ArticlePDF Available

Congenital Zika syndrome

Authors:

Abstract and Figures

In February 2016, the World Health Organization declared Zika virus (ZIKV) infection a public health emergency of international concern because it caused congenital Zika syndrome (CZS). The CZS is considered a specific pattern of birth defects caused by ZIKV infection, which is transmitted by the bite of the Aedes aegypti mosquito. The CZS clinical manifestations are broad and nonspecific, including microcephaly, subcortical calcifications, ocular alterations, congenital contractures, early hypertonia, and pyramidal as well as extrapyramidal symptoms. The ZIKV has gained great importance because it has affected a large percentage of the population worldwide during the last few years, despite the measures implemented by international organizations. The pathophysiology and non-vectorial transmission routes of the virus are still under study. The diagnosis is made upon suspicion of ZIKV infection, the patient’s clinical manifestations, and it is confirmed by molecular laboratory tests demonstrating the presence of viral particles. Unfortunately, there is no specific treatment or vaccine for this condition; however, patients receive multidisciplinary care and constant monitoring. Therefore, the strategies that have been implemented are directed toward preventive measures and vector control.
Content may be subject to copyright.
3
Congenital Zika syndrome
Israel E. Crisanto-López1,2, Pablo López-De Jesús2,3, Jacqueline López-Quecho4, and
Juan C. Flores-Alonso2*
1Dirección General de Calidad y Educación en Salud, Secretaría de Salud, Mexico City; 2Centro de Investigación Biomédica de Oriente, Instituto
Mexicano del Seguro Social, Atlixco, Puebla; 3Programa de Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana,
Mexico City; 4Universidad Popular Autónoma del Estado de Puebla, Facultad de Medicina, Puebla, Mexico
Boletín Médico del
Hospital Infantil de México
REVIEW ARTICLE
Abstract
In February 2016, the World Health Organization declared Zika virus (ZIKV) infection a public health emergency of interna-
tional concern because it caused congenital Zika syndrome (CZS). The CZS is considered a specific pattern of birth defects
caused by ZIKV infection, which is transmitted by the bite of the Aedes aegypti mosquito. The CZS clinical manifestations are
broad and nonspecific, including microcephaly, subcortical calcifications, ocular alterations, congenital contractures, early
hypertonia, and pyramidal as well as extrapyramidal symptoms. The ZIKV has gained great importance because it has affec-
ted a large percentage of the population worldwide during the last few years, despite the measures implemented by inter-
national organizations. The pathophysiology and non-vectorial transmission routes of the virus are still under study. The
diagnosis is made upon suspicion of ZIKV infection, the patient’s clinical manifestations, and it is confirmed by molecular
laboratory tests demonstrating the presence of viral particles. Unfortunately, there is no specific treatment or vaccine for this
condition; however, patients receive multidisciplinary care and constant monitoring. Therefore, the strategies that have been
implemented are directed toward preventive measures and vector control.
Keywords: Zika virus. Congenital Zika syndrome. Microcephaly.
Síndrome congénito por virus Zika
Resumen
En febrero de 2016, la Organización Mundial de la Salud declaró a la infección por virus Zika una emergencia de salud pública
de importancia internacional por ser la causa del síndrome congénito por virus Zika. Este síndrome es considerado un patrón
específico de defectos al nacimiento causados por la infección por virus Zika, que se transmite por la picadura del mosquito
vector Aedes aegypti y cuyas manifestaciones clínicas son amplias e inespecíficas, entre las que destacan microcefalia, calci-
ficaciones subcorticales, alteraciones oculares, contracturas congénitas, hipertonía temprana y síntomas de afectación piramidal
y extrapiramidal. Este virus ha tomado gran importancia debido a que durante los últimos años ha afectado a un gran porcen-
taje de la población en todo el mundo, a pesar de las medidas implementadas por organizaciones internacionales. La fisiopa-
tología y las vías de transmisión no vectorial de la infección aún siguen en estudio. El diagnóstico se realiza ante la sospecha
por las manifestaciones clínicas del paciente, y se confirma mediante pruebas moleculares de laboratorio en las que se
demuestre la presencia de partículas virales. Desafortunadamente, no existe aún un tratamiento ni una vacuna específica para
*Correspondence:
Juan C. Flores Alonso
E-mail:juan.oresal@imss.gob.mx
Available online: 27-02-2023
Bol Med Hosp Infant Mex. 2023;80(1):3-14
www.bmhim.com
Date of reception: 08-07-2022
Date of acceptance: 18-10-2022
DOI: 10.24875/BMHIM.22000110
1665-1146/© 2022 Hospital Infantil de México Federico Gómez. Published by Permanyer. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
4
Bol Med Hosp Infant Mex. 2023;80(1)
Introduction
Congenital Zika syndrome (CZS) is considered a
specic pattern of newborn defects caused by Zika
virus (ZIKV) infection, a microorganism that has been
considered a teratogenic agent1-3. Although the patho-
physiological pathways by which this infection causes
organ dysfunction are still under investigation, nervous
system, musculoskeletal and visual impairment have
been described in infected newborns. The following
clinical manifestations stand out: microcephaly with
subcortical calcications, retinal alterations, congenital
contractures, early hypertonia, and extrapyramidal
symptoms, which can occur together or separately in
each patient4,5.
ZIKV infection has become very important in recent
years because it is associated with signicant health
problems worldwide and has been classied as a public
health emergency of international concern6. This has
prompted various international organizations such as
the World Health Organization (WHO), the United
States Centers for Disease Control and Prevention
(CDC), and the Pan American Health Organization
(PAHO) to take preventive measures to avoid its further
progression as a pathological entity6 -8.
In October 2015, the Secretaría Estatal de Salud
(State Health Secretariat) of Pernambuco, Brazil,
reported 26 cases of microcephaly associated with
ZIKV infection8,9. Subsequently, countries worldwide
began to report cases of patients with microcephaly
and infection with serum ZIKV viral load. ZIKV infection
during pregnancy was found to cause CZS and com-
plications during pregnancy, such as preterm labor or
even spontaneous abortion5,10.
In Mexico, epidemiological surveillance has been
conducted through the Secretaría de Prevención y
Promoción a la Salud (Secretariat of Prevention and
Health Promotion). According to the information bulletin
“Conrmed cases of ZIKV disease” for the 52ndepide-
miological week of 2021, 12,991cases of ZIKV infection
were reported from 2015 to December 31, 2021, with
Veracruz, Yucatán, and Nuevo León being the states
with the highest number of conrmed cases. In addi-
tion, it was reported that 7152 pregnant patients pre-
sented ZIKV infection, with Yucatán having the highest
number of cases with 926, followed by Veracruz with
888 cases and Tamaulipas with 692 cases11. In addi-
tion, 44 cases conrmed by epidemiological associa-
tion of CZS and 56 laboratory-conrmed cases of CZS
were reported12,13.
At present, the diagnosis of ZIKV disease in rst-contact
health services is complicated due to the similarity of the
clinical picture with diseases caused by other viruses of
the same genus, such as Dengue and Chikungunya
virus14-16 (Table1). Symptoms are generally mild, lasting
2-7 days, and may consist of fever, rash, conjunctivitis,
muscle and joint pain, malaise, and headache; however,
most infected patients are asymptomatic8.
ZIKV
The virus was named after the Zika forest in Entebbe,
Uganda, Africa, where it was discovered in 1947 in
rhesus monkeys, in which transmission was only enzo-
otic, restricted to primate-mosquito circulation17,18.
Subsequently, the rst case of human transmission
was reported in 1954 in a 10-year-old Nigerian girl who
had a serum viral load of ZIKV. However, it was difficult
to relate her clinical presentation to ZIKV infection
because parasites of the genus Plasmodium were also
found in her blood samples19. In the same year, two
more cases of ZIKV infection in humans were reported,
conrmed by increased serum-neutralizing antibodies20.
In 1966, ZIKV infection was massively detected in the
Asian population, which increased research on human
transmission. As a result, the ability of the virus to infect
humans through a vector was reported, and its wide
geographical distribution and the presence of different
viral lineages, both from the African and Asian conti-
nents, were demonstrated21. Subsequent studies
showed that ZIKV is an arbovirus of the genus
Flavivirus, whose main vector is the Aedes aegypti
mosquito1. Since then, cases of ZIKV infections in
humans with diverse clinical manifestations have been
reported; over time, cases with more severe alterations
have appeared5,15,22.
Subsequently, a high incidence of cases was reported
in 2007 on the islands of Yap in Micronesia23, and in
2008 the rst cases of sexual transmission were
reported24. In 2013 and 2014 in French Polynesia, the
rst cases of ZIKV infection were reported with
este padecimiento; sin embargo, los pacientes reciben cuidados multidisciplinarios y monitorización constante. Las estrategias
que se han implementado se dirigen hacia medidas preventivas de la infección y el control de los vectores.
Palabras clave: Virus Zika. Síndrome congénito por virus Zika. Microcefalia.
5
I.E. Crisanto-López et al. Congenital Zika syndrome
post-infectious onset of Guillain-Barré syndrome,
maternal-fetal transmission, and the presence of viral
load in semen samples from asymptomatic donors25.
In the Americas, it is estimated that ZIKV was intro-
duced between May and December 2013; however, the
rst reported cases of CZS were in May 2015 in Brazil,
attributed to globalization and migratory movements
due to social and sporting events5,26,27.
The worldwide spread of the virus was rapidly pro-
gressive, and cases began to be documented in sub-
tropical areas, Central and North America, and 87
countries of different geographic distribution. More than
500,000cases were reported worldwide. This prompted
the WHO to declare ZIKV infection as a Public Health
Emergency of International Concern on February 1,
2016, and temporarily conrmed the association of
ZIKV infection with microcephaly, Guillain-Barré syn-
drome, sexual, blood and vertical transmission, as well
as the occurrence of severe birth defects in neonates
with parents with a history of ZIKV infection5-7,20.
ZIKV belongs to the Flavivirus genus of the
Flaviviridae family and measures approximately 40
nanometers (nm) in diameter. Its genetic material
corresponds to a single-stranded ribonucleic acid chain
(single-stranded RNA) of positive polarity, which
encodes a polyprotein whose translation gives rise to
three structural proteins: capsid protein (C), membrane
precursor (prM) and envelope protein (E), and 7
non-structural proteins (NS): NS1, NS2A, NS2B, NS3,
NS4A, NS4B, and NS528. The function of each of these
is described in Ta b le2.
The viral particle consists of a symmetric icosahedral
structure with a viral envelope containing the E and M
proteins as molecules for attachment and host cell rec-
ognition (Fig.1A)26,29. The entry of ZIKV into the host
cell is mediated by the E protein, which allows attach-
ment and fusion to the specic receptors TIM1, TYRO-
3, and AXL, thus activating the endocytosis processes
and the entry of the virion into the cell cytoplasm29,30.
Inside the cell, non-structural proteins bind to the
endoplasmic reticulum (ER), forming a complex that
allows replication and assembly of viral RNA called
viroplasm31. This involves NS4A and the formation of
ER invaginations in which NS1 subsequently acts to
initiate the viral genome replication processes, and
NS3 RNA helicase and NS5 polymerase act, assisted
Table1. Main clinical manifestations in Zika, dengue and Chikungunya virus infections
Signs and symptoms Zika Dengue Chikungunya
Reason for consultation Exantema o prurito Fever, myalgia Joint pain, fever
Fever Mild Moderate Intense
Very infrequent Very frequent Very frequent
Duration: 1‑3 days Duration: 5‑7 days Duration: 5‑7 days
Exanthema and features Typically from day 1 Appears from 5th to 7th day Appears on the 2nd or 3rd day
Maculopapular, cephalo‑caudal Not characteristic Not characteristic
Pruritus Moderate to intense Mild to severe Mild to moderate
Conjuntivitis Very frequent Infrequent Adults: Very rare/Children: Very common
Neurological
manifestations
Possible and serious Infrequent Infrequent (can be frequent and severe
in newborns)
Headache Mild to moderate Intense and frequent Mild to moderate
Retro‑orbital pain Infrequent Intense and frequent Infrequent
Polyarthralgia Frequent Absent Very frequent
Polyarthritis Frequent Absent Frequent
Myalgia Infrequent Very frequent and intense Frequent, moderate to severe
Diarrhea Very infrequent Frequent Very infrequent
Skin bleeding Very infrequent Frequent Very infrequent
Mucosal bleeding Very infrequent Alarm sign Very infrequent (severe when present)
6
Bol Med Hosp Infant Mex. 2023;80(1)
by NS4B. Viral packaging requires the action of NS2A
and NS2B through viral RNA packaging vesicles. In
these vesicles, the structural proteins are integrated, a
process necessary for their transport into the cell,
mainly through the interaction of the E protein and prM
with the Golgi apparatus28,31,32. At the same time, ZIKV
generates modications in centrosomes to reorganize
microtubules and facilitate their transport and intracel-
lular propagation33,34. Subsequently, for viral release
into the extracellular environment, it was described that
ZIKV can induce a process of apoptosis mediated by
autophagy, processes of degranulation of viral RNA
packaging vesicles, or processes of exocytosis34. The
NS5 viral protein antagonizes the type I interferon
response to favor viral dissemination. This leads to
proteasomal degradation of STAT2 (a transcription fac-
tor that generates chain responses of the immune sys-
tem), allowing ZIKV to inltrate the lymphatic and
circulatory system, which leads to the dissemination of
the virus throughout the body and the development of
clinical manifestations (Fig. 1B )30,35.
ZIKV transmission can be vector-borne or non-vector-borne1.
Vector-borne transmission occurs when the virus infects the
host through mosquito bites36. This explains the sylvatic
transmission cycle, where non-human primates and endemic
mosquitoes (enzootic vectors) are involved, as well as
human-mosquito-human transmission in urban and subur-
ban regions1,37. On the other hand, the mechanisms of
non-vectorial infection that has been described are sexual38,
vertical39, perinatal40, blood transfusion, and organ trans-
plantation41, as well as contact with body secretions such
as tears, saliva, urine, or sweat42.
Vertical, maternal-fetal transmission of Flaviviruses is
unusual. It has been proposed that these viruses follow
different pathways that vary according to gestational
age and the route of maternal infection (vectorial or
non-vectorial) to invade and replicate viral proteins in
fetal tissue43,44.
Pathophysiology
ZIKV has a strong tropism to ectodermal derivatives,
which confers greater dissemination potential with a
wide variety of clinical manifestations and specic alter-
ations according to the infected tissue35,45. It has been
reported that ZIKV presents neuronal tropism and
towards other organs, such as the liver, kidney, heart,
spleen, testicle, and ovary since viral RNA has been
found in these tissues30,46,47.
ZIKV tropism is mediated by type I and III interfer-
on-stimulated genes, which induce signaling for the pro-
duction of membrane proteins AXL and Tyro-3, members
of the receptor tyrosine kinase-like protein family, which
are expressed mainly in neuronal progenitor cells and
play an important role in embryonic developmental
homeostasis. In addition, the TIM1 glycoprotein, which
interacts with ZIKV, has been found in multiple target
organs, such as Hofbauer cells or placental macro-
phages, endothelium, and cytotrophoblast30,47.
Moreover, detection of ZIKV RNA in fetal tissues has
also been reported in the umbilical cord, placental cells
such as macrophages, amniotic uid, and brain, as well
as in tissues from spontaneous abortions of rst and
second-trimester human fetuses, mainly in placenta and
brain8,30,47. In vitro studies suggest that viral replication
occurs mainly in Hofbauer cells, trophoblast cells, and
fetal endothelium48-50.
Vertical transmission of ZIKV can develop through
the interaction of the virus in maternal blood or uterine
Table2. Functions of Zika virus proteins
Type Protein Function
Structural Protein C Forms the viral capsid
Protein E Enables membrane binding and
fusion with the host cell,
penetration and
hemagglutination during the
viral replication cycle
Protein
prM
Generates M protein, which
releases mature virion out of
the cell forming complexes
with E protein
Non‑structural NS1 Initiates the process of viral
RNA genome synthesis
NS2A Interacts with NS3 and NS5 for
viral packaging and replication
NS2B Interacts with NS3 for viral
packaging and replication
NS3 Harbors RNA protease and
helicase activity, essential for
viral replication and survival
NS4A Participates in the localization
of the replication complex to
the membrane
NS4B Assists NS3 protein
NS5 Participates in the inhibition of
the host innate immune system
and is a coadjuvant of the NS3
protein in the nuclear
localization of the virus
Adapted from Panwar28.
7
I.E. Crisanto-López et al. Congenital Zika syndrome
broblasts with embryonic cells35,51. For this, two
hypotheses are considered: direct transfer or placental
mediated response on placental precursor tissue such
as trophoblast, chorioamniotic villi and membranes,
Hofbauer cells, umbilical cord endothelial cells, and
amniotic epithelial cells44,50-52. The direct transfer the-
ory stipulates that viral contact occurs within the rst
10weeks of gestation through the presence of ZIKV in
maternal blood or uterine broblasts, thus infecting
embryonic precursor cells of neuronal tissue due to its
neurotropism. On the other hand, the theory of placental
response can be considered complementary to the
pathophysiology since the inammatory response
caused by ZIKV infection produces a disruption of
placental signals that mediate neuronal development
and changes the prole of inammatory markers in fetal
organs, which eventually generates the clinical manifes-
tations depending on the specic tissue affected49,52-54.
According to the CDC, approximately 5% of pregnant
patients with maternal ZIKV infection either lose the
pregnancy or develop birth defects50,51. Gestational age
and host genetic variation are important determinants
of placental infection and immune system function35. In
addition, infection acquired during the rst trimester
increases the risk of developing CZS in the embryo51,55.
Statistically, infection in the rst two trimesters is asso-
ciated with severe alterations in target organs such as
the brain, eye, and male and female genital tract;
Protein E
Protein M
Protein C
Viral RNA
E protein-
mediated
ZIKV entry
ZIKV-
specific
receptor
Viroplasma
NS protein
binding
to ER
NS4A - ER
Viral RNA replication
NS1-NS3-MS5-NS4B
Endoplasmic
reticulum
Nucleus
Golgi apparatus
Viral RNA
packaging
vesicles
NS2A-NS2B
Structural
proteins
ZIKV
release
ZIKV
B
A
Figure1. Zika virus. A: structure. B: viral cycle.
8
Bol Med Hosp Infant Mex. 2023;80(1)
however, infection in any trimester of pregnancy is
associated with adverse fetal outcomes35,55-57.
During the 3rdweek of embryonic development, gas-
trulation and, thus, organogenesis begins. The external
epithelium of the body and the neural tube are formed
from the ectoderm. The external body epithelium gives
rise to organs and structures that are in contact with
the outside world (e.g., sensory epithelium of the ear,
nose, and eye, epidermis, skin, nails, mammary, sweat,
and sebaceous glands), anterior pituitary, and tooth
enamel. At the same time, from the neural tube, the
components of the nervous system are formed from the
neural plate (notochord-induced thickening of the ecto-
derm) and neural crest cells (NCC) (Fig.2)58,59.
NCCs undergo an epithelial-mesenchymal transition
for their active movement and migration into the meso-
derm50, directed by the expression of the bone morpho-
genetic protein, WNT signaling pathways, and by
broblast stimulating factor 8 expressed by the meso-
derm. In addition, they are regulated by the expression
of cell adhesion molecules and extracellular matrix mol-
ecules (bronectin, laminin, and typeIV collagen), lo-
podia, guidance molecules (neuropilin/semaphorin),
and by planar cell polarity. It should be noted that NCCs
can be differentiated by prior programming and by inter-
action with nearby tissues59. NCCs are divided into
three regions for their study: cranial, circumpharyngeal,
and truncal, each with specic derivatives according to
its region59. NCCs contribute to the formation of the
peripheral nervous system, meninges, melanocytes,
odontoblasts, medullary chromaffin cells, and a large
part of the connective tissues that form the cervical and
craniofacial territory, including bone, cartilage, and
muscle tissue60. This is why an agent with ectodermal
tropism can present diverse clinical manifestations61.
Clinical, embryonic, and fetal
manifestations of CZS
The clinical features of CZS are a consequence of
direct neurological involvement and intracranial volume
loss, causing structural, and functional alterations4,62,63.
Structural alterations include cranial morphology, cere-
bral and ocular anomalies, congenital contractures, and
mainly microcephaly (Fig. 3A-C)64, while functional
alterations are exclusively related to neurological
impairment55,65,66.
Chronic cerebral neuropathy in ZIKV-infected children
consists of intracranial calcications, typically subcortical
(Fig.4A and B), increased uid spaces (ventricular and
extra-axial) (Fig. 4C and D), cortical thinning with
abnormalities in the convolutions (polymicrogyria, pachy-
gyria, and agyria), hypoplasia or absence of the corpus
callosum, decreased myelin, and hypoplasia of the cer-
ebellum or cerebellar vermis67-69.
Calcications have also been identied in the basal
ganglia and brainstem. Ultrasound or magnetic
C
B
A
Figure3. Newborn with clinical features of microcephaly.
Female patient at 39weeks of gestation. Aand B: frontal
view. C: lateral view. Image obtained from Aviña‑Padilla et
al.64.
Epiblast
Ectoderm
External body epithelium Neural tube
Mammary glands,
sebaceous glands,
sweat glands, hair,
nails, proctodermal
epithelium,
auditory vesicle,
lens and cornea
Optic vesicle, epiphysis,
posterior pituitary,
cerebrum and
spinal cord
Neural crest cells
Figure2. Ectodermal germinal layer derivatives.
9
I.E. Crisanto-López et al. Congenital Zika syndrome
resonance imaging (MRI) studies can detect these
brain abnormalities during the prenatal period. In
cases of microcephaly, damage of the central nervous
system caused by ZIKV occurs by direct and indirect
cellular injury through various pathways such as apop-
tosis, regulation of the immune response, ubiquitina-
tion, and viral replication, mostly in neuronal progenitor
cells35,70. This apoptotic capacity may explain the
alterations in the embryonic neurodevelopment of
infected products. In addition, experiments have been
reported in animal models, where infection in postna-
tal products causes dysfunction of cell death path-
ways, decreased proliferation of stem cells in the
periventricular zone, and disruption of corticospinal
pyramidal neurons71,72. These are not the only factors
contributing to microcephaly caused by ZIKV; NCCs
are also affected by this virus. Consequently, they
produce pro-inammatory cytokines that induce apop-
tosis via the paracrine pathway affecting neuronal pro-
genitor cells35.
Neurological alterations caused by ZIKV infection
include hypertonia, hypotonia, spasticity, hyperreexia,
severe irritability, and seizures. Some patients with con-
rmed or probable infection present fetal disruption
sequence phenotype, characterized by severe micro-
cephaly, marked craniofacial disproportion, occipital
bone prominence, altered hair implantation, short neck,
redundant nuchal and cranial folds, retrognathia, con-
tractures, and multiple arthrogryposes73,74.
In the eyes, viral RNA has been detected in the cor-
nea, optic nerve, and neurosensory retina. The most
frequent ocular structural anomalies are, in particular,
microphthalmia, coloboma, congenital cataracts, and
intraocular calcications. Likewise, cases of chorioreti-
nal atrophy and retinitis pigmentosa have been reported,
generally affecting the macular region and producing
atrophy of the optic nerve75.
In the male reproductive tract, the infection damages
spermatogonia and Sertoli cells, destroys testicular
architecture, decreases sperm motility, and thus
reduces fertility76. In parallel, in the female genital tract,
viral RNA has been reported in the cervical mucosa up
to 11days after the onset of symptoms. In vitro studies
showed that uterine broblasts have increased suscep-
tibility to ZIKV, contributing to fetal developmental
damage35,77.
On the other hand, congenital contractures of one or
multiple joints (arthrogryposis multiplex congenita)
have been described in fetuses and newborns with
congenital ZIKV infection. The clinical picture of con-
genital contractures varies according to proximal or
distal location, laterality, upper or lower limb, and sever-
ity, as reected by neurological damage78,79.
Neurogenic factors affecting the corticospinal tract
and motor neurons or their interactions may cause fetal
motor abnormalities, resulting in decreased movements
and contractures. However, the specic mechanism
causing contractures in prenatal ZIKV infection is not
yet fully understood80.
To date, the means of non-vectorial transmission and
the pathophysiology at the molecular level of this con-
dition are still under investigation.
Diagnosis
Due to the severe effects caused by ZIKV infection
on embryonic and fetal development, it is necessary to
identify risk factors and make a timely diagnosis. As
has been described, CZS can present diverse clinical
manifestations, both mild and severe; in addition, the
clinical data presented are nonspecic since different
infections can also cause them during pregnancy81.
Therefore, it is important to detect pregnant patients
D
C
B
A
Figure4. CT scan of newborn with severe microcephaly.
A: profound craniofacial disproportion, corpus callosum
(short white arrow), brainstem (long white arrow) and
cerebellar hypoplasia (short black arrow), enlarged
cisterna magna (long black arrow). B: calcifications in
frontal lobe. C: severe ventriculomegaly. D: increased
thickness of pachymeninges. Image obtained from de
Fatima Vasco Aragao et al.68.
10
Bol Med Hosp Infant Mex. 2023;80(1)
with probable ZIKV infection through adequate prenatal
screening, where all pregnant women should be eval-
uated to identify possible risk factors82.
Risk factors for ZIKV exposure include81-84:
Living in or having visited areas with active ZIKV trans-
mission in the past 15days or during pregnancy.
Having had unprotected sex in the 2 weeks before
conception with a partner who has traveled or lived
in areas with active virus transmission in the previous
6months, regardless of whether or not symptomatol-
ogy was present.
The clinical presentation of CZS is generally charac-
terized by disruption in brain development, both embry-
onically and at the fetal level20,85, characterized by
direct neurological disturbance followed by the collapse
of the fetal cranial vault due to decreased intracranial
hydrostatic pressure4,62,63.
For its study and diagnosis, PAHO has typied CZS
under the following criteria86:
A case of congenital syndrome suspected to be
associated with ZIKV infection is determined when the
live newborn presents:
Microcephaly, whose occipitofrontal head circumfer-
ence measurement is below the third percentile ac-
cording to standard growth charts for age, sex, and
gestational age at 24h after birth.
Maternal history of residence or travel to an area with
circulation or suspected circulation of ZIKV.
Unprotected sex with a partner with a history of res-
idence or travel to an area with ZIKV circulation or
suspected ZIKV circulation.
A case of congenital syndrome probably associated
with ZIKV infection is determined when the live new-
born meets the criteria for a case of congenital syn-
drome suspected to be associated with ZIKV infection
and with any of the following features:
Intracranial morphological alterations diagnosed by
any imaging method, excluding other known possible
causes.
Maternal history of having presented with exanthema
during pregnancy.
A case of congenital syndrome conrmed to be asso-
ciated with ZIKV infection:
When the live newborn of any gestational age
meets the criteria for a case of congenital syndrome
suspected of being associated with ZIKV infection
and in whom ZIKV infection has been laboratory
conrmed, regardless of the detection of other
agents.
Infection by vertical transmission can occur in any
trimester of pregnancy, regardless of whether the
mother has symptoms or not20,55,59,60. However, not all
products exposed to ZIKV become affected. It is esti-
mated that 70-80% of exposed products do not present
alterations suggestive of ZIKV infection. Of the 20-30%
who become infected, only 5-14% develop CZS. On the
other hand, about 4-7% culminate in fetal loss; the rest
are asymptomatic20.
The general malformations associated with CZS
are81:
Severe microcephaly, where the skull has partially
collapsed.
Decreased brain tissue with calcications in the sub-
cortical region.
Damage of the fundus, manifesting with macular
scarring and focal retinal pigmentary staining.
Congenital contractures such as clubfoot (talipes) or
arthrogryposis.
Congenital hypertonia limiting body movement short-
ly after birth.
It should be noted that microcephaly caused by ZIKV
is diagnosed under the following parameters81:
Dened dongenital microcephaly:
For live births:
Occipitofrontal circumference at birth less than the
3rdpercentile for gestational age and sex within the
rst 2weeks of life.
For stillbirths and elective terminations
• Occipitofrontalcircumferenceatbirthlessthanthe
3rdpercentile for gestational age and sex.
Possible congenital microcephaly
For live births
• Occipitofrontalcircumferencelessthanthe3rdper-
centile for age and sex after 6weeks of age.
For all pregnancy outcomes
• Microcephaly diagnosedorsuspectedbyprenatal
ultrasound in the absence of available postnatal
occipitofrontal circumference measurements.
The imaging methods for CZS diagnosis are ultraso-
nography (USG), computed tomography (CT), and
MRI87. It is important to remember that during preg-
nancy, ultrasounds should be performed every trimes-
ter for fetal and maternal risk screening during prenatal
control visits. Likewise, during the intrauterine stage, it
is also advisable to use USG and MRI; while in the
extrauterine stage, CT can be performed82,88.
Cranial alterations such as microcephaly and intracra-
nial calcications can be identied mainly at the end of
the second and beginning of the third trimester89. On the
other hand, images taken by CT or MRI can identify some
of the ndings that have been described in patients with
CZS, such as intracranial calcications, ventriculomegaly,
11
I.E. Crisanto-López et al. Congenital Zika syndrome
increased extra-axial uid, polymicrogyria (alterations of
neuronal migrations resulting in excessive cortical folds
and shallow sulci), reduction of brain parenchymal vol-
ume, cortical atrophy or malformation, hypoplasia of the
cerebellum or cerebellar vermis, delayed myelination, and
hypoplasia of the corpus callosum40,89-91.
In addition, laboratory diagnosis of CZS should be
made on clinical suspicion and identication of risk
factors by molecular testing to conrm or rule out the
presence of ZIKV, such as real-time polymerase chain
reaction or time-dependent neutralization of antibodies
(NNT), with blood samples from both the newborn and
the mother, or embryonic annexes such as the umbili-
cal cord or placenta, to prevent possible cases of
CZS92,93. Detection of immunoglobulin (Ig)M antibodies
is possible by enzyme-linked immunoadsorbent assay
and immunouorescence in cerebrospinal uid sam-
ples or by amniocentesis73,86,93. IgM antibodies can be
detected in serum from the 5th or 6th day after symp-
toms onset, and a positive serology requires a positive
NNT test to conrm infection. For this purpose, it is
important to strictly follow-up prenatal and postnatal
control, both for the mother and the newborn40,75.
Laboratory criteria for ZIKV infection in pregnant
patients are as follows. For conrmed cases, virus isola-
tion in a clinical specimen (blood/urine), nucleic acid
detection in a clinical specimen (blood/urine), and detec-
tion of NNT in IgM-positive specimens. For probable
cases, the presence of IgM antibodies, not conrmed by
NNT in a serum sample, seroconversion of virus-specic
IgG antibodies, or a four-fold increase in titer between
samples taken in the acute and convalescent phase, the
rst serum is collected at the onset of the disease, and
the second 10-14 days later. Furthermore, the detection
of NNT in samples with negative IgM and positive IgG
markers73. When congenital infection is suspected, it is
advisable to perform laboratory tests to determine the
presence of cytomegalovirus, herpes simplex, rubella,
human immunodeciency virus, toxoplasmosis, and syph-
ilis infection to rule out other possible etiologies54,69.
Treatment
Given the severe alterations caused by CZS, there is
no specic treatment, and therapeutic measures are
symptomatic, aimed at maintaining an adequate hydric
status for the patient, as well as the use of analgesics
and antipyretics. At present, some specic antiviral
treatments and the development of anti-Zika vaccines
are under study20,35,73,94.
Patients with SCZ require constant monitoring, mul-
tidisciplinary care, referral to early intervention and
stimulation centers, and family counseling20. Therefore,
preventive measures are of major importance to avoid
cases of CZS.
Preventive measures
According to PAHO and WHO, the best way to prevent
the infection of communicable diseases is to control their
vectors. Therefore, it is recommended to increase efforts
to implement effective strategies to reduce vector den-
sity, mainly of the transmitting mosquito, which is widely
distributed in the American continent, and the danger of
exposure increases due to the emerging risk of popula-
tion movement and migration95.
In countries without local cases of ZIKV infection, it
is recommended that patients with suspected ZIKV
infection be tested for the virus to identify the circulating
viral strains, the appropriate characterization of the out-
break, and the implementation of an adequate response.
In countries with local cases of ZIKV infection, it is
recommended to monitor the geographic spread of the
virus to delimit the affected area, assess the clinical
severity and public health impact, identify risk factors
associated with ZIKV infection, and identify circulating
ZIKV lineages. Any changes detected through surveil-
lance should be promptly reported to the national pre-
vention and control authorities to ensure that appropriate
action is taken on time73,95,96. It is also necessary to
take personal preventive measures, including wearing
appropriate clothing (cotton and neutral colors) that
covers most of the body (long-sleeved shirts, pants,
socks, or caps, especially during peak mosquito activity
hours). It is recommended to use mosquito nets on
windows, doors, and at night, on the child’s bed or crib.
Furthermore, use mosquito repellents containing dieth-
yltoluamide, picaridin, or icaridin according to the prod-
uct manufacturer’s instructions. In the house, look for
and destroy possible sources of mosquito breeding
sites by avoiding accumulations of stagnant water. For
couples who wish to conceive, and either partner has
traveled to high-risk areas, it is recommended that
pregnancy be postponed for at least 8weeks after the
onset of symptoms or last possible exposure to the
virus for women (symptomatic or not) or at least
6months after the onset of symptoms or last possible
exposure to the virus for men (symptomatic or not).
Sexual partners of pregnant women returning from
areas with local ZIKV transmission should maintain
barrier-protected sex throughout pregnancy95,96.
12
Bol Med Hosp Infant Mex. 2023;80(1)
In conclusion, CZS is a pathological entity of wide
distribution and easy transmission that can produce
severe affectations. Its clinical presentation varies from
asymptomatic to severe alterations that compromise the
quality of life and even the course of life. The main neo-
natal clinical manifestations are microcephaly, ventricu-
lomegaly, intracranial calcications, ocular alterations,
and congenital contractures. Its diagnosis is complex
due to the variety of its clinical manifestations and its
unspecicity; in addition, there is no specic treatment.
As health personnel, it is important to be constantly
updated on this subject to identify risk factors, make a
timely diagnosis and thus avoid both the effects during
embryological development and the spread of this virus
to more people in the population. Although there is no
treatment or vaccine, personal prevention measures
are straightforward and inexpensive, so it is essential
to know them and promote them to avoid further com-
plications. It is also important to continue with lines of
research to better understand the pathogenic pathways
through which it acts, optimize the diagnostic protocol,
and produce specic therapeutic measures.
Ethical disclosures
Protection of human and animal subjects. The
authors declare that no experiments were performed
on humans or animals for this study.
Confidentiality of data. The authors declare that
they have followed the protocols of their work center on
the publication of patient data.
Right to privacy and informed consent. The
authors have obtained the written informed consent of
the patients or subjects mentioned in the article. The
corresponding author has this document.
Conflicts of interest
The authors declare that they have no conicts of
interest.
Funding
No funding.
References
1. Petersen LR, Jamieson DJ, Powers AM, Honein MA. Zika virus. NEngl
J Med. 2016;374:1552-63.
2. Sharma V, Sharma M, Dhull D, Sharma Y, Kaushik S, Kaushik S. Zika
virus: an emerging challenge to public health worldwide. Can J Microbiol.
2020;66:87-98.
3. Honein MA. Recognizing the global impact of Zika virus infection during
pregnancy. NEngl J Med. 2018;378:1055-6.
4. Matos-Alviso LJ, Santos-Calderón LA, Reyes-Hernández KL, Reyes GU,
Santamaria AC, López CG, et al. Síndrome congénito por virus Zika,
conceptos básicos. Rev Salud Quintana Roo. 2017;10:33-6.
5. Freitas DA, Souza-Santos R, Carvalho LM, Barros WB, Neves LM,
Brasil P, et al. Congenital Zika syndrome: a systematic review. PLoS
One. 2020;15:e0242367.
6. World Health Organization. WHO Statement on the First Meeting of the
International Health Regulations (2005) (IHR 2005) Emergency Committee
on Zika Virus and Observed Increase in Neurological Disorders and Neo-
natal Malformations. Geneva: World Health Organization; 2016. Available
from: https://www.who.int/news/item/01-02-2016-who-statement-on-the-rst-
meeting-of-the-international-health-regulations-(2005)-(ihr-2005)-emergen-
cy-committee-on-zika-virus-and-observed-increase-in-neurological-disor-
ders-and-neonatal-malformations
7. Gulland A. Zika virus is a global public health emergency, declares WHO.
BMJ. 2016;352:i657.
8. World Health Organization. Zika Virus. Geneva: World Health Organiza-
tion; 2022. Available from: https://www.who.int/news-room/fact-sheets/
detail/zika-virus
9. World Health Organization. Assessment of Infants with Microcephaly in
the Context of Zika Virus-Interim Guidance-4March 2016. Geneva: World
Health Organization; 2016. Available from: https://www.apps.who.int/iris/
bitstream/10665/204475/1/who_zikv_moc_16.3_eng.pdf?ua=1
10. Collins MC, Waggoner JJ. Detecting vertical Zika transmission: emerging
diagnostic approaches for an emerged Flavivirus. ACS Infect Dis.
2019;5:1055-69.
11. SINAVE/DGE/SS: Sistema de Vigilancia Epidemiológica. Sistema de
Vigilancia Epidemiológica de Enfermedad Por Virus del Zika. Mexico:
Secretaría de Salud; 2021.
12. SINAVE/DGE/SS: Sistema de Vigilancia Epidemiológica. Defectos del
Tubo Neural y Craneofaciales. Casos Conrmados Por Asociación Epi-
demiológica de Síndrome Congénito Asociado a Zika, Mexico 2016-2021.
Mexico: Secretaría de Salud; 2021.
13. SINAVE/DGE/SS: Sistema de Vigilancia Epidemiológica. Dirección Ge-
neral de Epidemiología. Casos Conrmados Por Laboratorio de Síndro-
me Congénito Asociado a Zika, Mexico 2016-2021. Mexico: Secretaría
de Salud; 2021.
14. Organización Panamericana de la Salud. Instrumento Para el Diagnós-
tico y la Atención a Pacientes Con Sospecha de Arbovirosis. Whashin-
gton, DC: OPS/OMS; 2016.
15. Espinal MA, Andrus JK, Jauregui B, Waterman SH, Morens DM,
Santos JI, et al. Emerging and reemerging Aedes-transmitted arbovirus
infections in the region of the Americas: implications for health policy. Am
J Public Health. 2019;109:387-92.
16. Martins MM, Prata-Barbosa A, da Cunha AJ. Arboviral diseases in pe-
diatrics. JPediatr (Rio J). 2020;96 Suppl1(Suppl 1):2-11.
17. Dick GW, Kitchen SF, Haddow AJ. Zika virus. I. isolations and serologi-
cal specicity. Trans R Soc Trop Med Hyg. 1952;46:509-20.
18. Dick GW, Zika virus. II. pathogenicity and physical properties. Trans R
Soc Trop Med Hyg. 1952;46:521-34.
19. Macnamara FN. Zika virus: a report on three cases of human infection
during an epidemic of jaundice in Nigeria. Trans R Soc Trop Med Hyg.
1954;48:139-45.
20. Musso D, Ko AI, Baud D. Zika virus infection-after the pandemic. NEngl
J Med. 2019;381:1444-57.
21. Musso D, Gubler DJ. Zika virus. Clin Microbiol Rev. 2016;29:487-524.
22. Cortes-Escamilla A, Roche B, Rodríguez-López MH, López Gatell RH,
Alpuche-Aranda CM. Spatiotemporal patterns of dengue and Zika inci-
dence during the 2015-2018 outbreak of Zika in Mexico. Salud Publica
Mex. 2022;64:478-87.
23. Duffy MR, Chen TH, Hancock WT, Powers AM, Kool JL, Lanciotti RS,
etal. Zika virus outbreak on Yap Island, federated States of Micronesia.
NEngl J Med. 2009;360:2536-43.
24. Baud D, Gubler DJ, Schaub B, Lanteri MC, Musso D. An update on Zika
virus infection. Lancet. 2017;390:2099-109.
25. Musso D, Bossin H, Mallet HP, Besnard M, Broult J, Baudouin L, et al.
Zika virus in French Polynesia 2013-14: anatomy of a completed out-
break. Lancet Infect Dis. 2018;18:e172-82.
26. Faria NR, do Socorro da Silva Azevedo R, Kraemer MU, Souza R, Se-
quetin CM, Hill SC, et al. Zika virus in the Americas: early epidemiologi-
cal and genetic ndings. Science. 2016;352:345-9.
27. Baker RE, Mahmud AS, Miller IF, Rajeev M, Rasambainarivo F, Rice BL,
et al. Infectious disease in an era of global change. Nat Rev Microbiol.
2022;20:193-205.
28. Panwar U, Singh SK. An overview on Zika virus and the importance of
computational drug discovery. JExplor Res Pharmacol. 2018;3:43-51.
29. Guo M, Hui L, Nie Y, Tefsen B, Wu Y. ZIKV viral proteins and their roles
in virus-host interactions. Sci China Life Sci. 2021;64:709-19.
30. Laureti M, Narayanan D, Rodriguez-Andres J, Fazakerley JK, Kedzierski L.
Flavivirus receptors: diversity, identity, and cell entry. Front Immunol.
2018;9:2180.
31. Omasta B, Tomaskova J. Cellular lipids-hijacked victims of viruses. Viru-
ses. 2022;14:1896.
13
I.E. Crisanto-López et al. Congenital Zika syndrome
32. Sager G, Gabaglio S, Sztul E, Belov GA. Role of host cell secretory
machinery in Zika virus life cycle. Viruses. 2018;10:559.
33. Ropidi MI, Khazali AS, Rashid NN, Yusof R. Endoplasmic reticulum: a
focal point of Zika virus infection. JBiomed Sci. 2020;27:27.
34. Buchwalter RA, Ogden SC, York SB, Sun L, Zheng C, Hammack C, etal.
Coordination of Zika virus infection and viroplasm organization by micro-
tubules and microtubule-organizing centers. Cells. 2021;10:3335.
35. Kim S, Shin HY. Understanding the tissue specicity of ZIKV infection in
various animal models for vaccine development. Vaccines (Basel).
2022;10:1517.
36. Ramos LF, Martins M, Murillo JR, Barbosa DG, Perpétua OD, Sousa FN,
et al. Interspecies isobaric labeling-based quantitative proteomics reveals
protein changes in the ovary of Aedes aegypti coinfected with ZIKV and
Wolbachia. Front Cell Infect Microbiol. 2022;12:900608.
37. Vue D, Tang Q. Zika virus overview: transmission, origin, pathogenesis,
animal model and diagnosis. Zoonoses (Burlingt). 2021;1:1-29.
38. Mungin JW Jr., Chen X, Liu B. Interferon epsilon signaling confers atte-
nuated Zika replication in human vaginal epithelial cells. Pathogens.
2022;11:853.
39. Estévez-Herrera J, Pérez-Yanes S, Cabrera-Rodríguez R, Márquez AD,
Trujillo GR, Machado JD, et al. Zika virus pathogenesis: a battle for im-
mune evasion. Vaccines (Basel). 2021;9:294.
40. Desgraupes S, Hubert M, Gessain A, Ceccaldi PE, Vidy A. Mother-to-
child transmission of arboviruses during breastfeeding: from epidemiolo-
gy to cellular mechanisms. Viruses. 2021;13:1312.
41. Hasan S, Saeed S, Panigrahi R, Choudhary P. Zika virus: a global public
health menace: a comprehensive update. J Int Soc Prev Community
Dent. 2019;9:316-27.
42. Espinoza MM. Aspectos clínicos de la infección por el virus Zika. An Fac
Med. 2017;78:79-82.
43. Hussain T, Murtaza G, Kalhoro DH, Saleem KM, Yin Y, Ismail CM, et al.
Understanding the immune system in fetal protection and maternal infec-
tions during pregnancy. JImmunol Res. 2022;2022:7567708.
44. Tabata T, Petitt M, Puerta-Guardo H, Michlmayr D, Harris E, Pereira L.
Zika virus replicates in proliferating cells in explants from rst-trimester
human placentas, potential sites for dissemination of infection. JInfect
Dis. 2018;217:1202-13.
45. Miner JJ, Diamond MS. Zika virus pathogenesis and tissue tropism. Cell
Host Microbe. 2017;21:134-42.
46. Caldwell M, Boruah AP, Thakur KT. Acute neurologic emerging aviviru-
ses. Ther Adv Infect Dis. 2022;9:20499361221102664.
47. Salinas S, Foulongne V, Loustalot F, Fournier WC, Moles JP, Briant L,
et al. Le virus Zika-L’émergence d’une menace. Med Sci (Paris).
2016;32:378-86.
48. York SB, Sun L, Cone AS, Duke LC, Cheerathodi MR, Meckes DG Jr.
Zika virus hijacks extracellular vesicle tetraspanin pathways for cell-to-cell
transmission. mSphere. 2021;6:e0019221.
49. Zhao Z, Li Q, Ashraf U, Yang M, Zhu W, Gu J, et al. Zika virus causes
placental pyroptosis and associated adverse fetal outcomes by activating
GSDME. ELife. 2022;11:e73792.
50. Quicke K, Bowen JR, Johnson EL, McDonald CE, Ma H, O´Neal JT, etal.
Zika virus infects human placental macrophages. Cell Host Microbe.
2016;20:83-90.
51. Honein MA, Dawson AL, Petersen EE, Jones AM, Lee EH, Yazdy MM,
et al. Birth defects among fetuses and infants of us women with evi-
dence of possible Zika virus infection during pregnancy. JAMA.
2017;317:59-68.
52. Pereira L. Congenital viral infection: traversing the uterine-placental in-
terface. Annu Rev Virol. 2018;5:273-99.
53. Labib BA, Chigbu DI. Pathogenesis and manifestations of Zika
virus-associated ocular diseases. Trop Med Infect Dis. 2022;7:106.
54. Yu W, Hu X, Cao B. Viral infections during pregnancy: the big cha-
llenge threatening maternal and fetal health. Matern Fetal Med. 2021;
4:72-86.
55. Schuler-Faccini L, Del Campo M, García-Alix A, Ventura LO,
Bouquett JA, van der Linden V, et al. Neurodevelopment in children
exposed to Zika in utero: clinical and molecular aspects. Front Genet.
2022;13:758715.
56. Brasil P, Pereira JP Jr., Moreira ME, Ribeiro Nogueira RM, Damasceno L,
Wakimoto M, et al. Zika virus infection in pregnant women in Rio de
Janeiro. NEngl J Med. 2016;375:2321-34.
57. Pacheco O, Beltrán M, Nelson CA, Valencia D, Tolosa N, Farr SL, et al.
Zika virus disease in Colombia-preliminary report. N Engl J Med.
2020;383:e44.
58. Sadler TW. Embriología Médica con Orientación Clínica. Buenos Aires:
Médica Panamericana; 2019.
59. Carlson BM. Embriología Humana y Biología del Desarrollo. Madrid:
Saunders; 2019.
60. Arteaga M, García MI. Embriología Humana y Biología del Desarrollo.
Mexico City: Panamericana; 2021.
61. Gandhi S, Ezin M, Bronner ME. Reprogramming axial level identity to
rescue neural-crest-related congenital heart defects. Dev Cell.
2020;53:300-15.e4.
62. Xie X, Zeng J. Neuroimmune evasion of Zika virus to facilitate viral pa-
thogenesis. Front Cell Infect Microbiol. 2021;11:662447.
63. Gonik L, da Fonseca Oliveira AT, de Carvalho Chagas PS, da Silva
Frônio J. Auditory and language development assessment of newborns
aged one to four years exposed to gestational Zika virus infection: a case
series. Int J Environ Res Public Health. 2021;18:6562.
64. Aviña-Padilla N, López-Ortiz E, Aviña-Padilla K. Análisis epidemiológico
sobre el síndrome congénito asociado a Zika (CZS) en México: reporte
de caso clínico en Sinaloa. Arch Neurocien. 2019;24:34-45.
65. Faccini LS, Friedrich L, de Moura SK, Maria FD, da Silva Inácio de Bone S.
Neurological evaluation of microcephalic children with Zika syndrome
and congenital cytomegalovirus infection. eNeurologicalSci. 2022;
29:100417.
66. Hazin AN, Poretti A, Di Cavalcanti Souza Cruz D, Tenorio M, van der
Linden A, Pena LJ, et al. Computed tomographic ndings in microcepha-
ly associated with Zika virus. NEngl J Med. 2016;374:2193-5.
67. De Aguiar EB, Pone SM, Dos Santos Gomes Junior SC, Mendes SF,
Araujo ZA, Meira VZ, et al. Anthropometric parameters of children with
congenital Zika virus exposure in the rst three years of life. Viruses.
2022;14:876.
68. de Fatima Vasco Aragao M, van der Linden V, Brainer-Lima AM,
Ramos CR, Rocha MA, da Silva PS, et al. Clinical features and neuroi-
maging (CT and MRI) ndings in presumed Zika virus related congenital
infection and microcephaly: retrospective case series study. BMJ.
2016;353:i1901.
69. Lucignani G, Guarnera A, Rossi-Espagnet MC, Moltoni G, Antonelli A,
Figa TL, et al. From fetal to neonatal neuroimaging in TORCH infections:
a pictorial review. Children (Basel). 2022;9:1210.
70. Polonio CM, da Silva P, Russo FB, Hyppolito BR, Zanluqui NG, Benaz-
zato C, et al. microRNAs control antiviral immune response, cell death
and chemotaxis pathways in human neuronal precursor cells (NPCs)
during Zika virus infection. Int J Mol Sci. 2022;23:10282.
71. Turpin J, El Safadi D, Lebeau G, Krejbich M, Chatelain C, Despres P,
et al. Apoptosis during ZIKA virus infection: too soon or too late? Int J
Mol Sci. 2022;23:1287.
72. Huang WC, Abraham R, Shim BS, Choe H, Page DT. Zika virus infec-
tion during the period of maximal brain growth causes microcephaly
and corticospinal neuron apoptosis in wild type mice. Sci
Rep.2016;6:34793.
73. Troyano Luque JM, Suy A, Fernández MM, Viñuela MC, Bernabéu R,
Rodó C, et al. Procedimiento de Manejo de la Infección Por Virus Zika
Durante el Embarazo y en Recién Nacidos. Madrid: Asociación Españo-
la de Pediatría; 2017.
74. Secretaría de Salud. Atención y Seguimiento del Embarazo y su Rela-
ción con el Virus Zika. Lineamiento Técnico. Mexico: Secretaría de
Salud; 2016.
75. Da Cruz Costa CB, Freitas D. Ocular ndings of congenital Zika virus
infection with microcephaly. Int Ophthalmol. 2022;42:3117-27.
76. de Albuquerque BH, de Oliveira MT, Aderaldo JF, de Medeiros Garcia
Torres M, Lanza DC. Human seminal virome: a panel based on recent
literature. Basic Clin Androl. 2022;32:16.
77. Miner JJ, Diamond MS. Zika virus pathogenesis and tissue tropism. Cell
Host Microbe. 2017;21:134-42.
78. Martins MM, Alves da Cunha AJ, Robaina JR, Raymundo CE, Barbosa AP,
de Andrade Medronho R. Fetal, neonatal, and infant outcomes associa-
ted with maternal Zika virus infection during pregnancy: a systematic
review and meta-analysis. PLoS One. 2021;16:e0246643.
79. Coronell-Rodríguez W, Arteta-Acosta C, Suárez-Fuentes MA,
Burgos-Rolon MC, Rubio-Sotomayor MT, Sarmiento-Gutiérrez M, et al.
Infección por virus del Zika en el embarazo, impacto fetal y neonatal.
Rev Chil Infectol. 2016;33:665-73.
80. Di Villarosa do Amaral YN, Malacarne J, Brandão PG, Brasil P,
Nielsen-Saines K, Moreira ME. Time to evaluate the clinical repercus-
sions of Zika virus vertical transmission? a systematic review. Front
Psychiatry. 2021;12:699115.
81. Centers for Disease Control and Prevention. Congenital Zika Syndrome
and Other Birth Defects. Atlanta: Centers for Disease Control and Pre-
vention; 2022.
82. Secretaría de Salud. Norma Ocial Mexicana NOM-007-SSA2-2016, Para
la Atención de la Mujer Durante el Embarazo, Parto y Puerperio, y de la
Persona Recién Nacida. Mexico: Secretaría de Salud; 2016.
83. Centers for Disease Control and Prevention. Zika Prevención y Transmi-
sión. Atlanta: Centers for Disease Control and Prevention; 2018.
84. Halani S, Tombindo PE, O’Reilly R, Miranda RN, Erdman LK,
Whitehead C, et al. Clinical manifestations and health outcomes asso-
ciated with Zika virus infections in adults: a systematic review. PLoS Negl
Trop Dis. 2021;15:e0009516.
85. Paixao ES, Cardim LL, Costa MC, Brickley EB, Carvalho-Sauer RC,
Carmo EH, et al. Mortality from congenital Zika syndrome-nationwide
cohort study in Brazil. NEngl J Med. 2022;386:757-67.
86. Pan American Health Organization. Guía Para la Vigilancia de la Enfer-
medad por el Virus del Zika y Sus Complicaciones. Washington, DC: Pan
American Health Organization; 2018.
14
Bol Med Hosp Infant Mex. 2023;80(1)
87. Vhp L, Aragão MM, Pinho RS, Hazin AN, Paciorkowski AR, de Oliveira AC,
et al. Congenital Zika virus infection: a review with emphasis on the
spectrum of brain abnormalities. Curr Neurol Neurosci Rep.2020;20:49.
88. Kim E, Boyd B. Diagnostic imaging of pregnant women and fetuses: li-
terature review. Bioengineering (Basel). 2022;9:236.
89. Viens LJ, Fleck-Derderian S, Baez-Santiago MA, Oduyebo T,
Broussard CS, Khan S, et al. Role of prenatal ultrasonography and
amniocentesis in the diagnosis of congenital Zika syndrome: a systema-
tic review. Obstet Gynecol. 2020;135:1185-97.
90. Driggers RW, Ho CY, Korhonen EM, Kuivanen S, Jääskeläinen AJ,
Smura T, et al. Zika virus infection with prolonged maternal viremia and
fetal brain abnormalities. NEngl J Med. 2016;374:2142-51.
91. Ramos RC, de Barros Miranda-Filho D, Martelli CM, de Araújo TV,
Rocha MA, van der Linden V, et al. Characteristics of children of the
microcephaly epidemic research group pediatric cohort who developed
postnatal microcephaly. Sci Rep.2022;12:15778.
92. Center for Disease Control and Prevention. Guidance for U.S. Laborato-
ries Testing for Zika Virus Infection. Atlanta: Center for Disease Control
and Prevention; 2019.
93. Oduyebo T, Polen KD, Walke HT, Steiner SR, Larthrop E, Rabe IB, etal.
Update: interim guidance for health care providers caring for pregnant
women with possible Zika virus exposure-United States (Including
U.S. territories), July 2017. MMWR Morb Mortal Wkly Rep. 2017;
66:781-93.
94. Yeasmin M, Molla MM, Masud HM, Saif-Ur-Rahman KM. Safety and
immunogenicity of Zika virus vaccine: a systematic review of clinical trials.
Rev Med Virol. 2023;33:e2385.
95. World Health Organization. Zika Virus Disease. Geneva: World Health
Organization; 2016.
96. Protocolos de la Red Nacional de Vigilancia Epidemiológica. Protocolo
de Vigilancia de la Enfermedad por Virus Zika. Madrid: Red Nacional de
Vigilancia Epidemiológica; 2019.
... The virus's ability to cross the placental barrier and affect fetal neurodevelopment became a focal point of research and public health efforts. Pregnant women infected with Zika faced an elevated risk of transmitting the virus to the fetus, leading to a spectrum of congenital abnormalities collectively known as congenital Zika syndrome [2]. The Zika virus is a member of the Flaviviridae family and is primarily transmitted to humans through the bite of infected mosquitoes, particularly Aedes mosquitoes [3]. ...
Article
Full-text available
Background. Congenital Zika Syndrome presents a significant public health concern, particularly due to its association with severe neurological complications in infants born to maternal infected with the Zika virus during pregnancy. This article offers a comprehensive overview of the risks, complications, and preventive measures associated with Zika virus infection during pregnancy. Methods. A thorough literature search was conducted, encompassing studies, case reports, and clinical observations related to neurological complications in infants with congenital Zika syndrome. The review focuses on elucidating the intricate relationship between maternal Zika virus infection and the subsequent neurological challenges faced by newborns. Results. The neurological impacts of congenital Zika syndrome are multifaceted, ranging from the well-documented microcephaly to less explored manifestations such as brain abnormalities, ventriculomegaly, intracranial calcifications, seizures , and sensory impairments. Additionally, joint and limb contractures contribute to the complexity of congenital Zika syndrome-related neurological deficits. This narrative review synthesizes current knowledge on the subject, shedding light on the diverse ways in which Zika virus infection during pregnancy can affect the developing fetal nervous system. Conclusion. Understanding the intricate neurological complications associated with congenital Zika syndrome is crucial for healthcare practitioners, researchers, and policymakers. This review provides a comprehensive overview of the current state of knowledge, emphasizing the need for continued research, surveillance, and public health interventions to mitigate the impact of the Zika virus on maternal and child health.
... There was little attention paid to ZIKV infection until the first documented human outbreak in the Pacific islands in the late 2000s and the later epidemic in South America in 2015-2016 [8][9][10][11]. The ZIKV strains in the recent outbreaks belong to the Asian lineage and are associated with congenital Zika syndrome (microcephaly, cerebral calcifications, and macular scarring) and Guillain-Barre syndrome (GBS) [11][12][13][14][15][16]. There is no approved effective treatment or vaccine to control ZIKV infection [17]. ...
Article
Full-text available
Zika virus (ZIKV) is a mosquito-borne flavivirus and causes an infection associated with congenital Zika syndrome and Guillain–Barre syndrome. The mechanism of ZIKV-mediated neuropathogenesis is not well understood. In this study, we discovered that ZIKV induces degradation of the Numb protein, which plays a crucial role in neurogenesis by allowing asymmetric cell division during embryonic development. Our data show that ZIKV reduced the Numb protein level in a time- and dose-dependent manner. However, ZIKV infection appears to have minimal effect on the Numb transcript. Treatment of ZIKV-infected cells with a proteasome inhibitor restores the Numb protein level, which suggests the involvement of the ubiquitin–proteasome pathway. In addition, ZIKV infection shortens the half-life of the Numb protein. Among the ZIKV proteins, the capsid protein significantly reduces the Numb protein level. Immunoprecipitation of the Numb protein co-precipitates the capsid protein, indicating the interaction between these two proteins. These results provide insights into the ZIKV–cell interaction that might contribute to its impact on neurogenesis.
... The virus did not draw much attention until the first documented human outbreak in the Pacific islands in the late 2000s and the later epidemic in South America in 2015-2016 [3][4][5][6]. It is a mosquito-borne flavivirus associated with severe manifestations, including congenital Zika syndrome and Guillain-Barre syndrome (GBS) [6][7][8][9][10][11]. ZIKV infects neural precursor cells derived from pluripotent stem cells and causes apoptotic cell death and cell-cycle dysregulation [12][13][14]. ...
Preprint
Full-text available
Zika virus (ZIKV) is a mosquito-borne flavivirus and causes an infection that is associated with neurological manifestations, including microcephaly and Guillain-Barre syndrome. The mechanism of ZIKV-mediated neuropathogenesis is not well understood. In this study, we discovered that ZIKV induces the degradation of the Numb protein, which plays a crucial role in neurogenesis by allowing asymmetric cell division during embryonic development. Our data show that ZIKV reduced the Numb protein level in a time- and dose-dependent manner. However, ZIKV infection appears to have minimal effect on the Numb transcript. Treatment of ZIKV-infected cells with a proteasome inhibitor restores the Numb protein level, which suggests the involvement of the ubiquitin-proteasome pathway. In addition, ZIKV infection shortens the half-life of the Numb protein. Among the ZIKV proteins, the capsid protein significantly reduces the Numb protein level. Immunoprecipitation of the Numb protein co-precipitates the capsid protein, indicating the interaction between these two proteins. These results provide insights into the ZIKV-cell interaction that might contribute to its impact on neurogenesis.
Article
Viral infectious diseases are important causes of reproductive disorders, as abortion, fetal mummification, embryonic mortality, stillbirth, and congenital abnormalities in animals and in humans. In this chapter, we provide an overview of some virus, as important agents in teratology. We begin by describing the Zika virus, whose infection in humans had a very significant impact in recent years and has been associated with major health problems worldwide. This virus is a teratogenic agent in humans and has been classified as a public health emergency of international concern (PHEIC). Then, some viruses associated with reproductive abnormalities on animals, which have a significant economic impact on livestock, are described, as bovine herpesvirus, bovine viral diarrhea virus, Schmallenberg virus, Akabane virus, and Aino virus. For all viruses mentioned in this chapter, the teratogenic effects and the congenital malformations associated with fetus and newborn are described, according to the most recent scientific publications.
Article
Full-text available
The number of studies published on postnatal microcephaly in children with Congenital Zika Syndrome is small, clinical presentations vary and aspects of the evolution of these children remain unclarified. The present case series examined clinical characteristics and assessed the growth velocity of the head circumference, weight and height Z-scores in 23 children who developed postnatal microcephaly during follow-up in the Microcephaly Epidemic Research Group Pediatric Cohort. To estimate the change in the head circumference, weight and height Z-scores over time and compare the mean difference between sexes, we used multilevel mixed-effects linear regressions with child-specific random effects. Among these children, 60.9% (n = 14/23) presented with craniofacial disproportion, 60.9% (n = 14/23) with strabismus, 47.8% (n = 11/23) with early onset seizures, 47.8% (n = 11/23) with dysphagia and 43.5% (n = 10/23) with arthrogryposis. Of the 82.7% (n = 19/23) children who underwent neuroimaging, 78.9% (n = 15/19) presented with alterations in the central nervous system. Monthly growth velocity, expressed in Z-scores, of the head circumference was − 0.098 (95% CI % − 0.117 to − 0.080), of weight was: − 0.010 (95%-CI − 0.033 to 0.014) and of height was: − 0.023 (95%-CI − 0.046 to 0.0001). Postnatal microcephaly occurred mainly in children who had already presented with signs of severe brain damage at birth; there was variability in weight and height development, with no set pattern.
Article
Full-text available
Zika virus (ZIKV) is an arthropod-borne virus that belongs to the Flavivirus genus and is principally transmitted by Aedes aegypti mosquitoes. ZIKV infection often causes no or only mild symptoms, but it can also trigger severe consequences, including microcephaly in infants and Guillain-Barré syndrome, uveitis, and neurologic manifestations in adults. There is no ZIKV vaccine or treatment currently approved for clinical use. The primary target of ZIKV infection has been recognized as the maternal placenta, with vertical transmission to the fetal brain. However, ZIKV can also spread to multiple tissues in adults, including the sexual organs, eyes, lymph nodes, and brain. Since numerous studies have indicated that there are slightly different tissue-specific pathologies in each animal model of ZIKV, the distinct ZIKV tropism of a given animal model must be understood to enable effective vaccine development. Here, we comprehensively discussed the tissue specificity of ZIKV reported in each animal model depending on the genetic background and route of administration. This review should facilitate the selection of appropriate animal models when studying the fundamental pathogenesis of ZIKV infection, thereby supporting the design of optimal preclinical and clinical studies for the development of vaccines and therapeutics.
Article
Full-text available
Viral infections have always been a serious burden to public health, increasing morbidity and mortality rates worldwide. Zika virus (ZIKV) is a flavivirus transmitted by the Aedes aegypti vector and the causative agent of severe fetal neuropathogenesis and microcephaly. The virus crosses the placenta and reaches the fetal brain, mainly causing the death of neuronal precursor cells (NPCs), glial inflammation, and subsequent tissue damage. Genetic differences, mainly related to the antiviral immune response and cell death pathways greatly influence the susceptibility to infection. These components are modulated by many factors, including microRNAs (miRNAs). MiRNAs are small noncoding RNAs that regulate post-transcriptionally the overall gene expression, including genes for the neurodevelopment and the formation of neural circuits. In this context, we investigated the pathways and target genes of miRNAs modulated in NPCs infected with ZIKV. We observed downregulation of miR-302b, miR-302c and miR-194, whereas miR-30c was upregulated in ZIKV infected human NPCs in vitro. The analysis of a public dataset of ZIKV-infected human NPCs evidenced 262 upregulated and 3 downregulated genes, of which 142 were the target of the aforementioned miRNAs. Further, we confirmed a correlation between miRNA and target genes affecting pathways related to antiviral immune response, cell death and immune cells chemotaxis, all of which could contribute to the establishment of microcephaly and brain lesions. Here, we suggest that miRNAs target gene expression in infected NPCs, directly contributing to the pathogenesis of fetal microcephaly.
Article
Full-text available
Objective: Evaluate spatially and temporally simultaneous presence of clusters of dengue and Zika clinical cases and their relationship with expected dengue transmission risk. Materials and methods: A classification of dengue risk transmission was carried out for whole country, and spatial autocorrelation analyses to identify clusters of confirmed clinical cases of dengue and Zika from 2015 to 2018 was conducted using Moran's Index statistics. Results: Clusters of both diseases were identified in dengue-high risk munici-palities at the beginning of the outbreak, but, at the end of the outbreak, Zika clusters occurred in dengue low-risk mu-nicipalities. Conclusion: This study identified Zika clusters in low-risk dengue areas suggesting participation of several factors that favor virus introduction and dissemination, such as differences in entomological and control interventions, and the possibility of cross-immunity in the population.
Article
Full-text available
Background The seminal virome and its implications for fertility remain poorly understood. To date, there are no defined panels for the detection of viruses of clinical interest in seminal samples. Results In this study, we characterized the human seminal virome based on more than 1,000 studies published over the last five years. Conclusions The number of studies investigating viruses that occur in human semen has increased, and to date, these studies have been mostly prospective or related to specific clinical findings. Through the joint analysis of all these studies, we have listed the viruses related to the worsening of seminal parameters and propose a new panel with the main viruses already described that possibly affect male fertility and health. This panel can assist in evaluating semen quality and serve as a tool for investigation in cases of infertility.
Article
Full-text available
Over the millions of years-long co-evolution with their hosts, viruses have evolved plenty of mechanisms through which they are able to escape cellular anti-viral defenses and utilize cellular pathways and organelles for replication and production of infectious virions. In recent years, it has become clear that lipids play an important role during viral replication. Viruses use cellular lipids in a variety of ways throughout their life cycle. They not only physically interact with cellular membranes but also alter cellular lipid metabolic pathways and lipid composition to create an optimal replication environment. This review focuses on examples of how different viruses exploit cellular lipids in different cellular compartments during their life cycles.
Article
Full-text available
Several phase‐1 clinical trials have been performed to evaluate the safety and efficacy of candidate anti‐Zika vaccines. In this systematic review, we systematically evaluated the safety and immunogenicity of candidate vaccines, which would aid researchers in formulating an effective vaccination strategy for phase‐2 trials based on current evidence. A literature search was conducted using the electronic databases MEDLINE through Pubmed, Web of Science, and Cochrane Database for relevant studies on candidate anti‐zika vaccines. Studies on animal models were excluded from our study. Healthy individuals who were administered candidate Zika vaccines to evaluate the immune response and adverse events (AEs) compared to placebo were considered. Data were extracted, tabulated, and analysed using Microsoft Excel, while the risk of bias plots were generated using tidyverse and Robvis packages in R‐studio. A total of five phase‐1 clinical trials were included in our analysis comprising of studies on inactivated, viral vector, and DNA vaccines. Immunogenicity ranged from 10% to 100% after vaccination with the lowest seroconversion rate (10%) and geometric mean titre (GMT) (6.3; 95% confidence interval (CI):3.7–10.8) observed among recipients of single‐dose inactivated anti‐zika vaccine (ZPIV). For DNA vaccines, the seroconversion rate ranged from 60% to 100% with the highest seroconversion rate (100%) and GMT (2871; 95% CI:705.3–11688) observed among recipients of three shots of high dose GLS‐5700 vaccine. For viral vector vaccine (Ad26.ZIKV.001) seroconversion rate (100%) and GMT peaked after two shots with both low and high‐dose vaccines. In all those studies AEs were mostly local including injection site pain, erythema, and itching. The most common systemic AEs included fever, myalgia, nausea, and fatigue. In phase‐1 clinical trials, all candidate vaccines were found to be highly immunogenic and relatively safe, especially when administered in higher doses and with the help of needle‐free devices.
Article
Full-text available
Zika virus (ZIKV) can be transmitted from mother to fetus during pregnancy, causing adverse fetal outcomes. Several studies have indicated that ZIKV can damage the fetal brain directly; however, whether the ZIKV-induced maternal placental injury contributes to adverse fetal outcomes is sparsely defined. Here, we demonstrated that ZIKV causes the pyroptosis of placental cells by activating the executor gasdermin E (GSDME) in vitro and in vivo. Mechanistically, TNF-α release is induced upon the recognition of viral genomic RNA by RIG-I, followed by activation of caspase-8 and caspase-3 to ultimately escalate the GSDME cleavage. Further analyses revealed that the ablation of GSDME or treatment with TNF-α receptor antagonist in ZIKV-infected pregnant mice attenuates placental pyroptosis, which consequently confers protection against adverse fetal outcomes. In conclusion, our study unveils a novel mechanism of ZIKV-induced adverse fetal outcomes via causing placental cell pyroptosis, which provides new clues for developing therapies for ZIKV-associated diseases.
Article
Full-text available
Congenital infections represent a challenging and varied clinical scenario in which the brain is frequently involved. Therefore, fetal and neonatal neuro-imaging plays a pivotal role in reaching an accurate diagnosis and in predicting the clinical outcome. Congenital brain infections are characterized by various clinical manifestations, ranging from nearly asymptomatic diseases to syndromic disorders, often associated with severe neurological symptoms. Brain damage results from the complex interaction among the infectious agent, its specific cellular tropism, and the stage of development of the central nervous system at the time of the maternal infection. Therefore, neuroradiological findings vary widely and are the result of complex events. An early detection is essential to establishing a proper diagnosis and prognosis, and to guarantee an optimal and prompt therapeutic perinatal management. Recently, emerging infective agents (i.e., Zika virus and SARS-CoV2) have been related to possible pre- and perinatal brain damage, thus expanding the spectrum of congenital brain infections. The purpose of this pictorial review is to provide an overview of the current knowledge on fetal and neonatal brain neuroimaging patterns in congenital brain infections used in clinical practice.
Article
Full-text available
Zika virus (ZIKV) is an emerging flavivirus that causes congenital birth defects and neurological compilations in the human host. Although ZIKV is primarily transmitted through infected mosquitos, recent studies reveal sexual contact as a potential transmission route. In vagina-bearing individuals, the vaginal epithelium constitutes the first line of defense against viruses. However, it is unclear how ZIKV interacts with the vaginal epithelium to initiate ZIKV transmission. In this study, we demonstrate that exposing ZIKV to human vaginal epithelial cells (hVECs) resulted in de novo viral RNA replication, increased envelope viral protein production, and a steady, extracellular release of infectious viral particles. Interestingly, our data show that, despite an increase in viral load, the hVECs did not exhibit significant cytopathology in culture as other cell types typically do. Furthermore, our data reveal that the innate antiviral state of hVECs plays a crucial role in preventing viral cytopathology. For the first time, our data show that interferon epsilon inhibits ZIKV replication. Collectively, our results in this study provide a novel perspective on the viral susceptibility and replication dynamics during ZIKV infection in the human vaginal epithelium. These findings will be instrumental towards developing therapeutic agents aimed at eliminating the pathology caused by the virus.