ArticlePDF AvailableLiterature Review

Abstract

Since its discovery in 1955, the incidence and geographical spread of reported Oropouche virus (OROV) infections have increased. Oropouche fever has been suggested to be one of the most important vector-borne diseases in Latin America. However, both literature on OROV and genomic sequence availability are scarce, with few contributing laboratories worldwide. Three reassortant OROV glycoprotein gene variants termed Iquitos, Madre de Dios, and Perdões virus have been described from humans and non-human primates. OROV predominantly causes acute febrile illness, but severe neurological disease such as meningoencephalitis can occur. Due to unspecific symptoms, laboratory diagnostics are crucial. Several laboratory tests have been developed but robust commercial tests are hardly available. Although OROV is mainly transmitted by biting midges, it has also been detected in several mosquito species and a wide range of vertebrate hosts, which likely facilitates its widespread emergence. However, potential non-human vertebrate reservoirs have not been systematically studied. Robust animal models to investigate pathogenesis and immune responses are not available. Epidemiology, pathogenesis, transmission cycle, cross-protection from infections with OROV reassortants, and the natural history of infection remain unclear. This Review identifies Oropouche fever as a neglected disease and offers recommendations to address existing knowledge gaps, enable risk assessments, and ensure effective public health responses.
1
Emergence of Oropouche fever in Latin America: a narrative review
THE LANCET Infectious Diseases | Review |
Konrad
M
Wesselmann
1
,
Ignacio
Postigo-Hidalgo
2
,
Laura
Pezzi
1,3
,
Edmilson
F
de
Oliveira-Filho
2
,
Carlo
Fischer
2
,
Xavier
de
Lamballerie
1,3
,
Jan Felix Drexler
2
1Unité des Virus Émergents (UVE: Aix-Marseille Univ-IRD 190-Inserm 1207), Marseille, France
2Institute of Virology, Charité- Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
3Centre National de Référence (CNR) des Arbovirus, Marseille, France (L Pezzi, Prof X de Lamballerie); German Centre for Infection Research (DZIF), Berlin, Germany
https://doi.org/10.1016/S1473-3099(23)00740-5
Since its discovery in 1955, the incidence and geographical spread of reported Oropouche virus (OROV) infections have increased. Oropouche
fever has been suggested to be one of the most important vector-borne diseases in Latin America. However, both literature on OROV and
genomic sequence availability are scarce, with few contributing laboratories worldwide. Three reassortant OROV glycoprotein gene variants
termed Iquitos, Madre de Dios, and Perdões virus have been described from humans and non-human primates. OROV predominantly causes
acute febrile illness, but severe neurological disease such as meningoencephalitis can occur. Due to unspecific symptoms, laboratory
diagnostics are crucial. Several laboratory tests have been developed but robust commercial tests are hardly available. Although OROV is
mainly transmitted by biting midges, it has also been detected in several mosquito species and a wide range of vertebrate hosts, which likely
facilitates its widespread emergence. However, potential non-human vertebrate reservoirs have not been systematically studied. Robust animal
models to investigate pathogenesis and immune responses are not available. Epidemiology, pathogenesis, transmission cycle, cross- protection
from infections with OROV reassortants, and the natural history of infection remain unclear. This Review identifies Oropouche fever as a
neglected disease and offers recommendations to address existing knowledge gaps, enable risk assessments, and ensure effective public health
responses.
Key messages
Over the past 70 years, a notable increase in the incidence and geographical spread of reported Oropouche virus (OROV) infections
has been observed, highlighting a growing public health concern.
The OROV genome is tri-segmented, which allows for reassortment. Three OROV reassortants are known, among which Madre de
Dios and Iquitos viruses have been associated with human diseases.
Human OROV infection occurs through the bites of midges, but its presence in various mosquito species and a wide range of
vertebrate hosts contributes to the potential for widespread emergence of OROV. However, the virus’ transmission cycle is poorly
understood.
OROV causes febrile symptoms indistinguishable from common pathogens endemic to the Americas. Occasionally OROV infection
can result in severe conditions, such as meningoencephalitis.
Due to the virus’ unspecific symptoms, laboratory diagnostics are crucial. Several in-house molecular and serological tests have
been developed, but robust commercial tests are scarcely available.
Animal models of OROV infection are restricted to rodent models. The pathogenesis of OROV and its reassortants is poorly
understood. The possibility and extent of repeated infection by different OROV glycoprotein reassortants are unknown.
The gaps in understanding the epidemiology, ecology, and pathogenesis of OROV, including its reassortants, are substantial.
Addressing these knowledge gaps is crucial for developing risk assessments and effective public health strategies to combat
Oropouche fever, a prototypical neglected disease.
2
Introduction
Vector-borne diseases cause up to 28·8% of all emerging infectious
diseases.1 Arthropod-borne viruses (ie, arboviruses) comprise 39% of
all pathogenic viruses discovered in humans between 1897 and 2010,
and are a main cause of vector-borne diseases.2,3 The importance of
arboviruses is best illustrated by the dengue virus (DENV), which is
the most important arbovirus infecting over 100 000 000 humans
annually.4 In contrast to vector- borne parasites, the global burden of
arboviruses increased between 2007 and 2017.3 The true burden of
arboviral disease is probably still underestimated.5,6 Poor knowledge of
transmission cycles, non-specific clinical presentations, and scarcity of
robust diagnostic tools are among the main problems that make
arboviral infections and their effect on patient and public health
difficult to identify.7
Latin America is considered a hot spot of emerging arboviruses due to
its large biodiversity including relevant vertebrate reservoirs such as
bats, rodents, and primates, and multiple invertebrate taxa.8,9
Moreover, key emergence factors are present in the region, comprising
(1) ecological and socioeconomic factors, especially rapid population
growth and often arbitrary urbanisation; (2) favourable climatic
conditions for vector establishment and proliferation, and large
diversity of ecosystems; and (3) intense human migration and major
changes in land use, such as deforestation, illegal mining, and
intensification of agriculture.5,10,11 Along with increased global
connectivity, the Americas have therefore seen the emergence (and re-
emergence) of several arboviruses, such as DENV, chikungunya virus
(CHIKV), West Nile virus, and Zika virus (ZIKV), during the past 50
years. Nonetheless, only DENV and CHIKV are officially listed as
pathogens causing a neglected tropical disease (NTD) by WHO.12
However, the Pan American Health Organization has begun to
acknowledge additional arboviruses as emergent and has started
issuing guidelines for their diagnosis, treatment, and laboratory
detection in the region.13,14
The Pan American Health Organization considers DENV to be the
biggest threat in the Americas because of its four serotypes and its well
established urban transmission cycle.15 In addition, since their
emergence on the American continent in 2013, CHIKV and ZIKV have
caused millions of infections in the region, incurring approximately
140 000 disability-adjusted life- years every year and, in the case of
ZIKV, congenital mal- formations.1620 Other arboviruses, such as
Oropouche virus (OROV), Mayaro virus,21 or Venezuelan equine
encephalitis virus,22,23 are also considered emergent in the region.
Before the recent emergence of CHIKV and ZIKV from 2013 onwards,
OROV was claimed to be the second most frequent arbovirus in Brazil
after DENV, allegedly accounting for more than half a million
estimated cases in Central and South America since its first
identification.24,25
Upon closer examination of OROV cases, estimates of its relevance
have relied on a few small-scale studies;2628 therefore, the true burden
of OROV-induced disease remains unknown. Although Oropouche
fever could be a prototypic NTD, incurring considerable health burden
in affected countries, knowledge of OROV and the disease it causes is
scarce.
This Review discusses the current state of research on OROV and
identifies research gaps requiring further investigation to enhance
patient care, enable risk assessments, and ensure effective public health
responses to Oropouche fever.
Genetic features of OROV and its reassortants
OROV’s genomic sequence availability
OROV belongs to the Simbu serogroup of the viral genus
Orthobunyavirus in the Peribunyaviridae family. The OROV genome
consists of three single-stranded, negative-sense RNA molecules:
small (S), medium (M), and large (L) segment. The S segment encodes
the nucleocapsid and a non-structural protein (NSs) in overlapping
open reading frames; the M segment encodes the two glycoproteins Gc
and Gn, and a non- structural protein (NSm); and the L segment
encodes the RNA-dependent RNA polymerase (figure 1A).2931
513 sequences, including 50 sequences of OROV reassortants, were
retrieved from GenBank. Most of those sequences corresponded to the
S segment (n=256); this could be because this segment is the most
conserved and the target of most PCR systems (compared with 142 M-
derived sequences and 115 L-derived sequences), which restricts our
knowledge and analysis of those genomic segments (figure 1B). This
small number of sequences probably represents only a fraction of the
existing OROV genetic diversity that is additionally biased towards
few sampling sites and dates.
OROV reassortants
OROV’s tri-segmented genome is susceptible to reassortment when
two viruses co-infect a single cell, resulting in progeny with mixed
genomic segments.3032 These events are important drivers of genetic
divergence, potentially altering vector competence and disease severity
and therefore facilitating viral emergence.31,32 Among
orthobunyaviruses circulating in South America, such as Fort Sherman
virus and Jatobal virus, reassortment events are frequent.30,33 Because
arboviral surveillance in the Americas is scarce, the number of OROV
reassortants is possibly underestimated.30,34 Furthermore, experimental
evidence shows possible reassortment between OROV and non-
American ortho- bunyaviruses, such as Schmallenberg, Bunyamwera,
and Oya viruses,32 underscoring the risk of new OROV reassortants
emerging in the future.
Three OROV reassortants termed Iquitos virus (IQTV), Madre de Dios
virus (MDDV), and Perdões virus (PDEV) have been identified. These
reassortants contain the S and L segments of OROV, the M segment of
unknown viruses, and, in the case of IQTV, an MDDV-related M
segment, as highlighted by the high sequence distances of the
reassortant M segments as compared with OROV (figure 2A) and the
common ancestor of MDDV and IQTV in an M-based pyhlogenetic
tree (figure 2B).29,34,37,38,39 The M segment’s genetic diversity could be
influenced by evolutionary pressure on the glycoproteins targeted by
antibody-mediated immune response29 and suggests that M segment-
based reassortment could provide evolutionary benefits for evading
population-level immunity, whereas S and L segments could be
functionally linked due to the interaction of derived proteins during
replication.29,40
Data concerning the serological relationship between OROV and its
reassortants, including IQTV, are scarce. Experiments have shown that
mouse antisera against IQTV only weakly neutralise OROV, indicating
distinct serotypes, with no reciprocal effect observed.38 IQTV infection
in two Peruvian patients triggered an increase in potentially pre-
existing OROV immune responses, suggesting conserved epitopes but
insufficient cross-protection.38 The potential absence of cross-
protection coupled with shared antigenicity among reassortants could
result in antibody-dependent enhancement (ADE), a process that likely
contributes to severe secondary DENV infections.41 Therefore, as
OROV and its reassortants coexist, investigating their serological
relationship is crucial to gauge potential ADE risks and any limitations
in cross-protection.41
Bibliographic analysis of publications on OROV
Literature on OROV is scarce. Only 192 peer-reviewed papers
published since July 1, 1961, to June 30, 2023, were retrieved from
PubMed, highlighting the sparse research interest in OROV (figure
3A). We also conducted a Boolean search on the Web of Science on
March 31, 2023, using the terms: “OROV” OR “virus del oropuche”
OR “Oropouche virus” OR “Oropouche” OR “Oropuche” OR “Iquitos
virus” OR “Madre de Dios virus” OR “Perdoes virus” (Title), OR
“OROV” OR “Virus del oropuche” OR “Oropouche virus” OR
“Oropouche” OR “Oropuche” OR “Iquitos virus” OR “Madre de Dios
virus” OR “Perdoes virus” (Abstract), AND NOT poecilla OR guppy
(Abstract) AND NOT trinidad (Abstract).
3
The search revealed that OROV research is conducted by a small
number of groups; the top five authors contributed 67 articles, which
represent 35% of all the articles on the subject. Further breakdown of
these data indicates that most of these articles come from Brazil (57%)
and the USA (38%). Bibliographic analysis of OROV literature
showed high bibliographic coupling (two given works sharing a
common reference) for articles published in the past decade (figure
3B). High bibliographic coupling can be explained by the low numbers
of publications and little heterogeneity in literature, with the few
researchers working on OROV citing the same papers because of
insufficient new evidence.
Geographical distribution of OROV and its reassortants
Since its initial detection in 1955 in Trinidad and Tobago,42 OROV has
been detected across Central and South America, spanning 5000 km
south and 2000 km west, and its incidence has increased over the past
several decades (figure 4). As of June, 2023, reports of OROV in
human and non-human hosts have emerged pre- dominantly from rural
and forest areas in Brazil,26,28,29,4465 Peru,6673 Ecuador,74,75,76,77
Argentina,43 Bolivia,74 Panama,26 Colombia,7880 and Venezuela.37 The
virus’ first Caribbean detection—after its initial discovery in Trinidad
and Tobago in 1955occurred in Haiti in 2014,81 followed by a 2016
outbreak in non-endemic coastal regions of Ecuador and Peru76,82 and
its emergence in a densely populated urban zone in northeastern
Brazil.63 In 2020, nearly half of a small rainforest village in French
Guiana experienced a dengue-like syndrome, with OROV diagnosed
in 43% (41/95) of inhabitants, suggesting OROV can present with high
attack rates.83 Colombian data from 2019 to 2021 attribute 11%
(87/791) of acute febrile disease during this time period to OROV, with
16% (92/568) of Colombian patients testing positive for OROV IgG
antibodies in chemiluminescent immunoassays.84 Altogether, the
available literature suggests a wide geographical expansion of OROV.
IQTV was first isolated in 1999 from a patient in Iquitos, Peru, and has
caused at least 160 human cases between 2005 and 2006.38 In 2006,
15·4% (160/1037) of patients with undifferentiated febrile illness from
Iquitos showed IQTV neutralising antibodies in plaque reduction
neutralisation tests, which is slightly higher than the 14·9% (154/1037)
found for OROV. Additionally, 3·4% (35/1037) had antibodies to both
viruses, suggesting cocirculation or substantial cross-neutralisation.38
MDDV was identified in a febrile patient in Peru39 in 2007, and in a
white-faced capuchin monkey in Venezuela in 2010, nearly 2000 km
away.37 PDEV has only been isolated once in 2012, from two black-
tufted marmosets in Minas Gerais, Brazil.29
Clinical presentation, treatment, and vaccine
Most information available on the clinical presentation of the infection
with OROV and its reassortants stems from case reports or small-scale
outbreaks. Therefore, the evidence available on clinical presentation
and sequelae is poor, and morbidity and mortality could be
underestimated.
Oropouche fever is generally mild and self-limiting and often begins
with a 310-day incubation period,44,62 after which patients develop a
febrile illness with headache, arthralgia, myalgia, nausea, vomiting,
chills, and photophobia.72,82,83,85 Some patients have less common
symptoms, such as rash, retro-orbital pain, anorexia, and haemorrhagic
manifestations.44,72,82 Severe clinical presentations, which are rare,
could involve CNS infections that result in aseptic meningitis or
meningoencephalitis.57,62,64,83,86,87 The disease is typically biphasic,
with an acute phase lasting 24 days, followed by a remission and a
resurgence of symptoms 710 days after onset.64,83 Most patients
recover without sequelae, although persistent myalgia and asthenia
lasting up to 1 month have been reported.83 No deaths have been
attributed to OROV infection yet. Similarly to OROV, IQTV infection
presents with fever, headache, eye pain, body pain, arthralgia,
diarrhoea, and chills.38 MDDV was reported to cause undifferentiated
febrile illness, and no human infection has been recorded for
PDEV.29,39 The rate of asymptomatic infections is difficult to estimate
as studies generally include febrile patients; a single study reported
symptoms in up to 63% of patients with OROV infection.65
Co-infections of OROV with reassortants have not been reported.
However, acute infections of OROV and other arboviruses can occur,
as observed in Peru and Colombia with DENV.72,73,84 Co-infections of
OROVZIKV and OROVCHIKV have been described at low
detection rates,72 as well as a triple co-infection of OROVDENV
CHIKV identified once in western Brazil,86 but the effect of co-
infections by OROV and other co-circulating arboviruses on disease
severity remains unclear.
Currently, no specific antiviral treatment against OROV infection is
available. Ribavirin, a broad-spectrum antiviral, was ineffective
against OROV in vivo in mice;88 mycophenolic acid, which has shown
in vitro activity against yellow fever virus, DENV, and Semliki Forest
virus, was also ineffective against OROV.89,90 Interferon alfa can
restrict viral replication in vitro and in vivo in mice when administrated
early or pre-infection, but its clinical relevance is unclear.91
Although no vaccine is currently available for OROV, a single study88
presented a candidate vaccine based on a replication-competent
chimeric vesicular stomatitis virus expressing complete or truncated
OROV glycoproteins, which showed a reduction in viral loads and
symptom severity in mice; moreover, another study predicted novel
epitope candidates for epitope-based peptide vaccine design against
OROV using computational methods.92 Other orthobunyavirus
vaccines could guide the development of OROV vaccines. A
chemically inactivated Schmallenberg virus (SBV) vaccine is licensed
for veterinary use in the EU,93 and an SBV nucleoprotein fragment-
based vaccine has been shown to reduce viraemia in infected mice.94
Evidence regarding vaccine-induced cross-protection in the Simbu
serogroup is scarce. A trivalent vaccine comprising two inactivated
Simbu serogroup viruses, Akabane virus (AKAV) and Aino virus
(AINOV), failed to prevent SBV infection in cows.95
Laboratory diagnosis
Identifying the causative agent of acute febrile illness without
laboratory tests is hardly feasible and sometimes inaccurate. Therefore,
many treatable diseases with non- specific symptoms are misdiagnosed
as dengue or malaria.7,96 Similarly, discrimination between OROV and
other pathogens causing acute febrile illness based solely on clinical
symptoms was shown to be unreliable.73 Consequently, patients are
being misdiagnosed or untreated and outbreaks could remain
unrecognised. To verify acute OROV infections, laboratories typically
employ three primary diagnostic criteria: a positive PCR result, the
identification of specific IgM, or the detection of seroconversion
through paired samples. Historically, serological methods were
predominant but have been gradually replaced by molecular
methods.24,27,40,85,97
Molecular diagnosis
Molecular detection of OROV is typically possible during the first
week on acute-phase specimens.24,40,61,85,98 Real- time RT-PCR in sera
collected during the first 5 days of illness has shown a 93% detection
rate.98 Serum viral loads can range from 10⁴ to 10⁸ genome copies per
mL,98 and the presence of low viraemia in recent cases highlights the
need for sensitive PCR assays.84 Real-time RT-PCR is preferred for
early-stage diagnosis due to its speed, ease, and reduced contamination
risk. Various in-house tests have been developed,51,61,77,84,98,99,100
including multiplexed formats for detecting co-circulating Mayaro
virus.101 Most assays target the OROV S segment,77,98,100,101 which does
not distinguish between OROV and its reassortants. Typing can be
enabled by including M segment-based primers.61,84,102 Conventional
RT-PCR is widely used for research51,61,77,98,99,100,102 alongside virus
isolation.76,79,83
Serum or plasma are the preferred samples for OROV
diagnosis.51,54,72,79,83,84 In patients with OROV infections involving the
4
CNS, viral RNA detection, viral isolation from cerebrospinal fluid
(CSF), and IgM detection in CSF have been reported.57,62,64
Furthermore, OROV has been detected by PCR in saliva and urine
collected from symptomatic patients within 5 days of disease onset,61,63
suggesting that these samples could serve as alternative specimens for
OROV detection during the acute phase of infection. Additionally,
although impractical for individual patient testing due to high cost and
reduced sensitivity, metagenomic sequencing could serve as an
additional environmental surveillance tool. Pooled vector-derived or
patient-derived samples can provide genetic information of circulating
virus strains. The use of samples from wastewater has also been
explored for flaviviruses,103 but further exploratory analysis is needed
to assess whether this approach can be employed for other arboviruses,
especially OROV. Further implementation of these methods will help
to clarify OROV genetic diversity, enabling the improvement of
diagnostic assays, vaccines, and antivirals.103,104
Serological diagnosis
After acute infection, molecular testing should be supplemented by
serological methods. Immune responses against OROV are not well
studied, but serological procedures can detect OROV-specific IgM and
IgG in serum, plasma, and CSF.24,40,56,59,64,67,71,84,85 Ideally, IgM testing
in CSF should be done together with IgM testing in paired serum
samples and supported by assessments of bloodbrain barrier leakage
to confirm intrathecal immunoglobulin synthesis.105
Existing in-house serological tools include enzyme immunoassays,
neutralisation tests, complement fixation tests, immunofluorescence
tests, and haemagglutination inhibition tests.74,75,28,42,45,48,70,106 Tests
based on the nucleocapsid (N) protein, which elicits a strong humoral
immune response, have been developed;84,107,108 however, these tests
show cross-reactivity due to the conservation of epitopes on N proteins
among many Simbu serogroup viruses.107110 Although commercial Gc
or N antigens are not available for OROV, robust commercial immuno-
assays are hardly available. Neutralisation tests are considered the gold
standard for arbovirus serology,13 but they are not often used for patient
diagnosis due to long turnaround times and the need for handling
infectious viruses under biosafety level 3 conditions.
Robustness of laboratory diagnosis in articles reporting infection
with OROV and its reassortants
55 articles published between 1960 and 2022 indicate a shift in
diagnostic tools used for OROV and reassortant infections (figure 5).
Before the 2010s, serological methods were commonly used,
particularly in retrospective studies.28,4550,53,68,70,106 However, in the
past 15 years, the use of robust techniques (eg, RT-PCR and
sequencing) has greatly enhanced the accuracy of detecting OROV
infections and differentiating them from their reassortants.29,38,39,44,57,60
64,66,67,72,76,79,8183,86 Nonetheless, OROV and its reassortants are most
probably still underdiagnosed or misdiagnosed due to symptoms
resembling those of classical arboviral diseases reported in Central and
South America, highlighting the need for effective differential
laboratory testing.72
Transmission cycles of OROV
Understanding the arboviral transmission cycle allows for the
identification of geographical areas and people at risk for infection.
OROV is probably maintained through a sylvatic and an urban cycle
(appendix p 1). The biting midge Culicoides paraensis is considered
the main OROV vector because of viral isolation during outbreaks and
vector competence observed in vivo under laboratory conditions.46,47
49,65,111 Humans are the suspected link between the sylvatic and urban
transmission cycles given that C paraensis is present in both urban and
rural settings.40 Some OROV outbreaks have also coincided with
human-driven environmental changes, such as deforestation, increased
agriculture, and infrastructure development.69,82,83,112 These changes
can alter vector distribution and increase contacts among vectors,
vertebrate reservoirs, and humans.113
Humans are the major vertebrate hosts in the urban cycle of
OROV.38,68,71 Whether urban OROV transmission is transient,
introduced via human movements, or permanent but at low levels and
therefore undetected is unclear, and the potential role of domestic
animals as amplifying hosts requires further investigation.34,46,102,114
The sylvatic cycle is less understood than the urban cycle, with
definitive reservoirs and amplifying hosts yet to be identified. OROV
antibodies have been found in wild birds, sloths, non-human primates,
and rodents,46,47,78,115,116,117 alongside sporadic isolations from sloths
and non-human primates in Brazil.45,53,102 Systematic studies of OROV
non-human reservoir hosts are scarce.
The detection of OROV in a coastal region of Ecuador where
C paraensis is absent (or yet to be found118) suggests the possibility of
other vectors involved in OROV transmission.76 Minor mutations can
enhance vector- borne transmission, as evidenced by CHIKV adaption
to Aedes albopictus.119 Culex quinquefasciatus, a primary and
widespread vector of West Nile virus,5 was suspected to be involved in
OROV transmissions in Brazil and French Guiana.44,83,120
Conversely, low efficiency in viral transmission has been observed
experimentally for C quinquefasciatus.121123 The main DENV vectors,
Aedes aegypti and A albopictus, have been found to be ineffective
OROV vectors,121 whereas other mosquitoes belonging to the species
Coquillettidia venezuelensis,42 Aedes serratus,42,45,124 Psorophora
cingulata, and Haemagogus tropicalis were found to be naturally
infected by OROV.124 However, vector competence studies for these
mosquito species are missing. If confirmed, this wide vector range
would be only comparable with few other clinically relevant
arboviruses, such as Japanese Encephalitis virus and Rift Valley fever
virus.5 Although OROV dissemination seems restricted to South and
Central American regions, both primary and supposed secondary
vectors are not,122,125 highlighting the risk of further dissemination and
emergence in other parts of the world. A study estimated that 25
million people were at direct risk of exposure to OROV in regions up
to the southern USA.126
Animal models
To date, hamster and mouse models have been described for OROV
infection but not for reassortants. In immunocompetent Syrian golden
hamster models, OROV causes systemic disease analogous to humans,
as well as liver injury and severe neurological symptoms, such as motor
deficits and paralysis.127,128 In immuno- competent mice, OROV
infection outcomes range from no clinical signs, to few,88,129131 and to
low mortality rates.132 OROV infection in mice lacking the receptor for
type I interferon (IFN) induces increased mortality rates and caused
extensive liver and spleen damage.129,130 Suckling mice develop severe
neurological symptoms before death with no observed
hepatotropism.89,91,133,134 OROV hepatotropism has been observed in
naturally infected non-human primates, with OROV and PDEV
isolated from marmosets,29,135 but non-human primate models that
better characterise infection by OROV and reassortants have not yet
been developed.
Pathogenesis
OROV infection elicits varied cellular and systemic responses,
involving multiple host factors. Peripheral blood mononuclear cells are
supposed initial targets of OROV, but their virus replication capacity
is relatively small.136 Plasmacytoid dendritic cells important for type I
IFN and proinflammatory cytokines production, are also probable
targets.130
A key aspect of OROV immune evasion is its NSs protein, which
antagonises IFN and could induce apoptosis as observed in other
pathogenic orthobunyaviruses, sidestepping immune responses.137,138
Additionally, microRNAs miR-217 and miR-5763p increase during
infection, which potentially inhibits the antiviral IFN beta response.139
5
For cellular entry, lipoprotein-related protein 1 is a key host factor
aiding OROV entry, as it is also for Rift Valley fever virus,
highlighting its role in bunyavirus infections.140
In severe cases, OROV can affect the CNS; however, the exact
infiltration mechanism remains unclear. An in vivo mouse study
indicated bloodbrain barrier leakage and a possible neural pathway
for initial CNS invasion.134 OROV disrupts IFN responses in human
primary astrocytes in vitro, potentially leading to neuropathology.141
However, ex vivo studies in adult human brains found no OROV-
infected astrocytes, whereas microglia were more commonly
infected.142
Gaps of knowledge and recommendations
Genetics and reassortants
No instances of OROV reinfection have been recorded, and examining
the serological relationship between OROV and its reassortants
remains crucial. Similarly, the impact of reassortment on OROV host
range and pathogenesis remains unknown. We recommend that in vitro
investigations are conducted to explore the potential for reassortment
between OROV and other American orthobunyaviruses. Furthermore,
orthobunyavirus surveillance capabilities should be enhanced to
distinguish and probe for reassortant viruses.
Disease surveillance and epidemiology
OROV surveillance is suboptimal, with incomplete diagnostic panel
inclusion of the virus and its reassortants. Clinical understanding is
based on minor outbreaks or isolated cases, necessitating longitudinal
studies to describe asymptomatic infection and define clinical
presentation and potential long-lasting sequelae. The correlation of
specific symptoms or their severity with OROV and reassortant
infection also requires further investigation. Existing seroprevalence
studies are scarce. Consequently, reliable data to estimate the true
extent of OROV circulation or to distinguish it from its reassortants are
currently lacking. We recommend that comprehensive longitudinal
studies and cross-sectional seroprevalence studies are implemented to
improve understanding of eventual long-lasting or chronic effects of
OROV fever, and to monitor the circulation of OROV and its
reassortants to assess the potential for reinfection or multiple infections
by different OROV reassortants. Metagenomics combined with vector
or patient pool- based analyses can improve genomic surveillance and
allow investigation of co-infections and vectors. The potential of
wastewater surveillance for the detection and monitoring of OROV and
other circulating arboviruses should also be explored.
Entomological and environmental surveillance
Despite isolating OROV from various species, the role of mosquitoes
in transmission cycles remains unclear.121 Should OROV better adapt
to the prevalent vectors A aegypti and A albopictus, its geographical
spread could be substanially widened and, in regions where OROV is
already present, infections could increase.119 The role of C
quinquefasciatus, given its wide geographical spread, needs
clarification.120 More vector competence studies are therefore
necessary. Furthermore, information on OROV’s transmission cycle is
either outdated or incomplete. Virus isolation from reservoirs has been
sporadic; only a handful of articles report isolation of OROV53,135 or
reassortants37,135 from non-human hosts. Additionally, the
dissemination of these hosts does not completely coincide with regions
reporting OROV outbreaks. For example, the sloth and capuchin
species, from which OROV and MDDV have been isolated, are native
solely to northwestern South America143,144far below the
geographical range of described OROV infections. Surveillance of
sylvatic reservoirs and emergence events needs to be increased, and the
potential for long-distance OROV dissemination (eg, by migratory
birds) requires investigation. We recommend that currently known
sylvatic reservoirs are confirmed and that further reservoirs and
amplifying hosts are identified, especially in regions with outbreaks
within the past decade or in regions with an apparent absence of known
reservoirs.
Detection tools
The evaluation of OROV diagnostics remains difficult due to the
absence of reference material, external quality assessments, and
comparative studies. Information about viral loads and immune
response kinetics in various body fluids stems only from isolated cases.
Longitudinal studies are essential to identify optimal specimens for
diagnosis and standardise sampling for diagnostics and research.
Additionally, exploring potential correlations between prolonged
viraemia or high viral loads and disease severity, akin to other
arboviruses, could be valuable.145 Current molecular and serological
assays often struggle to differentiate between OROV and its
reassortants, and commercial tests are mostly unavailable. Laboratory
diagnostics of OROV should therefore be improved. The development
of point-of-care rapid diagnostic tools could help implement OROV
diagnostics on a larger scale, especially in rural areas. Pathogenesis,
animal models, and cross-protection OROV pathogenesis is poorly
understood, and factors leading to severe disease remain to be
identified. Reverse genetics systems developed for OROV could be
exploited for this purpose.137,146 Animal models are restricted to
rodents, but the development of adequate animal models will be
essential for OROV pathogenesis and vaccine research, including
assessments of potential ADE. Safe human challenge models have
proven to be valuable for DENV research and could be explored for
OROV in the future.147 The amino acid divergence in the M segment
between OROV and IQTV or MDDV resembles that of other Simbu
serogroup viruses, for which an experimental vaccine failed to afford
cross-protection. The available genomic and serological data for
OROV and its reassortants suggest that development of vaccines
against Oropouche fever could be challenging. We recommend that
animal models for OROV and reassortants are expanded, that the cross-
protection potential of existing vaccines be explored for OROV, and
that the development of an effective, safe, stable, and affordable
OROV vaccine is prioritised.
Conclusion
OROV has the potential to emerge as a substantial threat given its wide
host and vector range, diverse environmental spread, potential for
severe course of disease, and existence of human-infecting reassortants
that could correspond to different serotypes. Despite the promising
increase in research, knowledge of the virus lags compared with other
American arboviruses. Intensifying international collaboration,
including between Latin American countries in which OROV is
endemic, and funding to bridge these knowledge gaps is imperative, as
this will simultaneously enhance our understanding of other neglected
diseases and inform public health policies. Therefore, OROV
represents a prototypic NTD that necessitates investigation to assess
and minimise its health burden.
Contributors
KMW, IP-H, and LP contributed to the literature search, figure conceptualisation and
realisation, and writing of the original draft. EFdO- F and CF contributed to figure
conceptualisation and data analysis. XdL contributed to t he study design. JFD contributed to
the study design and editing and writing of the final dra ft. All authors were involved in
reviewing the manuscript.
Declaration of interests
We declare no competing interests.
Acknowledgments
This study was supported by the Deutsche Forschungsgemeinschaft project COALITION
(project number DR 810/6–1), t he European Union’s Horizon 2020 Research and Innovation
programme through the ZIKAlliance pro ject (grant number 734548), and the Ho st Switching
Pathogens, Infectious Outbreaks and Zoonosis (HONOURS) innovative training network
(grant number 721367).
6
Figure 1: OROV structure and genomic sequence availability
(A) OROV structure and genomic organisation. (B) Nucleotide sequences available on GenBank for each segment up to June,
2023. A total of 15 sequences (segments: small [S], n=11; medium [M], n=2; and large [L], n=2) labelled “unverified” on
GenBank (ie, GenBank staff could not verify the accuracy of the submitted sequences) were excluded. For S and L segments,
sequences from OROV and reassortants are merged; for the M segment, sequences from OROV and reassortants are
represented separately. Information on the genomic sequences used for figure 1B is available in the appendix. IQTV=Iquitos
virus. MDDV=Madre de Dios virus. NSm=non-structural protein M. NSs=non-structural protein S. OROV=Oropouche virus.
PDEV=Perdões virus. RdRp=RNA-dependent RNA polymerase. UTR=untranslated region. Figure created with BioRender.com
(A) and R ggplot2 package (B).
7
Figure 2: Evolutionary characteristics of OROV and genetically related viruses
(A) Maximum amino acid sequence distance between OROV, selected members of the Simbu serogroup, and BUNV belonging
to a different serogroup. Sequences were aligned using MAFFT (https://mafft.cbrc.jp/; algorithm: auto; scoring matrix
BLOSUM62) with all available complete genomic sequences of OROV, IQTV, MDDV, and PDEV, and to the reference sequences
of FPV, JATV, MANV, SBV, SIMV, and UTIV. These viruses were selected to improve visualisation of the genetic diversity
between OROV and its reassortants by comparing the distance within Simbu and between Simbu and Bunyamwera
serogroups. The amino acid p-distance matrix was generated using MEGA X (www.megasoftware.net) using a pairwise
deletion option. JATV, originally considered an OROV reassortant,35 is now classified as a separate species by the International
Committee on Taxonomy of Viruses.36 Sequences labelled “unverified” in GenBank (ie, GenBank staff could not verify the
accuracy of the submitted sequences) were excluded. All sequences were retrieved from GenBank in June, 2023. (B)
Maximum likelihood phylogenetic trees done in MEGA (MEGA X) using a Whelan and Goldman substitution model and
complete deletion option based on translated coding sequences of the L, M, and S segments. Black circles at nodes represent
support values of at least 0·70 from 1000 bootstrap replicates. OROV sequences were selected to cover the variability of the
three genome segments and all available sequences of the reassortant viruses. Information on the genomic sequences used
for figure 2A and 2B is available in the appendix. OROV=Oropouche virus. L=large segment. M=medium segment. S=small
segment. IQTV=Iquitos virus. MDDV=Madre de Dios virus. PDEV=Perdões virus. JATV=Jatobal virus. UTIV=Utinga virus.
FPV=Facey’s Paddock virus. MANV=Manzanilla virus. SBV=Schmallenberg virus. SIMV=Simbu virus. BUNV=Bunyamwera virus.
AUS=Australia. BRA=Brazil. ECU=Ecuador. NLD=Netherlands. PER=Peru. TTO=Trinidad and Tobago. VEN=Venezuela.
ZAF=South Africa.
8
Figure 3: Literature on Oropouche virus (OROV) and its reassortants
(A) The number of publications in PubMed on OROV and reassortants up to June 30, 2023. (B) Bibliographic coupling in OROV
literature from a Web of Science search (accessed March 31, 2023). Each node represents a publication (indicated with name
of the first author and year of publication); connected nodes represent articles sharing at least 15 references. Nodes are
distributed in two dimensions by the number of references shared; clustered nodes therefore represent similar publications.
Node size represents the total link strength of the node (ie, the sum of all link scores from each publication pair). The bigger
the node, the higher the bibliographic coupling with the rest of the reference set. Colour scale represents the year of
publication. Figure created with RStudio (A) and VOSviewer (B; University of Leiden, 2010).
9
Figure 4: Oropouche virus detections in humans, other vertebrate hosts, midges, and mosquitoes
Confirmed cases were detected with sequencing or RT-PCR or neutralisation tests or viral isolation and complement fixation
test or seroconversion in paired samples with any type of serological tool. Probable infections were detected with ELISA,
immunofluorescence tests, or haemagglutination inhibition tests in non-paired samples. Estimated cases were taken from
the literature without laboratory evidence. Cases from Argentina in 200009, included in the map, were reported as personal
communication in a textbook of paediatric infectious diseases43 and not in a peer-reviewed article. Host and vector cases
detected with any method were included. Maps were created with QGIS (https://qgis.org/en/site/; long-term release version
3.22 Białowieża) on a regional level and by decade. Supporting information regarding the infections is available in the
appendix. IQTV=Iquitos virus. MDDV=Madre de Dios virus. PDEV=Perdões virus.
10
11
Figure 5: Robustness of laboratory diagnosis of human infections with Oropouche virus and reassortants
(A) Predominance of serodiagnosis and viral isolation. (B) Widespread use of molecular detection and sequencing. For both
panels, confirmed cases were detected with sequencing, RT-PCR, neutralisation tests, viral isolation combined with CF, or
seroconversion in paired samples with any type of serological tool. Probable cases were detected with ELISA,
immunofluorescence assays, or haemagglutination inhibition tests on a single sample. Studies with no black dots are based
solely on symptoms. CF=complement fixation. *Studies include different study groups, presented separately. ¤ Describes
cases of Iquitos virus. # Describes cases of Madre de Dios virus. For clarity of presentation only one surname is shown.
Details on the studies summarised are available in the appendix.
References
1
Jones KE, Patel NG, Levy MA, et al. Global trends in emerging infectious diseases. Nature 2008; 451: 99093.
2
Rosenberg R, Johansson MA, Powers AM, Miller BR. Search strategy has influenced the discovery rate of human viruses. Proc Natl Acad Sci
USA 2013; 110: 1396164.
3
Rocklöv J, Dubrow R. Climate change: an enduring challenge for vector-borne disease prevention and control. Nat Immunol 2020; 21: 47983.
4
Zeng Z, Zhan J, Chen L, Chen H, Cheng S. Global, regional, and national dengue burden from 1990 to 2017: a systematic analysis based on
the global burden of disease study 2017. EClinicalMedicine 2021; 32: 100712.
5
Gould E, Pettersson J, Higgs S, Charrel R, de Lamballerie X. Emerging arboviruses: why today? One Health 2017; 4: 113.
6
Labeaud AD, Bashir F, King CH. Measuring the burden of arboviral diseases: the spectrum of morbidity and mortality from four prevalent
infections. Popul Health Metr 2011; 9: 1.
7
Fischer C, Jo WK, Haage V, Moreira-Soto A, de Oliveira Filho EF, Drexler JF. Challenges towards serologic diagnostics of emerging
arboviruses. Clin Microbiol Infect 2021; 27: 122129.
8
Marcondes CB, Contigiani M, Gleiser RM. Emergent and reemergent arboviruses in South America and the Caribbean: why so many and why
now? J Med Entomol 2017; 54: 50932.
9
Bernstein AS, Ando AW, Loch-Temzelides T, et al. The costs and benefits of primary prevention of zoonotic pandemics. Sci Adv 2022; 8:
eabl4183.
10
Ortiz DI, Piche-Ovares M, Romero-Vega LM, Wagman J, Troyo A. The impact of deforestation, urbanization, and changing land use patterns
on the ecology of mosquito and tick-borne diseases in Central America. Insects 2021; 13: 20.
11
Liang G, Gao X, Gould EA. Factors responsible for the emergence of arboviruses; strategies, challenges and limitations for their control.
Emerg Microbes Infect 2015; 4: e18.
12
WHO. Ending the neglect to attain the Sustainable Development Goals: a road map for neglected tropical diseases 20212030. Geneva: World
Health Organization, 2020.
13
PAHO. Recommendations for laboratory detection and diagnosis of arbovirus infections in the region of the Americas. Washington, DC: Pan
American Health Organization, 2023.
14
PAHO. Tool for the diagnosis and care of patients with suspected arboviral diseases. Washington, DC: Pan American Health Organization,
2017.
15
Pan American Health Organization. Dengue. https://www.paho. org/en/topics/dengue (accessed May 11, 2022).
16
Pan American Health Organization. Chikungunya. https://www. paho.org/en/topics/chikungunya (accessed May 11, 2022).
17
Bartholomeeusen K, Daniel M, LaBeaud DA, et al. Chikungunya fever. Nat Rev Dis Primers 2023; 9: 17.
18
de Souza WM, de Lima STS, Simões Mello LM, et al. Spatiotemporal dynamics and recurrence of chikungunya virus in Brazil: an
epidemiological study. Lancet Microbe 2023; 4: e31929.
19
Pan American Health Organization. Zika. https://www.paho.org/ en/topics/zika (accessed May 11, 2022).
20
Puntasecca CJ, King CH, LaBeaud AD. Measuring the global burden of chikungunya and Zika viruses: a systematic review. PLoS Negl Trop
Dis 2021; 15: e0009055.
21
Pezzi L, Rodriguez-Morales AJ, Reusken CB, et al. GloPIDR report on chikungunya, o’nyong-nyong and Mayaro virus, part 3:
epidemiological distribution of Mayaro virus. Antiviral Res 2019; 172: 104610.
22
Fischer C, Pontier D, Filippi-Codaccioni O, et al. Venezuelan equine encephalitis complex alphavirus in bats, French Guiana.
Emerg
Infect
Dis
2021;
27:
114145.
23
Zaid A, Burt FJ, Liu X, et al. Arthritogenic alphaviruses: epidemiological and clinical perspective on emerging arboviruses. Lancet Infect Dis
2021; 21: e12333.
24
Sakkas H, Bozidis P, Franks A, Papadopoulou C. Oropouche fever: a review. Viruses 2018; 10: 16.
25
Figueiredo LTM. Emergent arboviruses in Brazil. Rev Soc Bras Med Trop 2007; 40: 22429.
12
26
Tesh RB. The emerging epidemiology of Venezuelan hemorrhagic fever and Oropouche fever in tropical South America. Ann N Y Acad Sci
1994; 740: 12937.
27
Pinheiro FP, Travassos da Rosa AP, Travassos da Rosa JF, et al. Oropouche virus. I. A review of clinical, epidemiological, and ecological
findings. Am J Trop Med Hyg 1981; 30: 14960.
28
Vasconcelos HB, Azevedo RSS, Casseb SM, et al. Oropouche fever epidemic in Northern Brazil: epidemiology and molecular characterization
of isolates. J Clin Virol 2009; 44: 12933.
29
Tilston-Lunel NL, Hughes J, Acrani GO, et al. Genetic analysis of members of the species Oropouche virus and identification of a novel M
segment sequence. J Gen Virol 2015; 96: 163650.
30
Briese T, Calisher CH, Higgs S. Viruses of the family Bunyaviridae: are all available isolates reassortants? Virology 2013; 446: 20716.
31
Elliott RM. Orthobunyaviruses: recent genetic and structural insights. Nat Rev Microbiol 2014; 12: 67385.
32
Tilston-Lunel NL, Shi X, Elliott RM, Acrani GO. The potential for reassortment between Oropouche and Schmallenberg orthobunyaviruses.
Viruses 2017; 9: E220.
33
de Oliveira Filho EF, Carneiro IO, Ribas JRL, et al. Identification of animal hosts of Fort Sherman virus, a New World zoonotic
orthobunyavirus. Transbound Emerg Dis 2020; 67: 143341.
34
Gutierrez B, Wise EL, Pullan ST, et al. Evolutionary dynamics of Oropouche Virus in South America. J Virol 2020; 94: e0112719.
35
Saeed MF, Wang H, Suderman M, et al. Jatobal virus is a reassortant containing the small RNA of Oropouche virus. Virus Res 2001;
77:
25
30.
36
International Committee on Taxonomy of Viruses. Current ICTV taxonomy release. https://ictv.global/taxonomy (accessed Nov 25, 2022).
37
Navarro JC, Giambalvo D, Hernandez R, et al. Isolation of Madre de Dios virus (Orthobunyavirus; Bunyaviridae), an Oropouche virus species
reassortant, from a monkey in Venezuela. Am J Trop Med Hyg 2016; 95: 32838.
38
Aguilar PV, Barrett AD, Saeed MF, et al. Iquitos virus: a novel reassortant Orthobunyavirus associated with human illness in Peru. PLoS Negl
Trop Dis 2011; 5: e1315.
39
Ladner JT, Savji N, Lofts L, et al. Genomic and phylogenetic characterization of viruses included in the Manzanilla and Oropouche species
complexes of the genus Orthobunyavirus, family Bunyaviridae. J Gen Virol 2014; 95: 105566.
40
Travassos da Rosa JF, de Souza WM, Pinheiro FP, et al. Oropouche virus: clinical, epidemiological, and molecular aspects of a neglected
Orthobunyavirus. Am J Trop Med Hyg 2017; 96: 101930.
41
Huang X, Yue Y, Li D, et al. Antibody-dependent enhancement of dengue virus infection inhibits RLR-mediated type-I IFN- independent
signalling through upregulation of cellular autophagy. Sci Rep 2016; 6: 22303.
42
Anderson CR, Spence L, Downs WG, Aitken TH. Oropouche virus: a new human disease agent from Trinidad, West Indies. Am J Trop Med
Hyg 1961; 10: 57478.
43
Cherry JD, Harrison GJ, Kaplan SL, Hotez PJ, Steinbach WJ, eds.
Feigin and Cherry’s textbook of pediatric infectious diseases, 7th edn.
Philadelphia, PA: Elsevier, Saunders, 2014.
44
Cardoso BF, Serra OP, Heinen LBS, et al. Detection of Oropouche virus segment S in patients and inCulex quinquefasciatus in the state of
Mato Grosso, Brazil. Mem Inst Oswaldo Cruz 2015;
110:
74554.
45
Pinheiro F, Pinheiro M, Bensabath G, Causey OR, Shope R. Epidemia de vírus Oropouche em Belém*. Nota Prévia Em Rev Serviço Espec
Saúde Pública 1962; 12: 1523.
46
Pinheiro FP, Travassos da Rosa AP, Travassos da Rosa JF, Bensabath G. An outbreak of Oropouche virus diease in the vicinity of Santarem,
Para, Brazil. Tropenmed Parasitol 1976; 27: 21323.
47
LeDuc JW, Hoch AL, Pinheiro FP, da Rosa AP. Epidemic Oropouche virus disease in northern Brazil. Bull Pan Am Health Organ 1981;
15:
97103.
48
Borborema CA, Pinheiro FP, Albuquerque BC, da Rosa AP, da Rosa JF, Dourado HV. Primeiro registro de epidemias causadas pelo vírus
Oropouche no Estado do Amazonas. Rev Inst Med Trop São Paulo 1982; 24: 13239.
49
Vasconcelos PFC, Travassos Da Rosa JF, Guerreiro SC, Dégallier N, Travassos Da Rosa ES, Travassos Da Rosa AP. Primeiro registro de
epidemias causadas pelo vírus Oropouche nos estados do Maranhão e Goiás, Brasil. Rev Inst Med Trop São Paulo 1989;
31:
27178.
50
Rosa APAT, Rodrigues SG, Nunes MRT, Magalhães MTF, Rosa JFST, Vasconcelos PFC. Epidemia de febre do Oropouche em Serra Pelada,
município de Curionópolis, Pará, 1994. Rev Soc Bras Med Trop 1996; 29: 53741.
51
Moreli ML, Aquino VH, Cruz ACR, Figueiredo LTM. Diagnosis of Oropouche virus infection by RT-nested-PCR. J Med Virol 2002; 66:
13942.
52
De Figueiredo RMP, Thatcher BD, de Lima ML, Almeida TC, Alecrim WD, Guerra MVF. Doenças exantemáticas e primeira epidemia de
dengue ocorrida em Manaus, Amazonas, no período de 19981999. Rev Soc Bras Med Trop 2004; 37: 47679.
53
Azevedo RSS, Nunes MR, Chiang JO, et al. Reemergence of Oropouche fever, northern Brazil. Emerg Infect Dis 2007; 13: 91215.
54
Bernardes-Terzian AC, de-Moraes-Bronzoni RV, Drumond BP, et al. Sporadic Oropouche virus infection, Acre, Brazil. Emerg Infect Dis
2009; 15: 34850.
55
Nunes MRT, Barbosa TFS, Casseb LMN, et al. Eco-epidemiologia dos arbovírus na área de influência da rodovia Cuiabá-Santarém (BR 163),
13
Estado do Pará, Brasil. Cad Saude Publica 2009; 25: 2583602.
56
Mourão MP, Bastos MS, Gimaqu JBL, et al. Oropouche fever outbreak, Manaus, Brazil, 20072008. Emerg Infect Dis 2009; 15: 206364.
57
Bastos MS, Figueiredo LTM, Naveca FG, et al. Identification of Oropouche Orthobunyavirus in the cerebrospinal fluid of three patients in the
Amazonas, Brazil. Am J Trop Med Hyg 2012;
86:
73235.
58
Bastos
MS,
Lessa
N,
Naveca
FG,
et al. Detection of herpesvirus, enterovirus, and arbovirus infection in patients with suspected central nervous
system viral infection in the western Brazilian Amazon. J Med Virol 2014; 86: 152227.
59
da Costa VG, de Rezende Féres VC, Saivish MV, de Lima Gimaque JB, Moreli ML. Silent emergence of Mayaro and Oropouche viruses in
humans in Central Brazil. Int J Infect Dis 2017; 62: 8485.
60
Naveca FG, Nascimento VA, Souza VC, de Figueiredo RMP. Human Orthobunyavirus infections, Tefé, Amazonas, Brazil. PLoS Curr 2018;
published online March 22. https://doi.org/10.1371%2Fcurrents.outb reaks.7d65e5eb6ef75664da68905c5582f7f7.
61
Nascimento VAD, Santos JHA, Monteiro DCDS, et al. Oropouche virus detection in saliva and urine. Mem Inst Oswaldo Cruz 2020; 115:
e190338.
62
Vernal S, Martini CCR, da Fonseca BAL. Oropouche virus- associated aseptic meningoencephalitis, southeastern Brazil. Emerg Infect Dis
2019; 25: 38082.
63
Fonseca LMDS, Carvalho RH, Bandeira AC, Sardi SI, Campos GS. Oropouche virus detection in febrile patients’ saliva and urine samples
in Salvador, Bahia, Brazil. Jpn J Infect Dis 2020;
73:
16465.
64
Chiang
JO,
Azevedo RS, Justino MCA, et al. Neurological disease caused by Oropouche virus in northern Brazil: should it be included in the
scope of clinical neurological diseases? J Neurovirol 2021;
27:
62630.
65
Freitas B, Pinheiro FP, Santos MAV. Epidemia de vírus Oropouche no Leste do Estado da Pará, 1979. https://patua.iec.gov.br/items/ cc5ac320-
39b1-4a9c-b0d5-40472caf0b9d (accessed March 8, 2023).
66
Phan TG, Del Valle Mendoza J, Sadeghi M, Altan E, Deng X, Delwart E. Sera of Peruvians with fever of unknown origins include viral
nucleic acids from non-vertebrate hosts. Virus Genes 2018;
54:
3340.
67
García MP, Merino NS, Figueroa D, et al. Detección de la circulación del virus Oropuche en la región Madre de Dios, Perú, (Diciembre
2015Enero 2016). Rev Peru Med Exp Salud Publica 2016; 33: 38081.
68
Baisley KJ, Watts DM, Munstermann LE, Wilson ML. Epidemiology of endemic Oropouche virus transmission in upper Amazonian Peru.
Am J Trop Med Hyg 1998; 59: 71016.
69
Alvarez-Falconi PP, Ruiz BAR. Brote de fiebre de Oropuche en Bagazán, San MartínPerú: evaluación epidemiológica. Manifestaciones
Gastrointestinales y Hemorrágicas 2010; 30: 33440.
70
Chavez R, Colan E, Philips II. Fiebre de Oropuche en Iquitos: reporte preliminar de 5 casos. Rev Farm Ter 1992; 2: 1214.
71
Watts DM, Russell KL, Wooster MT, et al. Etiologies of acute undifferentiated febrile illnesses in and near Iquitos from 1993 to 1999 in the
Amazon River Basin of Peru. Am J Trop Med Hyg
2022;
107:
111428.
72
Martins-Luna J, Del Valle-Mendoza J, Silva-Caso W, et al. Oropouche infection a neglected arbovirus in patients with acute febrile illness
from the Peruvian coast. BMC Res Notes 2020;
13:
67.
73
Durango-Chavez HV, Toro-Huamanchumo CJ, Silva-Caso W, et al. Oropouche virus infection in patients with acute febrile syndrome: is a
predictive model based solely on signs and symptoms useful? PLoS One 2022; 17: e0270294.
74
Forshey BM, Guevara C, Laguna-Torres VA, et al. Arboviral etiologies of acute febrile illnesses in Western South America, 20002007. PLoS
Negl Trop Dis 2010; 4: e787.
75
Manock SR, Jacobsen KH, de Bravo NB, et al. Etiology of acute undifferentiated febrile illness in the Amazon basin of Ecuador. Am J Trop
Med Hyg 2009; 81: 14651.
76
Wise EL, Pullan ST, Márquez S, et al. Isolation of Oropouche virus from febrile patient, Ecuador. Emerg Infect Dis 2018; 24: 93537.
77
Wise EL, Márquez S, Mellors J, et al. Oropouche virus cases identified in Ecuador using an optimised qRT-PCR informed by metagenomic
sequencing. PLoS Negl Trop Dis 2020; 14: e0007897.
78
Groot Liévano H. Estudios sobre virus transmitidos por artrópodos en Colombia. Rev Acad Colomb Cienc Exactas Fis Nat 1964;
41
(suppl)
:
12.
79
Gómez-Camargo DE, Egurrola-Pedraza JA, Cruz CD, et al. Evidence of Oropouche Orthobunyavirus infection, Colombia, 2017.
Emerg
Infect
Dis
2021;
27:
175658.
80
Gil-Mora J, Acevedo-Gutiérrez LY, Betancourt-Ruiz PL, et al. Arbovirus antibody seroprevalence in the human population from Cauca,
Colombia. Am J Trop Med Hyg 2022; 107: 121825.
81
Elbadry MA, Durães-Carvalho R, Blohm GM, et al. Orthobunyaviruses in the Caribbean: Melao and Oropouche virus infections in school
children in Haiti in 2014. PLoS Negl Trop Dis 2021; 15: e0009494.
82
Silva-Caso W, Aguilar-Luis MA, Palomares-Reyes C, et al. First outbreak of Oropouche Fever reported in a non-endemic western region of
the Peruvian Amazon: Molecular diagnosis and clinical characteristics. Int J Infect Dis 2019; 83: 13944.
83
Gaillet M, Pichard C, Restrepo J, et al. Outbreak of Oropouche virus in French Guiana. Emerg Infect Dis 2021; 27: 271114.
84
Ciuoderis KA, Berg MG, Perez LJ, et al. Oropouche virus as an emerging cause of acute febrile illness in Colombia.
Emerg
Microbes
Infect
2022;
11:
264557.
14
85
Romero-Alvarez D, Escobar LE. Oropouche fever, an emergent disease from the Americas. Microbes Infect 2018; 20: 13546.
86
Pavon JAR, Neves NADS, Silva LCF, et al. Neurological infection by chikungunya and a triple Arbovirus co-infection in Mato Grosso,
central western Brazil during 2019. J Clin Virol 2022; 146: 105056.
87
Pinheiro FP, Rocha AG, Freitas RB, et al. Meningite associada às infecções por vírus Oropouche. Rev Inst Med Trop São Paulo 1982; 24: 246
51.
88
Stubbs SH, Cornejo Pontelli M, Mishra N, et al. Vesicular stomatitis virus chimeras expressing the Oropouche virus glycoproteins elicit
protective immune responses in mice. MBio 2021; 12: e0046321.
89
Livonesi MC, De Sousa RLM, Badra SJ, Figueiredo LTM. In vitro and in vivo studies of ribavirin action on Brazilian Orthobunyavirus. Am
J Trop Med Hyg 2006; 75: 101116.
90
Livonesi MC, Moro de Sousa RL, Moraes Figueiredo LT. In vitro study of antiviral activity of mycophenolic acid on Brazilian
orthobunyaviruses. Intervirology 2007; 50: 20408.
91
Livonesi MC, de Sousa RLM, Badra SJ, Figueiredo LTM. In vitro and in vivo studies of the interferon-alpha action on distinct
Orthobunyavirus. Antiviral Res 2007; 75: 12128.
92
Adhikari UK, Tayebi M, Rahman MM. Immunoinformatics approach for epitope-based peptide vaccine design and active site prediction against
polyprotein of emerging Oropouche virus. J Immunol Res 2018; 2018: 6718083.
93
Wernike K, Beer M. Schmallenberg virus: to vaccinate, or not to vaccinate? Vaccines (Basel) 2020; 8: 287.
94
Guerra GS, Barriales D, Lorenzo G, et al. Immunization with a small fragment of the Schmallenberg virus nucleoprotein highly conserved
across the Orthobunyaviruses of the Simbu serogroup reduces viremia in SBV challenged IFNAR/ mice. Vaccine 2023; 41: 327584.
95
Hechinger S, Wernike K, Beer M. Evaluating the protective efficacy of a trivalent vaccine containing Akabane virus, Aino virus and Chuzan
virus against Schmallenberg virus infection. Vet Res 2013; 44: 114.
96
Senn N, Luang-Suarkia D, Manong D, Siba PM, McBride WJ. Contribution of dengue fever to the burden of acute febrile illnesses in Papua
New Guinea: an age-specific prospective study. Am J Trop Med Hyg 2011; 85: 13237.
97
Texeira Nunes MR, Vasconcelos HB, de Almeida Medeiros DB, et al. A febre do Oropouche: Uma Revisao dos aspectos epidemiologicos e
moleculares na Amazonia Brasileira. Cad Saude Publica 2007; 15: 30318.
98
Weidmann M, Rudaz V, Nunes MRT, Vasconcelos PFC, Hufert FT. Rapid detection of human pathogenic orthobunyaviruses. J Clin Microbiol
2003; 41: 3299305.
99
Camarão AAR, Swanepoel R, Boinas F, Quan M. Development and analytical validation of a group-specific RT-qPCR assay for the detection
of the Simbu serogroup orthobunyaviruses. J Virol Methods 2019; 271: 113685.
100
Rojas A, Stittleburg V, Cardozo F, et al. Real-time RT-PCR for the detection and quantitation of Oropouche virus. Diagn Microbiol Infect Dis
2020; 96: 114894.
101
Naveca FG, Nascimento VAD, Souza VC, Nunes BTD,
Rodrigues DSG, Vasconcelos PFDC. Multiplexed reverse transcription real-time
polymerase chain reaction for simultaneous detection of Mayaro, Oropouche, and Oropouche-like viruses. Mem Inst Oswaldo Cruz 2017; 112:
51013.
102
Nunes MRT, de Souza WM, Savji N, et al. Oropouche orthobunyavirus: genetic characterization of full-length genomes and development of
molecular methods to discriminate natural reassortments. Infect Genet Evol 2019; 68: 1622.
103
Souza JVC, Santos HO, Leite AB, et al. Viral metagenomics for the identification of emerging infections in clinical samples with inconclusive
Dengue, Zika, and Chikungunya viral amplification. Viruses 2022; 14: 1933.
104
Batovska J, Mee PT, Lynch SE, Sawbridge TI, Rodoni BC. Sensitivity and specificity of metatranscriptomics as an arbovirus surveillance tool.
Sci Rep 2019; 9: 19398.
105
Reiber H, Lange P. Quantification of virus-specific antibodies in cerebrospinal fluid and serum: sensitive and specific detection of antibody
synthesis in brain. Clin Chem 1991; 37: 115360.
106
Watts DM, Phillips I, Callahan JD, Griebenow W, Hyams KC,
Hayes CG. Oropouche virus transmission in the Amazon River
basin of
Peru. Am J Trop Med Hyg 1997;
56:
14852.
107
Saeed MF, Nunes M, Vasconcelos PF, et al. Diagnosis of Oropouche virus infection using a recombinant nucleocapsid protein-based enzyme
immunoassay. J Clin Microbiol 2001; 39: 244552.
108
Bréard E, Lara E, Comtet L, et al. Validation of a commercially available indirect ELISA using a nucleocapside recombinant protein for
detection of Schmallenberg virus antibodies. PLoS One 2013; 8: e53446.
109
Kinney RM, Calisher CH. Antigenic relationships among Simbu serogroup (Bunyaviridae) viruses. Am J Trop Med Hyg 1981;
30:
130718.
110
Wernike K, Aebischer A, Sick F, Szillat KP, Beer M. Differentiation of antibodies against selected Simbu serogroup viruses by a glycoprotein
Gc-based triplex ELISA. Vet Sci 2021; 8: 12.
111
Pinheiro FP, Travassos Da Rosa AP, Duc JL, Gomes MLC, Hoch AL. Transmission of Oropouche virus from man to hamster by the midge
Culicoides paraensis. Science 1982; 215: 125153.
112
Romero-Alvarez D, Escobar LE. Vegetation loss and the 2016 Oropouche fever outbreak in Peru. Mem Inst Oswaldo Cruz
2017;
112:
29298.
113
Estrada-Peña A, Ostfeld RS, Peterson AT, Poulin R, de la Fuente J. Effects of environmental change on zoonotic disease risk: an ecological
15
primer. Trends Parasitol 2014; 30: 20514.
114
Pauvolid-Corrêa A, Campos Z, Soares R, Nogueira RMR, Komar N. Neutralizing antibodies for orthobunyaviruses in Pantanal, Brazil. PLoS
Negl Trop Dis 2017; 11: e0006014.
115
Batista PM, Andreotti R, Chiang JO, et al. Seroepidemiological monitoring in sentinel animals and vectors as part of arbovirus surveillance
in the state of Mato Grosso do Sul, Brazil. Rev Soc Bras Med Trop 2012; 45: 16873.
116
Gibrail MM, Fiaccadori FS, Souza M, et al. Detection of antibodies to Oropouche virus in non-human primates in Goiânia City, Goiás. Rev
Soc Bras Med Trop 2016; 49: 35760.
117
Batista PM, Andreotti R, Silva de Almeida P, et al. Detection of arboviruses of public health interest in free-living New World primates
(Sapajus spp.; Alouatta caraya) captured in Mato Grosso do Sul, Brazil. Rev Soc Bras Med Trop 2013; 46: 68490.
118
Mosquera JD, Zapata S, Spinelli G, Gualapuro M, León R, Augot D. An updated list of the Culicoides (Diptera, Ceratopogonidae) fauna from
Ecuador. Parasite 2022; 29: 63.
119
Arias-Goeta C, Mousson L, Rougeon F, Failloux AB. Dissemination and transmission of the E1-226V variant of chikungunya virus in Aedes
albopictus are controlled at the midgut barrier level. PLoS One 2013; 8: e57548.
120
Samy AM, Elaagip AH, Kenawy MA, Ayres CF, Peterson AT, Soliman DE. Climate change influences on the global potential distribution of
the mosquito Culex quinquefasciatus, vector of West Nile virus and lymphatic filariasis. PLoS One 2016; 11: e0163863.
121
de Mendonça SF, Rocha MN, Ferreira FV, et al. Evaluation of Aedes aegypti, Aedes albopictus, and Culex quinquefasciatus mosquitoes competence
to Oropouche virus infection. Viruses 2021; 13: 755.
122
McGregor BL, Connelly CR, Kenney JL. Infection, dissemination, and transmission potential of North American Culex quinquefasciatus,
Culex tarsalis, and Culicoides sonorensis for Oropouche virus. Viruses 2021; 13: 226.
123
Hoch AL, Pinheiro FP, Roberts DR, Gomes ML. Laboratory transmission of Oropouche virus by Culex quinquefasciatus say. Bull Pan Am
Health Organ 1987; 21: 5561.
124
Pereira-Silva JW, Ríos-Velásquez CM, Lima GR, et al. Distribution and diversity of mosquitoes and Oropouche-like virus infection rates in
an Amazonian rural settlement. PLoS One 2021;
16:
e0246932.
125
CABI Digital Library. Culicoides paraensis. https://www.cabi.org/ isc/datasheet/87062 (accessed May 9, 2022).
126
Romero-Alvarez D, Escobar LE, Auguste AJ, Del Valle SY, Manore CA. Transmission risk of Oropouche fever across the Americas. Infect
Dis Poverty 2023; 12: 47.
127
Rodrigues AH, Santos RI, Arisi GM, et al. Oropouche virus experimental infection in the golden hamster (Mesocrisetus auratus). Virus Res
2011; 155: 3541.
128
Araújo R, Dias LB, Araújo MT, Pinheiro F, Oliva OF. Ultrastructural changes in the hamster liver after experimental inoculation with
Oropouche arbovirus (type BeAn 19991). Rev Inst Med Trop São Paulo 1978; 20: 4554.
129
Proenca-Modena JL, Sesti-Costa R, Pinto AK, et al. Oropouche virus infection and pathogenesis are restricted by MAVS, IRF-3, IRF-7, and
type I interferon signaling pathways in nonmyeloid cells. J Virol 2015; 89: 472037.
130
Proenca-Modena JL, Hyde JL, Sesti-Costa R, et al. Interferon- regulatory factor 5-dependent signaling restricts Orthobunyavirus dissemination
to the central nervous system. J Virol 2015;
90:
189205.
131
da Silva Menegatto MB, Ferraz AC, Lima RLS, et al. Oropouche virus infection induces ROS production and oxidative stress in liver and
spleen of mice. J Gen Virol 2023; 104.
132
Pinto AK, Williams GD, Szretter KJ, et al. Human and murine IFIT1 proteins do not restrict infection of negative-sense RNA viruses of the
Orthomyxoviridae, Bunyaviridae, and Filoviridae families. J Virol 2015; 89: 946576.
133
Santos RI, Almeida MFP, Paula FE, et al. Experimental infection of suckling mice by subcutaneous inoculation with Oropouche virus. Virus
Res 2012; 170: 2533.
134
Santos RI, Bueno-Júnior LS, Ruggiero RN, et al. Spread of Oropouche virus into the central nervous system in mouse. Viruses 2014; 6: 3827
36.
135
Nunes MR, Martins LC, Rodrigues SG, et al. Oropouche virus isolation, southeast Brazil. Emerg Infect Dis 2005; 11: 161013.
136
Ribeiro Amorim M, Cornejo Pontelli M, Fabiano de Souza G, et al. Oropouche virus infects, persists and induces IFN response in human
peripheral blood mononuclear cells as identified by RNA PrimeFlow and qRT-PCR assays. Viruses 2020; 12: E785.
137
Tilston-Lunel NL, Acrani GO, Randall RE, Elliott RM. Generation of recombinant oropouche viruses lacking the nonstructural protein NSm
or NSs. J Virol 2015; 90: 261627.
138
Acrani GO, Gomes R, Proença-Módena JL, et al. Apoptosis induced by Oropouche virus infection in HeLa cells is dependent on virus protein
expression. Virus Res 2010; 149: 5663.
139
Geddes VEV, de Oliveira AS, Tanuri A, Arruda E, Ribeiro-Alves M, Aguiar RS. MicroRNA and cellular targets profiling reveal miR-217 and
miR-576-3p as proviral factors during Oropouche infection. PLoS Negl Trop Dis 2018; 12: e0006508.
140
Schwarz MM, Price DA, Ganaie SS, et al. Oropouche orthobunyavirus infection is mediated by the cellular host factor Lrp1. Proc Natl Acad
Sci USA 2022; 119: e2204706119.
16
141
Geddes VEV, Brustolini OJB, Cavalcante LTF, et al. Common dysregulation of innate immunity pathways in human primary astrocytes
infected with Chikungunya, Mayaro, Oropouche, and Zika viruses. Front Cell Infect Microbiol 2021; 11: 641261.
142
Almeida GM, Souza JP, Mendes ND, et al. Neural infection by Oropouche virus in adult human brain slices induces an inflammatory and toxic
response. Front Neurosci 2021; 15: 674576.
143
Martins-Junior AMG, Carneiro J, Sampaio I, Ferrari SF, Schneider H. Phylogenetic relationships among Capuchin
(Cebidae, Platyrrhini)
lineages: an old event of sympatry explains the current distribution of Cebus and Sapajus. Genet Mol Biol 2018;
41:
699712.
144
Kaup M, Trull S, Hom EFY. On the move: sloths and their epibionts as model mobile ecosystems. Biol Rev Camb Philos Soc 2021;
96:
2638
60.
145
Vaughn DW, Green S, Kalayanarooj S, et al. Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease severity.
J Infect Dis 2000; 181: 29.
146
Kirkpatrick BD, Whitehead SS, Pierce KK, et al. The live attenuated dengue vaccine TV003 elicits complete protection against dengue in a
human challenge model. Sci Transl Med 2016; 8: 330ra36.
147
Files MA, Hansen CA, Herrera VC, et al. Baseline mapping of Oropouche virology, epidemiology, therapeutics, and vaccine research and
development. npj Vaccines 2022; 7: 38.
148
Moreli ML, Aquino VH, Figueiredo LT. Identification of Simbu, California and Bunyamwera serogroup bunyaviruses by nested RT-PCR.
Trans R Soc Trop Med Hyg 2001; 95: 10813.
... Furthermore, human mobility significantly contributes to the dissemination of the virus, underscoring the intricate interplay between environmental and social factors in OF's expansion. [1][2][3] OF's incubation period ranges from 4 to 8 days, with clinical manifestations closely mimicking those of dengue and Chikungunya, including high fever, headache, arthralgia, myalgia, skin rashes, malaise, nausea, and vomiting. While most cases resolve within two weeks, severe complications, such as meningitis or encephalitis, may develop. ...
... 4 Currently, treatment is primarily supportive, focusing on symptomatic relief, which underscores the importance of prevention and control strategies aimed at reducing vector mosquito populations and educating the public about transmission routes. [1][2][3] The recent confirmation of OF's first case in Rio de Janeiro on February 29, 2024, involving a 42-year-old man returning from Amazonas, signals the virus's spread beyond traditional hotspots such as Amazonas, Acre, and Rondônia. Additionally, the record of 1674 OF cases in Amazonas in 2024, a threefold increase from the previous year, underscores the disease's alarming escalation. ...
... CSF microfilariae resembling those of MP in Mansonellosis [271] have been reported once in 2 subjects in what has been described as a "very tentative report" [272]; one subject was diagnosed with acute encephalomyelitis (headaches, vomiting, blurred vision, stupor, and cord-like sensory signs), the other complained of headache, drowsiness and fatigue that cleared spontaneously. MP-carrying Culicoides biting midges also carry >50 arboviruses, but the only known significant viral pathogen of humans (Oropouche virus) produces a non-NS, dengue-like disorder (fever, chills, headache, arthralgia, myalgia, anorexia, vomiting, photophobia, dizziness and, occasionally, meningoencephalitis) in Latin America [273,274]. In conclusion, neither MP nor OV appears to be a probable cause of NS; however, infection with the first nematode, but not the second, is associated with the only recent-onset NS cases ever studied [53,54]. ...
Article
Full-text available
Background Vector-borne diseases (VBDs) are important contributors to the global burden of infectious diseases due to their epidemic potential, which can result in significant population and economic impacts. Oropouche fever, caused by Oropouche virus (OROV), is an understudied zoonotic VBD febrile illness reported in Central and South America. The epidemic potential and areas of likely OROV spread remain unexplored, limiting capacities to improve epidemiological surveillance. Methods To better understand the capacity for spread of OROV, we developed spatial epidemiology models using human outbreaks as OROV transmission-locality data, coupled with high-resolution satellite-derived vegetation phenology. Data were integrated using hypervolume modeling to infer likely areas of OROV transmission and emergence across the Americas. Results Models based on one-support vector machine hypervolumes consistently predicted risk areas for OROV transmission across the tropics of Latin America despite the inclusion of different parameters such as different study areas and environmental predictors. Models estimate that up to 5 million people are at risk of exposure to OROV. Nevertheless, the limited epidemiological data available generates uncertainty in projections. For example, some outbreaks have occurred under climatic conditions outside those where most transmission events occur. The distribution models also revealed that landscape variation, expressed as vegetation loss, is linked to OROV outbreaks. Conclusions Hotspots of OROV transmission risk were detected along the tropics of South America. Vegetation loss might be a driver of Oropouche fever emergence. Modeling based on hypervolumes in spatial epidemiology might be considered an exploratory tool for analyzing data-limited emerging infectious diseases for which little understanding exists on their sylvatic cycles. OROV transmission risk maps can be used to improve surveillance, investigate OROV ecology and epidemiology, and inform early detection.
Article
Full-text available
Schmallenberg Virus (SBV), an arbovirus from the Peribunyaviridae family and Orthobunyavirus genus, was discovered in late 2011 in Germany and has been circulating in Europe, Asia and Africa ever since. The virus causes a disease associated with ruminants that includes fever, fetal malformation, drop in milk production, diarrhoea and stillbirths, becoming a burden for small and large farms. Building on previous studies on SBV nucleoprotein (SBV-N) as a promising vaccine candidate, we have investigated the possible protein regions responsible for protection. Based on selective truncation of domains designed from the available crystal structure of the SBV-N, we identified both the N-terminal domain (N-term; Met1 - Thr133) and a smaller fragment within (C4; Met1 - Ala58) as vaccine prototypes. Two injections of the N-term and C4 polypeptides protected mice knockout for type I interferon (IFN) receptors (IFNAR-/-) challenged with virulent SBV, opposite to control groups that presented severe signs of morbidity and weight loss. Viremia analyses along with the presence of IFN-γ secreted from splenocytes re-stimulated with the N-terminal region of the protein corroborate that these two portions of SBV-N can be employed as subunit vaccines. Apart from both proteinaceous fragments being easily produced in bacterial cells, the C4 polypeptide shares a high sequence homology (∼87.1 %) with the corresponding region of nucleoproteins of several viruses of the Simbu serogroup, a group of Orthobunyaviruses that comprises SBV and veterinary pathogens like Akabane virus and human infecting viruses like Oropouche. Thus, we propose that this smaller fragment is better suited for vaccine nanoparticle formulation, and it paves the way to further research with other related Orthobunyaviruses.
Article
Full-text available
Background: Chikungunya virus (CHIKV) is an Aedes mosquito-borne virus that has caused large epidemics linked to acute, chronic, and severe clinical outcomes. Currently, Brazil has the highest number of chikungunya cases in the Americas. We aimed to investigate the spatiotemporal dynamics and recurrence pattern of chikungunya in Brazil since its introduction in 2013. Methods: In this epidemiological study, we used CHIKV genomic sequencing data, CHIKV vector information, and aggregate clinical data on chikungunya cases from Brazil. The genomic data comprised 241 Brazilian CHIKV genome sequences from GenBank (n=180) and the 2022 CHIKV outbreak in Ceará state (n=61). The vector data (Breteau index and House index) were obtained from the Brazilian Ministry of Health for all 184 municipalities in Ceará state and 116 municipalities in Tocantins state in 2022. Epidemiological data on laboratory-confirmed cases of chikungunya between 2013 and 2022 were obtained from the Brazilian Ministry of Health and Laboratory of Public Health of Ceará. We assessed the spatiotemporal dynamics of chikungunya in Brazil via time series, mapping, age–sex distribution, cumulative case-fatality, linear correlation, logistic regression, and phylogenetic analyses. Findings: Between March 3, 2013, and June 4, 2022, 253545 laboratory-confirmed chikungunya cases were reported in 3316 (59·5%) of 5570 municipalities, mainly distributed in seven epidemic waves from 2016 to 2022. To date, Ceará in the northeast has been the most affected state, with 77 418 cases during the two largest epidemic waves in 2016 and 2017 and the third wave in 2022. From 2016 to 2022 in Ceará, the odds of being CHIKV-positive were higher in females than in males (odds ratio 0·87, 95% CI 0·85–0·89, p<0·0001), and the cumulative case-fatality ratio was 1·3 deaths per 1000 cases. Chikungunya recurrences in the states of Ceará, Tocantins (recurrence in 2022), and Pernambuco (recurrence in 2021) were limited to municipalities with few or no previously reported cases in the previous epidemic waves. The recurrence of chikungunya in Ceará in 2022 was associated with a new East-CentralSouth-African lineage. Population density metrics of the main CHIKV vector in Brazil, Aedes aegypti, were not correlated spatially with locations of chikungunya recurrence in Ceará and Tocantins. Interpretation: Spatial heterogeneity of CHIKV spread and population immunity might explain the recurrence pattern of chikungunya in Brazil. These results can be used to inform public health interventions to prevent future chikungunya epidemic waves in urban settings.
Article
Full-text available
An updated list of biting midges of the genus Culicoides inhabiting Ecuador is provided. Entomological investigations were carried out from July 2010 to May 2019 using CDC light traps in three Ecuadorian regions (Amazon basin, Andean (foothills and highlands) and Pacific Coast). A total of 12,073 Culicoides specimens from seven subgenera and nine species groups were collected. More species and higher variation were found in the Amazon basin than in either of the Andes regions or coastal sites. A total of 53 species were identified. Of these, 15 are herein reported as new species records for Ecuador: Culicoides acotylus Lutz, C. aitkeni Wirth & Blanton, C. benarrochi Ortiz & Mirsa, C. carvalhoi Wirth & Blanton , C. freitasi Wirth & Blanton, C. ginesi Ortíz, C. lopesi Barretto , C. lyrinotatus Wirth & Blanton , C. profundus Santarém, Felippe-Bauer & Trindade, C. pseudoreticulatus Santarém, Felippe-Bauer & Castellón , C. quasiparaensis Clastrier, C. vernoni Wirth & Blanton, C. youngi Wirth & Barreto and two new species. Our results show that the updated list of the Ecuadorian Culicoides fauna comprises 70 species. This inventory highlights the presence of species that have been incriminated as vectors of disease elsewhere in animals and humans, mainly C. insignis and C. paraensis .
Article
Full-text available
Several arboviruses have emerged or reemerged into the New World during the past several decades, causing outbreaks of significant proportion. In particular, the outbreaks of Dengue virus (DENV), Zika virus, and Chikungunya virus (CHIKV) have been explosive and unpredictable, and have led to significant adverse health effects. These viruses are considered the leading cause of acute undifferentiated febrile illnesses in Colombia. However, Venezuelan equine encephalitis virus (VEEV) is endemic in Colombia, and arboviruses such as the Mayaro virus (MAYV) and the Oropouche virus (OROV) cause febrile illnesses in neighboring countries. Yet, evidence of human exposure to MAYV and OROV in Colombia is scarce. In this study, we conducted a serosurvey study in healthy individuals from the Cauca Department in Colombia. We assessed the seroprevalence of antibodies against multiple arboviruses, including DENV serotype 2, CHIKV, VEEV, MAYV, and OROV. Based on serological analyses, we found that the overall seroprevalence for DENV serotype 2 was 30%, 1% for MAYV, 2.6% for CHIKV, 4.4% for VEEV, and 2% for OROV. This study provides evidence about the circulation of MAYV and OROV in Colombia, and suggests that they—along with VEEV and CHIKV—might be responsible for cases of acute undifferentiated febrile illnesses that remain undiagnosed in the region. The study results also highlight the need to strengthen surveillance programs to identify outbreaks caused by these and other vector-borne pathogens.
Article
Full-text available
Arbovirus infections are frequent causes of acute febrile illness (AFI) in tropical countries. We conducted health facility based AFI surveillance at four sites in Colombia (Cucuta, Cali, Villavicencio, Leticia) during 2019-2022. Demographic, clinical and risk factor data were collected from persons with AFI that consented to participate in the study (n=2,967). Serologic specimens were obtained and tested for multiple pathogens by RT-PCR and rapid test (Antigen/IgM), with 20.7% identified as dengue positive from combined testing. Oropouche virus (OROV) was initially detected in serum by metagenomic next generation sequencing (mNGS) and virus target capture in a patient from Cúcuta. Three additional infections from Leticia were confirmed by conventional PCR, sequenced, and isolated in tissue culture. Phylogenetic analysis determined there have been at least two independent OROV introductions into Colombia. To assess OROV spread, a RT-qPCR dual-target assay was developed which identified 87/791 (10.9%) viremic cases in AFI specimens from Cali (3/53), Cucuta (3/19), Villavicencio (38/566), and Leticia (43/153). In parallel, an automated anti-nucleocapsid antibody assay detected IgM in 27/503 (5.4%) and IgG in 92/568 (16.2%) patients screened, for which 24/68 (35.3%) of PCR positives had antibodies. Dengue was found primarily in people aged <18 years and linked to several clinical manifestations (weakness, skin rash and petechiae), whereas Oropouche cases were associated with the location, climate phase, and odynophagia symptom. Our results confirm OROV as an emerging pathogen and recommend increased surveillance to determine its burden as a cause of AFI in Colombia.
Article
Full-text available
The objective of this study was to determine the etiology of febrile illnesses among patients from October 1, 1993 through September 30, 1999, in the urban community of Iquitos in the Amazon River Basin of Peru. Epidemiological and clinical data as well as blood samples were obtained from consenting patients at hospitals, health clinics and private residences. Samples were tested for arboviruses in cell cultures and for IgM and IgG antibodies by ELISA. Blood smears were examined for malaria, and sera were tested for antibodies to Leptospira spp. by ELISA and microscopic agglutination. Among 6,607 febrile patients studied, dengue viruses caused 14.6% of the cases, and Venezuelan equine encephalitis virus caused 2.5%, Oropouche virus 1.0%, Mayaro virus 0.4%, and other arboviruses caused 0.2% of the cases. Also, 22.9% of 4,844 patients tested were positive for malaria, and of 400 samples tested, 9% had evidence of acute leptospirosis. Although the study was not designed to assess the importance of these pathogens as a cause of human morbidity in the total population, these results indicate that arboviruses, leptospirosis, and malaria were the cause of approximately 50% of the febrile cases. Although the arboviruses that were diagnosed can produce asymptomatic infections, our findings increased the overall understanding of the relative health burden of these infections, as well as baseline knowledge needed for designing and implementing further studies to better assess the health impact and threat of these pathogens in the Amazon Basin of Peru.
Article
Full-text available
Viral metagenomics is increasingly being used for the identification of emerging and re-emerging viral pathogens in clinical samples with unknown etiology. The objective of this study was to shield light on the metavirome composition in clinical samples obtained from patients with clinical history compatible with an arboviral infection, but that presented inconclusive results when tested using RT-qPCR. The inconclusive amplification results might be an indication of the presence of an emerging arboviral agent that is inefficiently amplified by conventional PCR techniques. A total of eight serum samples with inconclusive amplification results for the routinely tested arboviruses—dengue (DENV), Zika (ZIKV), and Chikungunya (CHIKV) obtained during DENV and CHIKV outbreaks registered in the state of Alagoas, Northeast Brazil between July and August 2021—were submitted to metagenomic next-generation sequencing assay using NextSeq 2000 and bioinformatic pipeline for viral discovery. The performed bioinformatic analysis revealed the presence of two arboviruses: DENV type 2 (DENV-2) and CHIKV with a high genome coverage. Further, the metavirome of those samples revealed the presence of multiple commensal viruses apparently without clinical significance. The phylogenetic analysis demonstrated that the DENV-2 genome belonged to the Asian/American genotype and clustered with other Brazilian strains. The identified CHIKV genome was taxonomically assigned as ECSA genotype, which is circulating in Brazil. Together, our results reinforce the utility of metagenomics as a valuable tool for viral identification in samples with inconclusive arboviral amplification. Viral metagenomics is one of the most potent methods for the identification of emerging arboviruses.
Article
Oropouche virus (OROV) is the aetiological agent of Oropouche fever, the symptoms of which are common to most arboviruses, such as fever, headache, malaise, nausea and vomiting. More than half a million people have been infected with OROV since its isolation in 1955. Although Oropouche fever is classified as a neglected and emerging disease, to date, there are no antiviral drugs or vaccines available against the infection and little is known about its pathogenicity. Therefore, it is essential to elucidate the possible mechanisms involved in its pathogenesis. Since oxidative stress plays a pivotal role in the progression of various viral diseases, in this study, redox homeostasis in the target organs of OROV infection was evaluated using an animal model. Infected BALB/c mice exhibited reduced weight gain, splenomegaly, leukopenia, thrombocytopenia, anaemia, development of anti-OROV neutralizing antibodies, increased liver transaminases, and serum levels of pro-inflammatory cytokines tumour necrosis factor (TNF-α) and interferon-γ (IFN-γ). The OROV genome and infectious particles were detected in the liver and spleen of infected animals, with liver inflammation and an increase in the number and total area of lymphoid nodules in the spleen. In relation to redox homeostasis in the liver and spleen, infection led to an increase in reactive oxygen species (ROS) levels, increased oxidative stress biomarkers malondialdehyde (MDA) and carbonyl protein, and decreased activity of the antioxidant enzymes superoxide dismutase (SOD) and catalase (CAT). Taken together, these results help elucidate some important aspects of OROV infection that may contribute to the pathogenesis of Oropouche.
Article
Chikungunya virus is widespread throughout the tropics, where it causes recurrent outbreaks of chikungunya fever. In recent years, outbreaks have afflicted populations in East and Central Africa, South America and Southeast Asia. The virus is transmitted by Aedes aegypti and Aedes albopictus mosquitoes. Chikungunya fever is characterized by severe arthralgia and myalgia that can persist for years and have considerable detrimental effects on health, quality of life and economic productivity. The effects of climate change as well as increased globalization of commerce and travel have led to growth of the habitat of Aedes mosquitoes. As a result, increasing numbers of people will be at risk of chikungunya fever in the coming years. In the absence of specific antiviral treatments and with vaccines still in development, surveillance and vector control are essential to suppress re-emergence and epidemics.