191 results on '"Foy, Brian D."'
Search Results
152. Perl 6 Now.
- Author
-
Foy, Brian D.
- Subjects
PERL (Computer program language) ,NONFICTION - Abstract
The article reviews the book "Perl 6 Now," by Scott Walters.
- Published
- 2005
153. The effect of oral anthelmintics on the survivorship and re-feeding frequency of anthropophilic mosquito disease vectors
- Author
-
Kobylinski, Kevin C., Deus, Kelsey M., Butters, Matthew P., Hongyu, Tan, Gray, Meg, da Silva, Ines Marques, Sylla, Massamba, and Foy, Brian D.
- Subjects
- *
MALARIA , *DENGUE , *ANOPHELES gambiae , *DRUG administration , *IVERMECTIN , *AEDES aegypti , *ANTHELMINTICS , *DISEASE vectors - Abstract
Abstract: In the Tropics, there is substantial temporal and spatial overlap of diseases propagated by anthropophilic mosquito vectors (such as malaria and dengue) and human helminth diseases (such as onchocerciasis and lymphatic filariasis) that are treated though mass drug administrations (MDA). This overlap will result in mosquito vectors imbibing significant quantities of these drugs when they blood feed on humans. Since many anthelmintic drugs have broad anti-invertebrate effects, the possibility of combined helminth control and mosquito-borne disease control through MDA is apparent. It has been previously shown that ivermectin can reduce mosquito survivorship when administered in a blood meal, but more detailed examinations are needed if MDA is to ever be developed into a tool for malaria or dengue control. We examined concentrations of drugs that follow human pharmacokinetics after MDA and that matched with mosquito feeding times, for effects against the anthropophilic mosquito vectors Anopheles gambiae s.s. and Aedes aegypti. Ivermectin was the only human-approved MDA drug we tested that affected mosquito survivorship, and only An. gambiae s.s. were affected at concentrations respecting human pharmacokinetics at indicated doses. Ivermectin also delayed An. gambiae s.s. re-feeding frequency and defecation rates, and two successive ivermectin-spiked blood meals following human pharmacokinetic concentrations compounded mortality effects compared to controls. These findings suggest that ivermectin MDA in Africa may be used to decrease malaria transmission if MDAs were administered more frequently. Such a strategy would broaden the current scope of polyparasitism control already afforded by MDAs, and which is needed in many African villages simultaneously burdened by many parasitic diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
154. Efficacy and risk of harms of repeat ivermectin mass drug administrations for control of malaria (RIMDAMAL): a cluster-randomised trial
- Author
-
Hannah C Slater, Florence Fournet, André B. Sagna, François Drabo, Roch K. Dabiré, Martina Wade, Sangeeta Rao, Sunil Parikh, Jonathan A. Seaman, Brian D. Foy, Roland W Bougma, Dieudonné Diloma Soma, Tereza Magalhaes, Noel Rouamba, Abdoulaye Diabaté, A Gafar V Coulidiaty, Haoues Alout, Department of Microbiology, Immunology, and Pathology, Arthropod-borne and Infectious Diseases Laboratory, Colorado State University [Fort Collins] (CSU), Animal, Santé, Territoires, Risques et Ecosystèmes (UMR ASTRE), Centre de Coopération Internationale en Recherche Agronomique pour le Développement (Cirad)-Institut National de la Recherche Agronomique (INRA), Department of Clinical Sciences [Fort Collins, CO, USA] (Orthopaedic Research Center), Department of Epidemiology of Microbial Diseases [New Haven], Yale School of Public Health (YSPH), Institut de Recherche en Sciences de la Santé Bobo Dioulasso (INSSA), Université Polytechnique Nazi Boni Bobo-Dioulasso (UNB), International Mixed Laboratory on Vector Diseases, Partenaires INRAE, Maladies infectieuses et vecteurs : écologie, génétique, évolution et contrôle (MIVEGEC), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD [France-Sud]), Diversity, ecology, evolution & Adaptation of arthropod vectors (MIVEGEC-DEEVA), Evolution des Systèmes Vectoriels (ESV), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD [France-Sud])-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD [France-Sud])-Maladies infectieuses et vecteurs : écologie, génétique, évolution et contrôle (MIVEGEC), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD [France-Sud])-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD [France-Sud]), Imperial College London, Ministère de la Santé [Burkina Faso], Institut de Recherche en Sciences de la Santé (IRSS), CNRST, Centre Muraz [Bobo-Dioulasso, Burkina Faso], Bill & Melinda Gates Foundation, Foy, Brian D., and MRC Centre for Global Infectious Disease Analysis (MRC-GIDA), Imperial College London
- Subjects
Adult ,Male ,Pediatrics ,medicine.medical_specialty ,Adolescent ,030204 cardiovascular system & hematology ,Albendazole ,Drug Administration Schedule ,law.invention ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Ivermectin ,Randomized controlled trial ,law ,Burkina Faso ,medicine ,Cluster Analysis ,Humans ,Cumulative incidence ,030212 general & internal medicine ,Malaria, Falciparum ,Adverse effect ,Child ,[SDV.EE.SANT]Life Sciences [q-bio]/Ecology, environment/Health ,Antiparasitic Agents ,business.industry ,Microbiology and Parasitology ,Absolute risk reduction ,General Medicine ,medicine.disease ,Microbiologie et Parasitologie ,3. Good health ,Treatment Outcome ,Relative risk ,Cohort ,Mass Drug Administration ,Female ,business ,Malaria ,medicine.drug - Abstract
Summary Background Ivermectin is widely used in mass drug administrations for controlling neglected parasitic diseases, and can be lethal to malaria vectors that bite treated humans. Therefore, it could be a new tool to reduce plasmodium transmission. We tested the hypothesis that frequently repeated mass administrations of ivermectin to village residents would reduce clinical malaria episodes in children and would be well tolerated with minimal harms. Methods We invited villages (clusters) in Burkina Faso to participate in a single-blind (outcomes assessor), parallel-assignment, two-arm, cluster-randomised trial over the 2015 rainy season. Villages were assigned (1:1) by random draw to either the intervention group or the control group. In both groups, all eligible participants who consented to the treatment and were at least 90 cm in height received single oral doses of ivermectin (150–200 μg/kg) and albendazole (400 mg), and those in the intervention group received five further doses of ivermectin alone at 3-week intervals thereafter over the 18-week treatment phase. The primary outcome was cumulative incidence of uncomplicated malaria episodes over 18 weeks (analysed on a cluster intention-to-treat basis) in an active case detection cohort of children aged 5 years or younger living in the study villages. This trial is registered with ClinicalTrials.gov, number NCT02509481. Findings Eight villages agreed to participate, and four were randomly assigned to each group. 2712 participants (1333 [49%] males and 1379 [51%] females; median age 15 years [IQR 6–34]), including 590 children aged 5 years or younger, provided consent and were enrolled between May 22 and July 20, 2015 (except for 77 participants enrolled after these dates because of unavailability before the first mass drug administration, travel into the village during the trial, or birth), with 1447 enrolled into the intervention group and 1265 into the control group. 330 (23%) participants in the intervention group and 233 (18%) in the control group met the exclusion criteria for mass drug administration. Most children in the active case detection cohort were not treated because of height restrictions. 14 (4%) children in the intervention group and 10 (4%) in the control group were lost to follow-up. Cumulative malaria incidence was reduced in the intervention group (648 episodes among 327 children; estimated mean 2·00 episodes per child) compared with the control group (647 episodes among 263 children; 2·49 episodes per child; risk difference −0·49 [95% CI −0·79 to −0·21], p=0·0009, adjusted for sex and clustering). The risk of adverse events among all participants did not differ between groups (45 events [3%] among 1447 participants in the intervention group vs 24 events [2%] among 1265 in the control group; risk ratio 1·63 [1·01 to 2·67]; risk difference 1·21 [0·04 to 2·38], p=0·060), and no adverse reactions were reported. Interpretation Frequently repeated mass administrations of ivermectin during the malaria transmission season can reduce malaria episodes among children without significantly increasing harms in the populace. Funding Bill & Melinda Gates Foundation.
- Published
- 2019
155. Sibling species of the major malaria vector Anopheles gambiae display divergent preferences for aquatic breeding sites in southern Nigeria.
- Author
-
Ebhodaghe FI, Sanchez-Vargas I, Isaac C, Foy BD, and Hemming-Schroeder E
- Subjects
- Animals, Adult, Humans, Mosquito Vectors, Nigeria, Water, Larva, Breeding, Anopheles genetics, Malaria epidemiology
- Abstract
Background: When integrated with insecticide-treated bed nets, larval control of Anopheles mosquitoes could fast-track reductions in the incidence of human malaria. However, larval control interventions may deliver suboptimal outcomes where the preferred breeding places of mosquito vectors are not well known. This study investigated the breeding habitat choices of Anopheles mosquitoes in southern Nigeria. The objective was to identify priority sites for mosquito larval management in selected urban and periurban locations where malaria remains a public health burden. METHODS: Mosquito larvae were collected in urban and periurban water bodies during the wet-dry season interface in Edo, Delta, and Anambra States. Field-collected larvae were identified based on PCR gel-electrophoresis and amplicon sequencing, while the associations between Anopheles larvae and the properties and locations of water bodies were assessed using a range of statistical methods., Results: Mosquito breeding sites were either man-made (72.09%) or natural (27.91%) and mostly drainages (48.84%) and puddles (25.58%). Anopheles larvae occurred in drainages, puddles, stream margins, and a concrete well, and were absent in drums, buckets, car tires, and a water-holding iron pan, all of which contained culicine larvae. Wild-caught Anopheles larvae comprised Anopheles coluzzii (80.51%), Anopheles gambiae sensu stricto (s.s.) (11.54%), and Anopheles arabiensis (7.95%); a species-specific PCR confirmed the absence of the invasive urban malaria vector Anopheles stephensi among field-collected larvae. Anopheles arabiensis, An. coluzzii, and An. gambiae s.s. displayed preferences for turbid, lowland, and partially sunlit water bodies, respectively. Furthermore, An. arabiensis preferred breeding sites located outside 500 m of households, whereas An. gambiae s.s. and An. coluzzii had increased detection odds in sites within 500 m of households. Anopheles gambiae s.s. and An. coluzzii were also more likely to be present in natural water bodies; meanwhile, 96.77% of An. arabiensis were in man-made water bodies. Intraspecific genetic variations were little in the dominant vector An. coluzzii, while breeding habitat choices of populations made no statistically significant contributions to these variations., Conclusion: Sibling malaria vectors in the An. gambiae complex display divergent preferences for aquatic breeding habitats in southern Nigeria. The findings are relevant for planning targeted larval control of An. coluzzii whose increasing evolutionary adaptations to urban ecologies are driving the proliferation of the mosquito, and An. arabiensis whose adults typically evade the effects of treated bed nets due to exophilic tendencies., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
156. Corrigendum: Intrinsic factors driving mosquito vector competence and viral evolution: a review.
- Author
-
Lewis J, Gallichotte EN, Randall J, Glass A, Foy BD, Ebel GD, and Kading RC
- Abstract
[This corrects the article DOI: 10.3389/fcimb.2023.1330600.]., (Copyright © 2024 Lewis, Gallichotte, Randall, Glass, Foy, Ebel and Kading.)
- Published
- 2024
- Full Text
- View/download PDF
157. Intrinsic factors driving mosquito vector competence and viral evolution: a review.
- Author
-
Lewis J, Gallichotte EN, Randall J, Glass A, Foy BD, Ebel GD, and Kading RC
- Subjects
- Animals, Arthropod Vectors, Saliva, Mosquito Vectors, Culicidae
- Abstract
Mosquitoes are responsible for the transmission of numerous viruses of global health significance. The term "vector competence" describes the intrinsic ability of an arthropod vector to transmit an infectious agent. Prior to transmission, the mosquito itself presents a complex and hostile environment through which a virus must transit to ensure propagation and transmission to the next host. Viruses imbibed in an infectious blood meal must pass in and out of the mosquito midgut, traffic through the body cavity or hemocoel, invade the salivary glands, and be expelled with the saliva when the vector takes a subsequent blood meal. Viruses encounter physical, cellular, microbial, and immunological barriers, which are influenced by the genetic background of the mosquito vector as well as environmental conditions. Collectively, these factors place significant selective pressure on the virus that impact its evolution and transmission. Here, we provide an overview of the current state of the field in understanding the mosquito-specific factors that underpin vector competence and how each of these mechanisms may influence virus evolution., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Lewis, Gallichotte, Randall, Glass, Foy, Ebel and Kading.)
- Published
- 2023
- Full Text
- View/download PDF
158. Colorado tick fever virus: a review of historical literature and research emphasis for a modern era.
- Author
-
Harris EK, Foy BD, and Ebel GD
- Subjects
- Humans, Animals, Canada, Colorado tick fever virus, Dermacentor
- Abstract
Colorado tick fever virus is an understudied tick-borne virus of medical importance that is primarily transmitted in the western United States and southwestern Canada. The virus is the type species of the genus Coltivirus (Spinareoviridae) and consists of 12 segments that remain largely uncharacterized. Patterns of viral distribution are driven by the presence of the primary vector, the Rocky Mountain wood tick, Dermacentor andersoni. Infection prevalence in D. andersoni can range from 3% to 58% across the geographic distribution of the tick. Infection in humans can be severe and often presents with fever relapses but is rarely fatal. Here, we review the literature from primary characterizations in the early 20th century to current virus/vector research being conducted and identify vacancies in current research., (© The Author(s) 2023. Published by Oxford University Press on behalf of Entomological Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
159. Evaluation of Vector-Enabled Xenosurveillance in Rural Guatemala.
- Author
-
McMinn RJ, Chacon A, Rückert C, Scorza V, Young MC, Worthington D, Lamb MM, Medrano RE, Harris EK, Arias K, Lopez MR, Asturias EJ, Foy BD, Stenglein MD, Olson D, and Ebel GD
- Subjects
- Animals, Humans, Guatemala epidemiology, Prospective Studies, Mosquito Vectors, Mammals, Chickens, Culex, Aedes, Viruses
- Abstract
Surveillance methods that permit rapid detection of circulating pathogens in low-resource settings are desperately needed. In this study, we evaluated a mosquito bloodmeal-based surveillance method ("xenosurveillance") in rural Guatemala. Twenty households from two villages (Los Encuentros and Chiquirines) in rural southwest Guatemala were enrolled and underwent weekly prospective surveillance from August 2019 to December 2019 (16 weeks). When febrile illness was reported in a household, recently blood-fed mosquitoes were collected from within dwellings and blood samples taken from each member of the household. Mosquitoes were identified to species and blood sources identified by sequencing. Shotgun metagenomic sequencing was used to identify circulating viruses. Culex pipiens (60.9%) and Aedes aegypti (18.6%) were the most abundant mosquitoes collected. Bloodmeal sources were most commonly human (32.6%) and chicken (31.6%), with various other mammal and avian hosts detected. Several mosquito-specific viruses were detected, including Culex orthophasma virus. Human pathogens were not detected. Therefore, xenosurveillance may require more intensive sampling to detect human pathogens in Guatemala and ecologically similar localities in Central America.
- Published
- 2023
- Full Text
- View/download PDF
160. Predicted reduction in transmission from deployment of ivermectin-treated birdfeeders for local control of West Nile virus.
- Author
-
Holcomb KM, Nguyen C, Komar N, Foy BD, Panella NA, Baskett ML, and Barker CM
- Subjects
- Animals, Humans, Ivermectin pharmacology, Ivermectin therapeutic use, Birds, West Nile virus, West Nile Fever prevention & control, West Nile Fever veterinary, Culicidae, Culex
- Abstract
Ivermectin (IVM)-treated birds provide the potential for targeted control of Culex mosquitoes to reduce West Nile virus (WNV) transmission. Ingestion of IVM increases mosquito mortality, which could reduce WNV transmission from birds to humans and in enzootic maintenance cycles affecting predominantly bird-feeding mosquitoes and from birds to humans. This strategy might also provide an alternative method for WNV control that is less hampered by insecticide resistance and the logistics of large-scale pesticide applications. Through a combination of field studies and modeling, we assessed the feasibility and impact of deploying IVM-treated birdfeed in residential neighborhoods to reduce WNV transmission. We first tracked 105 birds using radio telemetry and radio frequency identification to monitor their feeder usage and locations of nocturnal roosts in relation to five feeder sites in a neighborhood in Fort Collins, Colorado. Using these results, we then modified a compartmental model of WNV transmission to account for the impact of IVM on mosquito mortality and spatial movement of birds and mosquitoes on the neighborhood level. We found that, while the number of treated lots in a neighborhood strongly influenced the total transmission potential, the arrangement of treated lots in a neighborhood had little effect. Increasing the proportion of treated birds, regardless of the WNV competency status, resulted in a larger reduction in infection dynamics than only treating competent birds. Taken together, model results indicate that deployment of IVM-treated feeders could reduce local transmission throughout the WNV season, including reducing the enzootic transmission prior to the onset of human infections, with high spatial coverage and rates of IVM-induced mortality in mosquitoes. To improve predictions, more work is needed to refine estimates of daily mosquito movement in urban areas and rates of IVM-induced mortality. Our results can guide future field trials of this control strategy., Competing Interests: Declaration of Competing Interest BDF, though Colorado State University, has filed a patent application on aspects underpinning this control method. Declaration of competing interest BDF, though Colorado State University, has filed a patent application on aspects underpinning this control method., (Published by Elsevier B.V.)
- Published
- 2023
- Full Text
- View/download PDF
161. Tracking antimalarial drug resistance using mosquito blood meals: a cross-sectional study.
- Author
-
Ehrlich HY, Somé AF, Bazié T, Ebou CN, Dembélé EL, Balma R, Goodwin J, Wade M, Bei AK, Ouédraogo JB, Foy BD, Dabiré RK, and Parikh S
- Subjects
- Animals, Humans, Cross-Sectional Studies, Plasmodium falciparum genetics, Polymerase Chain Reaction, Antimalarials pharmacology, Antimalarials therapeutic use, Culicidae, Folic Acid Antagonists
- Abstract
Background: Strong surveillance systems with wide geographic coverage are needed to detect and respond to reports of antimalarial drug resistance on the African continent. We aimed to assess the utility and feasibility of using blood-fed mosquitos (xenomonitoring) to conduct rapid surveillance of molecular markers associated with resistance in human populations., Methods: We conducted three cross-sectional surveys in two rainy seasons and the interim dry season in southwest Burkina Faso between Oct 10, 2018, and Sept 17, 2019. We collected human blood samples and blood-fed mosquitos residing in household clusters across seven village sectors. Samples were assessed for Plasmodium falciparum with ultrasensitive quantitative PCR, genotyped for two markers of reduced drug susceptibility, pfmdr1 256A>T (Asn86Tyr) and pfcrt 227A>C (Lys76Thr), and sequenced for four markers of clonality. We assessed statistical equivalence using a 10% margin of equivalence., Findings: We identified 551 infections in 1483 human blood samples (mean multiplicity of infection [MOI] 1·94, SD 1·47) and 346 infections in 2151 mosquito blood meals (mean MOI 2·2, SD 1·67). The frequency of pfmdr1 Asn86Tyr was 4% in survey 1, 2% in survey 2, and 12% in survey 3 in human samples, and 3% in survey 1, 0% in survey 2, and 8% in survey 3 in mosquito blood meals, and inter-host frequencies were statistically equivalent in surveys 1 and 2 (p<0·0001) but not Survey 3 (p=0·062) within a tolerability of 0·10. The frequency of pfcrt Lys76Thr was 16% in survey 1, 55% in survey 2, and 11% in survey 3 in humans and 40% in survey 1, 72% in survey 2, and 13% in survey 3 in mosquitos, and inter-host frequencies were equivalent in survey 3 only (p=0·032) within a tolerability of 0·10. In simulations, multiple but not preferential feeding behaviour in mosquitos reduced the accuracy of frequency estimates between hosts, particularly for markers circulating at higher frequencies., Interpretation: Molecular markers in mosquito blood meals and in humans exhibited similar temporal trends but frequencies were not statistically equivalent in all scenarios. More work is needed to determine empirical and pragmatic thresholds of difference. Xenomonitoring might be an efficient tool to provide rapid information on emerging antimalarial resistance in regions with insufficient surveillance., Funding: National Institute of Allergy and Infectious Diseases., Translation: For the French translation of the abstract see Supplementary Materials section., Competing Interests: Declaration of interests We declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY-NC-ND 4.0 license. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
162. Repeat Ivermectin Mass Drug Administrations for Malaria Control II: Protocol for a Double-blind, Cluster-Randomized, Placebo-Controlled Trial for the Integrated Control of Malaria.
- Author
-
Foy BD, Some A, Magalhaes T, Gray L, Rao S, Sougue E, Jackson CL, Kittelson J, Slater HC, Bousema T, Da O, Coulidiaty AGV, Colt M, Wade M, Richards K, Some AF, Dabire RK, and Parikh S
- Abstract
Background: The gains made against malaria have stagnated since 2015, threatened further by increasing resistance to insecticides and antimalarials. Improvement in malaria control necessitates a multipronged strategy, which includes the development of novel tools. One such tool is mass drug administration (MDA) with endectocides, primarily ivermectin, which has shown promise in reducing malaria transmission through lethal and sublethal impacts on the mosquito vector., Objective: The primary objective of the study is to assess the impact of repeated ivermectin MDA on malaria incidence in children aged ≤10 years., Methods: Repeat Ivermectin MDA for Malaria Control II is a double-blind, placebo-controlled, cluster-randomized, and parallel-group trial conducted in a setting with intense seasonal malaria transmission in Southwest Burkina Faso. The study included 14 discrete villages: 7 (50%) randomized to receive standard measures (seasonal malaria chemoprevention [SMC] and bed net use for children aged 3 to 59 months) and placebo, and 7 (50%) randomized to receive standard measures and monthly ivermectin MDA at 300 μg/kg for 3 consecutive days, provided under supervision to all eligible village inhabitants, over 2 successive rainy seasons. Nonpregnant individuals >90 cm in height were eligible for ivermectin MDA, and cotreatment with ivermectin and SMC was not permitted. The primary outcome is malaria incidence in children aged ≤10 years, as assessed by active case surveillance. The secondary safety outcome of repeated ivermectin MDA was assessed through active and passive adverse event monitoring., Results: The trial intervention was conducted from July to November in 2019 and 2020, with additional sampling of humans and mosquitoes occurring through February 2022 to assess postintervention changes in transmission patterns. Additional human and entomological assessments were performed over the 2 years in a subset of households from 6 cross-sectional villages. A subset of individuals underwent additional sampling in 2020 to characterize ivermectin pharmacokinetics and pharmacodynamics. Analysis and unblinding will commence once the database has been completed, cleaned, and locked., Conclusions: Our trial represents the first study to directly assess the impact of a novel approach for malaria control, ivermectin MDA as a mosquitocidal agent, layered into existing standard-of-care interventions. The study was designed to leverage the current SMC deployment infrastructure and will provide evidence regarding the additional benefit of ivermectin MDA in reducing malaria incidence in children., Trial Registrations: ClinicalTrials.gov NCT03967054; https://clinicaltrials.gov/ct2/show/NCT03967054 and Pan African Clinical Trials Registry PACT201907479787308; https://pactr.samrc.ac.za/TrialDisplay.aspx?TrialID=8219., International Registered Report Identifier (irrid): DERR1-10.2196/41197., (©Brian D Foy, Anthony Some, Tereza Magalhaes, Lyndsey Gray, Sangeeta Rao, Emmanuel Sougue, Conner L Jackson, John Kittelson, Hannah C Slater, Teun Bousema, Ollo Da, A Gafar V Coulidiaty, McKenzie Colt, Martina Wade, Kacey Richards, A Fabrice Some, Roch K Dabire, Sunil Parikh. Originally published in JMIR Research Protocols (https://www.researchprotocols.org), 20.03.2023.)
- Published
- 2023
- Full Text
- View/download PDF
163. Back to the Future: Quantifying Wing Wear as a Method to Measure Mosquito Age.
- Author
-
Gray L, Asay BC, Hephaestus B, McCabe R, Pugh G, Markle ED, Churcher TS, and Foy BD
- Abstract
Vector biologists have long sought the ability to accurately quantify the age of wild mosquito populations, a metric used to measure vector control efficiency. This has proven difficult due to the difficulties of working in the field and the biological complexities of wild mosquitoes. Ideal age grading techniques must overcome both challenges while also providing epidemiologically relevant age measurements. Given these requirements, the Detinova parity technique, which estimates age from the mosquito ovary and tracheole skein morphology, has been most often used for mosquito age grading despite significant limitations, including being based solely on the physiology of ovarian development. Here, we have developed a modernized version of the original mosquito aging method that evaluated wing wear, expanding it to estimate mosquito chronological age from wing scale loss. We conducted laboratory experiments using adult Anopheles gambiae held in insectary cages or mesocosms, the latter of which also featured ivermectin bloodmeal treatments to change the population age structure. Mosquitoes were age graded by parity assessments and both human- and computational-based wing evaluations. Although the Detinova technique was not able to detect differences in age population structure between treated and control mesocosms, significant differences were apparent using the wing scale technique. Analysis of wing images using averaged left- and right-wing pixel intensity scores predicted mosquito age at high accuracy (overall test accuracy: 83.4%, average training accuracy: 89.7%). This suggests that this technique could be an accurate and practical tool for mosquito age grading though further evaluation in wild mosquito populations is required.
- Published
- 2022
- Full Text
- View/download PDF
164. Two-year Decay of Zika Virus Neutralizing Antibodies in People Living in an Endemic Region in Brazil.
- Author
-
Magalhaes T, Morais CNL, Azevedo EAN, Jacques IJAA, Castanha PMS, Cordeiro MT, Braga C, Jaenisch T, Marques ETA, and Foy BD
- Subjects
- Antibodies, Neutralizing, Antibodies, Viral, Brazil epidemiology, Cross Reactions, Humans, Dengue, Dengue Virus, Zika Virus, Zika Virus Infection
- Abstract
It is currently not clear whether humoral immunity to Zika virus (ZIKV) elicited upon natural ZIKV infection is long-lasting. In addition, cross-reactivity of anti-ZIKV antibodies with antigenically related dengue viruses (DENV) may have biological implications in nonnaive individuals who subsequently acquire a heterotypic infection. Cross-reactive humoral immunity between ZIKV and DENV also complicates the interpretation of serological tests to evaluate previous exposure to either virus. Here, we have measured the 2-year decay of ZIKV neutralizing antibodies in people living in a ZIKV/DENV endemic area in Brazil who were identified as having an acute infection (group 1) or past (but recent) infection (group 2) with ZIKV in 2015-16. The titers of neutralizing antibodies to ZIKV decreased 9.1 and 2.3 times in groups 1 and 2, respectively. We also show that the plaque reduction neutralization assay (PRNT) is a reliable method to measure past exposure to ZIKV in coendemic areas.
- Published
- 2022
- Full Text
- View/download PDF
165. Effects of ivermectin treatment of backyard chickens on mosquito dynamics and West Nile virus transmission.
- Author
-
Holcomb KM, Nguyen C, Foy BD, Ahn M, Cramer K, Lonstrup ET, Mete A, Tell LA, and Barker CM
- Subjects
- Animals, Chickens, Ivermectin pharmacology, Ivermectin therapeutic use, Mosquito Vectors, Culex, Culicidae, West Nile Fever drug therapy, West Nile Fever epidemiology, West Nile Fever veterinary, West Nile virus
- Abstract
Background: Vector control strategies typically rely on pesticides to target mosquitoes involved in enzootic and zoonotic transmission of West Nile virus (WNV). Nevertheless, increasing insecticide resistance and a desire to reduce pesticide usage provide the impetus for developing alternative strategies. Ivermectin (IVM), an antiparasitic drug which is widely used in human and veterinary medicine, is a potential alternative for targeted control because Culex mosquitoes experience increased mortality following ingestion of IVM in bloodmeals., Methodology/principal Findings: We conducted a randomized field trial to investigate the impact of treating backyard chicken flocks with IVM in urban neighborhoods across Davis, California on mosquito populations and WNV transmission dynamics. We observed a significant reduction in WNV seroconversions in treated vs. untreated chickens, suggesting a reduction in WNV transmission intensity around treated flocks. We also detected a reduction in parity rates of Cx. tarsalis near treated vs. untreated flocks and increased mortality in wild mosquitoes following a bloodmeal on treated chickens (IVM serum concentration > 5ng/mL) vs. chickens with IVM serum concentrations < 5 ng/mL. However, we did not find a significant difference in abundance or infection prevalence in mosquitoes between treatment groups associated with the reductions in seroconversions. Mosquito immigration from surrounding larval habitat, relatively low WNV activity in the study area, and variable IVM serum concentrations likely contributed to uncertainty about the impact., Conclusions/significance: Taken together, our results point to a reduction in WNV transmission due to the impact of IVM on Culex mosquito populations and support the ongoing investigation of oral administration of IVM to wild birds for local control of WNV transmission, although further work is needed to optimize dosing and understand effects on entomological endpoints., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: BDF, through Colorado State University, has filed a patent application for aspects underpinning the control methods evaluated in this study.
- Published
- 2022
- Full Text
- View/download PDF
166. Design and analysis of a 2-year parallel follow-up of repeated ivermectin mass drug administrations for control of malaria: Small sample considerations for cluster-randomized trials with count data.
- Author
-
Jackson CL, Colborn K, Gao D, Rao S, Slater HC, Parikh S, Foy BD, and Kittelson J
- Subjects
- Cluster Analysis, Follow-Up Studies, Humans, Mass Drug Administration, Randomized Controlled Trials as Topic, Research Design, Sample Size, Ivermectin, Malaria drug therapy, Malaria prevention & control
- Abstract
Background: Cluster-randomized trials allow for the evaluation of a community-level or group-/cluster-level intervention. For studies that require a cluster-randomized trial design to evaluate cluster-level interventions aimed at controlling vector-borne diseases, it may be difficult to assess a large number of clusters while performing the additional work needed to monitor participants, vectors, and environmental factors associated with the disease. One such example of a cluster-randomized trial with few clusters was the "efficacy and risk of harms of repeated ivermectin mass drug administrations for control of malaria" trial. Although previous work has provided recommendations for analyzing trials like repeated ivermectin mass drug administrations for control of malaria, additional evaluation of the multiple approaches for analysis is needed for study designs with count outcomes., Methods: Using a simulation study, we applied three analysis frameworks to three cluster-randomized trial designs (single-year, 2-year parallel, and 2-year crossover) in the context of a 2-year parallel follow-up of repeated ivermectin mass drug administrations for control of malaria. Mixed-effects models, generalized estimating equations, and cluster-level analyses were evaluated. Additional 2-year parallel designs with different numbers of clusters and different cluster correlations were also explored., Results: Mixed-effects models with a small sample correction and unweighted cluster-level summaries yielded both high power and control of the Type I error rate. Generalized estimating equation approaches that utilized small sample corrections controlled the Type I error rate but did not confer greater power when compared to a mixed model approach with small sample correction. The crossover design generally yielded higher power relative to the parallel equivalent. Differences in power between analysis methods became less pronounced as the number of clusters increased. The strength of within-cluster correlation impacted the relative differences in power., Conclusion: Regardless of study design, cluster-level analyses as well as individual-level analyses like mixed-effects models or generalized estimating equations with small sample size corrections can both provide reliable results in small cluster settings. For 2-year parallel follow-up of repeated ivermectin mass drug administrations for control of malaria, we recommend a mixed-effects model with a pseudo-likelihood approximation method and Kenward-Roger correction. Similarly designed studies with small sample sizes and count outcomes should consider adjustments for small sample sizes when using a mixed-effects model or generalized estimating equation for analysis. Although the 2-year parallel follow-up of repeated ivermectin mass drug administrations for control of malaria is already underway as a parallel trial, applying the simulation parameters to a crossover design yielded improved power, suggesting that crossover designs may be valuable in settings where the number of available clusters is limited. Finally, the sensitivity of the analysis approach to the strength of within-cluster correlation should be carefully considered when selecting the primary analysis for a cluster-randomized trial.
- Published
- 2021
- Full Text
- View/download PDF
167. Characterization of subclinical ZIKV infection in immune-competent guinea pigs and mice.
- Author
-
Westrich JA, McNulty EE, Edmonds MJ, Nalls AV, Miller MR, Foy BD, Rovnak J, Perera R, and Mathiason CK
- Subjects
- Animals, Chlorocebus aethiops, Female, Guinea Pigs, Humans, Infectious Disease Transmission, Vertical, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Pregnancy, Vero Cells, Fetus immunology, Fetus virology, Placenta immunology, Placenta virology, Pregnancy Complications, Infectious immunology, Pregnancy Complications, Infectious virology, Zika Virus immunology, Zika Virus pathogenicity, Zika Virus Infection virology
- Abstract
An infectious agent's pathogenic and transmission potential is heavily influenced by early events during the asymptomatic or subclinical phase of disease. During this phase, the presence of infectious agent may be relatively low. An important example of this is Zika virus (ZIKV), which can cross the placenta and infect the foetus, even in mothers with subclinical infections. These subclinical infections represent roughly 80 % of all human infections. Initial ZIKV pathogenesis studies were performed in type I interferon receptor (IFNAR) knockout mice. Blunting the interferon response resulted in robust infectivity, and increased the utility of mice to model ZIKV infections. However, due to the removal of the interferon response, the use of these models impedes full characterization of immune responses to ZIKV-related pathologies. Moreover, IFNAR-deficient models represent severe disease whereas less is known regarding subclinical infections. Investigation of the anti-viral immune response elicited at the maternal-foetal interface is critical to fully understand mechanisms involved in foetal infection, foetal development, and disease processes recognized to occur during subclinical maternal infections. Thus, immunocompetent experimental models that recapitulate natural infections are needed. We have established subclinical intravaginal ZIKV infections in mice and guinea pigs. We found that these infections resulted in: the presence of both ZIKV RNA transcripts and infectious virus in maternal and placental tissues, establishment of foetal infections and ZIKV-mediated CXCL10 expression. These models will aid in discerning the mechanisms of subclinical ZIKV mother-to-offspring transmission, and by extension can be used to investigate other maternal infections that impact foetal development.
- Published
- 2021
- Full Text
- View/download PDF
168. Three Immunocompetent Small Animal Models That Do Not Support Zika Virus Infection.
- Author
-
Miller MR, Fagre AC, Clarkson TC, Markle ED, and Foy BD
- Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that is primarily transmitted to humans through the bite of an infected mosquito. ZIKV causes disease in infected humans with added complications of Guillain-Barré syndrome and birth defects in infants born to mothers infected during pregnancy. There are several large immunocompetent animal models for ZIKV including non-human primates (NHPs). NHP models closely reflect human infection; however, due to sample size restrictions, investigations into the effects of transmission route and the impacts on disease dynamics have been understudied. Mice have been widely used for modeling ZIKV infection, yet there are few ZIKV-susceptible immunocompetent mouse models and none of these have been used to investigate sexual transmission. In an effort to identify a small immunocompetent animal model to characterize sexual transmission of ZIKV, we attempt experimental infection of multimammate mice, New Zealand white rabbits, and Hartley guinea pigs. The multimammate mouse is the natural reservoir of Lassa fever virus and has been identified to harbor other human pathogens. Likewise, while NZW rabbits are susceptible to West Nile virus, they have not yet been examined for their susceptibility to infection with ZIKV. Guinea pigs have been successfully used as models for ZIKV infection, but only in immunocompromised life stages (young or pregnant). Here, it was found that the multimammate mouse and New Zealand White (NZW) rabbits are not susceptible ZIKV infection as determined by a lack viral RNA in tissues and fluids collected. Sexually mature male Hartley guinea pigs were inoculated subcutaneously and by mosquito bite, but found to be refractory to ZIKV infection, contrary to findings of other studies in young and pregnant guinea pigs. Interestingly, here it is shown that adult male guinea pigs are not susceptible to ZIKV infection, even when infected by natural route (e.g., mosquito bite). Although a new small animal model for the sexual transmission for ZIKV was not established through this study, these findings provide information on outbred animal species that are not permissive to infection (NZW rabbits and multimammate mice) and new information surrounding limitations of a previously established animal model (guinea pigs).
- Published
- 2021
- Full Text
- View/download PDF
169. Nootkatone Is an Effective Repellent against Aedes aegypti and Aedes albopictus .
- Author
-
Clarkson TC, Janich AJ, Sanchez-Vargas I, Markle ED, Gray M, Foster JR, Black Iv WC, Foy BD, and Olson KE
- Abstract
We tested a nootkatone product for insecticide activity against the most prominent vectors of Zika virus (ZIKV), Aedes aegypti , and Aedes albopictus. We tested the permethrin-resistant (PERM-R) Vergel strain of A. aegypti and the permethrin-susceptible (PERM-S) New Orleans strain of A. aegypti to determine if insecticide resistance affected their susceptibility to nootkatone. Bottle bioassays showed that the PERM-S strain (New Orleans) was more susceptible to nootkatone than the confirmed A. aegypti permethrin-resistant (PERM-R) strain, Vergel. The A. albopictus strain ATM-NJ95 was a known PERM-S strain and Coatzacoalcos permethrin susceptibility was unknown but proved to be similar to the ATM-NJ95 PERM-S phenotype. The A. albopictus strains (ATM-NJ95 and Coatzacoalcos) were as susceptible to nootkatone as the New Orleans strain. Bottle bioassays conducted with ZIKV-infected mosquitoes showed that the New Orleans (PERM-S) strain was as susceptible to nootkatone as the mock-infected controls, but the PERM-R strain was less susceptible to nootkatone than the mock-infected controls. Repellency/irritancy and biting inhibition bioassays (RIBB) of A. aegypti determined whether the nootkatone-treated arms of three human subjects prevented uninfected A. aegypti mosquitoes from being attracted to the test subjects and blood-feeding on them. The RIBB analyses data calculated the spatial activity index (SAI) and biting inhibition factor (BI) of A. aegypti at different nootkatone concentrations and then compared the SAI and BI of existing repellency products. We concluded that nootkatone repelled mosquitoes at a rate comparable to 7% DEET or 5% picaridin and has the potential to be an efficacious repellent against adult A. aegypti mosquitoes.
- Published
- 2021
- Full Text
- View/download PDF
170. Characterizing and Quantifying Arbovirus Transmission by Aedes aegypti Using Forced Salivation and Analysis of Bloodmeals.
- Author
-
Miller MR, Sorensen MR, Markle ED, Clarkson TC, Knight AL, Savran MJ, and Foy BD
- Abstract
Arbovirus transmission studies are dependent on the ability to estimate the titer of virus transmitted from infectious mosquitoes to a host. There are several methods for estimating virus titer in mosquito saliva, including (1) using forced salivation (FS) whereby the infectious mosquito's proboscis is forced into a capillary tube containing media to collect and test their saliva for virus, and (2) by quantifying virus expectorated into host tissues or into the blood contained in an artificial feeder immediately after blood feeding. We studied FS and bloodmeals to estimate and compare titers of Zika virus and chikungunya virus transmitted by the mosquito vector Aedes aegypti . Infectious virus and viral genomes of both viruses were detected more often from individual mosquitoes using immersion oil for the FS media compared to fetal bovine serum (FBS) plus glycerol, but the FS media had no influence on virus quantification from positive samples. FS virus titers were equivalent when comparing individuals or groups of mosquitoes that never received a blood meal compared to those that were blood fed immediately prior, showing that blood feeding does not influence FS. This suggested that performing FS on mosquitoes after blood feeding might be an efficient way to estimate virus transmitted during blood feeding. However, detecting virus from the blood remaining in an artificial feeder post-blood feeding was mostly unsuccessful relative to quantifying virus from FS of the post-blood fed mosquitoes. In contrast, immunocompromised mice always became infected after being fed on by Zika-infected mosquitoes, even when no infectious virus was detected in their saliva by FS post-blood feed. Due to this discrepancy, we tested the ingested bloodmeals of individual mosquitoes that fed on artificial blood feeders for virus, and compared these to virus in their saliva harvested from FS and to virus in their bodies. These experiments revealed ~50-100 times higher virus titers in the dissected bloodmeals compared to those detected in the same mosquitoes' saliva, demonstrating how mosquitoes re-ingest much of their saliva during artificial blood feeding, and highlighting a large increase in virus transmission during Aedes aegypti blood feeding. Both FS and the dissected bloodmeals of artificially blood-fed mosquitoes showed that the quantity of viral RNA expectorated by mosquitoes was 2-5 logs more than the quantity of infectious virus. The results from this study add critical information to understanding and quantifying the transmission of Aedes aegypti arboviruses.
- Published
- 2021
- Full Text
- View/download PDF
171. Considerations for Human Blood-Feeding and Arthropod Exposure in Vector Biology Research: An Essential Tool for Investigations and Disease Control.
- Author
-
Harrington LC, Foy BD, and Bangs MJ
- Subjects
- Animals, Ethics Committees, Research, Humans, Research Personnel, Arthropod Vectors physiology, Arthropods physiology, Feeding Behavior physiology, Vector Borne Diseases blood, Vector Borne Diseases transmission
- Abstract
Eventually there may be a broadly acceptable, even perfected, substitute for the human host requirement for direct feeding experiments by arthropods, most notably mosquitoes. However, for now, direct and indirect feeding on human volunteers is an important, if not essential, tool in vector biology research (VBR). This article builds on the foundational publication by Achee et al. (2015) covering considerations for the use of human participants in VBR pursuits. The authors introduced methods involving human participation in VBR, while detailing human-landing collections (catches) as a prime example. Benedict et al. (2018) continued this theme with an overview of human participation and considerations for research that involves release of mosquito vectors into the environment. In this study, we discuss another important aspect of human use in VBR activities: considerations addressing studies that require an arthropod to feed on a live human host. Using mosquito studies as our principal example, in this study, we discuss the tremendous importance and value of this approach to support and allow study of a wide variety of factors and interactions related to our understanding of vector-borne diseases and their control. This includes establishment of laboratory colonies for test populations, characterization of essential nutrients that contribute to mosquito fitness, characterization of blood-feeding (biting) behavior and pathogen transmission, parameterization for modeling transmission dynamics, evaluation of human host attraction and/or agents that repel, and the effectiveness of antivector or parasite therapeutic drug studies.
- Published
- 2020
- Full Text
- View/download PDF
172. Partitiviruses Infecting Drosophila melanogaster and Aedes aegypti Exhibit Efficient Biparental Vertical Transmission.
- Author
-
Cross ST, Maertens BL, Dunham TJ, Rodgers CP, Brehm AL, Miller MR, Williams AM, Foy BD, and Stenglein MD
- Subjects
- Animals, Drosophila melanogaster, Female, Male, Aedes virology, Double Stranded RNA Viruses metabolism
- Abstract
Partitiviruses are segmented, multipartite double-stranded RNA (dsRNA) viruses that until recently were only known to infect fungi, plants, and protozoans. Metagenomic surveys have revealed that partitivirus-like sequences are also commonly associated with arthropods. One arthropod-associated partitivirus, galbut virus, is common in wild populations of Drosophila melanogaster To begin to understand the processes that underlie this virus's high global prevalence, we established colonies of wild-caught infected flies. Infection remained at stably high levels over 3 years, with between 63 and 100% of individual flies infected. Galbut virus infects fly cells and replicates in tissues throughout infected adults, including reproductive tissues and the gut epithelium. We detected no evidence of horizontal transmission via ingestion, but vertical transmission from either infected females or infected males was ∼100% efficient. Vertical transmission of a related partitivirus, verdadero virus, that we discovered in a laboratory colony of Aedes aegypti mosquitoes was similarly efficient. This suggests that efficient biparental vertical transmission may be a feature of at least a subset of insect-infecting partitiviruses. To study the impact of galbut virus infection free from the confounding effect of other viruses, we generated an inbred line of flies with galbut virus as the only detectable virus infection. We were able to transmit infection experimentally via microinjection of homogenate from these galbut-only flies. This sets the stage for experiments to understand the biological impact and possible utility of partitiviruses infecting model organisms and disease vectors. IMPORTANCE Galbut virus is a recently discovered partitivirus that is extraordinarily common in wild populations of the model organism Drosophila melanogaster Like for most viruses discovered through metagenomics, most of the basic biological questions about this virus remain unanswered. We found that galbut virus, along with a closely related partitivirus found in Aedes aegypti mosquitoes, is transmitted from infected females or males to offspring with ∼100% efficiency and can be maintained in laboratory colonies over years. This efficient transmission mechanism likely underlies the successful spread of these viruses through insect populations. We created Drosophila lines that contained galbut virus as the only virus infection and showed that these flies can be used as a source for experimental infections. This provides insight into how arthropod-infecting partitiviruses may be maintained in nature and sets the stage for exploration of their biology and potential utility., (Copyright © 2020 American Society for Microbiology.)
- Published
- 2020
- Full Text
- View/download PDF
173. Sexual Transmission of Arboviruses: A Systematic Review.
- Author
-
Blitvich BJ, Magalhaes T, Laredo-Tiscareño SV, and Foy BD
- Subjects
- Animals, Arboviruses classification, Genitalia virology, Humans, Semen virology, Arbovirus Infections transmission, Arbovirus Infections virology, Arboviruses isolation & purification, Sexually Transmitted Diseases transmission, Sexually Transmitted Diseases virology
- Abstract
Arthropod-borne viruses (arboviruses) are primarily maintained in nature in transmission cycles between hematophagous arthropods and vertebrate hosts, but an increasing number of arboviruses have been isolated from or indirectly detected in the urogenital tract and sexual secretions of their vertebrate hosts, indicating that further investigation on the possibility of sexual transmission of these viruses is warranted. The most widely recognized sexually-transmitted arbovirus is Zika virus but other arboviruses, including Crimean-Congo hemorrhagic fever virus and dengue virus, might also be transmitted, albeit occasionally, by this route. This review summarizes our current understanding on the ability of arboviruses to be sexually transmitted. We discuss the sexual transmission of arboviruses between humans and between vertebrate animals, but not arthropod vectors. Every taxonomic group known to contain arboviruses ( Asfarviridae , Bunyavirales , Flaviviridae , Orthomyxoviridae , Reoviridae , Rhabdoviridae and Togaviridae ) is covered.
- Published
- 2020
- Full Text
- View/download PDF
174. Investigating selected host and parasite factors potentially impacting upon seasonal malaria chemoprevention in Bama, Burkina Faso.
- Author
-
Somé FA, Bazié T, Ehrlich HY, Goodwin J, Lehane A, Neya C, Zachari K, Wade M, Ouattara JM, Foy BD, Dabiré RK, Parikh S, and Ouédraogo JB
- Subjects
- Amodiaquine blood, Amodiaquine therapeutic use, Antimalarials therapeutic use, Burkina Faso epidemiology, Chemoprevention, Child, Preschool, Cross-Sectional Studies, Female, Humans, Infant, Malaria, Falciparum epidemiology, Malaria, Falciparum parasitology, Male, Plasma chemistry, Amodiaquine analogs & derivatives, Antimalarials blood, Cytochrome P-450 CYP2C8 genetics, Drug Resistance genetics, Genes, Protozoan drug effects, Malaria, Falciparum prevention & control, Polymorphism, Genetic drug effects
- Abstract
Background: Since 2014, seasonal malaria chemoprevention (SMC) with amodiaquine-sulfadoxine-pyrimethamine (AQ-SP) has been implemented on a large scale during the high malaria transmission season in Burkina Faso. This paper reports the prevalence of microscopic and submicroscopic malaria infection at the outset and after the first round of SMC in children under 5 years old in Bama, Burkina Faso, as well as host and parasite factors involved in mediating the efficacy and tolerability of SMC., Methods: Two sequential cross-sectional surveys were conducted in late July and August 2017 during the first month of SMC in a rural area in southwest Burkina Faso. Blood smears and dried blood spots were collected from 106 to 93 children under five, respectively, at the start of SMC and again 3 weeks later. Malaria infection was detected by microscopy and by PCR from dried blood spots. For all children, day 7 plasma concentrations of desethylamodiaquine (DEAQ) were measured and CYP2C8 genetic variants influencing AQ metabolism were genotyped. Samples were additionally genotyped for pfcrt K76T and pfmdr1 N86Y, molecular markers associated with reduced amodiaquine susceptibility., Results: 2.8% (3/106) of children were positive for Plasmodium falciparum infection by microscopy and 13.2% (14/106) by nested PCR within 2 days of SMC administration. Three weeks after SMC administration, in the same households, 4.3% (4/93) of samples were positive by microscopy and 14.0% (13/93) by PCR (p = 0.0007). CYP2C8*2, associated with impaired amodiaquine metabolism, was common with an allelic frequency of 17.1% (95% CI 10.0-24.2). Day 7 concentration of DEAQ ranged from 0.48 to 362.80 ng/mL with a median concentration of 56.34 ng/mL. Pfmdr1 N86 predominated at both time points, whilst a non-significant trend towards a higher prevalence of pfcrt 76T was seen at week 3., Conclusion: This study showed a moderate prevalence of low-level malaria parasitaemia in children 3 weeks following SMC during the first month of administration. Day 7 concentrations of the active DEAQ metabolite varied widely, likely reflecting variability in adherence and possibly metabolism. These findings highlight factors that may contribute to the effectiveness of SMC in children in a high transmission setting.
- Published
- 2020
- Full Text
- View/download PDF
175. The Endless Challenges of Arboviral Diseases in Brazil.
- Author
-
Magalhaes T, Chalegre KDM, Braga C, and Foy BD
- Abstract
In this Editorial, we list and discuss some of the main challenges faced by the population and public health authorities in Brazil concerning arbovirus infections, including the occurrence of concurrent epidemics like the ongoing SARS-CoV-2/COVID-19 pandemic., Competing Interests: The authors declare no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
176. Ivermectin as a novel complementary malaria control tool to reduce incidence and prevalence: a modelling study.
- Author
-
Slater HC, Foy BD, Kobylinski K, Chaccour C, Watson OJ, Hellewell J, Aljayyoussi G, Bousema T, Burrows J, D'Alessandro U, Alout H, Ter Kuile FO, Walker PGT, Ghani AC, and Smit MR
- Subjects
- Animals, Antimalarials therapeutic use, Artemisinins therapeutic use, Burkina Faso epidemiology, Child, Female, Humans, Incidence, Insecticides, Male, Mass Drug Administration, Prevalence, Quinolines therapeutic use, Randomized Controlled Trials as Topic, Seasons, Ivermectin administration & dosage, Malaria epidemiology, Malaria prevention & control, Mosquito Vectors
- Abstract
Background: Ivermectin is a potential new vector control tool to reduce malaria transmission. Mosquitoes feeding on a bloodmeal containing ivermectin have a reduced lifespan, meaning they are less likely to live long enough to complete sporogony and become infectious. We aimed to estimate the effect of ivermectin on malaria transmission in various scenarios of use., Methods: We validated an existing population-level mathematical model of the effect of ivermectin mass drug administration (MDA) on the mosquito population and malaria transmission against two datasets: clinical data from a cluster- randomised trial done in Burkina Faso in 2015 wherein ivermectin was given to individuals taller than 90 cm and entomological data from a study of mosquito outcomes after ivermectin MDA for onchocerciasis or lymphatic filariasis in Burkina Faso, Senegal, and Liberia between 2008 and 2013. We extended the existing model to include a range of complementary malaria interventions (seasonal malaria chemoprevention and MDA with dihydroartemisinin-piperaquine) and to incorporate new data on higher doses of ivermectin with a longer mosquitocidal effect. We consider two ivermectin regimens: a single dose of 400 μg/kg (1 × 400 μg/kg) and three consecutive daily doses of 300 μg/kg per day (3 × 300 μg/kg). We simulated the effect of these two doses in a range of usage scenarios in different transmission settings (highly seasonal, seasonal, and perennial). We report percentage reductions in clinical incidence and slide prevalence., Findings: We estimate that MDA with ivermectin will reduce prevalence and incidence and is most effective in areas with highly seasonal transmission. In a highly seasonal moderate transmission setting, three rounds of ivermectin only MDA at 3 × 300 μg/kg (rounds spaced 1 month apart) and 70% coverage is predicted to reduce clinical incidence by 71% and prevalence by 34%. We predict that adding ivermectin MDA to seasonal malaria chemoprevention in this setting would reduce clinical incidence by an additional 77% in children younger than 5 years compared with seasonal malaria chemoprevention alone; adding ivermectin MDA to MDA with dihydroartemisinin-piperaquine in this setting would reduce incidence by an additional 75% and prevalence by an additional 64% (all ages) compared with MDA with dihydroartemisinin-piperaquine alone., Interpretation: Our modelling predictions suggest that ivermectin could be a valuable addition to the malaria control toolbox, both in areas with persistently high transmission where existing interventions are insufficient and in areas approaching elimination to prevent resurgence., Funding: Imperial College Junior Research Fellowship., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
177. Successive blood meals enhance virus dissemination within mosquitoes and increase transmission potential.
- Author
-
Armstrong PM, Ehrlich HY, Magalhaes T, Miller MR, Conway PJ, Bransfield A, Misencik MJ, Gloria-Soria A, Warren JL, Andreadis TG, Shepard JJ, Foy BD, Pitzer VE, and Brackney DE
- Subjects
- Aedes ultrastructure, Animals, Arbovirus Infections blood, Arbovirus Infections virology, Basic Reproduction Number, Chikungunya Fever transmission, Dengue transmission, Digestive System ultrastructure, Digestive System virology, Female, Host Microbial Interactions, Humans, Male, Mice, Microscopy, Electron, Scanning, Mosquito Vectors ultrastructure, Zika Virus Infection transmission, Aedes virology, Arbovirus Infections transmission, Models, Biological, Mosquito Vectors virology
- Abstract
The recent Zika virus (ZIKV) and chikungunya virus epidemics highlight the explosive nature of arthropod-borne viruses (arboviruses) transmitted by Aedes spp. mosquitoes
1,2 . Vector competence and the extrinsic incubation period (EIP) are two key entomological parameters used to assess the public health risk posed by arboviruses3 . These are typically measured empirically by offering mosquitoes an infectious blood meal and temporally sampling mosquitoes to determine the infection and transmission status. This approach has been used for the better part of a century; however, it does not accurately capture the biology and behaviour of many mosquito vectors that refeed frequently (every 2-3 d)4 . Here, we demonstrate that acquisition of a second non-infectious blood meal significantly shortens the EIP of ZIKV-infected Aedes aegypti by enhancing virus dissemination from the mosquito midgut. Similarly, a second blood meal increases the competence of this species for dengue virus and chikungunya virus as well as Aedes albopictus for ZIKV, suggesting that this phenomenon may be common among other virus-vector pairings and that A. albopictus might be a more important vector than once thought. Blood-meal-induced microperforations in the virus-impenetrable basal lamina that surrounds the midgut provide a mechanism for enhanced virus escape. Modelling of these findings reveals that a shortened EIP would result in a significant increase in the basic reproductive number, R0 , estimated from experimental data. This helps to explain how A. aegypti can sustain explosive epidemics such as ZIKV despite relatively poor vector competence in single-feed laboratory trials. Together, these data demonstrate a direct and unrecognized link between mosquito feeding behaviour, EIP and vector competence.- Published
- 2020
- Full Text
- View/download PDF
178. Analysis of the RIMDAMAL trial - Authors' reply.
- Author
-
Foy BD, Rao S, Parikh S, Slater HC, and Dabiré RK
- Subjects
- Humans, Ivermectin, Malaria, Mass Drug Administration
- Published
- 2019
- Full Text
- View/download PDF
179. Induction of RNA interference to block Zika virus replication and transmission in the mosquito Aedes aegypti.
- Author
-
Magalhaes T, Bergren NA, Bennett SL, Borland EM, Hartman DA, Lymperopoulos K, Sayre R, Borlee BR, Campbell CL, Foy BD, Olson KE, Blair CD, Black W 4th, and Kading RC
- Subjects
- Animals, Cattle, Chlorocebus aethiops, Mosquito Vectors virology, Pilot Projects, RNA, Double-Stranded, RNA, Small Interfering, Saliva virology, Sequence Analysis, RNA, Vero Cells, Viral Load, Virus Replication, Zika Virus physiology, Zika Virus Infection virology, Aedes virology, RNA Interference, Zika Virus genetics, Zika Virus Infection transmission
- Abstract
The yellow fever mosquito, Aedes aegypti, serves as the primary vector for epidemic transmission of yellow fever, dengue, Zika (ZIKV), and chikungunya viruses to humans. Control of Ae. aegypti is currently limited to insecticide applications and larval habitat management; however, to combat growing challenges with insecticide resistance, novel genetic approaches for vector population reduction or transmission interruption are being aggressively pursued. The objectives of this study were to assess the ability of the Ae. aegypti antiviral exogenous-small interfering RNA (exo-siRNA) response to inhibit ZIKV infection and transmission, and to identify the optimal RNA interference (RNAi) target region in the ZIKV genome. We accomplished these objectives by in vitro transcription of five long double-stranded RNAs (dsRNAs) from the genome region spanning the NS2B-NS3-NS4A genes, which were the most highly conserved among ZIKV RNA sequences representing both East and West African and Asian-American clades, and evaluation of the ability of these dsRNAs to trigger an effective antiviral exo-siRNA response after intrathoracic injection into Ae. aegypti. In a pilot study, five ZIKV dsRNAs were tested by intrathoracic inoculation of 250 ng dsRNA into groups of approximately 5-day-old mosquitoes. Three days post-inoculation, mosquitoes were provided an infectious blood-meal containing ZIKV strain PRVABC59 (Puerto Rico), MR766 (Uganda), or 41525 (Senegal). On days 7 and 14 post-infection individual whole mosquito bodies were assessed for ZIKV infectious titer by plaque assays. Based on the results of this initial assessment, three dsRNAs were selected for further evaluation of viral loads of matched body and saliva expectorants using a standardized infectious dose of 1 × 10
7 PFU/mL of each ZIKV strain. Fourteen days post-exposure to ZIKV, paired saliva and carcass samples were harvested from individual mosquitoes and assessed for ZIKV RNA load by qRT-PCR. Injection of each of the three dsRNAs resulted in significant inhibition of replication of all three strains of ZIKV in mosquito bodies and saliva. This study lays critical groundwork for pursuing ZIKV transmission-blocking strategies that exploit the Ae. aegypti exo-siRNA response for arbovirus suppression in natural populations., (Copyright © 2019 Elsevier Ltd. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
180. Efficacy and risk of harms of repeat ivermectin mass drug administrations for control of malaria (RIMDAMAL): a cluster-randomised trial.
- Author
-
Foy BD, Alout H, Seaman JA, Rao S, Magalhaes T, Wade M, Parikh S, Soma DD, Sagna AB, Fournet F, Slater HC, Bougma R, Drabo F, Diabaté A, Coulidiaty AGV, Rouamba N, and Dabiré RK
- Subjects
- Adolescent, Adult, Albendazole therapeutic use, Antiparasitic Agents adverse effects, Burkina Faso, Child, Cluster Analysis, Drug Administration Schedule, Female, Humans, Ivermectin adverse effects, Male, Treatment Outcome, Young Adult, Antiparasitic Agents administration & dosage, Ivermectin administration & dosage, Malaria, Falciparum drug therapy, Mass Drug Administration
- Abstract
Background: Ivermectin is widely used in mass drug administrations for controlling neglected parasitic diseases, and can be lethal to malaria vectors that bite treated humans. Therefore, it could be a new tool to reduce plasmodium transmission. We tested the hypothesis that frequently repeated mass administrations of ivermectin to village residents would reduce clinical malaria episodes in children and would be well tolerated with minimal harms., Methods: We invited villages (clusters) in Burkina Faso to participate in a single-blind (outcomes assessor), parallel-assignment, two-arm, cluster-randomised trial over the 2015 rainy season. Villages were assigned (1:1) by random draw to either the intervention group or the control group. In both groups, all eligible participants who consented to the treatment and were at least 90 cm in height received single oral doses of ivermectin (150-200 μg/kg) and albendazole (400 mg), and those in the intervention group received five further doses of ivermectin alone at 3-week intervals thereafter over the 18-week treatment phase. The primary outcome was cumulative incidence of uncomplicated malaria episodes over 18 weeks (analysed on a cluster intention-to-treat basis) in an active case detection cohort of children aged 5 years or younger living in the study villages. This trial is registered with ClinicalTrials.gov, number NCT02509481., Findings: Eight villages agreed to participate, and four were randomly assigned to each group. 2712 participants (1333 [49%] males and 1379 [51%] females; median age 15 years [IQR 6-34]), including 590 children aged 5 years or younger, provided consent and were enrolled between May 22 and July 20, 2015 (except for 77 participants enrolled after these dates because of unavailability before the first mass drug administration, travel into the village during the trial, or birth), with 1447 enrolled into the intervention group and 1265 into the control group. 330 (23%) participants in the intervention group and 233 (18%) in the control group met the exclusion criteria for mass drug administration. Most children in the active case detection cohort were not treated because of height restrictions. 14 (4%) children in the intervention group and 10 (4%) in the control group were lost to follow-up. Cumulative malaria incidence was reduced in the intervention group (648 episodes among 327 children; estimated mean 2·00 episodes per child) compared with the control group (647 episodes among 263 children; 2·49 episodes per child; risk difference -0·49 [95% CI -0·79 to -0·21], p=0·0009, adjusted for sex and clustering). The risk of adverse events among all participants did not differ between groups (45 events [3%] among 1447 participants in the intervention group vs 24 events [2%] among 1265 in the control group; risk ratio 1·63 [1·01 to 2·67]; risk difference 1·21 [0·04 to 2·38], p=0·060), and no adverse reactions were reported., Interpretation: Frequently repeated mass administrations of ivermectin during the malaria transmission season can reduce malaria episodes among children without significantly increasing harms in the populace., Funding: Bill & Melinda Gates Foundation., (Copyright © 2019 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
181. Evaluation of a novel West Nile virus transmission control strategy that targets Culex tarsalis with endectocide-containing blood meals.
- Author
-
Nguyen C, Gray M, Burton TA, Foy SL, Foster JR, Gendernalik AL, Rückert C, Alout H, Young MC, Boze B, Ebel GD, Clapsaddle B, and Foy BD
- Subjects
- Animals, Chickens, Colorado, Columbidae, Culex virology, Mosquito Vectors virology, Poultry Diseases drug therapy, Poultry Diseases mortality, Poultry Diseases transmission, West Nile Fever drug therapy, West Nile Fever mortality, West Nile Fever transmission, Communicable Disease Control methods, Culex drug effects, Insecticides pharmacology, Mosquito Vectors drug effects, Poultry Diseases prevention & control, West Nile Fever prevention & control, West Nile virus drug effects
- Abstract
Control of arbovirus transmission remains focused on vector control through application of insecticides directly to the environment. However, these insecticide applications are often reactive interventions that can be poorly-targeted, inadequate for localized control during outbreaks, and opposed due to environmental and toxicity concerns. In this study, we developed endectocide-treated feed as a systemic endectocide for birds to target blood feeding Culex tarsalis, the primary West Nile virus (WNV) bridge vector in the western United States, and conducted preliminary tests on the effects of deploying this feed in the field. In lab tests, ivermectin (IVM) was the most effective endectocide tested against Cx. tarsalis and WNV-infection did not influence mosquito mortality from IVM. Chickens and wild Eurasian collared doves exhibited no signs of toxicity when fed solely on bird feed treated with concentrations up to 200 mg IVM/kg of diet, and significantly more Cx. tarsalis that blood fed on these birds died (greater than 80% mortality) compared to controls (less than 25% mortality). Mosquito mortality following blood feeding correlated with IVM serum concentrations at the time of blood feeding, which dropped rapidly after the withdrawal of treated feed. Preliminary field testing over one WNV season in Fort Collins, Colorado demonstrated that nearly all birds captured around treated bird feeders had detectable levels of IVM in their blood. However, entomological data showed that WNV transmission was non-significantly reduced around treated bird feeders. With further development, deployment of ivermectin-treated bird feed might be an effective, localized WNV transmission control tool., Competing Interests: I have read the journal's policy and the authors of this manuscript have the following competing interests: BDF through the Colorado State University Research Foundation, declares a pending patent application on ideas presented within this manuscript. BC and TDA Research, Inc. have no competing interests. All other authors have no competing interests to declare.
- Published
- 2019
- Full Text
- View/download PDF
182. Mosquito-borne and sexual transmission of Zika virus: Recent developments and future directions.
- Author
-
Magalhaes T, Foy BD, Marques ETA, Ebel GD, and Weger-Lucarelli J
- Subjects
- Animals, Bodily Secretions virology, Disease Models, Animal, Humans, Sexually Transmitted Diseases, Viral virology, Zika Virus Infection virology, Aedes virology, Mosquito Vectors virology, Sexually Transmitted Diseases, Viral transmission, Zika Virus, Zika Virus Infection transmission
- Abstract
Zika virus (ZIKV; Genus Flavivirus, Family Flaviviridae) has recently emerged in Asia and the Americas to cause large outbreaks of human disease. The outbreak has been characterized by high attack rates, birth defects in infants and severe neurological complications in adults. ZIKV is transmitted to humans by Aedes mosquitoes, but recent evidence implicates sexual transmission as playing an important role as well. This review highlights the transmission of ZIKV in humans, with a focus on both mosquito and sexually-transmitted routes and their outcomes. We also discuss critical directions for future research., (Copyright © 2017 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
183. Endectocides for controlling transmission of mosquito-borne diseases.
- Author
-
Sylla M, Kobylinski KC, and Foy BD
- Abstract
Competing Interests: Competing interests: No competing interests declared.
- Published
- 2013
- Full Text
- View/download PDF
184. Ivermectin inhibits the sporogony of Plasmodium falciparum in Anopheles gambiae.
- Author
-
Kobylinski KC, Foy BD, and Richardson JH
- Subjects
- Animals, Survival Analysis, Anopheles drug effects, Anopheles parasitology, Antiparasitic Agents pharmacology, Insecticides pharmacology, Ivermectin pharmacology, Plasmodium falciparum drug effects, Plasmodium falciparum growth & development
- Abstract
Background: When ingested in a blood meal, ivermectin has been shown to reduce the survivorship of Anopheles gambiae in the laboratory and field. Furthermore, ivermectin mass drug administrations in Senegal have been shown to reduce the proportion of Plasmodium falciparum-sporozoite-containing An. gambiae. This study addresses whether ivermectin inhibits sporogony of P. falciparum in An. gambiae., Methods: Anophele gambiae s.s. G3 strain were fed two concentrations of ivermectin (LC25 and LC5) along with P. falciparum NF54 in human blood meals at staggered intervals. Mosquitoes ingested ivermectin concurrent with parasites (DPI 0), or at three (DPI 3), six (DPI 6), and nine (DPI 9) days post parasite ingestion, or three days prior (DPI -3) to parasite ingestion. Mosquitoes were dissected at seven, twelve or fourteen days post parasite ingestion and either oocyst or sporozoite prevalence was recorded. To determine if P. falciparum sporozoite-containing An. gambiae were more susceptible to ivermectin than uninfected controls, survivorship was recorded for mosquitoes which ingested P. falciparum or control blood meal on DPI 0 and then a second blood meal containing ivermectin (LC25) on DPI 14., Results: Ivermectin (LC25) co-ingested (DPI 0) with parasites reduced the proportion of An. gambiae that developed oocysts (χ2 = 15.4842, P = 0.0002) and sporozoites (χ2 = 19.9643, P < 0.0001). Ivermectin (LC25) ingested DPI 6 (χ2 = 8.5103, P = 0.0044) and 9 (χ2 = 14.7998, P < 0.0001) reduced the proportion of An. gambiae that developed sporozoites but not when ingested DPI 3 (χ2 = 0.0113, P = 1). Ivermectin (LC5) co-ingested (DPI 0) with parasites did not reduce the proportion of An. gambiae that developed oocysts (χ2 = 4.2518, P = 0.0577) or sporozoites (χ2 = 2.3636, P = 0.1540), however, when ingested DPI -3 the proportion of An. gambiae that developed sporozoites was reduced (χ2 = 8.4806, P = 0.0047). Plasmodium falciparum infection significantly reduced the survivorship of An. gambiae that ingested ivermectin (LC25) on DPI 14 compared to control mosquitoes that ingested a primary blood meal without parasites (χ2 = 4.97, P = 0.0257)., Conclusions: Ivermectin at sub-lethal concentrations inhibits the sporogony of P. falciparum in An. gambiae. These findings support the utility of ivermectin for P. falciparum transmission control.
- Published
- 2012
- Full Text
- View/download PDF
185. Mermithid nematodes found in adult Anopheles from southeastern Senegal.
- Author
-
Kobylinski KC, Sylla M, Black W 4th, and Foy BD
- Subjects
- Animals, DNA, Helminth chemistry, DNA, Helminth genetics, DNA, Ribosomal chemistry, DNA, Ribosomal genetics, Female, Humans, Larva parasitology, Mermithoidea classification, Mermithoidea genetics, Phylogeny, RNA, Ribosomal, 18S genetics, Senegal, Sequence Analysis, DNA, Anopheles parasitology, Mermithoidea isolation & purification
- Abstract
Background: Over two dozen mermithid nematodes have been described parasitizing mosquitoes worldwide, however, only two species were found in Africa. Mermithid nematodes kill their mosquito host upon emergence, which suggests that they could be developed as biological control agents of mosquitoes. Both Romanomermis culicivorax and Romanomermis iyengari have been reared for mass release to control numerous Anopheles species vector populations, and in one instance this may have led to reduced malaria prevalence in a human population., Methods: Anopheles mosquitoes were collected during a malaria study in southeastern Senegal. Two different adult blood fed mosquitoes had a single mermithid nematode emerge from their anus while they were being held post-capture. Primers from the 18 S rDNA were developed to sequence nematode DNA and screen mosquitoes for mermithid DNA. 18 S rDNA from the Senegalese mermithid and other mermithid entries in GenBank were used to create a Maximum Parsimony tree of the Mermithidae family., Results: The mermithid was present in 1.8% (10/551) of the sampled adult Anopheles species in our study area. The mermithid was found in An. gambiae s.s., An. funestus, and An. rufipes from the villages of Ndebou, Boundoucondi, and Damboucoye. Maximum parsimony analysis confirmed that the nematode parasites found in Anopheles were indeed mermithid parasites, and of the mermithid sequences available in GenBank, they are most closely related to Strelkovimermis spiculatus., Conclusions: To our knowledge, this is the first report of mermithids from adult Anopheles mosquitoes in Senegal. The mermithid appears to infect Anopheles mosquitoes that develop in diverse larval habitats. Although maximum parsimony analysis determined the mermithid was closely related to Strelkovimermis spiculatus, several characteristics of the mermithid were more similar to the Empidomermis genus. Future mermithid isolations will hopefully allow: formal taxonomic identification, laboratory colonization, determination of life history traits and species specificity, and characterize its usefulness as a biological control agent.
- Published
- 2012
- Full Text
- View/download PDF
186. Endectocides for malaria control.
- Author
-
Foy BD, Kobylinski KC, da Silva IM, Rasgon JL, and Sylla M
- Subjects
- Animals, Anthelmintics pharmacology, Disease Vectors, Drug Resistance, Female, Humans, Insecticides administration & dosage, Insecticides pharmacology, Ivermectin administration & dosage, Mosquito Control, Plasmodium growth & development, Plasmodium pathogenicity, Seasons, Anopheles drug effects, Disease Transmission, Infectious prevention & control, Ivermectin pharmacology, Malaria prevention & control
- Abstract
Systemic endectocidal drugs, used to control nematodes in humans and other vertebrates, can be toxic to Anopheles spp. mosquitoes when they take a blood meal from a host that has recently received one of these drugs. Recent laboratory and field studies have highlighted the potential of ivermectin to control malaria parasite transmission if this drug is distributed strategically and more often. There are important theoretical benefits to this strategy, as well as caveats. A better understanding of drug effects against vectors and malaria ecologies are needed. In the near future, ivermectin and other endectocides could serve as potent and novel malaria transmission control tools that are directly linked to the control of neglected tropical diseases in the same communities., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
187. Probable non-vector-borne transmission of Zika virus, Colorado, USA.
- Author
-
Foy BD, Kobylinski KC, Chilson Foy JL, Blitvich BJ, Travassos da Rosa A, Haddow AD, Lanciotti RS, and Tesh RB
- Subjects
- Adult, Animals, Chlorocebus aethiops, Colorado, Exanthema etiology, Female, Humans, Male, Mice, RNA, Viral genetics, Serologic Tests, Vero Cells, Zika Virus Infection complications, Zika Virus Infection diagnosis, Zika Virus genetics, Zika Virus pathogenicity, Zika Virus Infection transmission
- Abstract
Clinical and serologic evidence indicate that 2 American scientists contracted Zika virus infections while working in Senegal in 2008. One of the scientists transmitted this arbovirus to his wife after his return home. Direct contact is implicated as the transmission route, most likely as a sexually transmitted infection.
- Published
- 2011
- Full Text
- View/download PDF
188. Alphavirus transducing systems.
- Author
-
Foy BD and Olson KE
- Subjects
- Animals, DNA, Complementary, Gene Expression, Genetic Vectors adverse effects, Insecta genetics, RNA Interference, Recombination, Genetic, Alphavirus genetics, Transduction, Genetic
- Abstract
Alphavirus transducing systems (ATSs) are important tools for expressing genes of interest (GOI) in mosquitoes and nonvector insects. ATSs are derived from infectious cDNA clones of mosquito-borne RNA viruses (family Togaviridae). The most common ATSs in use are derived from Sindbis viruses; however, ATSs have been derived from other alphaviruses as well. ATSs generate viruses with genomes that contain GOI's that can be expressed from additional viral subgenomic promoters. ATSs in which an exogenous gene sequence is positioned 5' to the viral structural genes is used for stable protein expression in insects. ATSs in which a gene sequence is positioned 3' to the structural genes is used to trigger RNAi and silence expression of that gene in the insect. ATSs are proving to be invaluable tools for understanding vector-pathogen interactions, vector competence, and other components of vector-pathogen amplification and maintenance cycles in nature. These virus-based expression systems also facilitate the researcher's ability to decide which gene-based disease control strategies merit a further investment in time and resources in transgenic mosquitoes.
- Published
- 2008
- Full Text
- View/download PDF
189. Molecular evidence for trypanosomatids in Culex mosquitoes collected during a West Nile virus survey.
- Author
-
Van Dyken M, Bolling BG, Moore CG, Blair CD, Beaty BJ, Black WC 4th, and Foy BD
- Subjects
- Animals, Culex virology, Female, Insect Vectors virology, Phylogeny, Polymerase Chain Reaction methods, Trypanosoma classification, Trypanosomiasis transmission, West Nile Fever transmission, Culex parasitology, Insect Vectors parasitology, Trypanosoma isolation & purification, West Nile virus isolation & purification
- Abstract
Adult mosquitoes were previously collected and tested for West Nile virus during an intense WNV outbreak in 2003-2004 along the Cache la Poudre River in Colorado, USA. A subset of these mosquitoes was also tested for infection with trypanosomatids using nested PCR to amplify 18S rRNA. Of the 69 pools of Culex pipiens that were screened for both pathogens, 4.3% were positive for WNV and 11.6% tested positive for trypanosomes; no pools were found to be co-infected with both pathogens. One hundred and forty-three pools of Culex tarsalis, considered to be the principal WNV vector in this area, were tested in the same manner. 7.7% were positive for WNV and 20.3% of these pools tested positive for trypanosomes. Five pools of C. tarsalis were found to be co-infected with both pathogens, which was approximately 2.2 times more frequent than would be expected if these pathogens are independent of each other. Sequencing and maximum parsimony analysis of 18S rRNA revealed that four of the isolates arise in or near clades of described avian trypanosomes, likely indicating that these are vectored pathogens between birds and mosquitoes. Unexpectedly, the majority (24/28, 86%) of our positive samples form their own separate clade within the order Trypanosomatida with 100% bootstrap support. We have identified a potential new clade of trypanosomatids that exist within important mosquito vectors and discuss the potential ecological connections between these trypanosomes, arboviruses and mosquitoes.
- Published
- 2006
- Full Text
- View/download PDF
190. RNA interference acts as a natural antiviral response to O'nyong-nyong virus (Alphavirus; Togaviridae) infection of Anopheles gambiae.
- Author
-
Keene KM, Foy BD, Sanchez-Vargas I, Beaty BJ, Blair CD, and Olson KE
- Subjects
- Alphavirus immunology, Animals, Anopheles immunology, Injections, Insect Vectors, RNA, Double-Stranded immunology, RNA, Double-Stranded pharmacology, Virus Replication drug effects, Alphavirus Infections immunology, Anopheles virology, Immunity, Innate, RNA Interference immunology
- Abstract
RNA interference (RNAi) is triggered in eukaryotic organisms by double-stranded RNA (dsRNA), and it destroys any mRNA that has sequence identity with the dsRNA trigger. The RNAi pathway in Anopheles gambiae can be silenced by transfecting cells with dsRNA derived from exon sequence of the A. gambiae Argonaute2 (AgAgo2) gene. We hypothesized that RNAi may also act as an antagonist to alphavirus replication in A. gambiae because RNA viruses form dsRNA during replication. Silencing AgAgo2 expression would make A. gambiae mosquitoes more permissive to virus infection. To determine whether RNAi conditions the vector competence of A. gambiae for O'nyong-nyong virus (ONNV), we engineered a genetically modified ONNV that expresses enhanced GFP (eGFP) as a marker. After intrathoracic injection, ONNV-eGFP slowly spread to other A. gambiae tissues over a 9-day incubation period. Mosquitoes were then coinjected with virus and either control beta-galactosidase dsRNA (dsbetagal; note that "ds" is used as a prefix to indicate the dsRNA derived from a given gene throughout) or ONNV dsnsP3. Treatment with dsnsP3 inhibited virus spread significantly, as determined by eGFP expression patterns. ONNV-eGFP titers from mosquitoes coinjected with dsnsP3 were significantly lower at 3 and 6 days after injection than in mosquitoes coinjected with dsbetagal. Mosquitoes were then coinjected with ONNV-eGFP and dsAgAgo2. Mosquitoes coinjected with virus and AgAgo2 dsRNA displayed widespread eGFP expression and virus titers 16-fold higher than dsbetagal controls after 3 or 6 days after injection. These observations provide direct evidence that RNAi is an antagonist of ONNV replication in A. gambiae, and they suggest that the innate immune response conditions vector competence.
- Published
- 2004
- Full Text
- View/download PDF
191. Clitolactone: a banana slug antifeedant from Clitocybe flaccida.
- Author
-
Wood WF, Clark TJ, Bradshaw DE, Foy BD, Largent DL, and Thompson BL
- Abstract
Clitolactone, 5-(chloromethyl)-3-methyl-2(5H)-furanone, was isolated from sporocaps of the mushroom Clitocybe flaccida. The structure was determined by HRMS, EIMS, (1)H & (13)C NMR, 2D (1)H-(13)C COSY and (1)H-(1)H COSY. This mushroom is not eaten by the banana slug Ariolimax columbianus (Gould), a mycophagist from the temperate rain forests of the Pacific Northwest. Clitolactone acts as an antifeedant because these slugs rejected 1.0 cm(2) pieces of lettuce treated with 25 μg of clitolactone.
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.