Cargando…

Age and prior blood feeding of Anopheles gambiae influences their susceptibility and gene expression patterns to ivermectin-containing blood meals

BACKGROUND: Ivermectin has been proposed as a novel malaria transmission control tool based on its insecticidal properties and unique route of acquisition through human blood. To maximize ivermectin’s effect and identify potential resistance/tolerance mechanisms, it is important to understand its ef...

Descripción completa

Detalles Bibliográficos
Autores principales: Seaman, Jonathan A., Alout, Haoues, Meyers, Jacob I., Stenglein, Mark D., Dabiré, Roch K., Lozano-Fuentes, Saul, Burton, Timothy A., Kuklinski, Wojtek S., Black, William C., Foy, Brian D.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4608139/
https://www.ncbi.nlm.nih.gov/pubmed/26471037
http://dx.doi.org/10.1186/s12864-015-2029-8
_version_ 1782395617151025152
author Seaman, Jonathan A.
Alout, Haoues
Meyers, Jacob I.
Stenglein, Mark D.
Dabiré, Roch K.
Lozano-Fuentes, Saul
Burton, Timothy A.
Kuklinski, Wojtek S.
Black, William C.
Foy, Brian D.
author_facet Seaman, Jonathan A.
Alout, Haoues
Meyers, Jacob I.
Stenglein, Mark D.
Dabiré, Roch K.
Lozano-Fuentes, Saul
Burton, Timothy A.
Kuklinski, Wojtek S.
Black, William C.
Foy, Brian D.
author_sort Seaman, Jonathan A.
collection PubMed
description BACKGROUND: Ivermectin has been proposed as a novel malaria transmission control tool based on its insecticidal properties and unique route of acquisition through human blood. To maximize ivermectin’s effect and identify potential resistance/tolerance mechanisms, it is important to understand its effect on mosquito physiology and potential to shift mosquito population age-structure. We therefore investigated ivermectin susceptibility and gene expression changes in several age groups of female Anopheles gambiae mosquitoes. METHODS: The effect of aging on ivermectin susceptibility was analyzed in three age groups (2, 6, and 14-days) of colonized female Anopheles gambiae mosquitoes using standard survivorship assays. Gene expression patterns were then analyzed by transcriptome sequencing on an Illumina HiSeq 2500 platform. RT-qPCR was used to validate transcriptional changes and also to examine expression in a different, colonized strain and in wild mosquitoes, both of which blood fed naturally on an ivermectin-treated person. RESULTS: Mosquitoes of different ages and blood meal history died at different frequencies after ingesting ivermectin. Mortality was lowest in 2-day old mosquitoes exposed on their first blood meal and highest in 6-day old mosquitoes exposed on their second blood meal. Twenty-four hours following ivermectin ingestion, 101 and 187 genes were differentially-expressed relative to control blood-fed, in 2 and 6-day groups, respectively. Transcription patterns of select genes were similar in membrane-fed, colonized, and naturally-fed wild vectors. Transcripts from several unexpected functional classes were highly up-regulated, including Niemann-Pick Type C (NPC) genes, peritrophic matrix-associated genes, and immune-response genes, and these exhibited different transcription patterns between age groups, which may explain the observed susceptibility differences. Niemann-Pick Type 2 genes were the most highly up-regulated transcripts after ivermectin ingestion (up to 160 fold) and comparing phylogeny to transcriptional patterns revealed that NPCs have rapidly evolved and separate members respond to either blood meals or to ivermectin. CONCLUSION: We present evidence of increased ivermectin susceptibility in older An. gambiae mosquitoes that had previously bloodfed. Differential expression analysis suggests complex midgut interactions resulting from ivermectin ingestion that likely involve blood meal digestion physiological responses, midgut microflora, and innate immune responses. Thus, the transcription of certain gene families is consistently affected by ivermectin ingestion, and may provide important clues to ivermectin’s broad effects on malaria vectors. These findings contribute to the growing understanding of ivermectin’s potential as a transmission control tool. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s12864-015-2029-8) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-4608139
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-46081392015-10-17 Age and prior blood feeding of Anopheles gambiae influences their susceptibility and gene expression patterns to ivermectin-containing blood meals Seaman, Jonathan A. Alout, Haoues Meyers, Jacob I. Stenglein, Mark D. Dabiré, Roch K. Lozano-Fuentes, Saul Burton, Timothy A. Kuklinski, Wojtek S. Black, William C. Foy, Brian D. BMC Genomics Research Article BACKGROUND: Ivermectin has been proposed as a novel malaria transmission control tool based on its insecticidal properties and unique route of acquisition through human blood. To maximize ivermectin’s effect and identify potential resistance/tolerance mechanisms, it is important to understand its effect on mosquito physiology and potential to shift mosquito population age-structure. We therefore investigated ivermectin susceptibility and gene expression changes in several age groups of female Anopheles gambiae mosquitoes. METHODS: The effect of aging on ivermectin susceptibility was analyzed in three age groups (2, 6, and 14-days) of colonized female Anopheles gambiae mosquitoes using standard survivorship assays. Gene expression patterns were then analyzed by transcriptome sequencing on an Illumina HiSeq 2500 platform. RT-qPCR was used to validate transcriptional changes and also to examine expression in a different, colonized strain and in wild mosquitoes, both of which blood fed naturally on an ivermectin-treated person. RESULTS: Mosquitoes of different ages and blood meal history died at different frequencies after ingesting ivermectin. Mortality was lowest in 2-day old mosquitoes exposed on their first blood meal and highest in 6-day old mosquitoes exposed on their second blood meal. Twenty-four hours following ivermectin ingestion, 101 and 187 genes were differentially-expressed relative to control blood-fed, in 2 and 6-day groups, respectively. Transcription patterns of select genes were similar in membrane-fed, colonized, and naturally-fed wild vectors. Transcripts from several unexpected functional classes were highly up-regulated, including Niemann-Pick Type C (NPC) genes, peritrophic matrix-associated genes, and immune-response genes, and these exhibited different transcription patterns between age groups, which may explain the observed susceptibility differences. Niemann-Pick Type 2 genes were the most highly up-regulated transcripts after ivermectin ingestion (up to 160 fold) and comparing phylogeny to transcriptional patterns revealed that NPCs have rapidly evolved and separate members respond to either blood meals or to ivermectin. CONCLUSION: We present evidence of increased ivermectin susceptibility in older An. gambiae mosquitoes that had previously bloodfed. Differential expression analysis suggests complex midgut interactions resulting from ivermectin ingestion that likely involve blood meal digestion physiological responses, midgut microflora, and innate immune responses. Thus, the transcription of certain gene families is consistently affected by ivermectin ingestion, and may provide important clues to ivermectin’s broad effects on malaria vectors. These findings contribute to the growing understanding of ivermectin’s potential as a transmission control tool. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s12864-015-2029-8) contains supplementary material, which is available to authorized users. BioMed Central 2015-10-15 /pmc/articles/PMC4608139/ /pubmed/26471037 http://dx.doi.org/10.1186/s12864-015-2029-8 Text en © Seaman et al. 2015 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research Article
Seaman, Jonathan A.
Alout, Haoues
Meyers, Jacob I.
Stenglein, Mark D.
Dabiré, Roch K.
Lozano-Fuentes, Saul
Burton, Timothy A.
Kuklinski, Wojtek S.
Black, William C.
Foy, Brian D.
Age and prior blood feeding of Anopheles gambiae influences their susceptibility and gene expression patterns to ivermectin-containing blood meals
title Age and prior blood feeding of Anopheles gambiae influences their susceptibility and gene expression patterns to ivermectin-containing blood meals
title_full Age and prior blood feeding of Anopheles gambiae influences their susceptibility and gene expression patterns to ivermectin-containing blood meals
title_fullStr Age and prior blood feeding of Anopheles gambiae influences their susceptibility and gene expression patterns to ivermectin-containing blood meals
title_full_unstemmed Age and prior blood feeding of Anopheles gambiae influences their susceptibility and gene expression patterns to ivermectin-containing blood meals
title_short Age and prior blood feeding of Anopheles gambiae influences their susceptibility and gene expression patterns to ivermectin-containing blood meals
title_sort age and prior blood feeding of anopheles gambiae influences their susceptibility and gene expression patterns to ivermectin-containing blood meals
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4608139/
https://www.ncbi.nlm.nih.gov/pubmed/26471037
http://dx.doi.org/10.1186/s12864-015-2029-8
work_keys_str_mv AT seamanjonathana ageandpriorbloodfeedingofanophelesgambiaeinfluencestheirsusceptibilityandgeneexpressionpatternstoivermectincontainingbloodmeals
AT alouthaoues ageandpriorbloodfeedingofanophelesgambiaeinfluencestheirsusceptibilityandgeneexpressionpatternstoivermectincontainingbloodmeals
AT meyersjacobi ageandpriorbloodfeedingofanophelesgambiaeinfluencestheirsusceptibilityandgeneexpressionpatternstoivermectincontainingbloodmeals
AT stengleinmarkd ageandpriorbloodfeedingofanophelesgambiaeinfluencestheirsusceptibilityandgeneexpressionpatternstoivermectincontainingbloodmeals
AT dabirerochk ageandpriorbloodfeedingofanophelesgambiaeinfluencestheirsusceptibilityandgeneexpressionpatternstoivermectincontainingbloodmeals
AT lozanofuentessaul ageandpriorbloodfeedingofanophelesgambiaeinfluencestheirsusceptibilityandgeneexpressionpatternstoivermectincontainingbloodmeals
AT burtontimothya ageandpriorbloodfeedingofanophelesgambiaeinfluencestheirsusceptibilityandgeneexpressionpatternstoivermectincontainingbloodmeals
AT kuklinskiwojteks ageandpriorbloodfeedingofanophelesgambiaeinfluencestheirsusceptibilityandgeneexpressionpatternstoivermectincontainingbloodmeals
AT blackwilliamc ageandpriorbloodfeedingofanophelesgambiaeinfluencestheirsusceptibilityandgeneexpressionpatternstoivermectincontainingbloodmeals
AT foybriand ageandpriorbloodfeedingofanophelesgambiaeinfluencestheirsusceptibilityandgeneexpressionpatternstoivermectincontainingbloodmeals