A., Fernandez, S., Yoon, I.K.,
Adekolu-John, E.O., Fagbami, A.H., 1983. Arthropod-borne virus antibodies in sera of
Akrasewi, P., Plipat, T., 2015. Detection of Zika virus infection in Thailand,
residents of Kainji Lake Basin, Nigeria 1980. Trans. R. Soc. Trop. Med. Hyg. 77,
2012–2014. Am. J. Trop. Med. Hyg. 93, 380–383.
149–151.
Butler, D., 2016. First Zika-linked birth defects detected in Colombia. Nature 531, 153.
Adiga, R., 2016. Phylogenetic analysis of the NS5 gene of Zika virus. J. Med. Virol. 88,
Calvet, G., Aguiar, R.S., Melo, A.S., Sampaio, S.A., de Filippis, I., Fabri, A., Araujo, E.S., de
1821–1826.
Sequeira, P.C., de Mendonca, M.C., de Oliveira, L., Tschoeke, D.A., Schrago, C.G.,
AGDH, 2016. Zika Virus — Notifications of Zika Virus Infection (Zika). Australian Govern-
Thompson, F.L., Brasil, P., Dos Santos, F.B., Nogueira, R.M., Tanuri, A., de Filippis,
ment Department of Health, Canberra, Australia November 11, 2016. http://www.
A.M., 2016. Detection and sequencing of Zika virus from amniotic fluid of fetuses
health.gov.au/internet/main/publishing.nsf/Content/ohp-zika-notifications.htm.
with microcephaly in Brazil: a case study. Lancet Infect. Dis. 16, 653–660.
Akoua-Koffi, C., Diarrassouba, S., Benie, V.B., Ngbichi, J.M., Bozoua, T., Bosson, A., Akran, V.,
Campos, G.S., Bandeira, A.C., Sardi, S.I., 2015. Zika virus outbreak, Bahia, Brazil. Emerg. In-
Carnevale, P., Ehouman, A., 2001. Investigation surrounding a fatal case of yellow
fect. Dis. 21, 1885–1886.
fever in Cote d'Ivoire in 1999. Bull. Soc. Pathol. Exot. 94, 227–230.
Cao-Lormeau, V.M., Blake, A., Mons, S., Lastere, S., Roche, C., Vanhomwegen, J., Dub, T.,
Alcantara, D., O'Driscoll, M., 2014. Congenital microcephaly. Am. J. Med. Genet. C. Semin.
Baudouin, L., Teissier, A., Larre, P., Vial, A.L., Decam, C., Choumet, V., Halstead, S.K.,
Med. Genet. 166C, 124–139.
Willison, H.J., Musset, L., Manuguerra, J.C., Despres, P., Fournier, E., Mallet, H.P.,
Alera, M.T., Hermann, L., Tac-An, I.A., Klungthong, C., Rutvisuttinunt, W., Manasatienkij,
Musso, D., Fontanet, A., Neil, J., Ghawche, F., 2016. Guillain–Barre syndrome outbreak
W., Villa, D., Thaisomboonsuk, B., Velasco, J.M., Chinnawirotpisan, P., Lago, C.B.,
associated with Zika virus infection in French Polynesia: a case–control study. Lancet
Roque Jr., V.G., Macareo, L.R., Srikiatkhachorn, A., Fernandez, S., Yoon, I.K., 2015.
387, 1531–1539.
Zika virus infection, Philippines, 2012. Emerg. Infect. Dis. 21, 722–724.
Cao-Lormeau, V.M., Musso, D., 2014. Emerging arboviruses in the Pacific. Lancet 384,
Armstrong, P., Hennessey, M., Adams, M., Cherry, C., Chiu, S., Harrist, A., Kwit, N., Lewis, L.,
1571–1572.
McGuire, D.O., Oduyebo, T., Russell, K., Talley, P., Tanner, M., Williams, C., Zika Virus
Cao-Lormeau, V.M., Roche, C., Teissier, A., Robin, E., Berry, A.L., Mallet, H.P., Sall, A.A.,
Response Epidemiology and Laboratory Team, 2016. Travel-associated Zika virus dis-
Musso, D., 2014. Zika virus, French Polynesia, South Pacific, 2013. Emerg. Infect. Dis.
ease cases among U.S. residents — United States, January 2015–February 2016.
20, 1085–1086.
MMWR Morb. Mortal. Wkly. Rep. 65, 286–289.
Cardoso, C.W., Paploski, I.A., Kikuti, M., Rodrigues, M.S., Silva, M.M., Campos, G.S., Sardi,
Atkinson, B., Hearn, P., Afrough, B., Lumley, S., Carter, D., Aarons, E.J., Simpson, A.J., Brooks,
S.I., Kitron, U., Reis, M.G., Ribeiro, G.S., 2015. Outbreak of exanthematous illness asso-
T.J., Hewson, R., 2016. Detection of Zika virus in semen. Emerg. Infect. Dis. 22, 940.
ciated with Zika, chikungunya, and dengue viruses, Salvador, Brazil. Emerg. Infect.
Aubry, M., Finke, J., Teissier, A., Roche, C., Broult, J., Paulous, S., Despres, P., Cao-Lormeau,
Dis. 21, 2274–2276.
V.M., Musso, D., 2015. Seroprevalence of arboviruses among blood donors in French
Carod-Artal, F.J., Wichmann, O., Farrar, J., Gascon, J., 2013. Neurological complications of
Polynesia, 2011–2013. Int. J. Infect. Dis. 41, 11–12.
dengue virus infection. Lancet Neurol. 12, 906–919.
Aubry, M., Richard, V., Green, J., Broult, J., Musso, D., 2016. Inactivation of Zika virus in
Carteaux, G., Maquart, M., Bedet, A., Contou, D., Brugieres, P., Fourati, S., Cleret de
plasma with amotosalen and ultraviolet A illumination. Transfusion 56, 33–40.
Langavant, L., de Broucker, T., Brun-Buisson, C., Leparc-Goffart, I., Mekontso Dessap,
Barbier, A., Boivin, A., Yoon, W., Vallerand, D., Platt, R.W., Audibert, F., Barrington, K.J.,
A., 2016. Zika virus associated with meningoencephalitis. N. Engl. J. Med. 374,
Shah, P.S., Nuyt, A.M., Network, Canadian Neonatal, 2013. New reference curves for
1595–1596.
head circumference at birth, by gestational age. Pediatrics 131, e1158–e1167.
Cauchemez, S., Besnard, M., Bompard, P., Dub, T., Guillemette-Artur, P., Eyrolle-Guignot,
Baronti, C., Piorkowski, G., Charrel, R.N., Boubis, L., Leparc-Goffart, I., de Lamballerie, X.,
D., Salje, H., Van Kerkhove, M.D., Abadie, V., Garel, C., Fontanet, A., Mallet, H.P.,
2014. Complete coding sequence of Zika virus from a French Polynesia outbreak in
2016. Association between Zika virus and microcephaly in French Polynesia,
2013. Genome Announc. 2, e00500–14.
2013–15: a retrospective study. Lancet 387, 2125–2132.
Barzon, L., Pacenti, M., Berto, A., Sinigaglia, A., Franchin, E., Lavezzo, E., Brugnaro, P., Palu,
CDC, 2016a. All Countries and Territories With Active Zika Virus Transmission. Centers for
G., 2016a. Isolation of infectious Zika virus from saliva and prolonged viral RNA shed-
Disease Control and Prevention, Atlanta, GA November 21, 2016. https://www.cdc.
ding in a traveller returning from the Dominican Republic to Italy, January 2016. Euro
gov/zika/geo/active-countries.html.
Surveill. 21, 30159.
CDC, 2016b. Case Counts in the US. Centers for Disease Control and Prevention, Atlanta,
Barzon, L., Pacenti, M., Franchin, E., Lavezzo, E., Trevisan, M., Sgarabotto, D., Palu, G.,
GA November 23, 2016. http://www.cdc.gov/zika/geo/united-states.html.
2016b. Infection dynamics in a traveller with persistent shedding of Zika virus RNA
CDC, 2016c. Congenital Microcephaly Case Definitions. Centers for Disease Control and
in semen for six months after returning from Haiti to Italy, January 2016. Euro
Prevention, Atlanta, GA October 20, 2016. http://www.cdc.gov/zika/public-health-
Surveill. 21, 30316.
partners/microcephaly-case-definitions.html.
Bayer, A., Lennemann, N.J., Ouyang, Y., Bramley, J.C., Morosky, S., Marques Jr., E.T., Cherry,
CDC, 2016d. Outcomes of Pregnancies With Laboratory Evidence of Possible Zika Virus In-
S., Sadovsky, Y., Coyne, C.B., 2016. Type III interferons produced by human placental
fection in the United States, 2016. Centers for Disease Control and Prevention, Atlan-
trophoblasts confer protection against Zika virus infection. Cell Host Microbe 19,
ta, GA November 23, 2016. http://www.cdc.gov/zika/geo/pregnancy-outcomes.html.
705–712.
60 B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64

CDC, 2016e. Potential Range in US. Centers for Disease Control and Prevention, Atlanta, infection in a Belgian traveler returning from Guatemala, and the diagnostic chal-
GA August 2, 2016. http://www.cdc.gov/zika/vector/range.html. lenges of imported cases into Europe. J. Clin. Virol. 80, 8–11.
CDC, 2016f. Zika Cases Reported in the United States. Centers for Disease Control and Pre- Deckard, D.T., Chung, W.M., Brooks, J.T., Smith, J.C., Woldai, S., Hennessey, M., Kwit, N.,
vention, Atlanta, GA November 23, 2016. http://www.cdc.gov/zika/intheus/maps- Mead, P., 2016. Male-to-male sexual transmission of Zika virus — Texas, January
zika-us.html. 2016. MMWR Morb. Mortal. Wkly. Rep. 65, 372–374.
CDC, 2016g. Zika Virus. Centers for Disease Control and Prevention, Atlanta, GA December Diagne, C.T., Diallo, D., Faye, O., Ba, Y., Faye, O., Gaye, A., Dia, I., Faye, O., Weaver, S.C., Sall,
5, 2016. http://www.cdc.gov/zika/index.html. A.A., Diallo, M., 2015. Potential of selected Senegalese Aedes spp. mosquitoes (Dip-
Cerbino-Neto, J., Mesquita, E.C., Souza, T.M., Parreira, V., Wittlin, B.B., Durovni, B., Lemos, tera: Culicidae) to transmit Zika virus. BMC Infect. Dis. 15, 492.
M.C., Vizzoni, A., Bispo de Filippis, A.M., Sampaio, S.A., Goncalves Bde, S., Bozza, F.A., Diallo, D., Sall, A.A., Diagne, C.T., Faye, O., Faye, O., Ba, Y., Hanley, K.A., Buenemann, M.,
2016. Clinical manifestations of Zika virus infection, Rio de Janeiro, Brazil, 2015. Weaver, S.C., Diallo, M., 2014. Zika virus emergence in mosquitoes in southeastern
Emerg. Infect. Dis. 22, 1318–1320. Senegal, 2011. PLoS One 9, e109442.
Chan, J.F., Zhang, A.J., Chan, C.C., Yip, C.C., Mak, W.W., Zhu, H., Poon, V.K., Tee, K.M., Zhu, Z., Diaz-Menendez, M., de la Calle-Prieto, F., Montero, D., Antolin, E., Vazquez, A., Arsuaga, M.,
Cai, J.P., Tsang, J.O., Chik, K.K., Yin, F., Chan, K.H., Kok, K.H., Jin, D.Y., Au-Yeung, R.K., Trigo, E., Garcia-Bujalance, S., de la Calle, M., Sanchez Seco, P., de Ory, F., Arribas, J.R.,
Yuen, K.Y., 2016. Zika virus infection in dexamethasone-immunosuppressed mice 2016. Grupo de Trabajo Multidisciplinar del Hospital La Paz-Carlos III en Enfermedad
demonstrating disseminated infection with multi-organ involvement including por Virus Zika, Initial experience with imported Zika virus infection in Spain. Enferm.
orchitis effectively treated by recombinant type I interferons. EBioMedicine 14, Infecc. Microbiol. Clin. http://dx.doi.org/10.31016/j.eimc.32016.30208.30003 pii:
112–122. S0213-0005X(0216)30257-30259. (In Press).
Chen, J.C., Wang, Z., Huang, H., Weitz, S.H., Wang, A., Qiu, X., Baumeister, M.A., Uzgiris, A., Dick, G.W., 1952. Zika virus. II. Pathogenicity and physical properties. Trans. R. Soc. Trop.
2016. Infection of human uterine fibroblasts by Zika virus in vitro: implications for Med. Hyg. 46, 521–534.
viral transmission in women. Int. J. Infect. Dis. 51, 139–140. Dick, G.W., 1953. Epidemiological notes on some viruses isolated in Uganda; yellow fever,
Chen, Y., Maguire, T., Hileman, R.E., Fromm, J.R., Esko, J.D., Linhardt, R.J., Marks, R.M., 1997. Rift Valley fever, Bwamba fever, West Nile, Mengo, Semliki forest, Bunyamwera,
Dengue virus infectivity depends on envelope protein binding to target cell heparan Ntaya, Uganda S and Zika viruses. Trans. R. Soc. Trop. Med. Hyg. 47, 13–48.
sulfate. Nat. Med. 3, 866–871. Dick, G.W., Kitchen, S.F., Haddow, A.J., 1952. Zika virus. I. Isolations and serological spec-
Chouin-Carneiro, T., Vega-Rua, A., Vazeille, M., Yebakima, A., Girod, R., Goindin, D., ificity. Trans. R. Soc. Trop. Med. Hyg. 46, 509–520.
Dupont-Rouzeyrol, M., Lourenco-de-Oliveira, R., Failloux, A.B., 2016. Differential sus- Driggers, R.W., Ho, C.Y., Korhonen, E.M., Kuivanen, S., Jaaskelainen, A.J., Smura, T.,
ceptibilities of Aedes aegypti and Aedes albopictus from the Americas to Zika virus. Rosenberg, A., Hill, D.A., DeBiasi, R.L., Vezina, G., Timofeev, J., Rodriguez, F.J.,
PLoS Negl. Trop. Dis. 10, e0004543. Levanov, L., Razak, J., Iyengar, P., Hennenfent, A., Kennedy, R., Lanciotti, R., du
Chraibi, S., Najioullah, F., Bourdin, C., Pegliasco, J., Deligny, C., Resiere, D., Meniane, J.C., Plessis, A., Vapalahti, O., 2016. Zika virus infection with prolonged maternal viremia
2016. Two cases of thrombocytopenic purpura at onset of Zika virus infection. and fetal brain abnormalities. N. Engl. J. Med. 374, 2142–2151.
J. Clin. Virol. 83, 61–62. Duffy, M.R., Chen, T.H., Hancock, W.T., Powers, A.M., Kool, J.L., Lanciotti, R.S., Pretrick, M.,
Clyde, K., Kyle, J.L., Harris, E., 2006. Recent advances in deciphering viral and host deter- Marfel, M., Holzbauer, S., Dubray, C., Guillaumot, L., Griggs, A., Bel, M., Lambert, A.J.,
minants of dengue virus replication and pathogenesis. J. Virol. 80, 11418–11431. Laven, J., Kosoy, O., Panella, A., Biggerstaff, B.J., Fischer, M., Hayes, E.B., 2009. Zika
Coloma, J., Jain, R., Rajashankar, K.R., Garcia-Sastre, A., Aggarwal, A.K., 2016. Structures of virus outbreak on Yap Island, Federated States of Micronesia. N. Engl. J. Med. 360,
NS5 methyltransferase from Zika virus. Cell Rep. 16, 3097–3102. 2536–2543.
Cordeiro, M.T., Pena, L.J., Brito, C.A., Gil, L.H., Marques, E.T., 2016. Positive IgM for Zika Duijster, J.W., Goorhuis, A., van Genderen, P.J., Visser, L.G., Koopmans, M.P., Reimerink,
virus in the cerebrospinal fluid of 30 neonates with microcephaly in Brazil. Lancet J.H., Grobusch, M.P., van der Eijk, A.A., van den Kerkhof, J.H., Reusken, C.B., Hahne,
387, 1811–1812. S.J., Dutch ZIKV Study Team, 2016. Zika virus infection in 18 travellers returning
Cornet, M., Robin, Y., Adam, C., Valade, M., Calvo, M.A., 1979a. Comparison between ex- from Surinam and the Dominican Republic, the Netherlands, November 2015–
perimental transmission of yellow fever and Zika viruses in Aedes aegypti. Cah. March 2016. Infection 44, 797–802.
ORSTOM Ser. Ent. Med. Parasitol. 17, 47–53. Dupont-Rouzeyrol, M., Biron, A., O'Connor, O., Huguon, E., Descloux, E., 2016. Infectious
Cornet, M., Robin, Y., Chateau, R., Heme, G., Adam, C., 1979b. Isolement d'arbovirus au Zika viral particles in breastmilk. Lancet 387, 1051.
Senegal oriental a partir de moustiques (1972–1977) et note surl'epidemiologie des Dupont-Rouzeyrol, M., O'Connor, O., Calvez, E., Daures, M., John, M., Grangeon, J.P.,
virus transmis par les Aedes, en particulier du virus amaril. ORSTOM Entomol. Med. Gourinat, A.C., 2015. Co-infection with Zika and dengue viruses in 2 patients, New
Parasitol. 17, 149–163. Caledonia, 2014. Emerg. Infect. Dis. 21, 381–382.
Cox, B.D., Stanton, R.A., Schinazi, R.F., 2015. Predicting Zika virus structural biology: chal- ECDC, 2014. Rapid Risk Assessment: Zika Virus Infection Outbreak, French Polynesia.
lenges and opportunities for intervention. Antivir. Chem. Chemother. 24, 118–126. European Centre for Disease Prevention and Control, Stockholm, Sweden February
Creange, A., 2016. Guillain–Barre syndrome: 100 years on. Rev. Neurol. 172, 770–774. 14, 2014. http://ecdc.europa.eu/en/publications/Publications/Zika-virus-French-
Cugola, F.R., Fernandes, I.R., Russo, F.B., Freitas, B.C., Dias, J.L., Guimaraes, K.P., Benazzato, Polynesia-rapid-risk-assessment.pdf.
C., Almeida, N., Pignatari, G.C., Romero, S., Polonio, C.M., Cunha, I., Freitas, C.L., ECDC, 2016a. Current Zika Transmission. European Centre for Disease Prevention and
Brandao, W.N., Rossato, C., Andrade, D.G., Faria Dde, P., Garcez, A.T., Buchpigel, C.A., Control, Stockholm, Sweden November 23, 2016. http://ecdc.europa.eu/en/
Braconi, C.T., Mendes, E., Sall, A.A., Zanotto, P.M., Peron, J.P., Muotri, A.R., Beltrao- healthtopics/zika_virus_infection/zika-outbreak/pages/zika-countries-with-
Braga, P.C., 2016. The Brazilian Zika virus strain causes birth defects in experimental transmission.aspx.
models. Nature 534, 267–271. ECDC, 2016b. Main Diagnostic Morphological Characters for Adults of IMS. European Cen-
Cunha, M.S., Esposito, D.L., Rocco, I.M., Maeda, A.Y., Vasami, F.G., Nogueira, J.S., de Souza, tre for Disease Prevention and Control, Stockholm, Sweden December 2, 2016. http://
R.P., Suzuki, A., Addas-Carvalho, M., Barjas-Castro Mde, L., Resende, M.R., Stucchi, ecdc.europa.eu/en/healthtopics/vectors/surveillance-invasive-mosquitoes/Docu-
R.S., Boin Ide, F., Katz, G., Angerami, R.N., da Fonseca, B.A., 2016. First complete ge- ments/mosquito-identification-methods-table-1.pdf.
nome sequence of Zika virus (Flaviviridae, Flavivirus) from an autochthonous trans- Ellison, D.W., Ladner, J.T., Buathong, R., Alera, M.T., Wiley, M.R., Hermann, L.,
mission in Brazil. Genome Announc. 4, e00032-16. Rutvisuttinunt, W., Klungthong, C., Chinnawirotpisan, P., Manasatienkij, W.,
Dahlgren, L., Wilson, R.D., 2001. Prenatally diagnosed microcephaly: a review of etiolo- Melendrez, M.C., Maljkovic Berry, I., Thaisomboonsuk, B., Ong-Ajchaowlerd, P.,
gies. Fetal Diagn. Ther. 16, 323–326. Kaneechit, W., Velasco, J.M., Tac-An, I.A., Villa, D., Lago, C.B., Roque Jr., V.G., Plipat, T.,
Dang, J., Tiwari, S.K., Lichinchi, G., Qin, Y., Patil, V.S., Eroshkin, A.M., Rana, T.M., 2016. Zika Nisalak, A., Srikiatkhachorn, A., Fernandez, S., Yoon, I.K., Haddow, A.D., Palacios, G.F.,
virus depletes neural progenitors in human cerebral organoids through activation of Jarman, R.G., Macareo, L.R., 2016. Complete genome sequences of Zika virus strains
the innate immune receptor TLR3. Cell Stem Cell 19, 258–265. isolated from the blood of patients in Thailand in 2014 and the Philippines in 2012.
Darwish, M.A., Hoogstraal, H., Roberts, T.J., Ahmed, I.P., Omar, F., 1983. A sero- Genome Announc. 4, e00359-16.
epidemiological survey for certain arboviruses (Togaviridae) in Pakistan. Trans. R. Enfissi, A., Codrington, J., Roosblad, J., Kazanji, M., Rousset, D., 2016. Zika virus genome
Soc. Trop. Med. Hyg. 77, 442–445. from the Americas. Lancet 387, 227–228.
Davis, C.W., Nguyen, H.Y., Hanna, S.L., Sanchez, M.D., Doms, R.W., Pierson, T.C., 2006. West Fagbami, A., 1977. Epidemiological investigations on arbovirus infections at Igbo-Ora,
Nile virus discriminates between DC-SIGN and DC-SIGNR for cellular attachment and Nigeria. Trop. Geogr. Med. 29, 187–191.
infection. J. Virol. 80, 1290–1301. Fagbami, A.H., 1979. Zika virus infections in Nigeria: virological and seroepidemiological
de Araujo, T.V., Rodrigues, L.C., de Alencar Ximenes, R.A., de Barros Miranda-Filho, D., investigations in Oyo State. J. Hyg. 83, 213–219.
Montarroyos, U.R., de Melo, A.P., Valongueiro, S., de Albuquerque, M.F., Souza, W.V., Faria, N.R., Azevedo Rdo, S., Kraemer, M.U., Souza, R., Cunha, M.S., Hill, S.C., Theze, J.,
Braga, C., Filho, S.P., Cordeiro, M.T., Vazquez, E., Di Cavalcanti Souza Cruz, D., Bonsall, M.B., Bowden, T.A., Rissanen, I., Rocco, I.M., Nogueira, J.S., Maeda, A.Y.,
Henriques, C.M., Bezerra, L.C., da Silva Castanha, P.M., Dhalia, R., Marques-Junior, Vasami, F.G., Macedo, F.L., Suzuki, A., Rodrigues, S.G., Cruz, A.C., Nunes, B.T.,
E.T., Martelli, C.M., Investigators from the Microcephaly Epidemic Research Group, Medeiros, D.B., Rodrigues, D.S., Nunes Queiroz, A.L., da Silva, E.V., Henriques, D.F.,
Brazilian Ministry of Health, Pan American Health Organization, Instituto de Medicina Travassos da Rosa, E.S., de Oliveira, C.S., Martins, L.C., Vasconcelos, H.B., Casseb, L.M.,
Integral Professor Fernando Figueira, State Health Department of Pernambuco, 2016. Simith Dde, B., Messina, J.P., Abade, L., Lourenco, J., Carlos Junior Alcantara, L., de
Association between Zika virus infection and microcephaly in Brazil, January to May, Lima, M.M., Giovanetti, M., Hay, S.I., de Oliveira, R.S., Lemos Pda, S., de Oliveira, L.F.,
2016: preliminary report of a case–control study. Lancet Infect. Dis. 16, 1356–1363. de Lima, C.P., da Silva, S.P., de Vasconcelos, J.M., Franco, L., Cardoso, J.F., Vianez-Junior,
de Paula Freitas, B., de Oliveira Dias, J.R., Prazeres, J., Sacramento, G.A., Ko, A.I., Maia, M., J.L., Mir, D., Bello, G., Delatorre, E., Khan, K., Creatore, M., Coelho, G.E., de Oliveira,
Belfort Jr., R., 2016. Ocular findings in infants with microcephaly associated with pre- W.K., Tesh, R., Pybus, O.G., Nunes, M.R., Vasconcelos, P.F., 2016. Zika virus in the
sumed Zika virus congenital infection in Salvador, Brazil. JAMA Ophthalmol. 134, Americas: early epidemiological and genetic findings. Science 352, 345–349.
529–535. Fauci, A.S., Morens, D.M., 2016. Zika virus in the Americas — yet another arbovirus threat.
De Santis, M., Cavaliere, A.F., Straface, G., Caruso, A., 2006. Rubella infection in pregnancy. N. Engl. J. Med. 374, 601–604.
Reprod. Toxicol. 21, 390–398. Faye, O., Faye, O., Diallo, D., Diallo, M., Weidmann, M., Sall, A.A., 2013. Quantitative real-
De Smet, B., Van den Bossche, D., van de Werve, C., Mairesse, J., Schmidt-Chanasit, J., time PCR detection of Zika virus and evaluation with field-caught mosquitoes.
Michiels, J., Arien, K.K., Van Esbroeck, M., Cnops, L., 2016. Confirmed Zika virus Virol. J. 10, 311.
B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64 61

Faye, O., Freire, C.C., Iamarino, A., Faye, O., de Oliveira, J.V., Diallo, M., Zanotto, P.M., Sall, Huang, W.C., Abraham, R., Shim, B.S., Choe, H., Page, D.T., 2016. Zika virus infection during
A.A., 2014. Molecular evolution of Zika virus during its emergence in the 20th centu- the period of maximal brain growth causes microcephaly and corticospinal neuron
ry. PLoS Negl. Trop. Dis. 8, e2636. apoptosis in wild type mice. Sci. Rep. 6, 34793.
Fonseca, K., Meatherall, B., Zarra, D., Drebot, M., MacDonald, J., Pabbaraju, K., Wong, S., Jain, R., Coloma, J., Garcia-Sastre, A., Aggarwal, A.K., 2016. Structure of the NS3 helicase
Webster, P., Lindsay, R., Tellier, R., 2014. First case of Zika virus infection in a returning from Zika virus. Nat. Struct. Mol. Biol. 23, 752–754.
Canadian traveler. Am. J. Trop. Med. Hyg. 91, 1035–1038. Jan, C., Languillat, G., Renaudet, J., Robin, Y., 1978. A serological survey of arboviruses in
Foy, B.D., Kobylinski, K.C., Chilson Foy, J.L., Blitvich, B.J., Travassos da Rosa, A., Haddow, Gabon. Bull. Soc. Pathol. Exot. Filiales 71, 140–146.
A.D., Lanciotti, R.S., Tesh, R.B., 2011. Probable non-vector-borne transmission of Zika Jouannic, J.M., Friszer, S., Leparc-Goffart, I., Garel, C., Eyrolle-Guignot, D., 2016. Zika virus
virus, Colorado, USA. Emerg. Infect. Dis. 17, 880–882. infection in French Polynesia. Lancet 387, 1051–1052.
Frank, C., Cadar, D., Schlaphof, A., Neddersen, N., Gunther, S., Schmidt-Chanasit, J., Tappe, Karimi, O., Goorhuis, A., Schinkel, J., Codrington, J., Vreden, S.G., Vermaat, J.S., Stijnis, C.,
D., 2016. Sexual transmission of Zika virus in Germany, April 2016. Euro Surveill. 21, Grobusch, M.P., 2016. Thrombocytopenia and subcutaneous bleedings in a patient
30252. with Zika virus infection. Lancet 387, 939–940.
Freour, T., Mirallie, S., Hubert, B., Splingart, C., Barriere, P., Maquart, M., Leparc-Goffart, I., Kaufmann, B., Rossmann, M.G., 2011. Molecular mechanisms involved in the early steps of
2016. Sexual transmission of Zika virus in an entirely asymptomatic couple returning flavivirus cell entry. Microbes Infect. 13, 1–9.
from a Zika epidemic area, France, April 2016. Euro Surveill. 21, 30254. Kim, J.K., Kim, J.M., Song, B.H., Yun, S.I., Yun, G.N., Byun, S.J., Lee, Y.M., 2015. Profiling of
Garcez, P.P., Loiola, E.C., Madeiro da Costa, R., Higa, L.M., Trindade, P., Delvecchio, R., viral proteins expressed from the genomic RNA of Japanese encephalitis virus using
Nascimento, J.M., Brindeiro, R., Tanuri, A., Rehen, S.K., 2016. Zika virus impairs growth a panel of 15 region-specific polyclonal rabbit antisera: implications for viral gene ex-
in human neurospheres and brain organoids. Science 352, 816–818. pression. PLoS One 10, e0124318.
Gatherer, D., Kohl, A., 2016. Zika virus: a previously slow pandemic spreads rapidly Klase, Z.A., Khakhina, S., Schneider Ade, B., Callahan, M.V., Glasspool-Malone, J., Malone, R.,
through the Americas. J. Gen. Virol. 97, 269–273. 2016. Zika fetal neuropathogenesis: etiology of a viral syndrome. PLoS Negl. Trop. Dis.
Gaunt, M.W., Sall, A.A., de Lamballerie, X., Falconar, A.K., Dzhivanian, T.I., Gould, E.A., 2001. 10, e0004877.
Phylogenetic relationships of flaviviruses correlate with their epidemiology, disease Korhonen, E.M., Huhtamo, E., Smura, T., Kallio-Kokko, H., Raassina, M., Vapalahti, O., 2016.
association and biogeography. J. Gen. Virol. 82, 1867–1876. Zika virus infection in a traveller returning from the Maldives, June 2015. Euro
GC, 2016. Surveillance of Zika Virus. Government of Canada November 17, 2016. Surveill. 21, 30107.
http://www.healthycanadians.gc.ca/diseases-conditions-maladies-affections/ Kostyuchenko, V.A., Lim, E.X., Zhang, S., Fibriansah, G., Ng, T.S., Ooi, J.S., Shi, J., Lok, S.M.,
disease-maladie/zika-virus/surveillance-eng.php?id=zikacases%20-%20s1. 2016. Structure of the thermally stable Zika virus. Nature 533, 425–428.
Geser, A., Henderson, B.E., Christensen, S., 1970. A multipurpose serological survey in Kraemer, M.U., Sinka, M.E., Duda, K.A., Mylne, A.Q., Shearer, F.M., Barker, C.M., Moore, C.G.,
Kenya. 2. Results of arbovirus serological tests. Bull. World Health Organ. 43, Carvalho, R.G., Coelho, G.E., Van Bortel, W., Hendrickx, G., Schaffner, F., Elyazar, I.R.,
539–552. Teng, H.J., Brady, O.J., Messina, J.P., Pigott, D.M., Scott, T.W., Smith, D.L., Wint, G.R.,
Gillespie, L.K., Hoenen, A., Morgan, G., Mackenzie, J.M., 2010. The endoplasmic reticulum Golding, N., Hay, S.I., 2015. The global distribution of the arbovirus vectors Aedes
provides the membrane platform for biogenesis of the flavivirus replication complex. aegypti and Ae. albopictus. Elife 4, e08347.
J. Virol. 84, 10438–10447. Kramer, L.D., Ebel, G.D., 2003. Dynamics of flavivirus infection in mosquitoes. Adv. Virus
Giovanetti, M., Faria, N.R., Nunes, M.R., de Vasconcelos, J.M., Lourenco, J., Rodrigues, Res. 60, 187–232.
S.G., Vianez Jr., J.L., da Silva, S.P., Lemos, P.S., Tavares, F.N., Martin, D.P., do Kuno, G., Chang, G.J., 2007. Full-length sequencing and genomic characterization of
Rosario, M.S., Siqueira, I.C., Ciccozzi, M., Pybus, O.G., de Oliveira, T., Alcantara Jr., Bagaza, Kedougou, and Zika viruses. Arch. Virol. 152, 687–696.
L.C., 2016. Zika virus complete genome from Salvador, Bahia, Brazil. Infect. Genet. Kutsuna, S., Kato, Y., Takasaki, T., Moi, M., Kotaki, A., Uemura, H., Matono, T., Fujiya, Y.,
Evol. 41, 142–145. Mawatari, M., Takeshita, N., Hayakawa, K., Kanagawa, S., Ohmagari, N., 2014. Two
Goodfellow, J.A., Willison, H.J., 2016. Guillain–Barre syndrome: a century of progress. Nat. cases of Zika fever imported from French Polynesia to Japan, December 2013 to
Rev. Neurol. 12, 723–731. January 2014. Euro Surveill. 19, 20683.
Gourinat, A.C., O'Connor, O., Calvez, E., Goarant, C., Dupont-Rouzeyrol, M., 2015. Detection Kwong, J.C., Druce, J.D., Leder, K., 2013. Zika virus infection acquired during brief travel to
of Zika virus in urine. Emerg. Infect. Dis. 21, 84–86. Indonesia. Am. J. Trop. Med. Hyg. 89, 516–517.
Govero, J., Esakky, P., Scheaffer, S.M., Fernandez, E., Drury, A., Platt, D.J., Gorman, M.J., Ladner, J.T., Wiley, M.R., Prieto, K., Yasuda, C.Y., Nagle, E., Kasper, M.R., Reyes, D., Vasilakis,
Richner, J.M., Caine, E.A., Salazar, V., Moley, K.H., Diamond, M.S., 2016. Zika virus in- N., Heang, V., Weaver, S.C., Haddow, A., Tesh, R.B., Sovann, L., Palacios, G., 2016. Com-
fection damages the testes in mice. Nature 540, 438–442. plete genome sequences of five Zika virus isolates. Genome Announc. 4, e00377-16.
Grard, G., Caron, M., Mombo, I.M., Nkoghe, D., Mboui Ondo, S., Jiolle, D., Fontenille, D., Lanciotti, R.S., Kosoy, O.L., Laven, J.J., Velez, J.O., Lambert, A.J., Johnson, A.J., Stanfield, S.M.,
Paupy, C., Leroy, E.M., 2014. Zika virus in Gabon (Central Africa) — 2007: a new threat Duffy, M.R., 2008. Genetic and serologic properties of Zika virus associated with an
from Aedes albopictus? PLoS Negl. Trop. Dis. 8, e2681. epidemic, Yap State, Micronesia, 2007. Emerg. Infect. Dis. 14, 1232–1239.
Gubler, D.J., 2002. The global emergence/resurgence of arboviral diseases as public health Lanciotti, R.S., Lambert, A.J., Holodniy, M., Saavedra, S., Signor Ldel, C., 2016. Phylogeny of
problems. Arch. Med. Res. 33, 330–342. Zika virus in western hemisphere, 2015. Emerg. Infect. Dis. 22, 933–935.
Gubler, D.J., Kuno, G., Markoff, L., 2007. Flaviviruses. In: Knipe, D.M., Howley, P.M., Griffin, Lazear, H.M., Diamond, M.S., 2016. Zika virus: new clinical syndromes and its emergence
D.E., Lamb, R.A., Martin, M.A., Roizman, B., Straus, S.E. (Eds.), Fields Virology, fifth ed. in the western hemisphere. J. Virol. 90, 4864–4875.
Lippincott Williams & Wilkins Publishers, Philadelphia, PA, pp. 1153–1252. Leal, M.C., Muniz, L.F., Caldas Neto, S.D., van der Linden, V., Ramos, R.C., 2016a. Sensori-
Gubler, D.J., Nalim, S., Tan, R., Saipan, H., Sulianti Saroso, J., 1979. Variation in susceptibility neural hearing loss in a case of congenital Zika virus. Braz. J. Otorhinolaryngol.
to oral infection with dengue viruses among geographic strains of Aedes aegypti. Am. http://dx.doi.org/10.31016/j.bjorl.32016.30106.30001 pii: S1808-8694(1816)30127-
J. Trop. Med. Hyg. 28, 1045–1052. 30126. (In Press).
Gyurech, D., Schilling, J., Schmidt-Chanasit, J., Cassinotti, P., Kaeppeli, F., Dobec, M., 2016. Leal, M.C., Muniz, L.F., Ferreira, T.S., Santos, C.M., Almeida, L.C., Van Der Linden, V., Ramos,
False positive dengue NS1 antigen test in a traveller with an acute Zika virus infection R.C., Rodrigues, L.C., Neto, S.S., 2016b. Hearing loss in infants with microcephaly and
imported into Switzerland. Swiss Med. Wkly. 146, w14296. evidence of congenital Zika virus infection — Brazil, November 2015–May 2016.
Haddow, A.D., Schuh, A.J., Yasuda, C.Y., Kasper, M.R., Heang, V., Huy, R., Guzman, H., Tesh, MMWR Morb. Mortal. Wkly. Rep. 65, 917–919.
R.B., Weaver, S.C., 2012. Genetic characterization of Zika virus strains: geographic ex- Ledermann, J.P., Guillaumot, L., Yug, L., Saweyog, S.C., Tided, M., Machieng, P., Pretrick, M.,
pansion of the Asian lineage. PLoS Negl. Trop. Dis. 6, e1477. Marfel, M., Griggs, A., Bel, M., Duffy, M.R., Hancock, W.T., Ho-Chen, T., Powers, A.M.,
Haddow, A.J., Williams, M.C., Woodall, J.P., Simpson, D.I., Goma, L.K., 1964. Twelve isola- 2014. Aedes hensilli as a potential vector of chikungunya and Zika viruses. PLoS
tions of Zika virus from Aedes (Stegomyia) Africanus (Theobald) taken in and Negl. Trop. Dis. 8, e3188.
above a Uganda forest. Bull. World Health Organ. 31, 57–69. Lednicky, J., Beau De Rochars, V.M., El Badry, M., Loeb, J., Telisma, T., Chavannes, S., Anilis,
Hammon, W.M., Schrack Jr., W.D., Sather, G.E., 1958. Serological survey for a arthropod- G., Cella, E., Ciccozzi, M., Rashid, M., Okech, B., Salemi, M., Morris Jr., J.G., 2016. Zika
borne virus infections in the Philippines. Am. J. Trop. Med. Hyg. 7, 323–328. virus outbreak in Haiti in 2014: molecular and clinical data. PLoS Negl. Trop. Dis.
Harrison, S.C., 2008. Viral membrane fusion. Nat. Struct. Mol. Biol. 15, 690–698. 10, e0004687.
Hayes, E.B., 2009. Zika virus outside Africa. Emerg. Infect. Dis. 15, 1347–1350. Lei, J., Hansen, G., Nitsche, C., Klein, C.D., Zhang, L., Hilgenfeld, R., 2016. Crystal structure of
Hazin, A.N., Poretti, A., Turchi Martelli, C.M., Huisman, T.A., Microcephaly Epidemic Zika virus NS2B-NS3 protease in complex with a boronate inhibitor. Science 353,
Research Group, Di Cavalcanti Souza Cruz, D., Tenorio, M., van der Linden, A., Pena, 503–505.
L.J., Brito, C., Gil, L.H., de Barros Miranda-Filho, D., Marques, E.T., Alves, J.G., 2016. Lessler, J., Chaisson, L.H., Kucirka, L.M., Bi, Q., Grantz, K., Salje, H., Carcelen, A.C., Ott, C.T.,
Computed tomographic findings in microcephaly associated with Zika virus. N. Sheffield, J.S., Ferguson, N.M., Cummings, D.A., Metcalf, C.J., Rodriguez-Barraquer, I.,
Engl. J. Med. 374, 2193–2195. 2016. Assessing the global threat from Zika virus. Science 353 (aaf8160).
Heang, V., Yasuda, C.Y., Sovann, L., Haddow, A.D., Travassos da Rosa, A.P., Tesh, R.B., Leung, G.H., Baird, R.W., Druce, J., Anstey, N.M., 2015. Zika virus infection in Australia fol-
Kasper, M.R., 2012. Zika virus infection, Cambodia, 2010. Emerg. Infect. Dis. 18, lowing a monkey bite in Indonesia. SE Asian J. Trop. Med. Public Health 46, 460–464.
349–351. Li, C., Xu, D., Ye, Q., Hong, S., Jiang, Y., Liu, X., Zhang, N., Shi, L., Qin, C.F., Xu, Z., 2016. Zika
Hennessey, M., Fischer, M., Staples, J.E., 2016. Zika virus spreads to new areas — region of virus disrupts neural progenitor development and leads to microcephaly in mice. Cell
the Americas, May 2015–January 2016. MMWR Morb. Mortal. Wkly. Rep. 65, 55–58. Stem Cell 19, 120–126.
Hills, S.L., Russell, K., Hennessey, M., Williams, C., Oster, A.M., Fischer, M., Mead, P., 2016. Li, M.I., Wong, P.S., Ng, L.C., Tan, C.H., 2012. Oral susceptibility of Singapore Aedes
Transmission of Zika virus through sexual contact with travelers to areas of ongoing (Stegomyia) aegypti (Linnaeus) to Zika virus. PLoS Negl. Trop. Dis. 6, e1792.
transmission — continental United States, 2016. MMWR Morb. Mortal. Wkly. Rep. 65, Lindenbach, B.D., Murray, C.L., Thiel, H.J., Rice, C.M., 2013. Flaviviridae. In: Knipe, D.M.,
215–216. Howley, P.M., Cohen, J.I., Griffin, D.E., Lamb, R.A., Martin, M.A., Racaniello, V.R.,
Hsu, N.Y., Ilnytska, O., Belov, G., Santiana, M., Chen, Y.H., Takvorian, P.M., Pau, C., van der Roizman, B. (Eds.), Fields Virology. Wolters Kluwer Health, Philadelphia, PA,
Schaar, H., Kaushik-Basu, N., Balla, T., Cameron, C.E., Ehrenfeld, E., van Kuppeveld, F.J., pp. 712–746.
Altan-Bonnet, N., 2010. Viral reorganization of the secretory pathway generates dis- Lindenbach, B.D., Thiel, H.J., Rice, C.M., 2007. Flaviviridae: the viruses and their replication.
tinct organelles for RNA replication. Cell 141, 799–811. In: Knipe, D.M., Howley, P.M., Griffin, D.E., Lamb, R.A., Martin, M.A., Roizman, B.,
62 B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64

Straus, S.E. (Eds.), Fields Virology, fifth ed. Lippincott Williams & Wilkins Publishers, Murray, K.O., Gorchakov, R., Carlson, A.R., Berry, R., Lai, L., Natrajan, M., Garcia, M.N.,
Philadelphia, PA, pp. 1101–1152. Correa, A., Patel, S.M., Aagaard, K., Mulligan, M.J., 2017. Prolonged detection of Zika
Liu, L., Wu, W., Zhao, X., Xiong, Y., Zhang, S., Liu, X., Qu, J., Li, J., Nei, K., Liang, M., Shu, Y., virus in vaginal secretions and whole blood. Emerg. Infect. Dis. 23, 99–101.
Hu, G., Ma, X., Li, D., 2016. Complete genome sequence of Zika virus from the first Musso, D., Baud, D., 2016. Zika virus: time to move from case reports to case control. Lan-
imported case in mainland China. Genome Announc. 4, e00291-16. cet Infect. Dis. 16, 620–621.
Lorenz, I.C., Allison, S.L., Heinz, F.X., Helenius, A., 2002. Folding and dimerization of tick- Musso, D., Cao-Lormeau, V.M., Gubler, D.J., 2015a. Zika virus: following the path of dengue
borne encephalitis virus envelope proteins prM and E in the endoplasmic reticulum. and chikungunya? Lancet 386, 243–244.
J. Virol. 76, 5480–5491. Musso, D., Gubler, D.J., 2016. Zika virus. Clin. Microbiol. Rev. 29, 487–524.
Ma, W., Li, S., Ma, S., Jia, L., Zhang, F., Zhang, Y., Zhang, J., Wong, G., Zhang, S., Lu, X., Liu, M., Musso, D., Nhan, T., Robin, E., Roche, C., Bierlaire, D., Zisou, K., Shan Yan, A., Cao-Lormeau,
Yan, J., Li, W., Qin, C., Han, D., Qin, C., Wang, N., Li, X., Gao, G.F., 2016. Zika virus causes V.M., Broult, J., 2014a. Potential for Zika virus transmission through blood transfusion
testis damage and leads to male infertility in mice. Cell 167, 1511–1524. demonstrated during an outbreak in French Polynesia, November 2013 to February
Mackenzie, J.M., Westaway, E.G., 2001. Assembly and maturation of the flavivirus Kunjin 2014. Euro Surveill. 19, 20761.
virus appear to occur in the rough endoplasmic reticulum and along the secretory Musso, D., Nilles, E.J., Cao-Lormeau, V.M., 2014b. Rapid spread of emerging Zika virus in
pathway, respectively. J. Virol. 75, 10787–10799. the Pacific area. Clin. Microbiol. Infect. 20, O595–O596.
Mackenzie, J.S., Gubler, D.J., Petersen, L.R., 2004. Emerging flaviviruses: the spread and re- Musso, D., Roche, C., Nhan, T.X., Robin, E., Teissier, A., Cao-Lormeau, V.M., 2015b. Detec-
surgence of Japanese encephalitis, West Nile and dengue viruses. Nat. Med. 10, tion of Zika virus in saliva. J. Clin. Virol. 68, 53–55.
S98–109. Musso, D., Roche, C., Robin, E., Nhan, T., Teissier, A., Cao-Lormeau, V.M., 2015c. Potential
Macnamara, F.N., 1954. Zika virus: a report on three cases of human infection during an sexual transmission of Zika virus. Emerg. Infect. Dis. 21, 359–361.
epidemic of jaundice in Nigeria. Trans. R. Soc. Trop. Med. Hyg. 48, 139–145. Navarro-Sanchez, E., Altmeyer, R., Amara, A., Schwartz, O., Fieschi, F., Virelizier, J.L.,
Magalhaes-Barbosa, M.C., Prata-Barbosa, A., Robaina, J.R., Raymundo, C.E., Lima-Setta, F., Arenzana-Seisdedos, F., Despres, P., 2003. Dendritic-cell-specific ICAM3-grabbing
Cunha, A.J., 2016. Trends of the microcephaly and Zika virus outbreak in Brazil, non-integrin is essential for the productive infection of human dendritic cells by
January–July 2016. Travel Med. Infect. Dis. 14, 458–463. mosquito-cell-derived dengue viruses. EMBO Rep. 4, 723–728.
Mallet, H.P., Vial, A.L., Musso, D., 2015. Bilan de l’épidémie à virus Zika en Polynésie Nicastri, E., Castilletti, C., Liuzzi, G., Iannetta, M., Capobianchi, M.R., Ippolito, G., 2016a. Per-
française, 2013–2014. Bulletin d'information sanitaires, épidémiologiques et sistent detection of Zika virus RNA in semen for six months after symptom onset in a
statistiques (BISES). May 2015. http://www.hygiene-publique.gov.pf/IMG/pdf/ traveller returning from Haiti to Italy, February 2016. Euro Surveill. 21, 30314.
no13_-_mai_2015_-_zika.pdf. Nicastri, E., Pisapia, R., Corpolongo, A., Fusco, F.M., Cicalini, S., Scognamiglio, P., Castilletti,
Mansuy, J.M., Dutertre, M., Mengelle, C., Fourcade, C., Marchou, B., Delobel, P., Izopet, J., C., Bordi, L., Di Caro, A., Capobianchi, M.R., Puro, V., Ippolito, G., 2016b. Three cases of
Martin-Blondel, G., 2016. Zika virus: high infectious viral load in semen, a new sexu- Zika virus imported in Italy: need for a clinical awareness and evidence-based knowl-
ally transmitted pathogen? Lancet Infect. Dis. 16, 405. edge. BMC Infect. Dis. 16, 669.
Marchette, N.J., Garcia, R., Rudnick, A., 1969. Isolation of Zika virus from Aedes aegypti Oehler, E., Watrin, L., Larre, P., Leparc-Goffart, I., Lastere, S., Valour, F., Baudouin, L., Mallet,
mosquitoes in Malaysia. Am. J. Trop. Med. Hyg. 18, 411–415. H., Musso, D., Ghawche, F., 2014. Zika virus infection complicated by Guillain-Barre
Maria, A.T., Maquart, M., Makinson, A., Flusin, O., Segondy, M., Leparc-Goffart, I., Le Moing, syndrome — case report, French Polynesia, December 2013. Euro Surveill. 19, 20720.
V., Foulongne, V., 2016. Zika virus infections in three travellers returning from South Oliveira Melo, A.S., Malinger, G., Ximenes, R., Szejnfeld, P.O., Alves Sampaio, S., Bispo de
America and the Caribbean respectively, to Montpellier, France, December 2015 to Filippis, A.M., 2016. Zika virus intrauterine infection causes fetal brain abnormality
January 2016. Euro Surveill. 21, 30131. and microcephaly: tip of the iceberg? Ultrasound Obstet. Gynecol. 47, 6–7.
Martines, R.B., Bhatnagar, J., Keating, M.K., Silva-Flannery, L., Muehlenbachs, A., Gary, J., Olson, J.G., Ksiazek, T.G., Gubler, D.J., Lubis, S.I., Simanjuntak, G., Lee, V.H., Nalim, S., Juslis,
Goldsmith, C., Hale, G., Ritter, J., Rollin, D., Shieh, W.J., Luz, K.G., Ramos, A.M., Davi, K., See, R., 1983. A survey for arboviral antibodies in sera of humans and animals in
H.P., Kleber de Oliveria, W., Lanciotti, R., Lambert, A., Zaki, S., 2016. Notes from the Lombok, Republic of Indonesia. Ann. Trop. Med. Parasitol. 77, 131–137.
field: evidence of Zika virus infection in brain and placental tissues from two congen- Olson, J.G., Ksiazek, T.G., Suhandiman, Triwibowo, 1981. Zika virus, a cause of fever in Cen-
itally infected newborns and two fetal losses — Brazil, 2015. MMWR Morb. Mortal. tral Java, Indonesia. Trans. R. Soc. Trop. Med. Hyg. 75, 389–393.
Wkly. Rep. 65, 159–160. Onorati, M., Li, Z., Liu, F., Sousa, A.M., Nakagawa, N., Li, M., Dell'Anno, M.T., Gulden, F.O.,
McCrae, A.W., Kirya, B.G., 1982. Yellow fever and Zika virus epizootics and enzootics in Pochareddy, S., Tebbenkamp, A.T., Han, W., Pletikos, M., Gao, T., Zhu, Y., Bichsel, C.,
Uganda. Trans. R. Soc. Trop. Med. Hyg. 76, 552–562. Varela, L., Szigeti-Buck, K., Lisgo, S., Zhang, Y., Testen, A., Gao, X.B., Mlakar, J.,
Meaney-Delman, D., Hills, S.L., Williams, C., Galang, R.R., Iyengar, P., Hennenfent, A.K., Popovic, M., Flamand, M., Strittmatter, S.M., Kaczmarek, L.K., Anton, E.S., Horvath,
Rabe, I.B., Panella, A., Oduyebo, T., Honein, M.A., Zaki, S., Lindsey, N., Lehman, J.A., T.L., Lindenbach, B.D., Sestan, N., 2016. Zika virus disrupts phospho-TBK1 localization
Kwit, N., Bertolli, J., Ellington, S., Igbinosa, I., Minta, A.A., Petersen, E.E., Mead, P., and mitosis in human neuroepithelial stem cells and radial glia. Cell Rep. 16,
Rasmussen, S.A., Jamieson, D.J., 2016. Zika virus infection among U.S. pregnant 2576–2592.
travelers — August 2015–February 2016. MMWR Morb. Mortal. Wkly. Rep. 65, PAHO/WHO, 2016a. Zika — Epidemiological Update. Pan American Health Organization/
211–214. World Health Organization, Washington, D.C. November 17, 2016. http://www.
Mecharles, S., Herrmann, C., Poullain, P., Tran, T.H., Deschamps, N., Mathon, G., Landais, A., paho.org/hq/index.php?option=com_docman&task=doc_view&Itemid=270&gid=
Breurec, S., Lannuzel, A., 2016. Acute myelitis due to Zika virus infection. Lancet 387, 36943&lang=en.
1481. PAHO/WHO, 2016b. Zika Suspected and Confirmed Cases Reported by Countries and Ter-
Messina, J.P., Kraemer, M.U., Brady, O.J., Pigott, D.M., Shearer, F.M., Weiss, D.J., Golding, N., ritories in the Americas (Cumulative Cases), 2015–2016. Pan American Health Orga-
Ruktanonchai, C.W., Gething, P.W., Cohn, E., Brownstein, J.S., Khan, K., Tatem, A.J., nization/World Health Organization, Washington, D.C. November 17, 2016. http://
Jaenisch, T., Murray, C.J., Marinho, F., Scott, T.W., Hay, S.I., 2016. Mapping global envi- www.paho.org/hq/index.php?option=com_docman&task=doc_view&Itemid=
ronmental suitability for Zika virus. Elife 5, e15272. 270&gid=36937&lang=en.
Microcephaly Epidemic Research Group, 2016. Microcephaly in infants, Pernambuco Panchaud, A., Stojanov, M., Ammerdorffer, A., Vouga, M., Baud, D., 2016. Emerging role of
State, Brazil, 2015. Emerg. Infect. Dis. 22, 1090–1093. Zika virus in adverse fetal and neonatal outcomes. Clin. Microbiol. Rev. 29, 659–694.
Miller, B.R., Monath, T.P., Tabachnick, W.J., Ezike, V.I., 1989. Epidemic yellow fever caused Passemard, S., Kaindl, A.M., Verloes, A., 2013. Microcephaly. Handb. Clin. Neurol. 111,
by an incompetent mosquito vector. Trop. Med. Parasitol. 40, 396–399. 129–141.
Miner, J.J., Sene, A., Richner, J.M., Smith, A.M., Santeford, A., Ban, N., Weger-Lucarelli, J., Paupy, C., Delatte, H., Bagny, L., Corbel, V., Fontenille, D., 2009. Aedes albopictus, an arbo-
Manzella, F., Ruckert, C., Govero, J., Noguchi, K.K., Ebel, G.D., Diamond, M.S., Apte, virus vector: from the darkness to the light. Microbes Infect. 11, 1177–1185.
R.S., 2016. Zika virus infection in mice causes panuveitis with shedding of virus in Perera-Lecoin, M., Meertens, L., Carnec, X., Amara, A., 2013. Flavivirus entry receptors: an
tears. Cell Rep. 16, 3208–3218. update. Viruses 6, 69–88.
Miranda-Filho Dde, B., Martelli, C.M., Ximenes, R.A., Araujo, T.V., Rocha, M.A., Ramos, R.C., Perkasa, A., Yudhaputri, F., Haryanto, S., Hayati, R.F., Ma'roef, C.N., Antonjaya, U., Yohan, B.,
Dhalia, R., Franca, R.F., Marques Junior, E.T., Rodrigues, L.C., 2016. Initial description of Myint, K.S., Ledermann, J.P., Rosenberg, R., Powers, A.M., Sasmono, R.T., 2016. Isola-
the presumed congenital Zika syndrome. Am. J. Public Health 106, 598–600. tion of Zika virus from febrile patient, Indonesia. Emerg. Infect. Dis. 22, 924–925.
Mlakar, J., Korva, M., Tul, N., Popovic, M., Poljsak-Prijatelj, M., Mraz, J., Kolenc, M., Resman Petersen, L.R., Jamieson, D.J., Powers, A.M., Honein, M.A., 2016. Zika virus. N. Engl. J. Med.
Rus, K., Vesnaver Vipotnik, T., Fabjan Vodusek, V., Vizjak, A., Pizem, J., Petrovec, M., 374, 1552–1563.
Avsic Zupanc, T., 2016. Zika virus associated with microcephaly. N. Engl. J. Med. Phoo, W.W., Li, Y., Zhang, Z., Lee, M.Y., Loh, Y.R., Tan, Y.B., Ng, E.Y., Lescar, J., Kang, C., Luo,
374, 951–958. D., 2016. Structure of the NS2B-NS3 protease from Zika virus after self-cleavage. Nat.
Monaghan, A.J., Morin, C.W., Steinhoff, D.F., Wilhelmi, O., Hayden, M., Quattrochi, D.A., Commun. 7, 13410.
Reiskind, M., Lloyd, A.L., Smith, K., Schmidt, C.A., Scalf, P.E., Ernst, K., 2016. On the sea- Pierson, T.C., Diamond, M.S., 2012. Degrees of maturity: the complex structure and biolo-
sonal occurrence and abundance of the Zika virus vector mosquito Aedes aegypti in gy of flaviviruses. Curr. Opin. Virol. 2, 168–175.
the contiguous United States. PLoS Curr. 8. http://dx.doi.org/10.46791371/currents. Pierson, T.C., Kielian, M., 2013. Flaviviruses: braking the entering. Curr. Opin. Virol. 3,
outbreaks.46798650dfc46798677f46798675fc46798663e46798677d46798677 3–12.
da46798563da46798676 (pii: ecurrents.outbreaks.50dfc57f46798675fc46798663e Plosa, E.J., Esbenshade, J.C., Fuller, M.P., Weitkamp, J.H., 2012. Cytomegalovirus infection.
46798677d46798677da46798563da46798676). Pediatr. Rev. 33, 156–163.
Monath, T.P., Vasconcelos, P.F., 2015. Yellow fever. J. Clin. Virol. 64, 160–173. Pokidysheva, E., Zhang, Y., Battisti, A.J., Bator-Kelly, C.M., Chipman, P.R., Xiao, C., Gregorio,
Monlun, E., Zeller, H., Le Guenno, B., Traore-Lamizana, M., Hervy, J.P., Adam, F., Ferrara, G.G., Hendrickson, W.A., Kuhn, R.J., Rossmann, M.G., 2006. Cryo-EM reconstruction of
L., Fontenille, D., Sylla, R., Mondo, M., 1993. Surveillance of the circulation of arbo- dengue virus in complex with the carbohydrate recognition domain of DC-SIGN. Cell
virus of medical interest in the region of eastern Senegal. Bull. Soc. Pathol. Exot. 86, 124, 485–493.
21–28. Pond, W.L., 1963. Arthropod-borne virus antibodies in sera from residents of South-East
Moore, D.L., Causey, O.R., Carey, D.E., Reddy, S., Cooke, A.R., Akinkugbe, F.M., David-West, Asia. Trans. R. Soc. Trop. Med. Hyg. 57, 364–371.
T.S., Kemp, G.E., 1975. Arthropod-borne viral infections of man in Nigeria, 1964–1970. Ponlawat, A., Harrington, L.C., 2005. Blood feeding patterns of Aedes aegypti and Aedes
Ann. Trop. Med. Parasitol. 69, 49–64. albopictus in Thailand. J. Med. Entomol. 42, 844–849.
B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64 63

Prisant, N., Bujan, L., Benichou, H., Hayot, P.H., Pavili, L., Lurel, S., Herrmann, C., Janky, E., Smithburn, K.C., Kerr, J.A., Gatne, P.B., 1954a. Neutralizing antibodies against certain virus-
Joguet, G., 2016. Zika virus in the female genital tract. Lancet Infect. Dis. 16, es in the sera of residents of India. J. Immunol. 72, 248–257.
1000–1001. Smithburn, K.C., Taylor, R.M., Rizk, F., Kader, A., 1954b. Immunity to certain arthropod-
Pyke, A.T., Daly, M.T., Cameron, J.N., Moore, P.R., Taylor, C.T., Hewitson, G.R., Humphreys, borne viruses among indigenous residents of Egypt. Am. J. Trop. Med. Hyg. 3, 9–18.
J.L., Gair, R., 2014. Imported Zika virus infection from the Cook Islands into Australia, Stadler, K., Allison, S.L., Schalich, J., Heinz, F.X., 1997. Proteolytic activation of tick-borne
2014. PLoS Curr. 6. http://dx.doi.org/10.1371/currents.outbreaks.4635a4654dbffb encephalitis virus by furin. J. Virol. 71, 8475–8481.
a2156fb4632fd4676dc4649f4665e (pii: ecurrents.outbreaks.4635a4654dbffba2156f Stiasny, K., Fritz, R., Pangerl, K., Heinz, F.X., 2011. Molecular mechanisms of flavivirus
b4632fd4676dc4649f4665e). membrane fusion. Amino Acids 41, 1159–1163.
Pyke, A.T., Moore, P.R., Hall-Mendelin, S., McMahon, J.L., Harrower, B.J., Constantino, Sullivan, J.C., Tavassoli, T., Armstrong, K., Baron-Cohen, S., Humphrey, A., 2014. Reli-
T.R., van den Hurk, A.F., 2016. Isolation of Zika virus imported from Tonga into ability of self, parental, and researcher measurements of head circumference.
Australia. PLoS Curr. 8. http://dx.doi.org/10.4536695281371/currents.outbreaks. Mol. Autism 5, 2.
4536695281849adc4536695281700ad4536695281716beec4536695281707f4536 Sun, J., Wu, D., Zhong, H., Guan, D., Zhang, H., Tan, Q., Ke, C., 2016. Presence of Zika virus in
695281785 (pii: ecurrents.outbreaks.849adc840ad816beec4536695281707f4536 conjunctival fluid. JAMA Ophthalmol. 134, 1330–1332.
695281785). Swaminathan, S., Schlaberg, R., Lewis, J., Hanson, K.E., Couturier, M.R., 2016. Fatal Zika
Qian, X., Nguyen, H.N., Song, M.M., Hadiono, C., Ogden, S.C., Hammack, C., Yao, B., virus infection with secondary nonsexual transmission. N. Engl. J. Med. 375,
Hamersky, G.R., Jacob, F., Zhong, C., Yoon, K.J., Jeang, W., Lin, L., Li, Y., Thakor, J., 1907–1909.
Berg, D.A., Zhang, C., Kang, E., Chickering, M., Nauen, D., Ho, C.Y., Wen, Z., Christian, Swanson, E.C., Schleiss, M.R., 2013. Congenital cytomegalovirus infection: new prospects
K.M., Shi, P.Y., Maher, B.J., Wu, H., Jin, P., Tang, H., Song, H., Ming, G.L., 2016. Brain-re- for prevention and therapy. Pediatr. Clin. North Am. 60, 335–349.
gion-specific organoids using mini-bioreactors for modeling ZIKV exposure. Cell 165, Tabata, T., Petitt, M., Puerta-Guardo, H., Michlmayr, D., Wang, C., Fang-Hoover, J., Harris, E.,
1238–1254. Pereira, L., 2016. Zika virus targets different primary human placental cells, suggest-
Quicke, K.M., Bowen, J.R., Johnson, E.L., McDonald, C.E., Ma, H., O'Neal, J.T., Rajakumar, ing two routes for vertical transmission. Cell Host Microbe 20, 155–166.
A., Wrammert, J., Rimawi, B.H., Pulendran, B., Schinazi, R.F., Chakraborty, R., Suthar, Tang, B.L., 2016. Zika virus as a causative agent for primary microencephaly: the evidence
M.S., 2016. Zika virus infects human placental macrophages. Cell Host Microbe 20, so far. Arch. Microbiol. 198, 595–601.
83–90. Tang, H., Hammack, C., Ogden, S.C., Wen, Z., Qian, X., Li, Y., Yao, B., Shin, J., Zhang, F., Lee,
Ralapanawa, D.M., Kularatne, S.A., Jayalath, W.A., 2015. Guillain–Barre syndrome follow- E.M., Christian, K.M., Didier, R.A., Jin, P., Song, H., Ming, G.L., 2016. Zika virus infects
ing dengue fever and literature review. BMC Res. Notes 8, 729. human cortical neural progenitors and attenuates their growth. Cell Stem Cell 18,
Rasmussen, S.A., Jamieson, D.J., Honein, M.A., Petersen, L.R., 2016. Zika virus and birth de- 587–590.
fects — reviewing the evidence for causality. N. Engl. J. Med. 374, 1981–1987. Tappe, D., Nachtigall, S., Kapaun, A., Schnitzler, P., Gunther, S., Schmidt-Chanasit, J., 2015.
Robin, Y., Mouchet, J., 1975. Serological and entomological study on yellow fever in Sierra Acute Zika virus infection after travel to Malaysian Borneo, September 2014. Emerg.
Leone. Bull. Soc. Pathol. Exot. Filiales 68, 249–258. Infect. Dis. 21, 911–913.
Roth, A., Mercier, A., Lepers, C., Hoy, D., Duituturaga, S., Benyon, E., Guillaumot, L., Souares, Tappe, D., Rissland, J., Gabriel, M., Emmerich, P., Gunther, S., Held, G., Smola, S., Schmidt-
Y., 2014. Concurrent outbreaks of dengue, chikungunya and Zika virus infections — an Chanasit, J., 2014. First case of laboratory-confirmed Zika virus infection imported
unprecedented epidemic wave of mosquito-borne viruses in the Pacific 2012–2014. into Europe, November 2013. Euro Surveill. 19, 20685.
Euro Surveill. 19, 20929. Tassaneetrithep, B., Burgess, T.H., Granelli-Piperno, A., Trumpfheller, C., Finke, J., Sun, W.,
Roze, B., Najioullah, F., Ferge, J.L., Apetse, K., Brouste, Y., Cesaire, R., Fagour, C., Fagour, L., Eller, M.A., Pattanapanyasat, K., Sarasombath, S., Birx, D.L., Steinman, R.M.,
Hochedez, P., Jeannin, S., Joux, J., Mehdaoui, H., Valentino, R., Signate, A., Cabie, A., Schlesinger, S., Marovich, M.A., 2003. DC-SIGN (CD209) mediates dengue virus infec-
GBS Zika Working Group, 2016. Zika virus detection in urine from patients with tion of human dendritic cells. J. Exp. Med. 197, 823–829.
Guillain-Barre syndrome on Martinique, January 2016. Euro Surveill. 21, 30154. Thomas, D.L., Sharp, T.M., Torres, J., Armstrong, P.A., Munoz-Jordan, J., Ryff, K.R., Martinez-
Saitou, N., Nei, M., 1987. The neighbor-joining method: a new method for reconstructing Quinones, A., Arias-Berrios, J., Mayshack, M., Garayalde, G.J., Saavedra, S., Luciano, C.A.,
phylogenetic trees. Mol. Biol. Evol. 4, 406–425. Valencia-Prado, M., Waterman, S., Rivera-Garcia, B., 2016. Local transmission of Zika
Saluzzo, J.F., Gonzalez, J.P., Herve, J.P., Georges, A.J., 1981. Serological survey for the prev- virus — Puerto Rico, November 23, 2015–January 28, 2016. MMWR Morb. Mortal.
alence of certain arboviruses in the human population of the south-east area of Wkly. Rep. 65, 154–158.
Central African Republic. Bull. Soc. Pathol. Exot. Filiales 74, 490–499. Thomas, S.M., Obermayr, U., Fischer, D., Kreyling, J., Beierkuhnlein, C., 2012. Low-
Saluzzo, J.F., Ivanoff, B., Languillat, G., Georges, A.J., 1982. Serological survey for arbovirus temperature threshold for egg survival of a post-diapause and non-diapause
antibodies in the human and simian populations of the South-East of Gabon. Bull. Soc. European aedine strain, Aedes albopictus (Diptera: Culicidae). Parasit. Vectors 5,
Pathol. Exot. Filiales 75, 262–266. 100.
Sarno, M., Sacramento, G.A., Khouri, R., do Rosario, M.S., Costa, F., Archanjo, G., Santos, L.A., Thompson, J.D., Gibson, T.J., Plewniak, F., Jeanmougin, F., Higgins, D.G., 1997. The
Nery Jr., N., Vasilakis, N., Ko, A.I., de Almeida, A.R., 2016. Zika virus infection and still- CLUSTAL_X windows interface: flexible strategies for multiple sequence alignment
births: a case of hydrops fetalis, hydranencephaly and fetal demise. PLoS Negl. Trop. aided by quality analysis tools. Nucleic Acids Res. 25, 4876–4882.
Dis. 10, e0004517. Tognarelli, J., Ulloa, S., Villagra, E., Lagos, J., Aguayo, C., Fasce, R., Parra, B., Mora, J.,
Schuler-Faccini, L., Ribeiro, E.M., Feitosa, I.M., Horovitz, D.D., Cavalcanti, D.P., Pessoa, A., Becerra, N., Lagos, N., Vera, L., Olivares, B., Vilches, M., Fernandez, J., 2016. A report
Doriqui, M.J., Neri, J.I., Neto, J.M., Wanderley, H.Y., Cernach, M., El-Husny, A.S., Pone, on the outbreak of Zika virus on Easter Island, South Pacific, 2014. Arch. Virol. 161,
M.V., Serao, C.L., Sanseverino, M.T., Brazilian Medical Genetics Society–Zika 665–668.
Embryopathy Task Force, 2016. Possible association between Zika virus infection Ventura, C.V., Maia, M., Bravo-Filho, V., Gois, A.L., Belfort Jr., R., 2016a. Zika virus in Brazil
and microcephaly — Brazil, 2015. MMWR Morb. Mortal. Wkly. Rep. 65, 59–62. and macular atrophy in a child with microcephaly. Lancet 387, 228.
Scott, T.W., Takken, W., 2012. Feeding strategies of anthropophilic mosquitoes result in Ventura, C.V., Maia, M., Dias, N., Ventura, L.O., Belfort Jr., R., 2016b. Zika: neurological and
increased risk of pathogen transmission. Trends Parasitol. 28, 114–121. ocular findings in infant without microcephaly. Lancet 387, 2502.
Sejvar, J.J., Baughman, A.L., Wise, M., Morgan, O.W., 2011. Population incidence of Ventura, C.V., Maia, M., Travassos, S.B., Martins, T.T., Patriota, F., Nunes, M.E., Agra, C.,
Guillain–Barre syndrome: a systematic review and meta-analysis. Torres, V.L., van der Linden, V., Ramos, R.C., Rocha, M.A., Silva, P.S., Ventura, L.O.,
Neuroepidemiology 36, 123–133. Belfort Jr., R., 2016c. Risk factors associated with the ophthalmoscopic findings iden-
Sharp, T.M., Munoz-Jordan, J., Perez-Padilla, J., Bello-Pagan, M.I., Rivera, A., Pastula, D.M., tified in infants with presumed Zika virus congenital infection. JAMA Ophthalmol.
Salinas, J.L., Martinez Mendez, J.H., Mendez, M., Powers, A.M., Waterman, S., Rivera- 134, 912–918.
Garcia, B., 2016. Zika virus infection associated with severe thrombocytopenia. Clin. Ventura, C.V., Maia, M., Ventura, B.V., Linden, V.V., Araujo, E.B., Ramos, R.C., Rocha, M.A.,
Infect. Dis. 63, 1198–1201. Carvalho, M.D., Belfort Jr., R., Ventura, L.O., 2016d. Ophthalmological findings in in-
Shinohara, K., Kutsuna, S., Takasaki, T., Moi, M.L., Ikeda, M., Kotaki, A., Yamamoto, K., fants with microcephaly and presumable intra-uterus Zika virus infection. Arq. Bras.
Fujiya, Y., Mawatari, M., Takeshita, N., Hayakawa, K., Kanagawa, S., Kato, Y., Oftalmol. 79, 1–3.
Ohmagari, N., 2016. Zika fever imported from Thailand to Japan, and diagnosed by Venturi, G., Zammarchi, L., Fortuna, C., Remoli, M.E., Benedetti, E., Fiorentini, C., Trotta, M.,
PCR in the urine. J. Travel Med. 23 (tav011). Rizzo, C., Mantella, A., Rezza, G., Bartoloni, A., 2016. An autochthonous case of Zika
Simmonds, P., Becher, P., Collett, M.S., Gould, E.A., Heinz, F.X., Meyers, G., Monath, T., due to possible sexual transmission, Florence, Italy, 2014. Euro Surveill. 21, 30148.
Pletnev, A., Rice, C.M., Stiasny, K., Thiel, H.J., Weiner, A., Bukh, J., 2012. Family Verma, R., Sahu, R., Holla, V., 2014. Neurological manifestations of dengue infection: a re-
Flaviviridae. In: King, A.M.Q., Adams, M.J., Carstens, E.B., Lefkowitz, E.J. (Eds.), Virus view. J. Neurol. Sci. 346, 26–34.
Taxonomy: Ninth Report of the International Committee on Taxonomy of Viruses. Victora, C.G., Schuler-Faccini, L., Matijasevich, A., Ribeiro, E., Pessoa, A., Barros, F.C., 2016.
Elsevier Academic Press, San Diego, CA, pp. 1003–1020. Microcephaly in Brazil: how to interpret reported numbers? Lancet 387, 621–624.
Simon, O., Billot, S., Guyon, D., Daures, M., Descloux, E., Gourinat, A.C., Molko, N., Dupont- Villamil-Gomez, W.E., Mendoza-Guete, A., Villalobos, E., Gonzalez-Arismendy, E., Uribe-
Rouzeyrol, M., 2016. Early Guillain–Barre syndrome associated with acute dengue Garcia, A.M., Castellanos, J.E., Rodriguez-Morales, A.J., 2016. Diagnosis, management
fever. J. Clin. Virol. 77, 29–31. and follow-up of pregnant women with Zika virus infection: a preliminary report
Simpson, D.I., 1964. Zika virus infection in man. Trans. R. Soc. Trop. Med. Hyg. 58, of the ZIKERNCOL cohort study on Sincelejo, Colombia. Travel Med. Infect. Dis. 14,
335–338. 155–158.
Sirohi, D., Chen, Z., Sun, L., Klose, T., Pierson, T.C., Rossmann, M.G., Kuhn, R.J., 2016. The Villordo, S.M., Gamarnik, A.V., 2009. Genome cyclization as strategy for flavivirus RNA
3.8 A resolution cryo-EM structure of Zika virus. Science 352, 467–470. replication. Virus Res. 139, 230–239.
Smit, J.M., Moesker, B., Rodenhuis-Zybert, I., Wilschut, J., 2011. Flavivirus cell entry and von der Hagen, M., Pivarcsi, M., Liebe, J., von Bernuth, H., Didonato, N., Hennermann, J.B.,
membrane fusion. Viruses 3, 160–171. Buhrer, C., Wieczorek, D., Kaindl, A.M., 2014. Diagnostic approach to microcephaly in
Smithburn, K.C., 1952. Neutralizing antibodies against certain recently isolated viruses in childhood: a two-center study and review of the literature. Dev. Med. Child Neurol.
the sera of human beings residing in East Africa. J. Immunol. 69, 223–234. 56, 732–741.
Smithburn, K.C., 1954. Neutralizing antibodies against arthropod-borne viruses in the sera Waehre, T., Maagard, A., Tappe, D., Cadar, D., Schmidt-Chanasit, J., 2014. Zika virus infec-
of long-time residents of Malaya and Borneo. Am. J. Hyg. 59, 157–163. tion after travel to Tahiti, December 2013. Emerg. Infect. Dis. 20, 1412–1414.
64 B.-H. Song et al. / Journal of Neuroimmunology 308 (2017) 50–64

Wang, L., Valderramos, S.G., Wu, A., Ouyang, S., Li, C., Brasil, P., Bonaldo, M., Coates, T., Yin, Y., Xu, Y., Su, L., Zhu, X., Chen, M., Zhu, W., Xia, H., Huang, X., Gong, S., 2016. Epidemi-
Nielsen-Saines, K., Jiang, T., Aliyari, R., Cheng, G., 2016. From mosquitos to humans: ologic investigation of a family cluster of imported ZIKV cases in Guangdong, China:
genetic evolution of Zika virus. Cell Host Microbe 19, 561–565. probable human-to-human transmission. Emerg. Microbes Infect. 5, e100.
Watemberg, N., Silver, S., Harel, S., Lerman-Sagie, T., 2002. Significance of microcephaly Yu, I.M., Zhang, W., Holdaway, H.A., Li, L., Kostyuchenko, V.A., Chipman, P.R., Kuhn, R.J.,
among children with developmental disabilities. J. Child Neurol. 17, 117–122. Rossmann, M.G., Chen, J., 2008. Structure of the immature dengue virus at low pH
Weaver, S.C., Barrett, A.D., 2004. Transmission cycles, host range, evolution and emer- primes proteolytic maturation. Science 319, 1834–1837.
gence of arboviral disease. Nat. Rev. Microbiol. 2, 789–801. Yun, S.I., Lee, Y.M., 2006. Japanese encephalitis virus: molecular biology and vaccine de-
Weaver, S.C., Costa, F., Garcia-Blanco, M.A., Ko, A.I., Ribeiro, G.S., Saade, G., Shi, P.Y., velopment. In: Kalitzky, M., Borowski, P. (Eds.), Molecular Biology of the Flavivirus.
Vasilakis, N., 2016. Zika virus: history, emergence, biology, and prospects for control. Horizon Scientific Press, Norwich, pp. 225–271.
Antiviral Res. 130, 69–80. Yun, S.I., Lee, Y.M., 2014. Japanese encephalitis: the virus and vaccines. Hum. Vaccin.
Webster, W.S., 1998. Teratogen update: congenital rubella. Teratology 58, 13–23. Immunother. 10, 263–279.
Weinbren, M.P., Williams, M.C., 1958. Zika virus: further isolations in the Zika area, and Yun, S.I., Song, B.H., Frank, J.C., Julander, J.G., Polejaeva, I.A., Davies, C.J., White, K.L., Lee,
some studies on the strains isolated. Trans. R. Soc. Trop. Med. Hyg. 52, 263–268. Y.M., 2016. Complete genome sequences of three historically important, spatiotem-
Welsch, S., Miller, S., Romero-Brey, I., Merz, A., Bleck, C.K., Walther, P., Fuller, S.D., Antony, porally distinct, and genetically divergent strains of Zika virus: MR-766, P6-740,
C., Krijnse-Locker, J., Bartenschlager, R., 2009. Composition and three-dimensional ar- and PRVABC-59. Genome Announc. 4, e00800–16.
chitecture of the dengue virus replication and assembly sites. Cell Host Microbe 5, Zammarchi, L., Stella, G., Mantella, A., Bartolozzi, D., Tappe, D., Gunther, S., Oestereich, L.,
365–375. Cadar, D., Munoz-Fontela, C., Bartoloni, A., Schmidt-Chanasit, J., 2015a. Zika virus in-
Westaway, E.G., Mackenzie, J.M., Khromykh, A.A., 2002. Replication and gene function in fections imported to Italy: clinical, immunological and virological findings, and public
Kunjin virus. Curr. Top. Microbiol. Immunol. 267, 323–351. health implications. J. Clin. Virol. 63, 32–35.
WHO, 2015. Zika virus outbreaks in the Americas. Wkly. Epidemiol. Rec. 90, 609–610. Zammarchi, L., Tappe, D., Fortuna, C., Remoli, M.E., Gunther, S., Venturi, G., Bartoloni, A.,
WHO, 2016a. Situation Report: Zika Virus Microcephaly and Guillain–Barré Syndrome. Schmidt-Chanasit, J., 2015b. Zika virus infection in a traveller returning to Europe
World Health Organization, Geneva, Switzerland March 17, 2016. http://apps.who. from Brazil, March 2015. Euro Surveill. 20, 21153.
int/iris/bitstream/10665/204633/1/zikasitrep_17Mar2016_eng.pdf. Zanluca, C., Melo, V.C., Mosimann, A.L., Santos, G.I., Santos, C.N., Luz, K., 2015. First report
WHO, 2016b. WHO Confirms Zika Virus Strain Imported From the Americas to Cabo of autochthonous transmission of Zika virus in Brazil. Mem. Inst. Oswaldo Cruz 110,
Verde. World Health Organization, Geneva, Switzerland May 20, 2016. http://www. 569–572.
who.int/mediacentre/news/releases/2016/zika-cabo-verde/en/. Ze-Ze, L., Prata, M.B., Teixeira, T., Marques, N., Mondragao, A., Fernandes, R., Saraiva da
WHO, 2016c. WHO Statement on the First Meeting of the International Health Regula- Cunha, J., Alves, M.J., 2016. Zika virus infections imported from Brazil to Portugal,
tions 2005 (IHR 2005) Emergency Committee on Zika Virus and Observed Increase 2015. IDCases 4, 46–49.
in Neurological Disorders and Neonatal Malformations. World Health Organization, Zhang, C., Feng, T., Cheng, J., Li, Y., Yin, X., Zeng, W., Jin, X., Li, Y., Guo, F., Jin, T., 2016a.
Geneva, Switzerland February 1, 2016. http://www.who.int/mediacentre/news/ Structure of the NS5 methyltransferase from Zika virus and implications in inhibitor
statements/2016/1st-emergency-committee-zika/en/. design. Biochem. Biophys. Res. Commun. http://dx.doi.org/10.31016/j.bbrc.32016.
WHO, 2016d. WHO Statement: Fifth Meeting of the Emergency Committee under the In- 31911.31098 (pii: S0006-0291X(0016)31963-31965).
ternational Health Regulations (2005) Regarding Microcephaly, Other Neurological Zhang, J., Jin, X., Zhu, Z., Huang, L., Liang, S., Xu, Y., Liao, R., Zhou, L., Zhang, Y., Wilder-
Disorders and Zika Virus. World Health Organization, Geneva, Switzerland November Smith, A., 2016b. Early detection of Zika virus infection among travellers from areas
18, 2016. http://www.who.int/mediacentre/news/statements/2016/zika-fifth-ec/en/. of ongoing transmission in China. J. Travel Med. 23 (taw047).
Wielanek, A.C., Monredon, J.D., Amrani, M.E., Roger, J.C., Serveaux, J.P., 2007. Guillain– Zhang, Y., Corver, J., Chipman, P.R., Zhang, W., Pletnev, S.V., Sedlak, D., Baker, T.S., Strauss,
Barre syndrome complicating a chikungunya virus infection. Neurology 69, J.H., Kuhn, R.J., Rossmann, M.G., 2003. Structures of immature flavivirus particles.
2105–2107. EMBO J. 22, 2604–2613.
Wong, P.S., Li, M.Z., Chong, C.S., Ng, L.C., Tan, C.H., 2013. Aedes (Stegomyia) albopictus Zhang, Y., Kaufmann, B., Chipman, P.R., Kuhn, R.J., Rossmann, M.G., 2007. Structure of im-
(Skuse): a potential vector of Zika virus in Singapore. PLoS Negl. Trop. Dis. 7, e2348. mature West Nile virus. J. Virol. 81, 6141–6145.
Woods, C.G., 2004. Human microcephaly. Curr. Opin. Neurobiol. 14, 112–117. Zhu, Z., Chan, J.F., Tee, K.M., Choi, G.K., Lau, S.K., Woo, P.C., Tse, H., Yuen, K.Y., 2016. Com-
Wu, K.Y., Zuo, G.L., Li, X.F., Ye, Q., Deng, Y.Q., Huang, X.Y., Cao, W.C., Qin, C.F., Luo, Z.G., parative genomic analysis of pre-epidemic and epidemic Zika virus strains for viro-
2016. Vertical transmission of Zika virus targeting the radial glial cells affects cortex logical factors potentially associated with the rapidly expanding epidemic. Emerg.
development of offspring mice. Cell Res. 26, 645–654. Microbes Infect. 5, e22.

You might also like