Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 288
Filter
1.
Pan Afr Med J ; 47: 120, 2024.
Article in English | MEDLINE | ID: mdl-38828420

ABSTRACT

Introduction: Aedes albopictus, like Aedes aegypti, is a virulent vector of arboviruses especially the well-documented spread of yellow fever around the world. Although yellow fever is prevalent in Nigeria, there is a paucity of information in the Niger Delta region on the distribution of Aedes mosquito vectors and molecular detection of the virus in infected mosquitoes. This study sampled Aedes mosquitoes around houses associated with farms from four communities (Otolokpo, Ute-Okpu, Umunede, and Ute Alohen) in Ika North-East Local Government Area of Delta State, Nigeria. Methods: various sampling methods were used in Aedes mosquito collection to test their efficacy in the survey. Mosquitoes in holding cages were killed by freezing and morphologically identified. A pool of 15 mosquitoes per Eppendorf tube was preserved in RNAi later for yellow fever virus screening. Two samples were molecularly screened for each location. Results: seven hundred and twenty-five (725) mosquitoes were obtained from the various traps. The mean abundance of the mosquitoes was highest in m-HLC (42.9) compared to the mosquitoes sampled using other techniques (p<0.0001). The mean abundance of mosquitoes was lowest in Center for Disease Control (CDC) light traps without attractant (0.29). No yellow fever virus strain was detected in all the mosquitoes sampled at the four locations. Conclusion: this study suggests that Aedes albopictus are the mosquitoes commonly biting around houses associated with farms. More so, yellow fever virus was not detected in the mosquitoes probably due to the mass vaccination exercise that was carried out the previous year in the study area. More studies are required using the m-HLC to determine the infection rate in this endemic area.


Subject(s)
Aedes , Mosquito Vectors , Yellow Fever , Yellow fever virus , Animals , Aedes/virology , Nigeria , Yellow fever virus/isolation & purification , Mosquito Vectors/virology , Yellow Fever/transmission , Yellow Fever/epidemiology , Yellow Fever/virology , Humans
2.
Emerg Infect Dis ; 29(9): 1818-1826, 2023 09.
Article in English | MEDLINE | ID: mdl-37610174

ABSTRACT

Yellow fever virus, transmitted by infected Aedes spp. mosquitoes, causes an acute viral hemorrhagic disease. During October 2021-February 2022, a yellow fever outbreak in some communities in Ghana resulted in 70 confirmed cases with 35 deaths (case-fatality rate 50%). The outbreak started in a predominantly unvaccinated nomadic community in the Savannah region, from which 65% of the cases came. The molecular amplification methods we used for diagnosis produced full-length DNA sequences from 3 confirmed cases. Phylogenetic analysis characterized the 3 sequences within West Africa genotype II; strains shared a close homology with sequences from Cote d'Ivoire and Senegal. We deployed more sensitive advanced molecular diagnostic techniques, which enabled earlier detection, helped control spread, and improved case management. We urge increased efforts from health authorities to vaccinate vulnerable groups in difficult-to-access areas and to educate the population about potential risks for yellow fever infections.


Subject(s)
Yellow Fever , Yellow fever virus , Yellow fever virus/classification , Yellow fever virus/isolation & purification , Yellow Fever/virology , Disease Outbreaks , Ghana/epidemiology , Humans , Phylogeny , Sequence Analysis, RNA , RNA, Viral/analysis
3.
PLoS One ; 17(1): e0262312, 2022.
Article in English | MEDLINE | ID: mdl-34995319

ABSTRACT

Yellow fever is endemic in Ghana and outbreaks occur periodically. The prodromal signs due to Yellow Fever Virus (YFV) infection are non-specific, making clinical signs unreliable as the sole criteria for diagnosis. Accurate laboratory confirmation of suspected yellow fever cases is therefore vital in surveillance programs. Reporting of ELISA IgM testing results by laboratories can delay due to late arrival of samples from the collection sites as well as limited availability of ELISA kits. In this study, the diagnostic performance characteristics of a rapid immunochromatographic Standard Q Yellow Fever IgM test kit (SD Biosensor) was evaluated for the rapid diagnosis of Yellow Fever infection in Ghana. A panel of 275 sera, comprising 81 confirmed YFV positives and 194 negatives were re-tested in this study using the Standard Q Yellow Fever IgM test kit. Using the CDC/WHO Yellow Fever IgM capture ELISA as a benchmark, the sensitivity, specificity and accuracy of the Standard Q Yellow Fever test kit were 96.3%, 97.9% and 97.5%, respectively. The false positivity rate was 5.1% and there was no cross-reactivity when the Standard Q Yellow Fever test kit was tested against dengue, malaria and hepatitis B and C positive samples. In addition, inter-reader variability and invalid rate were both zero. The results indicate that the diagnostic performance of the Standard Q Yellow Fever IgM test kit on serum or plasma is comparable to the serum IgM detection by ELISA and can be used as a point of care rapid diagnostic test kit for YFV infection in endemic areas.


Subject(s)
Biosensing Techniques/instrumentation , Chromatography, Affinity/instrumentation , Immunoglobulin M/immunology , Reagent Kits, Diagnostic , Yellow Fever/diagnosis , Yellow fever virus/immunology , Biosensing Techniques/economics , Chromatography, Affinity/economics , Equipment Design , Humans , Immunoglobulin M/blood , Limit of Detection , Reagent Kits, Diagnostic/economics , Time Factors , Yellow Fever/blood , Yellow Fever/immunology , Yellow fever virus/isolation & purification
4.
PLoS Negl Trop Dis ; 16(1): e0010171, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35073317

ABSTRACT

Aedes simpsoni complex has a wide distribution in Africa and comprises at least three described sub-species including the yellow fever virus (YFV) vector Ae. bromeliae. To date, the distribution and relative contributions of the sub-species and/or subpopulations including bionomic characteristics in relation to YF transmission dynamics remain poorly studied. In this study conducted in two areas with divergent ecosystems: peri-urban (coastal Rabai) and rural (Rift Valley Kerio Valley) in Kenya, survival rate was estimated by parity in Ae. simpsoni s.l. mosquitoes sampled using CO2-baited BG Sentinel traps. We then applied PCR targeting the nuclear internal transcribed spacer 2 (ITS2), region followed by sequencing and phylogenetic analytics to identify the sibling species in the Ae. simpsoni complex among parous and blood fed cohorts. Our results show that Ae. bromeliae was the most dominant sub-species in both areas, exhibiting high survival rates, human blood-feeding, and potentially, high vectorial capacity for pathogen transmission. We document for the first time the presence of Ae. lilii in Kenya and potentially yet-to-be described species in the complex displaying human feeding tendencies. We also infer a wide host feeding range on rodents, reptile, and domestic livestock besides humans especially for Ae. bromeliae. This feeding trend could likely expose humans to various zoonotic pathogens. Taken together, we highlight the utility of genotype-based analyses to generate precision surveillance data of vector populations for enhanced disease risk prediction and to guide cost-effective interventions (e.g. YF vaccinations).


Subject(s)
Aedes/classification , Aedes/virology , Arbovirus Infections/transmission , Arboviruses/isolation & purification , Mosquito Vectors/virology , Yellow Fever/transmission , Aedes/physiology , Africa, Eastern/epidemiology , Animals , Arbovirus Infections/epidemiology , Arboviruses/classification , Ecosystem , Environment , Feeding Behavior , Female , Host Specificity , Yellow Fever/epidemiology , Yellow fever virus/classification , Yellow fever virus/isolation & purification
5.
Viruses ; 13(8)2021 07 28.
Article in English | MEDLINE | ID: mdl-34452343

ABSTRACT

Yellow fever virus remains a major threat in low resource countries in South America and Africa despite the existence of an effective vaccine. In Senegal and particularly in the eastern part of the country, periodic sylvatic circulation has been demonstrated with varying degrees of impact on populations in perpetual renewal. We report an outbreak that occurred from October 2020 to February 2021 in eastern Senegal, notified and managed through the synergistic effort yellow fever national surveillance implemented by the Senegalese Ministry of Health in collaboration with the World Health Organization, the countrywide 4S network set up by the Ministry of Health, the Institut Pasteur de Dakar, and the surveillance of arboviruses and hemorrhagic fever viruses in human and vector populations implemented since mid 2020 in eastern Senegal. Virological analyses highlighted the implication of sylvatic mosquito species in virus transmission. Genomic analysis showed a close relationship between the circulating strain in eastern Senegal, 2020, and another one from the West African lineage previously detected and sequenced two years ago from an unvaccinated Dutch traveler who visited the Gambia and Senegal before developing signs after returning to Europe. Moreover, genome analysis identified a 6-nucleotide deletion in the variable domain of the 3'UTR with potential impact on the biology of the viral strain that merits further investigations. Integrated surveillance of yellow fever virus but also of other arboviruses of public health interest is crucial in an ecosystem such as eastern Senegal.


Subject(s)
Yellow Fever/epidemiology , Yellow Fever/virology , Yellow fever virus/physiology , Adolescent , Adult , Aedes/classification , Aedes/physiology , Aedes/virology , Amino Acid Sequence , Animals , Child , Disease Outbreaks , Female , Humans , Male , Mosquito Vectors/classification , Mosquito Vectors/physiology , Mosquito Vectors/virology , Phylogeny , Senegal/epidemiology , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/genetics , Yellow Fever/transmission , Yellow fever virus/classification , Yellow fever virus/genetics , Yellow fever virus/isolation & purification , Young Adult
6.
Pan Afr Med J ; 38: 248, 2021.
Article in French | MEDLINE | ID: mdl-34104296

ABSTRACT

Yellow fever (YF) is a viral haemorrhagic fever caused by yellow fever virus transmitted by Aedes mosquitoes. Since 2013, in Chad, four cases of yellow fever have been detected and confirmed as part of the national fever surveillance program. We here report the last clinical case confirmed in the health district of Lai. The patient was a 57-year-old man with no significant medical and surgical history and unknown immunisation status. He consulted on April 21st, 2020 for fever, moderate to low abundance jaundice and epistaxis (nosebleed) and painful hepatomegaly. Paraclinical examinations, such as RT-PCR, objectified yellow fever virus in post-mortem tissue sample. Thus, confirmed yellow fever cases in this district, the low level of vaccination coverage, the circulation of the virus and the presence of vector in the country should warn of a real threat of reemergence of yellow fever in Chad.


Subject(s)
Yellow Fever/diagnosis , Yellow fever virus/isolation & purification , Aedes/virology , Animals , Chad , Humans , Male , Middle Aged , Mosquito Vectors/virology , Recurrence , Yellow Fever/transmission , Yellow Fever/virology , Yellow Fever Vaccine/administration & dosage
7.
Mem Inst Oswaldo Cruz ; 115: e190501, 2020.
Article in English | MEDLINE | ID: mdl-33174908

ABSTRACT

BACKGROUND: Non-human primates contribute to the spread of the yellow fever virus (YFV) and the establishment of transmission cycles in endemic areas. OBJECTIVE: To describe the severe histopathological aspects of YFV infection, 10 squirrel monkeys were infected with YFV and blood, brain, liver, kidney, spleen, heart, lung, lymph node and stomach were collected at 1-7, 10, 20 and 30 days post-infection (dpi). METHODS: Histopathological analysis and detection of the genome and viral antigens and neutralising antibodies were performed by RT-PCR, immunohistochemistry and neutralisation test, respectively. FINDINGS: Only one animal died from the experimental infection. The genome and viral antigens were detected in all investigated organs (1-30 dpi) and the neutralising antibodies from seven to 30 dpi. The brain contained perivascular haemorrhage (6 dpi); in the liver, midzonal haemorrhage and lytic necrosis (6 dpi) were observed. The kidney had bleeding in the Bowman's capsule and tubular necrosis (6 dpi). Pyknotic lymphocytes were observed in the spleen (1-20 dpi), the lung had haemorrhage (2-6 dpi), in the endocardium it contained nuclear pyknosis and necrosis (2-3 dpi) and the stomach contained blood in the lumen (6 dpi). MAIN FINDINGS: Squirrel monkeys reliably reproduced the responses observed in human cases of yellow fever and, therefore, constitute an excellent experimental model for studies on the pathophysiology of the disease.


Subject(s)
Saimiri/virology , Yellow Fever/diagnosis , Yellow fever virus/isolation & purification , Animals , Disease Models, Animal
8.
Parasit Vectors ; 13(1): 350, 2020 Jul 14.
Article in English | MEDLINE | ID: mdl-33019944

ABSTRACT

BACKGROUND: Mozambique, same as many other tropical countries, is at high risk of arthropod-borne virus (arbovirus) diseases and recently two dengue virus (DENV) outbreaks occurred in the northern part of the country. The occurrence of some important vector species, such as Aedes (Stegomyia) aegypti (Linnaeus) and Ae. (Stg.) albopictus (Skuse), besides several other sylvatic vectors, have been reported in the country, which may indicate that the transmission of some arboviruses of public health importance may involve multiple-vector systems. Therefore, knowing the occurrence and distribution of existing and the new important vectors species, is crucial for devising systematic transmission surveillance and vector control approaches. The aim of this study was to map the occurrence and distribution of mosquito species with potential for transmitting arboviruses of human and veterinary relevance in Niassa Province, Northern Mozambique. METHODS: Field entomological surveys were undertaken in April 2016 in Lago District, Niassa Province, northern Mozambique. Breeding sites of mosquitoes were inspected and immature stages were collected and reared into adult. Mosquitoes in the adult stages were morphologically identified using taxonomic keys. Morphological identification of Aedes (Stegomyia) luteocephalus (Newstead) were later confirmed using dissected male genitalia and molecular based on the phylogenetic analyses of the sequenced barcode (cox1 mtDNA) gene. RESULTS: A total of 92 mosquito larvae collected developed into adults. Of these, 16 (17.39%) were morphologically identified as Ae. luteocephalus. The remaining specimens belonged to Ae. (Stg.) aegypti (n = 4, 4.35%), Ae. (Aedimorphus) vittatus (n = 24, 26.09%), Anopheles garnhami (n = 1, 1.09%), Culex (Culiciomyia) nebulosus (n = 28, 30.43%), Eretmapodites subsimplicipes (n = 18, 19.57%) and Toxorhynchites brevipalpis (n = 1, 1.09%), taxa already known to the country. Male genitalia and phylogenetic analyses confirmed the identity of Ae. luteocephalus specimens collected in this study. CONCLUSIONS: To our knowledge, this is the first detection of Ae. luteocephalus in Mozambican territory, a vector species of yellow fever virus (YFV), Zika virus (ZIKV) and dengue virus (DENV) in Africa. Further studies are encouraged to investigate the role of Ae. luteocephalus in the transmission of arboviral diseases in Mozambique.


Subject(s)
Aedes/classification , Aedes/physiology , Mosquito Vectors/classification , Mosquito Vectors/physiology , Yellow Fever/transmission , Zika Virus Infection/transmission , Aedes/anatomy & histology , Aedes/virology , Animal Distribution , Animals , Dengue Virus/genetics , Dengue Virus/isolation & purification , Female , Humans , Male , Mosquito Vectors/anatomy & histology , Mosquito Vectors/virology , Mozambique , Phylogeny , Yellow Fever/virology , Yellow fever virus/genetics , Yellow fever virus/isolation & purification , Zika Virus Infection/virology
9.
PLoS Negl Trop Dis ; 14(10): e0008658, 2020 10.
Article in English | MEDLINE | ID: mdl-33017419

ABSTRACT

BACKGROUND: From the end of 2016 until the beginning of 2019, Brazil faced a massive sylvatic yellow fever (YF) outbreak. The 2016-2019 YF epidemics affected densely populated areas, especially the Southeast region, causing thousands of deaths of humans and non-human primates (NHP). METHODOLOGY/PRINCIPAL FINDINGS: We conducted a molecular investigation of yellow fever virus (YFV) RNA in 781 NHP carcasses collected in the urban, urban-rural interface, and rural areas of Minas Gerais state, from January 2017 to December 2018. Samples were analyzed according to the period of sampling, NHP genera, sampling areas, and sampling areas/NHP genera to compare the proportions of YFV-positive carcasses and the estimated YFV genomic loads. YFV infection was confirmed in 38.1% of NHP carcasses (including specimens of the genera Alouatta, Callicebus, Callithrix, and Sapajus), from the urban, urban-rural interface, and rural areas. YFV RNA detection was positively associated with epidemic periods (especially from December to March) and the rural environment. Higher median viral genomic loads (one million times) were estimated in carcasses collected in rural areas compared to urban ones. CONCLUSIONS/SIGNIFICANCE: The results showed the wide occurrence of YF in Minas Gerais in epidemic and non-epidemic periods. According to the sylvatic pattern of YF, a gradient of viral dissemination from rural towards urban areas was observed. A high YF positivity was observed for NHP carcasses collected in urban areas with a widespread occurrence in 67 municipalities of Minas Gerais, including large urban centers. Although there was no documented case of urban/Aedes YFV transmission to humans in Brazil during the 2016-2019 outbreaks, YFV-infected NHP in urban areas with high infestation by Aedes aegypti poses risks for YFV urban/Aedes transmission and urbanization.


Subject(s)
Yellow Fever/epidemiology , Yellow Fever/prevention & control , Yellow Fever/transmission , Zoonoses/virology , Aedes/virology , Alouatta/virology , Animals , Brazil/epidemiology , Callicebus/virology , Callithrix/virology , Disease Reservoirs/virology , Epidemics , Genome, Viral , Humans , Mosquito Vectors/virology , Primates/virology , Sapajus/virology , Yellow fever virus/isolation & purification , Yellow fever virus/pathogenicity , Zoonoses/epidemiology , Zoonoses/transmission
10.
PLoS Negl Trop Dis ; 14(9): e0008549, 2020 09.
Article in English | MEDLINE | ID: mdl-32881913

ABSTRACT

BACKGROUND: Chikungunya (CHIK) and yellow fever (YF) are becoming major public health threats in East African countries including Ethiopia. In Ethiopia, there is no reliable information about the epidemiology of CHIK. This study aimed to assess a community-based sero-prevalence of CHIK and YF in the South Omo Valley, an endemic area for YF. METHODS: Between February and June 2018, blood samples were collected from study participants and screened for IgG antibody against CHIK virus (CHIKV) and YF virus (YFV) infections using ELISA. Data were computerized using Epi Data Software v.3.1 and analyzed using SPSS. RESULTS: A total of 360 participants (51.7% males, age range from 6 to 80, mean age ± SD = 31.95 ± 14.05 years) participated in this study. The overall sero-prevalence of IgG antibody was 43.6% (157/360) against CHIKV, while it was 49.5% (155/313) against YFV. Out of 155 samples which were positive for IgG antibody to YFV, 93 (60.0%) were positive for IgG antibody to CHIKV. Out of 158 samples which were negative for IgG antibody to YFV, 64(40.5%) were positive for IgG antibody to CHIKV. There was a significant positive correlation between IgG antibodies to CHIKV and YFV (sr = 0.82; P<0.01). Residency in the Debub Ari district (AOR = 8.47; 95% CI: 1.50, 47.74) and travel history to sylvatic areas (AOR = 2.21; 95% CI: 1.02, 4.81) were significantly and positively associated with high sero-prevalence of IgG antibody to CHIKV and YFV, respectively. CONCLUSION: High sero-prevalence of IgG antibody to CHIKV shows the circulation of the virus in the present study area. A low sero-prevalence of IgG antibody to YFV in YF vaccine received individuals is highly concerning from a public health point of view as waning of immune response to YFV infection could result in a periodic outbreaks of YF in endemic areas.Nevertheless, the present study has not investigated for possible cross-reactivity of antibody to CHIKV with other alphaviruses like O'nyong-nyong virus and antibody to YFV with other flaviviruses like Dengue fever virus and this warrants further studies in the present study area.


Subject(s)
Antibodies, Viral/blood , Chikungunya Fever/blood , Yellow Fever/blood , Adolescent , Adult , Aged , Aged, 80 and over , Chikungunya Fever/epidemiology , Chikungunya Fever/virology , Chikungunya virus/immunology , Chikungunya virus/isolation & purification , Child , Cross-Sectional Studies , Ethiopia/epidemiology , Female , Humans , Male , Middle Aged , Residence Characteristics , Seroepidemiologic Studies , Yellow Fever/epidemiology , Yellow Fever/virology , Yellow fever virus/immunology , Yellow fever virus/isolation & purification , Young Adult
11.
Acta Trop ; 212: 105702, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32971069

ABSTRACT

Yellow fever (YF) surveillance in Brazil is focused mainly on the detection of epizootic events regarding New World non-human primates (NWNHP). We present a challenging case of a Callitrichidae (Callithrix spp) kept as a domiciliated pet that lived in the urban area of São Paulo municipality and was positive to YF virus by RT-qPCR and immunohistochemistry. After investigation, it was the first occurrence of non-autochthonous YF case of NWNHP described, with probable place of infection in the North shore of São Paulo state. This case illustrates the importance of coordinated laboratorial and field actions, and risks posed by transit of wildlife.


Subject(s)
Callithrix/virology , Yellow Fever/veterinary , Animals , Male , Yellow Fever/diagnosis , Yellow fever virus/genetics , Yellow fever virus/isolation & purification
12.
Antiviral Res ; 183: 104939, 2020 11.
Article in English | MEDLINE | ID: mdl-32980446

ABSTRACT

Yellow fever virus (YFV), a member of the Flaviviridae family, is an arthropod-borne virus that can cause severe disease in humans with a lethality rate of up to 60%. Since 2017, increases in YFV activity in areas of South America and Africa have been described. Although a vaccine is available, named strain 17D (Theiler and Smith, 1937), it is contraindicated for use in the elderly, expectant mothers, immunocompromised people, among others. To this day there is no antiviral treatment against YFV to reduce the severity of viral infection. Here, we used a circular polymerase extension reaction (CPER)-based reverse genetics approach to generate a full-length reporter virus (YFVhb) by introducing a small HiBit tag in the NS1 protein. The reporter virus replicates at a similar rate to the parental YFV in HuH-7 cells. Using YFVhb, we designed a high throughput antiviral screening luciferase-based assay to identify inhibitors that target any step of the viral replication cycle. We validated our assay by using a range of inhibitors including drugs, immune sera and neutralizing single chain variable fragments (scFv). In light of the recent upsurge in YFV and a potential spread of the virus, this assay is a further tool in the development of antiviral therapy against YFV.


Subject(s)
Antiviral Agents/pharmacology , High-Throughput Screening Assays/methods , Reverse Genetics/methods , Yellow fever virus/drug effects , Yellow fever virus/genetics , Animals , Cell Line , Drug Discovery/methods , Genes, Reporter , Humans , Mice , Mice, Inbred BALB C , Virus Replication/drug effects , Yellow fever virus/isolation & purification , Yellow fever virus/physiology
13.
PLoS Pathog ; 16(8): e1008699, 2020 08.
Article in English | MEDLINE | ID: mdl-32764827

ABSTRACT

São Paulo, a densely inhabited state in southeast Brazil that contains the fourth most populated city in the world, recently experienced its largest yellow fever virus (YFV) outbreak in decades. YFV does not normally circulate extensively in São Paulo, so most people were unvaccinated when the outbreak began. Surveillance in non-human primates (NHPs) is important for determining the magnitude and geographic extent of an epizootic, thereby helping to evaluate the risk of YFV spillover to humans. Data from infected NHPs can give more accurate insights into YFV spread than when using data from human cases alone. To contextualise human cases, identify epizootic foci and uncover the rate and direction of YFV spread in São Paulo, we generated and analysed virus genomic data and epizootic case data from NHPs in São Paulo. We report the occurrence of three spatiotemporally distinct phases of the outbreak in São Paulo prior to February 2018. We generated 51 new virus genomes from YFV positive cases identified in 23 different municipalities in São Paulo, mostly sampled from NHPs between October 2016 and January 2018. Although we observe substantial heterogeneity in lineage dispersal velocities between phylogenetic branches, continuous phylogeographic analyses of generated YFV genomes suggest that YFV lineages spread in São Paulo at a mean rate of approximately 1km per day during all phases of the outbreak. Viral lineages from the first epizootic phase in northern São Paulo subsequently dispersed towards the south of the state to cause the second and third epizootic phases there. This alters our understanding of how YFV was introduced into the densely populated south of São Paulo state. Our results shed light on the sylvatic transmission of YFV in highly fragmented forested regions in São Paulo state and highlight the importance of continued surveillance of zoonotic pathogens in sentinel species.


Subject(s)
Genome, Viral , Primate Diseases/virology , Yellow Fever/veterinary , Yellow Fever/virology , Yellow fever virus/genetics , Zoonoses/virology , Animals , Brazil/epidemiology , Disease Outbreaks , Genomics , Humans , Phylogeny , Phylogeography , Primate Diseases/epidemiology , Primate Diseases/transmission , Primates/virology , Yellow Fever/epidemiology , Yellow Fever/transmission , Yellow fever virus/classification , Yellow fever virus/isolation & purification , Zoonoses/epidemiology , Zoonoses/transmission
14.
Mem Inst Oswaldo Cruz ; 115: e200218, 2020.
Article in English | MEDLINE | ID: mdl-32696917

ABSTRACT

BACKGROUND: Southeast Brazil has recently experienced a Yellow Fever virus (YFV) outbreak where the mosquito Haemagogus leucocelaenus was a primary vector. Climatic factors influence the abundance of mosquito vectors and arbovirus transmission. OBJECTIVES: We aimed at describing the population dynamics of Hg. leucocelaenus in a county touched by the recent YFV outbreak. METHODS: Fortnightly egg collections with ovitraps were performed from November 2012 to February 2017 in a forest in Nova Iguaçu, Rio de Janeiro, Brazil. The effects of mean temperature and rainfall on the Hg. leucocelaenus population dynamics were explored. FINDINGS: Hg. leucocelaenus eggs were continuously collected throughout the study, with a peak in the warmer months (December-March). The climatic variables had a time-lagged effect and four weeks before sampling was the best predictor for the positivity of ovitraps and total number of eggs collected. The probability of finding > 50% positive ovitraps increased when the mean temperature was above 24ºC. The number of Hg. leucocelaenus eggs expressively increase when the mean temperature and accumulated precipitation surpassed 27ºC and 100 mm, respectively, although the effect of rainfall was less pronounced. MAIN CONCLUSIONS: Monitoring population dynamics of Hg. leucocelaenus and climatic factors in YFV risk areas, especially mean temperature, may assist in developing climate-based surveillance procedures to timely strengthening prophylaxis and control.


Subject(s)
Culicidae/virology , Forests , Insect Vectors/virology , Population Dynamics , Yellow Fever , Yellow fever virus/isolation & purification , Animals , Brazil , Culicidae/classification , Insect Vectors/classification , Seasons , Temperature , Yellow fever virus/genetics
15.
BMC Microbiol ; 20(1): 181, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32590939

ABSTRACT

BACKGROUND: Chikungunya (CHIKV), yellow fever (YFV) and Zika (ZIKV) viruses circulate in sylvatic transmission cycles in southeastern Senegal, where they share common hosts and vectors. All three viruses undergo periodic amplifications, during which they are detected in mosquitoes and sometimes in hosts. However, little is known about their spatio-temporal patterns in years in which they undergo concurrent amplification. The aim of this study was to describe the co-amplification of ZIKV, CHIKV, and YFV, and the daily dynamics of these arboviruses and theirs vectors within villages in southeastern Senegal. RESULTS: Mosquitoes were collected monthly from July to December 2015. Each evening, from 6 to 9 PM, landing collections were performed by teams of 3 persons working simultaneously in 70 sites situated in forest (canopy and ground), savannah, agriculture, barren, and village (indoor and outdoor) land covers. Collections within villages were continued until 6 AM. Mosquitoes were tested for virus infection by virus isolation and RT-PCR. Seventy-five mosquito pools comprising 10 mosquito species contained at least one virus. Ae. furcifer and Ae. luteocephalus were infected by all three viruses, Ae. taylori by YFV and ZIKV, and remaining seven species by only, only YFV or only ZIKV. No single mosquito pool contained more than one virus. CHIKV was the only virus detected in all land cover classes and was found in the greatest number of sampling sites (32.9%, n = 70). The proportion of sites in which more than one virus was detected was less than 6%. Ae. aegypti formosus, Ae. furcifer, Ae. luteocephalus, Ae. minutus, Ae. vittatus, and An. gambiae were found within villages. These vectors were mainly active around dusk but Ae. furcifer was collected until dawn. All viruses save ZIKV were detected indoors and outdoors, mainly around dusk. Virus positive pools were detected over 2, 3 and 4 months for YFV, CHIKV and ZIKV, respectively. CONCLUSION: Our data indicate that the distribution of different vector species and different arboviruses vary substantially between sites, suggesting that CHIKV, YFV, and ZIKV may have different transmission cycles in Southeastern Senegal.


Subject(s)
Chikungunya virus/isolation & purification , Culicidae/virology , Yellow fever virus/isolation & purification , Zika Virus/isolation & purification , Animals , Chikungunya virus/genetics , Culicidae/classification , Female , Male , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Senegal , Time Factors , Yellow fever virus/genetics , Zika Virus/genetics
16.
Virol J ; 17(1): 9, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31973727

ABSTRACT

Yellow fever (YF) is an acute viral disease, affecting humans and non-human primates (NHP), caused by the yellow fever virus (YFV). Despite the existence of a safe vaccine, YF continues to cause morbidity and mortality in thousands of people in Africa and South America. Since 2016, massive YF outbreaks have taken place in Brazil, reaching YF-free zones, causing thousands of deaths of humans and NHP. Here we reviewed the main epidemiological aspects, new clinical findings in humans, and issues regarding YFV infection in vectors and NHP in Brazil. The 2016-2019 YF epidemics have been considered the most significant outbreaks of the last 70 years in the country, and the number of human cases was 2.8 times higher than total cases in the previous 36 years. A new YFV lineage was associated with the recent outbreaks, with persistent circulation in Southeast Brazil until 2019. Due to the high number of infected patients, it was possible to evaluate severity and death predictors and new clinical features of YF. Haemagogus janthinomys and Haemagogus leucocelaenus were considered the primary vectors during the outbreaks, and no human case suggested the occurrence of the urban transmission cycle. YFV was detected in a variety of NHP specimens presenting viscerotropic disease, similar to that described experimentally. Further studies regarding NHP sensitivity to YFV, YF pathogenesis, and the duration of the immune response in NHP could contribute to YF surveillance, control, and future strategies for NHP conservation.


Subject(s)
Yellow Fever , Yellow fever virus , Aedes/virology , Animals , Brazil/epidemiology , Culicidae/virology , Disease Outbreaks , Disease Reservoirs/virology , Epidemics , Humans , Mosquito Vectors/virology , Primates/virology , Virus Diseases/epidemiology , Yellow Fever/epidemiology , Yellow Fever/prevention & control , Yellow Fever/transmission , Yellow fever virus/immunology , Yellow fever virus/isolation & purification , Yellow fever virus/pathogenicity , Zoonoses/epidemiology , Zoonoses/transmission , Zoonoses/virology
17.
J Infect Dis ; 221(12): 2026-2034, 2020 06 11.
Article in English | MEDLINE | ID: mdl-31545372

ABSTRACT

BACKGROUND: Yellow fever (YF) is a vector-borne viral hemorrhagic disease endemic in Africa and Latin America. In 2016, the World Health Organization (WHO) developed the Eliminate YF Epidemics strategy aiming at eliminating YF epidemics by 2026. METHODS: We developed a spatiotemporal model of YF, accounting for the impact of temperature, vector distribution, and socioeconomic factors on disease transmission. We validated our model against previous estimates of YF basic reproductive number (R0). We used the model to estimate global risk of YF outbreaks and vaccination efforts needed to achieve elimination of YF epidemics. RESULTS: We showed that the global risk of YF outbreaks is highly heterogeneous. High-risk transmission areas (R0 > 6) are mainly found in West Africa and the Equatorial region of Latin America. We showed that vaccination coverage needed to eliminate YF epidemics in an endemic country varies substantially between districts. In many endemic countries, a 90% vaccination coverage is needed to achieve elimination. However, in some high-risk districts in Africa, a 95% coverage may be required. CONCLUSIONS: Global elimination of YF epidemics requires higher population-level immunity than the 80% coverage recommended by the WHO. Optimal YF vaccination strategy should be tailored to the risk profile of each endemic country.


Subject(s)
Disease Eradication , Endemic Diseases/prevention & control , Epidemics/prevention & control , Yellow Fever Vaccine/administration & dosage , Yellow Fever/epidemiology , Africa , Americas , Humans , Latin America , Models, Statistical , Mosquito Vectors/virology , Risk Assessment , Seasons , Spatio-Temporal Analysis , Vaccination Coverage/standards , World Health Organization , Yellow Fever/prevention & control , Yellow Fever/transmission , Yellow Fever/virology , Yellow fever virus/immunology , Yellow fever virus/isolation & purification
18.
Talanta ; 208: 120338, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31816752

ABSTRACT

Arboviruses have been emerging as a significant global health problem due to the recurrent epidemics. Arboviruses require the development of new diagnostic devices due to the nonspecific clinical manifestations. Herein, we report a biosensor based on cysteine (Cys), zinc oxide nanoparticles (ZnONp), and Concanavalin A (ConA) lectin to differentiate between arboviruses infections. ConA is capable of interacting with the saccharide components of the viral capsid. In this study, we evaluated the reproducibility, sensitivity, and specificity of the sensor for the virus of Dengue type 2 (DENV2), Zika (ZIKV), Chikungunya (CHIKV), and Yellow fever (YFV). Atomic force microscopy measurements confirmed the electrode surface modification and revealed a heterogeneous topography during the biorecognition process. Cyclic voltammetry (CV) and impedance spectroscopy (EIS) were used to characterize the biosensor. The blockage of the oxidation-reduction process is related to the formation of Cys-ZnONp-ConA system on the electroactive area and its subsequent interaction with viral glycoproteins. The sensor exhibited a linear response to different concentrations of the studied arboviruses. Our study demonstrates that ConA lectin recognizes the structural glycoproteins of the DENV2, ZIKV, CHIKV, and YFV. DENV2 is the most structurally similar to ZIKV. Our results have shown that the impedimetric response correlates with the structural glycoproteins, as follow: DENV2 (18.6 kΩ) > ZIKV (14.6 kΩ) > CHIKV (6.86 kΩ) > YFV (5.98 kΩ). The homologous structural regions contribute to ConA-arboviruses recognition. Our results demonstrate the use of the proposed system for the development of biosensors for arboviruses infections.


Subject(s)
Arbovirus Infections/diagnosis , Arboviruses/metabolism , Biosensing Techniques/methods , Concanavalin A/chemistry , Electrochemistry/methods , Electrodes , Metal Nanoparticles/chemistry , Arbovirus Infections/blood , Arbovirus Infections/virology , Arboviruses/isolation & purification , Chikungunya Fever/blood , Chikungunya Fever/diagnosis , Chikungunya Fever/virology , Chikungunya virus/isolation & purification , Chikungunya virus/metabolism , Cysteine/chemistry , Dengue/blood , Dengue/diagnosis , Dengue/virology , Dengue Virus/isolation & purification , Dengue Virus/metabolism , Diagnosis, Differential , Glucose/analysis , Humans , Mannose/analysis , Yellow Fever/blood , Yellow Fever/diagnosis , Yellow Fever/virology , Yellow fever virus/isolation & purification , Yellow fever virus/metabolism , Zika Virus/isolation & purification , Zika Virus/metabolism , Zika Virus Infection/blood , Zika Virus Infection/diagnosis , Zika Virus Infection/virology , Zinc Oxide/chemistry
19.
Methods Mol Biol ; 2065: 65-77, 2020.
Article in English | MEDLINE | ID: mdl-31578688

ABSTRACT

The recent resurgence of yellow fever virus (YFV) activity in the tropical regions of Africa and South America has sparked renewed interest in this infamous arboviral disease. Also, the development and production of viral vaccines involve several steps that need the monitoring of viral load throughout the process (antigen production, purification, and inactivation). Currently, these steps are followed by plaque lysis titration assay, whose results take about 7-10 days to come out and thus resulting in a laborious and time-consuming approach. With the advent of quantitative real-time PCR (qPCR), we have a faster method to be applied during vaccine production and also to be effectively used for the diagnosis of YFV infection. The technique herein standardized proved to be effective for determining YF viral load both in vivo and in vitro, thus becoming a very important tool for laboratory analysis to verify the vaccination status of individuals, beyond acting as a quality control for vaccine production and diagnosis.


Subject(s)
Real-Time Polymerase Chain Reaction/methods , Viral Load/methods , Yellow Fever/diagnosis , Yellow fever virus/isolation & purification , Humans , Immunogenicity, Vaccine , RNA, Viral/genetics , RNA, Viral/isolation & purification , Viral Load/immunology , Yellow Fever/immunology , Yellow Fever/prevention & control , Yellow Fever/virology , Yellow Fever Vaccine/administration & dosage , Yellow Fever Vaccine/immunology , Yellow fever virus/genetics , Yellow fever virus/immunology
20.
Mem. Inst. Oswaldo Cruz ; 115: e200218, 2020. tab, graf
Article in English | LILACS, Sec. Est. Saúde SP | ID: biblio-1135274

ABSTRACT

BACKGROUND Southeast Brazil has recently experienced a Yellow Fever virus (YFV) outbreak where the mosquito Haemagogus leucocelaenus was a primary vector. Climatic factors influence the abundance of mosquito vectors and arbovirus transmission. OBJECTIVES We aimed at describing the population dynamics of Hg. leucocelaenus in a county touched by the recent YFV outbreak. METHODS Fortnightly egg collections with ovitraps were performed from November 2012 to February 2017 in a forest in Nova Iguaçu, Rio de Janeiro, Brazil. The effects of mean temperature and rainfall on the Hg. leucocelaenus population dynamics were explored. FINDINGS Hg. leucocelaenus eggs were continuously collected throughout the study, with a peak in the warmer months (December-March). The climatic variables had a time-lagged effect and four weeks before sampling was the best predictor for the positivity of ovitraps and total number of eggs collected. The probability of finding > 50% positive ovitraps increased when the mean temperature was above 24ºC. The number of Hg. leucocelaenus eggs expressively increase when the mean temperature and accumulated precipitation surpassed 27ºC and 100 mm, respectively, although the effect of rainfall was less pronounced. MAIN CONCLUSIONS Monitoring population dynamics of Hg. leucocelaenus and climatic factors in YFV risk areas, especially mean temperature, may assist in developing climate-based surveillance procedures to timely strengthening prophylaxis and control.


Subject(s)
Animals , Yellow Fever , Yellow fever virus/isolation & purification , Forests , Population Dynamics , Insect Vectors/virology , Culicidae/virology , Seasons , Temperature , Yellow fever virus/genetics , Brazil , Insect Vectors/classification , Culicidae/classification
SELECTION OF CITATIONS
SEARCH DETAIL
...