1
|
Dong Y, Kang S, Sandiford SL, Pike A, Simões ML, Ubalee R, Kobylinski K, Dimopoulos G. Targeting the mosquito prefoldin-chaperonin complex blocks Plasmodium transmission. Nat Microbiol 2025; 10:841-854. [PMID: 40050397 DOI: 10.1038/s41564-025-01947-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/27/2025] [Indexed: 03/16/2025]
Abstract
The Plasmodium infection cycle in mosquitoes relies on numerous host factors in the vector midgut, which can be targeted with therapeutics. The mosquito prefoldin complex is needed to fold proteins and macromolecular complexes properly. Here we show that the conserved Anopheles mosquito prefoldin (PFDN)-chaperonin system is a potent transmission-blocking target for multiple Plasmodium species. Silencing any prefoldin subunit or its CCT/TRiC partner via RNA interference reduces Plasmodium falciparum oocyst loads in the mosquito midgut, as does co-feeding mosquitoes with PFDN6-specific antibody and gametocytes. Inhibition of the PFDN-CCT/TRiC chaperonin complex results in the loss of epithelial and extracellular matrix integrity, which triggers microorganism-mediated anti-Plasmodium immune priming and compromises the parasite's laminin-based immune evasion. Mouse malaria transmission-blocking vaccine and antibody co-feeding assays support its potential as a multispecies transmission-blocking target for P. falciparum and Plasmodium vivax. Further study is needed to determine the potential of this system as a transmission-blocking vaccine target.
Collapse
Affiliation(s)
- Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Seokyoung Kang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Simone L Sandiford
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew Pike
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Ratawan Ubalee
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Kevin Kobylinski
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
2
|
Yadav M, Dahiya N, Janjoter S, Kataria D, Dixit R, Sehrawat N. A review on RNA interference studies in Anophelines to reveal candidate genes for malaria transmission blocking vaccine. Life Sci 2024; 351:122822. [PMID: 38866221 DOI: 10.1016/j.lfs.2024.122822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024]
Abstract
Malaria is a major public health concern. The development of parasite-based vaccine RTS/AS01 has some therapeutic value but its lower efficacy is one of the major limitations. Mosquito-based transmission-blocking vaccines could have a higher potential for parasite inhibition within the mosquitoes. Several genes of mosquito midgut, salivary gland, hemolymph, etc. get activate in response to the Plasmodium-infected blood and helps in parasite invasion directly or indirectly inside the mosquito. The studies of such genes provided a new insight into developing the more efficient vaccines. In the field of malaria genetics research, RNAi has become an innovative strategy used to identify mosquito candidate genes for transmission-blocking vaccines. This review targeted the gene studies that have been conducted in the period 2000-2023 in different malaria vectors against different malarial parasites using the RNAi approach to reveal mosquito novel gene candidates for vaccine development.
Collapse
Affiliation(s)
- Mahima Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Nisha Dahiya
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Sangeeta Janjoter
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Divya Kataria
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, India
| | | | - Neelam Sehrawat
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, India.
| |
Collapse
|
3
|
Zeineddine S, Jaber S, Saab SA, Nakhleh J, Dimopoulos G, Osta MA. Late sporogonic stages of Plasmodium parasites are susceptible to the melanization response in Anopheles gambiae mosquitoes. Front Cell Infect Microbiol 2024; 14:1438019. [PMID: 39149419 PMCID: PMC11324593 DOI: 10.3389/fcimb.2024.1438019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024] Open
Abstract
The malaria-causing parasites have to complete a complex infection cycle in the mosquito vector that also involves attack by the insect's innate immune system, especially at the early stages of midgut infection. However, Anopheles immunity to the late Plasmodium sporogonic stages, such as oocysts, has received little attention as they are considered to be concealed from immune factors due to their location under the midgut basal lamina and for harboring an elaborate cell wall comprising an external layer derived from the basal lamina that confers self-properties to an otherwise foreign structure. Here, we investigated whether Plasmodium berghei oocysts and sporozoites are susceptible to melanization-based immunity in Anopheles gambiae. Silencing of the negative regulator of melanization response, CLIPA14, increased melanization prevalence without significantly increasing the numbers of melanized oocysts, while co-silencing CLIPA14 with CLIPA2, a second negative regulator of melanization, resulted in a significant increase in melanized oocysts and melanization prevalence. Only late-stage oocysts were found to be melanized, suggesting that oocyst rupture was a prerequisite for melanization-based immune attack, presumably due to the loss of the immune-evasive features of their wall. We also found melanized sporozoites inside oocysts and in the hemocoel, suggesting that sporozoites at different maturation stages are susceptible to melanization. Silencing the melanization promoting factors TEP1 and CLIPA28 rescued oocyst melanization in CLIPA2/CLIPA14 co-silenced mosquitoes. Interestingly, silencing of CTL4, that protects early stage ookinetes from melanization, had no effect on oocysts and sporozoites, indicating differential regulation of immunity to early and late sporogonic stages. Similar to previous studies addressing ookinete stage melanization, the melanization of Plasmodium falciparum oocysts was significantly lower than that observed for P. berghei. In summary, our results provide conclusive evidence that late sporogonic malaria parasite stages are susceptible to melanization, and we reveal distinct regulatory mechanisms for ookinete and oocyst melanization.
Collapse
Affiliation(s)
- Suheir Zeineddine
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Sana Jaber
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Sally A. Saab
- Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Johnny Nakhleh
- Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - George Dimopoulos
- Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Mike A. Osta
- Department of Biology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
4
|
Chen TY, Raduwan H, Marín-López A, Cui Y, Fikrig E. Zika virus exists in enterocytes and enteroendocrine cells of the Aedes aegypti midgut. iScience 2024; 27:110353. [PMID: 39055935 PMCID: PMC11269924 DOI: 10.1016/j.isci.2024.110353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/21/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The Aedes aegypti midgut is crucial for blood digestion, nutrition, reproduction, and pathogen interaction. Using single-cell RNA sequencing, we explored virus infection and transcriptomic changes at the cellular level. We identified 12 distinct cell clusters in the Ae. aegypti midgut post-Zika virus infection, including intestinal stem cells, enteroblasts, enteroendocrine cells (EE), and enterocytes (ECs). The virus was found mainly in specific subsets of ECs and EE. Infection altered transcriptional profiles related to metabolism, signaling, and immune responses. Functional studies highlighted three significantly differentially expressed genes in infected cells. Notably, silencing apolipophorin III reduced virus RNA copy number in the midgut, emphasizing the role of specific genes in viral infection. These findings enhance our understanding of mosquito midgut cell processes during Zika virus infection and suggest potential targets for vector control.
Collapse
Affiliation(s)
- Tse-Yu Chen
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Hamidah Raduwan
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Alejandro Marín-López
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Yingjun Cui
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
5
|
Zeineddine S, Jaber S, Saab SA, Nakhleh J, Dimopoulos G, Osta MA. Late sporogonic stages of Plasmodium parasites are susceptible to the melanization response in Anopheles gambiae mosquitoes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596773. [PMID: 38853990 PMCID: PMC11160730 DOI: 10.1101/2024.05.31.596773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The malaria-causing parasites have to complete a complex infection cycle in the mosquito vector that also involves attack by the insect's innate immune system, especially at the early stages of midgut infection. However, Anopheles immunity to the late Plasmodium sporogonic stages, such as oocysts, has received little attention as they are considered to be concealed from immune factors due to their location under the midgut basal lamina and for harboring an elaborate cell wall comprising an external layer derived from the basal lamina that confers self-properties to an otherwise foreign structure. Here, we investigated whether Plasmodium berghei oocysts and sporozoites are susceptible to melanization-based immunity in Anopheles gambiae. Silencing of the negative regulator of melanization response, CLIPA14, increased melanization prevalence without significantly increasing the numbers of melanized oocysts, while co-silencing CLIPA14 with CLIPA2, a second negative regulator of melanization, resulted in a significant increase in melanized oocysts and melanization prevalence. Only late-stage oocysts were found to be melanized, suggesting that oocyst rupture was a prerequisite for melanization-based immune attack, presumably due to the loss of the immune-evasive features of their wall. We also found melanized sporozoites inside oocysts and in the hemocoel, suggesting that sporozoites at different maturation stages are susceptible to melanization. Silencing the melanization promoting factors TEP1 and CLIPA28 rescued oocyst melanization in CLIPA2/CLIPA14 co-silenced mosquitoes. Interestingly, silencing of CTL4, that protects early stage ookinetes from melanization, had no effect on oocysts and sporozoites, indicating differential regulation of immunity to early and late sporogonic stages. Similar to previous studies addressing ookinete stage melanization, the melanization of Plasmodium falciparum oocysts was significantly lower than that observed for P. berghei. In summary, our results provide conclusive evidence that late sporogonic malaria parasite stages are susceptible to melanization, and we reveal distinct regulatory mechanisms for ookinete and oocyst melanization.
Collapse
Affiliation(s)
- Suheir Zeineddine
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Sana Jaber
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Sally A. Saab
- Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Johnny Nakhleh
- Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - George Dimopoulos
- Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mike A. Osta
- Department of Biology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
6
|
Saab SA, Zhang X, Zeineddine S, Morejon B, Michel K, Osta MA. Insight into the structural hierarchy of the protease cascade that regulates the mosquito melanization response. Microbes Infect 2024; 26:105245. [PMID: 37918462 PMCID: PMC10872705 DOI: 10.1016/j.micinf.2023.105245] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
Serine protease cascades regulate important insect immune responses, including melanization and Toll pathway activation. In the context of melanization, central components of these cascades are clip domain serine proteases (CLIPs) including the catalytic, clip domain serine proteases (cSPs) and their non-catalytic homologs (cSPHs). Here, we define partially the structural hierarchy of An. gambiae cSPs of the CLIPB family, central players in melanization, and characterize their relative contributions to bacterial melanization and to mosquito susceptibility to bacterial infections. Using in vivo genetic analysis we show that the protease cascade branches downstream of the cSPs CLIPB4 and CLIPB17 into two branches one converging on CLIPB10 and the second on CLIPB8. We also show that the contribution of key cSPHs to melanization in vivo in response to diverse microbial challenges is more significant than any of the individual cSPs, possibly due to partial functional redundancy among the latter. Interestingly, we show that the key cSPH CLIPA8 which is essential for the efficient activation cleavage of CLIPBs in vivo is efficiently cleaved itself by several CLIPBs in vitro, suggesting that cSPs and cSPHs regulate signal amplification and propagation in melanization cascades by providing positive reinforcement upstream and downstream of each other.
Collapse
Affiliation(s)
- Sally A Saab
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Xiufeng Zhang
- Division of Biology, Kansas State University, Manhattan, KS, USA
| | - Suheir Zeineddine
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Bianca Morejon
- Division of Biology, Kansas State University, Manhattan, KS, USA
| | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS, USA
| | - Mike A Osta
- Department of Biology, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
7
|
Saab SA, Zhang X, Zeineddine S, Morejon B, Michel K, Osta MA. Insight into the structural hierarchy of the protease cascade that regulates the mosquito melanization response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.13.548954. [PMID: 37503117 PMCID: PMC10369957 DOI: 10.1101/2023.07.13.548954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Serine protease cascades regulate important insect immune responses, including melanization and Toll pathway activation. In the context of melanization, central components of these cascades are clip domain serine proteases (CLIPs) including the catalytic, clip domain serine proteases (cSPs) and their non-catalytic homologs (cSPHs). Here, we define partially the structural hierarchy of An. gambiae cSPs of the CLIPB family, central players in melanization, and characterize their relative contributions to bacterial melanization and to mosquito susceptibility to bacterial infections. Using in vivo genetic analysis we show that the protease cascade branches downstream of the cSPs CLIPB4 and CLIPB17 into two branches one converging on CLIPB10 and the second on CLIPB8. We also show that the contribution of key cSPHs to melanization in vivo in response to diverse microbial challenges is more significant than any of the individual cSPs, possibly due to partial functional redundancy among the latter. Interestingly, we show that the key cSPH CLIPA8 which is essential for the efficient activation cleavage of CLIPBs in vivo is efficiently cleaved itself by several CLIPBs in vitro, suggesting that cSPs and cSPHs regulate signal amplification and propagation in melanization cascades by providing positive reinforcement upstream and downstream of each other.
Collapse
Affiliation(s)
- Sally A. Saab
- Department of Biology, American University of Beirut, Beirut, Lebanon
- Present address: Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, USA
| | - Xiufeng Zhang
- Division of Biology, Kansas State University, Manhattan, KS, USA
| | - Suheir Zeineddine
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Bianca Morejon
- Division of Biology, Kansas State University, Manhattan, KS, USA
| | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS, USA
| | - Mike A. Osta
- Department of Biology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
8
|
Pujhari S, Hughes GL, Pakpour N, Suzuki Y, Rasgon JL. Wolbachia-induced inhibition of O'nyong nyong virus in Anopheles mosquitoes is mediated by Toll signaling and modulated by cholesterol. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.31.543096. [PMID: 37397989 PMCID: PMC10312510 DOI: 10.1101/2023.05.31.543096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Enhanced host immunity and competition for metabolic resources are two main competing hypotheses for the mechanism of Wolbachia-mediated pathogen inhibition in arthropods. Using an Anopheles mosquito - somatic Wolbachia infection - O'nyong nyong virus (ONNV) model, we demonstrate that the mechanism underpinning Wolbachia-mediated virus inhibition is up-regulation of the Toll innate immune pathway. However, the viral inhibitory properties of Wolbachia were abolished by cholesterol supplementation. This result was due to Wolbachia-dependent cholesterol-mediated suppression of Toll signaling rather than competition for cholesterol between Wolbachia and virus. The inhibitory effect of cholesterol was specific to Wolbachia-infected Anopheles mosquitoes and cells. These data indicate that both Wolbachia and cholesterol influence Toll immune signaling in Anopheles mosquitoes in a complex manner and provide a functional link between the host immunity and metabolic competition hypotheses for explaining Wolbachia-mediated pathogen interference in mosquitoes. In addition, these results provide a mechanistic understanding of the mode of action of Wolbachia-induced pathogen blocking in Anophelines, which is critical to evaluate the long-term efficacy of control strategies for malaria and Anopheles-transmitted arboviruses.
Collapse
Affiliation(s)
- Sujit Pujhari
- The Department of Entomology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Grant L Hughes
- The Department of Entomology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
- Departments of Vector Biology and Tropical Disease Biology, Centre for Neglected Tropical Disease, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Yasutsugu Suzuki
- The Department of Entomology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
- Center for Marine Environmental Studies, Ehime University, Matsuyama, Japan
| | - Jason L Rasgon
- The Department of Entomology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
9
|
Ngou CM, Bayibéki AN, Abate L, Makinde OS, Feufack-Donfack LB, Sarah-Matio EM, Bouopda-Tuedom AG, Taconet P, Moiroux N, Awono-Ambéné PH, Talman A, Ayong LS, Berry A, Nsango SE, Morlais I. Influence of the sickle cell trait on Plasmodium falciparum infectivity from naturally infected gametocyte carriers. BMC Infect Dis 2023; 23:317. [PMID: 37165325 PMCID: PMC10173526 DOI: 10.1186/s12879-023-08134-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 03/03/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Sickle cell trait (SCT) refers to the carriage of one abnormal copy of the β-globin gene, the HbS allele. SCT offers protection against malaria, controlling parasite density and preventing progression to symptomatic malaria. However, it remains unclear whether SCT also affects transmission stages and mosquito infection parameters. Deciphering the impact of the SCT on human to mosquito malaria transmission is key to understanding mechanisms that maintain the trait in malaria endemic areas. METHODS The study was conducted from June to July 2017 among asymptomatic children living in the locality of Mfou, Cameroon. Blood samples were collected from asymptomatic children to perform malaria diagnosis by microscopy, Plasmodium species by PCR and hemoglobin typing by RFLP. Infectiousness of gametocytes to mosquitoes was assessed by membrane feeding assays using blood from gametocyte carriers of HbAA and HbAS genotypes. A zero-inflated model was fitted to predict distribution of oocysts in mosquitoes according to hemoglobin genotype of the gametocyte source. RESULTS Among the 1557 children enrolled in the study, 314 (20.16%) were of the HbAS genotype. The prevalence of children with P. falciparum gametocytes was 18.47% in HbAS individuals and 13.57% in HbAA, and the difference is significant (χ2 = 4.61, P = 0.032). Multiplicity of infection was lower in HbAS gametocyte carriers (median = 2 genotypes/carrier in HbAS versus 3.5 genotypes/carrier in HbAA, Wilcoxon sum rank test = 188, P = 0.032). Gametocyte densities in the blood donor significantly influenced mosquito infection prevalence in both HbAS and HbAA individuals. The HbAS genotype had no significant effect on mosquito infection outcomes when using immune or naïve serum in feeding assays. In AB replacement feeding experiments, the odds ratio of mosquito infection for HbAA blood as compared to HbAS was 0.56 (95% CI 0.29-1.10), indicating a twice higher risk of infection in mosquitoes fed on gametocyte-containing blood of HbAS genotype. CONCLUSION Plasmodium transmission stages were more prevalent in SCT individuals. This may reflect the parasite's enhanced investment in the sexual stage to increase their survival rate when asexual replication is impeded. The public health impact of our results points the need for intensive malaria control interventions in areas with high prevalence of HbAS. The similar infection parameters in feeding experiments where mosquitoes received the original serum from the blood donor indicated that immune responses to gametocyte surface proteins occur in both HbAS and HbAA individuals. The higher risk of infection in mosquitoes fed on HbAS blood depleted of immune factors suggests that changes in the membrane properties in HbAS erythrocytes may impact on the maturation process of gametocytes within circulating red blood cells.
Collapse
Affiliation(s)
- Christelle M Ngou
- Institut de Recherche pour le Développement, MIVEGEC, Univ. Montpellier, CNRS, IRD, 91 Avenue Agropolis, BP 64501, 34394, Montpellier, France
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroon
| | | | - Luc Abate
- Institut de Recherche pour le Développement, MIVEGEC, Univ. Montpellier, CNRS, IRD, 91 Avenue Agropolis, BP 64501, 34394, Montpellier, France
| | - Olesula S Makinde
- Department of Statistics, Federal University of Technology, P.M.B 704, Akure, Nigeria
| | | | - Elangwe M Sarah-Matio
- Institut de Recherche pour le Développement, MIVEGEC, Univ. Montpellier, CNRS, IRD, 91 Avenue Agropolis, BP 64501, 34394, Montpellier, France
- Institute for Parasitology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Aline G Bouopda-Tuedom
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroon
- Department of Biological Sciences, Faculté de Médecine et des Sciences Pharmaceutiques, Université de Douala, Douala, Cameroon
| | - Paul Taconet
- Institut de Recherche pour le Développement, MIVEGEC, Univ. Montpellier, CNRS, IRD, 91 Avenue Agropolis, BP 64501, 34394, Montpellier, France
| | - Nicolas Moiroux
- Institut de Recherche pour le Développement, MIVEGEC, Univ. Montpellier, CNRS, IRD, 91 Avenue Agropolis, BP 64501, 34394, Montpellier, France
| | | | - Arthur Talman
- Institut de Recherche pour le Développement, MIVEGEC, Univ. Montpellier, CNRS, IRD, 91 Avenue Agropolis, BP 64501, 34394, Montpellier, France
| | - Lawrence S Ayong
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroon
| | - Antoine Berry
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université Toulouse, CNRS UMR5051, INSERM UMR1291, UPS, Toulouse, France
- Service de Parasitologie_Mycologie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Sandrine E Nsango
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroon
- Department of Biological Sciences, Faculté de Médecine et des Sciences Pharmaceutiques, Université de Douala, Douala, Cameroon
| | - Isabelle Morlais
- Institut de Recherche pour le Développement, MIVEGEC, Univ. Montpellier, CNRS, IRD, 91 Avenue Agropolis, BP 64501, 34394, Montpellier, France.
| |
Collapse
|
10
|
Adipokinetic hormone signaling in the malaria vector Anopheles gambiae facilitates Plasmodium falciparum sporogony. Commun Biol 2023; 6:171. [PMID: 36782045 PMCID: PMC9924834 DOI: 10.1038/s42003-023-04518-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 01/23/2023] [Indexed: 02/15/2023] Open
Abstract
An obligatory step in the complex life cycle of the malaria parasite is sporogony, which occurs during the oocyst stage in adult female Anopheles mosquitoes. Sporogony is metabolically demanding, and successful oocyst maturation is dependent on host lipids. In insects, lipid energy reserves are mobilized by adipokinetic hormones (AKHs). We hypothesized that Plasmodium falciparum infection activates Anopheles gambiae AKH signaling and lipid mobilization. We profiled the expression patterns of AKH pathway genes and AgAkh1 peptide levels in An. gambiae during starvation, after blood feeding, and following infection and observed a significant time-dependent up-regulation of AKH pathway genes and peptide levels during infection. Depletion of AgAkh1 and AgAkhR by RNAi reduced salivary gland sporozoite production, while synthetic AgAkh1 peptide supplementation rescued sporozoite numbers. Inoculation of uninfected female mosquitoes with supernatant from P. falciparum-infected midguts activated AKH signaling. Clearly, identifying the parasite molecules mediating AKH signaling in P. falciparum sporogony is paramount.
Collapse
|
11
|
Dong X, Chen J, Xu R, Li X, Wang Y, Pan X, Zhang C, Li Y, Wang F, Li C. Molecular identification and lipid mobilization role of adipokinetic hormone receptor in Spodoptera litura (F.). BULLETIN OF ENTOMOLOGICAL RESEARCH 2022; 112:758-765. [PMID: 35431022 DOI: 10.1017/s0007485322000141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Energy homeostasis is essential for organisms to maintain fluctuation in energy accumulation, mobilization. Lipids as the main energy reserve in insects, their metabolism is under the control of many physiological program. This study aimed to determine whether the adipokinetic hormone receptor (AKHR) was involved in the lipid mobilization in the Spodoptera litura. A full-length cDNA encoding AKHR was isolated from S. litura. The SlAKHR protein has a conserved seven-transmembrane domain which is the character of a putative G protein receptor. Expression profile investigation revealed that SlAKHR mRNA was highly expressed in immatural stage and abundant in fat body in newly emerged female adults. Knockdown of SlAKHR expression was achieved through RNAi by injecting double-stranded RNA (dsRNA) into the 6th instar larvae. The content of triacylgycerol (TAG) in the fat body increased significantly after the SlAKHR gene was knockdown. And decrease of TAG releasing to hemolymph with increase of free fatty acid (FFA) in hemolymph were observed when the SlAKHR gene was knowned-down. In addition, lipid droplets increased in fat body was also found. These results suggested that SlAKHR is critical for insects to regulate lipids metabolism.
Collapse
Affiliation(s)
- Xiaolin Dong
- Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
- Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Junyuan Chen
- Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Rubing Xu
- Tobacco Research Institute of Hubei Province, Wuhan 430002, Hubei, China
| | - Xihong Li
- Tobacco Research Institute of Hubei Province, Wuhan 430002, Hubei, China
| | - Yang Wang
- Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Xue Pan
- Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Cuici Zhang
- Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Yanyan Li
- Tobacco Research Institute of Hubei Province, Wuhan 430002, Hubei, China
| | - Fulian Wang
- Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| | - Chuanren Li
- Hubei Engineering Research Center for Pest Forewarning and Management, College of Agriculture, Yangtze University, Jingzhou 434025, Hubei, China
| |
Collapse
|
12
|
Zakhia R, Osta MA. CLIPA7 Exhibits Pleiotropic Roles in the Anopheles gambiae Immune Response. J Innate Immun 2022; 15:317-332. [PMID: 36423593 PMCID: PMC10643895 DOI: 10.1159/000526486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/09/2022] [Indexed: 10/30/2023] Open
Abstract
Clip domain serine proteases and clip domain serine protease homologs (cSPHs) are key components of serine protease cascades that drive the melanization response. Despite lacking catalytic activity, cSPHs play essential roles in regulating melanization, but the spectrum of functions they catalyze within and outside these cascades is not fully understood. Aside from their classical role as cofactors for PPO activation, we have previously revealed an unprecedented complexity in the function and molecular organization of these cSPHs in the immune response of the malaria vector Anopheles gambiae. Here, we add yet another dimension to the complex roles underpinning the contributions of cSPHs to mosquito immunity by showing that CLIPA7, a member of the expanded cSPH family, defines a novel branch within the cSPH network that is essential for the melanization of Escherichia coli but not Plasmodium ookinetes or Gram-positive bacteria. Despite its dispensability for the melanization of Gram-positive bacteria, we show that CLIPA7 is required for the clearance of systemic infections with Staphylococcus aureus. CLIPA7 is produced by hemocytes and associates with the surfaces of live E. coli and S. aureus cells in vivo as well as with those of melanized cells. Based on its RNAi phenotypes and its unique domain architecture among A. gambiae cSPHs including the presence of an RGD motif, we propose that CLIPA7 exhibits pleiotropic roles in mosquito immunity that extend beyond the regulation of melanization to microbial clearance.
Collapse
Affiliation(s)
| | - Mike A. Osta
- Department of Biology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
13
|
Shaw WR, Marcenac P, Catteruccia F. Plasmodium development in Anopheles: a tale of shared resources. Trends Parasitol 2022; 38:124-135. [PMID: 34548252 PMCID: PMC8758519 DOI: 10.1016/j.pt.2021.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
Interactions between the Anopheles mosquito vector and Plasmodium parasites shape how malaria is transmitted in endemic regions. The long association of these two organisms has led to evolutionary processes that minimize fitness costs of infection and benefit both players through shared nutrient resources, parasite immune suppression, and mosquito tolerance to infection. In this review we explore recent data describing how Plasmodium falciparum, the deadliest malaria parasite, associates with one of its most important natural mosquito hosts, Anopheles gambiae, and we discuss the implications of these findings for parasite transmission and vector control strategies currently in development.
Collapse
Affiliation(s)
- W Robert Shaw
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Perrine Marcenac
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
14
|
Tapanelli S, Inghilterra MG, Cai J, Philpott J, Capriotti P, Windbichler N, Christophides GK. Assessment of Plasmodium falciparum Infection and Fitness of Genetically Modified Anopheles gambiae Aimed at Mosquito Population Replacement. FRONTIERS IN TROPICAL DISEASES 2021. [DOI: 10.3389/fitd.2021.806880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Genetically modified (GM) mosquitoes expressing anti-plasmodial effectors propagating through wild mosquito populations by means of gene drive is a promising tool to support current malaria control strategies. The process of generating GM mosquitoes involves genetic transformation of mosquitoes from a laboratory colony and, often, interbreeding with other GM lines to cross in auxiliary traits. These mosquito colonies and GM lines thus often have different genetic backgrounds and GM lines are invariably highly inbred, which in conjunction with their independent rearing in the laboratory may translate to differences in their susceptibility to malaria parasite infection and life history traits. Here, we show that laboratory Anopheles gambiae colonies and GM lines expressing Cas9 and Cre recombinase vary greatly in their susceptibility to Plasmodium falciparum NF54 infection. Therefore, the choice of mosquitoes to be used as a reference when conducting infection or life history trait assays requires careful consideration. To address these issues, we established an experimental pipeline involving genetic crosses and genotyping of mosquitoes reared in shared containers throughout their lifecycle. We used this protocol to examine whether GM lines expressing the antimicrobial peptide (AMP) Scorpine in the mosquito midgut interfere with parasite infection and mosquito survival. We demonstrate that Scorpine expression in the Peritrophin 1 (Aper1) genomic locus reduces both P. falciparum sporozoite prevalence and mosquito lifespan; both these phenotypes are likely to be associated with the disturbance of the midgut microbiota homeostasis. These data lead us to conclude that the Aper1-Sco GM line could be used in proof-of-concept experiments aimed at mosquito population replacement, although the impact of its reduced fitness on the spread of the transgene through wild populations requires further investigation.
Collapse
|
15
|
Hun LV, Cheung KW, Brooks E, Zudekoff R, Luckhart S, Riehle MA. Increased insulin signaling in the Anopheles stephensi fat body regulates metabolism and enhances the host response to both bacterial challenge and Plasmodium falciparum infection. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 139:103669. [PMID: 34666189 PMCID: PMC8647039 DOI: 10.1016/j.ibmb.2021.103669] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/09/2021] [Accepted: 10/10/2021] [Indexed: 05/06/2023]
Abstract
In vertebrates and invertebrates, the insulin/insulin-like growth factor 1 (IGF1) signaling (IIS) cascade is highly conserved and plays a vital role in many different physiological processes. Among the many tissues that respond to IIS in mosquitoes, the fat body has a central role in metabolism, lifespan, reproduction, and innate immunity. We previously demonstrated that fat body specific expression of active Akt, a key IIS signaling molecule, in adult Anopheles stephensi and Aedes aegypti activated the IIS cascade and extended lifespan. Additionally, we found that transgenic females produced more vitellogenin (Vg) protein than non-transgenic mosquitoes, although this did not translate into increased fecundity. These results prompted us to further examine how IIS impacts immunity, metabolism, growth and development of these transgenic mosquitoes. We observed significant changes in glycogen, trehalose, triglycerides, glucose, and protein in young (3-5 d) transgenic mosquitoes relative to non-transgenic sibling controls, while only triglycerides were significantly changed in older (18 d) transgenic mosquitoes. More importantly, we demonstrated that enhanced fat body IIS decreased both the prevalence and intensity of Plasmodium falciparum infection in transgenic An. stephensi. Additionally, challenging transgenic An. stephensi with Gram-positive and Gram-negative bacteria altered the expression of several antimicrobial peptides (AMPs) and two anti-Plasmodium genes, nitric oxide synthase (NOS) and thioester complement-like protein (TEP1), relative to non-transgenic controls. Increased IIS in the fat body of adult female An. stephensi had little to no impact on body size, growth or development of progeny from transgenic mosquitoes relative to non-transgenic controls. This study both confirms and expands our understanding of the critical roles insulin signaling plays in regulating the diverse functions of the mosquito fat body.
Collapse
Affiliation(s)
- Lewis V Hun
- Department of Entomology, University of California Riverside, Riverside, CA, USA; Department of Entomology, University of Arizona, Tucson, AZ, USA
| | - Kong Wai Cheung
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Elizabeth Brooks
- Department of Entomology, University of Arizona, Tucson, AZ, USA
| | - Rissa Zudekoff
- Department of Entomology, University of Arizona, Tucson, AZ, USA
| | - Shirley Luckhart
- Departrment of Entomology, Plant Pathology and Nematology and Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - Michael A Riehle
- Department of Entomology, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
16
|
Dong S, Dong Y, Simões ML, Dimopoulos G. Mosquito transgenesis for malaria control. Trends Parasitol 2021; 38:54-66. [PMID: 34483052 DOI: 10.1016/j.pt.2021.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022]
Abstract
Malaria is one of the deadliest diseases. Because of the ineffectiveness of current malaria-control methods, several novel mosquito vector-based control strategies have been proposed to supplement existing control strategies. Mosquito transgenesis and gene drive have emerged as promising tools for preventing the spread of malaria by either suppressing mosquito populations by self-destructing mosquitoes or replacing mosquito populations with disease-refractory populations. Here we review the development of mosquito transgenesis and its application for malaria control, highlighting the transgenic expression of antiparasitic effector genes, inactivation of host factor genes, and manipulation of miRNAs and lncRNAs. Overall, from a malaria-control perspective, mosquito transgenesis is not envisioned as a stand-alone approach; rather, its use is proposed as a complement to existing vector-control strategies.
Collapse
Affiliation(s)
- Shengzhang Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
17
|
Patil G. Evolution of fibrinogen domain related proteins in Aedes aegypti: Their expression during Arbovirus infections. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
18
|
Ferdous Z, Fuchs S, Behrends V, Trasanidis N, Waterhouse RM, Vlachou D, Christophides GK. Anopheles coluzzii stearoyl-CoA desaturase is essential for adult female survival and reproduction upon blood feeding. PLoS Pathog 2021; 17:e1009486. [PMID: 34015060 PMCID: PMC8171932 DOI: 10.1371/journal.ppat.1009486] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 06/02/2021] [Accepted: 03/19/2021] [Indexed: 01/08/2023] Open
Abstract
Vitellogenesis and oocyte maturation require anautogenous female Anopheles mosquitoes to obtain a bloodmeal from a vertebrate host. The bloodmeal is rich in proteins that are readily broken down into amino acids in the midgut lumen and absorbed by the midgut epithelial cells where they are converted into lipids and then transported to other tissues including ovaries. The stearoyl-CoA desaturase (SCD) plays a pivotal role in this process by converting saturated (SFAs) to unsaturated (UFAs) fatty acids; the latter being essential for maintaining cell membrane fluidity amongst other housekeeping functions. Here, we report the functional and phenotypic characterization of SCD1 in the malaria vector mosquito Anopheles coluzzii. We show that RNA interference (RNAi) silencing of SCD1 and administration of sterculic acid (SA), a small molecule inhibitor of SCD1, significantly impact on the survival and reproduction of female mosquitoes following blood feeding. Microscopic observations reveal that the mosquito thorax is quickly filled with blood, a phenomenon likely caused by the collapse of midgut epithelial cell membranes, and that epithelial cells are depleted of lipid droplets and oocytes fail to mature. Transcriptional profiling shows that genes involved in protein, lipid and carbohydrate metabolism and immunity-related genes are the most affected by SCD1 knock down (KD) in blood-fed mosquitoes. Metabolic profiling reveals that these mosquitoes exhibit increased amounts of saturated fatty acids and TCA cycle intermediates, highlighting the biochemical framework by which the SCD1 KD phenotype manifests as a result of a detrimental metabolic syndrome. Accumulation of SFAs is also the likely cause of the potent immune response observed in the absence of infection, which resembles an auto-inflammatory condition. These data provide insights into mosquito bloodmeal metabolism and lipid homeostasis and could inform efforts to develop novel interventions against mosquito-borne diseases. Female mosquitoes can become infected with malaria parasites upon ingestion of blood from an infected person and can transmit the disease when they bite another person some days later. The bloodmeal is rich in proteins which female mosquitoes use to develop their eggs after converting them first to saturated and then to unsaturated fatty acids inside their gut cells. Here, we present the characterization of the enzyme that mosquitoes use to convert saturated to unsaturated fatty acids and show that when this enzyme is eliminated or inhibited mosquitoes cannot produce eggs and die soon after they feed on blood. The mosquito death appears to be primarily associated with the collapse of their gut epithelial barrier due to the loss of cell membrane integrity, leading to their inner body cavity being filled with the ingested blood. These mosquitoes also suffer from an acute and detrimental auto-inflammatory condition due to mounting of a potent immune response in the absence of any infection. We conclude that this enzyme and the mechanism of converting blood-derived proteins to unsaturated fatty acids as a whole can be a good target of interventions aiming at limiting the mosquito abundance and blocking malaria transmission.
Collapse
Affiliation(s)
- Zannatul Ferdous
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Silke Fuchs
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Volker Behrends
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Health Science Research Centre, University of Roehampton, London, United Kingdom
| | - Nikolaos Trasanidis
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Robert M. Waterhouse
- Department of Ecology and Evolution, University of Lausanne, Lausanne, Switzerland
| | - Dina Vlachou
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | |
Collapse
|
19
|
Soumare HM, Guelbeogo WM, van de Vegte-Bolmer M, van Gemert GJ, Soumanaba Z, Ouedraogo A, Ouattara MS, Abdullahi A, Jadama L, Camara MM, Gaye PM, Mendy M, Davis N, Tiono AB, D'Alessandro U, Drakeley C, Bousema T, Moreno M, Collins KA. Maintaining Plasmodium falciparum gametocyte infectivity during blood collection and transport for mosquito feeding assays in the field. Malar J 2021; 20:191. [PMID: 33879163 PMCID: PMC8056727 DOI: 10.1186/s12936-021-03725-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/04/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Mosquito feeding assays using venous blood are commonly used for evaluating the transmission potential of malaria infected individuals. To improve the accuracy of these assays, care must be taken to prevent premature activation or inactivation of gametocytes before they are fed to mosquitoes. This can be challenging in the field where infected individuals and insectary facilities are sometimes very far apart. In this study, a simple, reliable, field applicable method is presented for storage and transport of gametocyte infected blood using a thermos flask. METHODS The optimal storage conditions for maintaining the transmissibility of gametocytes were determined initially using cultured Plasmodium falciparum gametocytes in standard membrane feeding assays (SMFAs). The impact of both the internal thermos water temperature (35.5 to 37.8 °C), and the external environmental temperature (room temperature to 42 °C) during long-term (4 h) storage, and the impact of short-term (15 min) temperature changes (room temp to 40 °C) during membrane feeding assays was assessed. The optimal conditions were then evaluated in direct membrane feeding assays (DMFAs) in Burkina Faso and The Gambia where blood from naturally-infected gametocyte carriers was offered to mosquitoes immediately and after storage in thermos flasks. RESULTS Using cultured gametocytes in SMFAs it was determined that an internal thermos water temperature of 35.5 °C and storage of the thermos flask between RT (~ 21.3 °C) and 32 °C was optimal for maintaining transmissibility of gametocytes for 4 h. Short-term storage of the gametocyte infected blood for 15 min at temperatures up to 40 °C (range: RT, 30 °C, 38 °C and 40 °C) did not negatively affect gametocyte infectivity. Using samples from natural gametocyte carriers (47 from Burkina Faso and 16 from The Gambia), the prevalence of infected mosquitoes and the intensity of oocyst infection was maintained when gametocyte infected blood was stored in a thermos flask in water at 35.5 °C for up to 4 h. CONCLUSIONS This study determines the optimal long-term (4 h) storage temperature for gametocyte infected blood and the external environment temperature range within which gametocyte infectivity is unaffected. This will improve the accuracy, reproducibility, and utility of DMFAs in the field, and permit reliable comparative assessments of malaria transmission epidemiology in different settings.
Collapse
Affiliation(s)
- Harouna M Soumare
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | | | | | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Zongo Soumanaba
- Centre National de Recherche Et de Formation Sur Le Paludisme, Ouagadougou, Burkina Faso
| | - Alphonse Ouedraogo
- Centre National de Recherche Et de Formation Sur Le Paludisme, Ouagadougou, Burkina Faso
| | - Maurice S Ouattara
- Centre National de Recherche Et de Formation Sur Le Paludisme, Ouagadougou, Burkina Faso
| | - Ahmad Abdullahi
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Lamin Jadama
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Muhammed M Camara
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Pa Modou Gaye
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Michael Mendy
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Nwakanma Davis
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Alfred B Tiono
- Centre National de Recherche Et de Formation Sur Le Paludisme, Ouagadougou, Burkina Faso
| | - Umberto D'Alessandro
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Chris Drakeley
- Department of Biology of Infection, London School of Hygiene and Tropical Medicine, Faculty of Infectious and Tropical Diseases, London, UK
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marta Moreno
- Department of Biology of Infection, London School of Hygiene and Tropical Medicine, Faculty of Infectious and Tropical Diseases, London, UK
| | - Katharine A Collins
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
20
|
Plasmodium oocysts respond with dormancy to crowding and nutritional stress. Sci Rep 2021; 11:3090. [PMID: 33542254 PMCID: PMC7862253 DOI: 10.1038/s41598-021-81574-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Malaria parasites develop as oocysts in the mosquito for several days before they are able to infect a human host. During this time, mosquitoes take bloodmeals to replenish their nutrient and energy reserves needed for flight and reproduction. We hypothesized that these bloodmeals are critical for oocyst growth and that experimental infection protocols, typically involving a single bloodmeal at the time of infection, cause nutritional stress to the developing oocysts. Therefore, enumerating oocysts disregarding their growth and differentiation state may lead to erroneous conclusions about the efficacy of transmission blocking interventions. Here, we examine this hypothesis in Anopheles coluzzii mosquitoes infected with the human and rodent parasites Plasmodium falciparum and Plasmodium berghei, respectively. We show that oocyst growth and maturation rates decrease at late developmental stages as infection intensities increase; an effect exacerbated at very high infection intensities but fully restored with post infection bloodmeals. High infection intensities and starvation conditions reduce RNA Polymerase III activity in oocysts unless supplemental bloodmeals are provided. Our results suggest that oocysts respond to crowding and nutritional stress with a dormancy-like strategy, which urges the development of alternative methods to assess the efficacy of transmission blocking interventions.
Collapse
|
21
|
Shaw WR, Holmdahl IE, Itoe MA, Werling K, Marquette M, Paton DG, Singh N, Buckee CO, Childs LM, Catteruccia F. Multiple blood feeding in mosquitoes shortens the Plasmodium falciparum incubation period and increases malaria transmission potential. PLoS Pathog 2020; 16:e1009131. [PMID: 33382824 PMCID: PMC7774842 DOI: 10.1371/journal.ppat.1009131] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/08/2020] [Indexed: 12/15/2022] Open
Abstract
Many mosquito species, including the major malaria vector Anopheles gambiae, naturally undergo multiple reproductive cycles of blood feeding, egg development and egg laying in their lifespan. Such complex mosquito behavior is regularly overlooked when mosquitoes are experimentally infected with malaria parasites, limiting our ability to accurately describe potential effects on transmission. Here, we examine how Plasmodium falciparum development and transmission potential is impacted when infected mosquitoes feed an additional time. We measured P. falciparum oocyst size and performed sporozoite time course analyses to determine the parasite's extrinsic incubation period (EIP), i.e. the time required by parasites to reach infectious sporozoite stages, in An. gambiae females blood fed either once or twice. An additional blood feed at 3 days post infection drastically accelerates oocyst growth rates, causing earlier sporozoite accumulation in the salivary glands, thereby shortening the EIP (reduction of 2.3 ± 0.4 days). Moreover, parasite growth is further accelerated in transgenic mosquitoes with reduced reproductive capacity, which mimic genetic modifications currently proposed in population suppression gene drives. We incorporate our shortened EIP values into a measure of transmission potential, the basic reproduction number R0, and find the average R0 is higher (range: 10.1%-12.1% increase) across sub-Saharan Africa than when using traditional EIP measurements. These data suggest that malaria elimination may be substantially more challenging and that younger mosquitoes or those with reduced reproductive ability may provide a larger contribution to infection than currently believed. Our findings have profound implications for current and future mosquito control interventions.
Collapse
Affiliation(s)
- W. Robert Shaw
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Inga E. Holmdahl
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Center for Communicable Disease Dynamics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Maurice A. Itoe
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Kristine Werling
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Meghan Marquette
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Douglas G. Paton
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Naresh Singh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Caroline O. Buckee
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Center for Communicable Disease Dynamics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Lauren M. Childs
- Department of Mathematics, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| |
Collapse
|
22
|
Taylor DM, Haney RS, Luckhart S. Aquatic Exposure to Abscisic Acid Transstadially Enhances Anopheles stephensi Resistance to Malaria Parasite Infection. Genes (Basel) 2020; 11:E1393. [PMID: 33255333 PMCID: PMC7761407 DOI: 10.3390/genes11121393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 12/13/2022] Open
Abstract
The ancient stress signaling molecule abscisic acid (ABA) is ubiquitous in animals and plants but is perhaps most well-known from its early discovery as a plant hormone. ABA can be released into water by plants and is found in nectar, but is also present in mammalian blood, three key contexts for mosquito biology. We previously established that addition of ABA to Anopheles stephensi larval rearing water altered immature development and life history traits of females derived from treated larvae, while addition of ABA to an infected bloodmeal increased resistance of adult female A. stephensi to human malaria parasite infection. Here we sought to determine whether larval treatment with ABA could similarly impact resistance to parasite infection in females derived from treated larvae and, if so, whether resistance could be extended to another parasite species. We examined nutrient levels and gene expression to demonstrate that ABA can transstadially alter resistance to a rodent malaria parasite with hallmarks of previously observed mechanisms of resistance following provision of ABA in blood to A. stephensi.
Collapse
Affiliation(s)
- Dean M. Taylor
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID 83844, USA; (D.M.T.); (R.S.H.)
| | - Reagan S. Haney
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID 83844, USA; (D.M.T.); (R.S.H.)
| | - Shirley Luckhart
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID 83844, USA; (D.M.T.); (R.S.H.)
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA
| |
Collapse
|
23
|
Tao D, McGill B, Hamerly T, Kobayashi T, Khare P, Dziedzic A, Leski T, Holtz A, Shull B, Jedlicka AE, Walzer A, Slowey PD, Slowey CC, Nsango SE, Stenger DA, Chaponda M, Mulenga M, Jacobsen KH, Sullivan DJ, Ryan SJ, Ansumana R, Moss WJ, Morlais I, Dinglasan RR. A saliva-based rapid test to quantify the infectious subclinical malaria parasite reservoir. Sci Transl Med 2020; 11:11/473/eaan4479. [PMID: 30602535 PMCID: PMC6441545 DOI: 10.1126/scitranslmed.aan4479] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/27/2018] [Accepted: 11/30/2018] [Indexed: 01/01/2023]
Abstract
A large proportion of ongoing malaria parasite transmission is attributed to low-density subclinical infections not readily detected by available rapid diagnostic tests (RDTs) or microscopy. Plasmodium falciparum gametocyte carriage is subclinical, but gametocytemic individuals comprise the parasite reservoir that leads to infection of mosquitoes and local transmission. Effective detection and quantification of these carriers can help advance malaria elimination strategies. However, no point-of-need (PON) RDTs for gametocyte detection exist, much less one that can perform noninvasive sampling of saliva outside a clinical setting. Here, we report on the discovery of 35 parasite markers from which we selected a single candidate for use in a PON RDT. We performed a cross-sectional, multi-omics study of saliva from 364 children with subclinical infection in Cameroon and Zambia and produced a prototype saliva-based PON lateral flow immunoassay test for P. falciparum gametocyte carriers. The test is capable of identifying submicroscopic carriage in both clinical and nonclinical settings and is compatible with archived saliva samples.
Collapse
Affiliation(s)
- Dingyin Tao
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.,Johns Hopkins Malaria Research Institute, Baltimore, MD 21205, USA
| | - Brent McGill
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.,Johns Hopkins Malaria Research Institute, Baltimore, MD 21205, USA
| | - Timothy Hamerly
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.,Johns Hopkins Malaria Research Institute, Baltimore, MD 21205, USA.,Emerging Pathogens Institute and Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Tamaki Kobayashi
- Johns Hopkins Malaria Research Institute, Baltimore, MD 21205, USA.,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Prachi Khare
- Emerging Pathogens Institute and Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Amanda Dziedzic
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Tomasz Leski
- United States Naval Research Laboratory (NRL), Center for Biomolecular Science and Engineering, Washington, DC 20375, USA
| | - Andrew Holtz
- College of Science, George Mason University, Fairfax, VA 22030, USA
| | - Bruce Shull
- Thermo Fisher Scientific, Fremont, CA 94538, USA
| | - Anne E Jedlicka
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | | | | - Sandrine E Nsango
- Laboratoire de Recherche sur le Paludisme, Institut de Recherche pour le Développement-Organisation de Coordination et de Coopération pour la Lutte Contre les Grandes Endémies en Afrique Centrale (IRD-OCEAC), Yaoundé, Cameroon.,Faculty of Medicine and Pharmaceutical Sciences, University of Douala, PO Box 2701, Douala, Cameroon
| | - David A Stenger
- United States Naval Research Laboratory (NRL), Center for Biomolecular Science and Engineering, Washington, DC 20375, USA
| | | | | | - Kathryn H Jacobsen
- College of Health and Human Services, George Mason University, Fairfax, VA 22030, USA
| | - David J Sullivan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Sadie J Ryan
- Emerging Pathogens Institute and Department of Geography, College of Liberal Arts and Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Rashid Ansumana
- Mercy Hospital Research Laboratory, Kulanda Town, Bo, Sierra Leone
| | - William J Moss
- Johns Hopkins Malaria Research Institute, Baltimore, MD 21205, USA.,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Isabelle Morlais
- Laboratoire de Recherche sur le Paludisme, Institut de Recherche pour le Développement-Organisation de Coordination et de Coopération pour la Lutte Contre les Grandes Endémies en Afrique Centrale (IRD-OCEAC), Yaoundé, Cameroon
| | - Rhoel R Dinglasan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA. .,Johns Hopkins Malaria Research Institute, Baltimore, MD 21205, USA.,Emerging Pathogens Institute and Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
24
|
Yang J, Schleicher TR, Dong Y, Park HB, Lan J, Cresswell P, Crawford J, Dimopoulos G, Fikrig E. Disruption of mosGILT in Anopheles gambiae impairs ovarian development and Plasmodium infection. J Exp Med 2020; 217:e20190682. [PMID: 31658986 PMCID: PMC7037243 DOI: 10.1084/jem.20190682] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/06/2019] [Accepted: 10/25/2019] [Indexed: 11/04/2022] Open
Abstract
Plasmodium infection in Anopheles is influenced by mosquito-derived factors. We previously showed that a protein in saliva from infected Anopheles, mosquito gamma-interferon-inducible lysosomal thiol reductase (mosGILT), inhibits the ability of sporozoites to traverse cells and readily establish infection of the vertebrate host. To determine whether mosGILT influences Plasmodium within the mosquito, we generated Anopheles gambiae mosquitoes carrying mosaic mutations in the mosGILT gene using CRISPR/CRISPR associated protein 9 (Cas9). Here, we show that female mosaic mosGILT mutant mosquitoes display defects in ovarian development and refractoriness to Plasmodium. Following infection by either Plasmodium berghei or Plasmodium falciparum, mutant mosquitoes have significantly reduced oocyst numbers as a result of increased thioester-containing protein 1 (TEP1)-dependent parasite killing. Expression of vitellogenin (Vg), the major yolk protein that can reduce the parasite-killing efficiency of TEP1, is severely impaired in mutant mosquitoes. MosGILT is a mosquito factor that is essential for ovarian development and indirectly protects both human and rodent Plasmodium species from mosquito immunity.
Collapse
Affiliation(s)
- Jing Yang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Tyler R. Schleicher
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Yuemei Dong
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Hyun Bong Park
- Department of Chemistry, Yale University, New Haven, CT
- Chemical Biology Institute, Yale University, West Haven, CT
| | - Jiangfeng Lan
- College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Peter Cresswell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Jason Crawford
- Department of Chemistry, Yale University, New Haven, CT
- Chemical Biology Institute, Yale University, West Haven, CT
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT
| | - George Dimopoulos
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Chevy Chase, MD
| |
Collapse
|
25
|
Abstract
The composition of insect hemolymph can change depending on many factors, e.g. access to nutrients, stress conditions, and current needs of the insect. In this chapter, insect immune-related polypeptides, which can be permanently or occasionally present in the hemolymph, are described. Their division into peptides or low-molecular weight proteins is not always determined by the length or secondary structure of a given molecule but also depends on the mode of action in insect immunity and, therefore, it is rather arbitrary. Antimicrobial peptides (AMPs) with their role in immunity, modes of action, and classification are presented in the chapter, followed by a short description of some examples: cecropins, moricins, defensins, proline- and glycine-rich peptides. Further, we will describe selected immune-related proteins that may participate in immune recognition, may possess direct antimicrobial properties, or can be involved in the modulation of insect immunity by both abiotic and biotic factors. We briefly cover Fibrinogen-Related Proteins (FREPs), Down Syndrome Cell Adhesion Molecules (Dscam), Hemolin, Lipophorins, Lysozyme, Insect Metalloproteinase Inhibitor (IMPI), and Heat Shock Proteins. The reader will obtain a partial picture presenting molecules participating in one of the most efficient immune strategies found in the animal world, which allow insects to inhabit all ecological land niches in the world.
Collapse
Affiliation(s)
- Iwona Wojda
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland.
| | - Małgorzata Cytryńska
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Agnieszka Zdybicka-Barabas
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Jakub Kordaczuk
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
26
|
Wolanin K, Fontinha D, Sanches-Vaz M, Nyboer B, Heiss K, Mueller AK, Prudêncio M. A crucial role for the C-terminal domain of exported protein 1 during the mosquito and hepatic stages of the Plasmodium berghei life cycle. Cell Microbiol 2019; 21:e13088. [PMID: 31364224 PMCID: PMC6771729 DOI: 10.1111/cmi.13088] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/25/2019] [Accepted: 07/28/2019] [Indexed: 12/19/2022]
Abstract
Intracellular Plasmodium parasites develop inside a parasitophorous vacuole (PV), a specialised compartment enclosed by a membrane (PVM) that contains proteins of both host and parasite origin. Although exported protein 1 (EXP1) is one of the earliest described parasitic PVM proteins, its function throughout the Plasmodium life cycle remains insufficiently understood. Here, we show that whereas the N-terminus of Plasmodium berghei EXP1 (PbEXP1) is essential for parasite survival in the blood, parasites lacking PbEXP1's entire C-terminal (CT) domain replicate normally in the blood but cause less severe pathology than their wild-type counterparts. Moreover, truncation of PbEXP1's CT domain not only impairs parasite development in the mosquito but also abrogates PbEXP1 localization to the PVM of intrahepatic parasites, severely limiting their replication and preventing their egress into the blood. Our findings highlight the importance of EXP1 during the Plasmodium life cycle and identify this protein as a promising target for antiplasmodial intervention.
Collapse
Affiliation(s)
- Kamil Wolanin
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Margarida Sanches-Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Britta Nyboer
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Kirsten Heiss
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany.,German Centre for Infection Research, Heidelberg Division, Heidelberg, Germany.,PEPperPRINT GmbH, Research & Development Division, Heidelberg, Germany
| | - Ann-Kristin Mueller
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany.,German Centre for Infection Research, Heidelberg Division, Heidelberg, Germany
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
27
|
Reyes Ruiz VM, Sousa GL, Sneed SD, Farrant KV, Christophides GK, Povelones M. Stimulation of a protease targeting the LRIM1/APL1C complex reveals specificity in complement-like pathway activation in Anopheles gambiae. PLoS One 2019; 14:e0214753. [PMID: 30958840 PMCID: PMC6453449 DOI: 10.1371/journal.pone.0214753] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/19/2019] [Indexed: 12/15/2022] Open
Abstract
The complement-like pathway of the African malaria mosquito Anopheles gambiae provides protection against infection by diverse pathogens. A functional requirement for a core set of proteins during infections by rodent and human malaria parasites, bacteria, and fungi suggests a similar mechanism operates against different pathogens. However, the extent to which the molecular mechanisms are conserved is unknown. In this study we probed the biochemical responses of complement-like pathway to challenge by the Gram-positive bacterium Staphyloccocus aureus. Western blot analysis of the hemolymph revealed that S. aureus challenge activates a TEP1 convertase-like activity and promotes the depletion of the protein SPCLIP1. S. aureus challenge did not lead to an apparent change in the abundance of the LRIM1/APL1C complex compared to challenge by the Gram-negative bacterium, Escherichia coli. Following up on this observation using a panel of LRIM1 and APL1C antibodies, we found that E. coli challenge, but not S. aureus, specifically activates a protease that cleaves the C-terminus of APL1C. Inhibitor studies in vivo and in vitro protease assays suggest that a serine protease is responsible for APL1C cleavage. This study reveals that despite different challenges converging on activation of a TEP1 convertase-like activity, the mosquito complement-like pathway also includes pathogen-specific reactions.
Collapse
Affiliation(s)
- Valeria M. Reyes Ruiz
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gregory L. Sousa
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sarah D. Sneed
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Katie V. Farrant
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | | - Michael Povelones
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
28
|
Costa G, Gildenhard M, Eldering M, Lindquist RL, Hauser AE, Sauerwein R, Goosmann C, Brinkmann V, Carrillo-Bustamante P, Levashina EA. Non-competitive resource exploitation within mosquito shapes within-host malaria infectivity and virulence. Nat Commun 2018; 9:3474. [PMID: 30150763 PMCID: PMC6110728 DOI: 10.1038/s41467-018-05893-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 08/01/2018] [Indexed: 11/22/2022] Open
Abstract
Malaria is a fatal human parasitic disease transmitted by a mosquito vector. Although the evolution of within-host malaria virulence has been the focus of many theoretical and empirical studies, the vector’s contribution to this process is not well understood. Here, we explore how within-vector resource exploitation would impact the evolution of within-host Plasmodium virulence. By combining within-vector dynamics and malaria epidemiology, we develop a mathematical model, which predicts that non-competitive parasitic resource exploitation within-vector restricts within-host parasite virulence. To validate our model, we experimentally manipulate mosquito lipid trafficking and gauge within-vector parasite development and within-host infectivity and virulence. We find that mosquito-derived lipids determine within-host parasite virulence by shaping development (quantity) and metabolic activity (quality) of transmissible sporozoites. Our findings uncover the potential impact of within-vector environment and vector control strategies on the evolution of malaria virulence. The evolution of within-host malaria virulence has been studied, but the vector’s contribution isn’t well understood. Here, Costa et al. show that non-competitive parasitic resource exploitation within-vector, in particular lipid trafficking, restricts within-host infectivity and virulence of the parasite.
Collapse
Affiliation(s)
- G Costa
- Vector Biology Unit, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany
| | - M Gildenhard
- Vector Biology Unit, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany
| | - M Eldering
- Vector Biology Unit, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany.,Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - R L Lindquist
- Immunodynamics, German Rheumatism Research Centre (DRFZ), 10117, Berlin, Germany
| | - A E Hauser
- Immunodynamics, German Rheumatism Research Centre (DRFZ), 10117, Berlin, Germany.,Immune Dynamics and Intravital Microscopy, Charité-Universitätsmedizin, 10117, Berlin, Germany
| | - R Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - C Goosmann
- Microscopy Core Facility, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany
| | - V Brinkmann
- Microscopy Core Facility, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany
| | - P Carrillo-Bustamante
- Vector Biology Unit, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany
| | - E A Levashina
- Vector Biology Unit, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany.
| |
Collapse
|
29
|
Assessing Plasmodium falciparum transmission in mosquito-feeding assays using quantitative PCR. Malar J 2018; 17:249. [PMID: 29976199 PMCID: PMC6034226 DOI: 10.1186/s12936-018-2382-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/09/2018] [Indexed: 12/29/2022] Open
Abstract
Background Evaluating the efficacy of transmission-blocking interventions relies on mosquito-feeding assays, with transmission typically assessed by microscopic identification of oocysts in mosquito midguts; however, microscopy has limited throughput, sensitivity and specificity. Where low prevalence and intensity mosquito infections occur, as observed during controlled human malaria infection studies or natural transmission, a reliable method for detection and quantification of low-level midgut infection is required. Here, a semi-automated, Taqman quantitative PCR (qPCR) assay sufficiently sensitive to detect a single-oocyst midgut infection is described. Results Extraction of genomic DNA from Anopheles stephensi midguts using a semi-automated extraction process was shown to have equivalent extraction efficiency to manual DNA extraction. An 18S Plasmodium falciparum qPCR assay was adapted for quantitative detection of P. falciparum midgut oocyst infection using synthetic DNA standards. The assay was validated for sensitivity and specificity, and the limit of detection was 0.7 genomes/µL (95% CI 0.4–1.6 genomes/µL). All microscopy-confirmed oocyst infected midgut samples were detected by qPCR, including all single-oocyst positive midguts. The genome number per oocyst was assessed 8–9 days after feeding assay using both qPCR and droplet digital PCR and was 3722 (IQR: 2951–5453) and 3490 (IQR: 2720–4182), respectively. Conclusions This semi-automated qPCR method enables accurate detection of low-level P. falciparum oocyst infections in mosquito midguts, and may improve the sensitivity, specificity and throughput of assays used to evaluate candidate transmission-blocking interventions. Electronic supplementary material The online version of this article (10.1186/s12936-018-2382-6) contains supplementary material, which is available to authorized users.
Collapse
|
30
|
Simões ML, Caragata EP, Dimopoulos G. Diverse Host and Restriction Factors Regulate Mosquito-Pathogen Interactions. Trends Parasitol 2018; 34:603-616. [PMID: 29793806 DOI: 10.1016/j.pt.2018.04.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 12/12/2022]
Abstract
Mosquitoes transmit diseases that seriously impact global human health. Despite extensive knowledge of the life cycles of mosquito-borne parasites and viruses within their hosts, control strategies have proven insufficient to halt their spread. An understanding of the relationships established between such pathogens and the host tissues they inhabit is therefore paramount for the development of new strategies that specifically target these interactions, to prevent the pathogens' maturation and transmission. Here we present an updated account of the antagonists and host factors that affect the development of Plasmodium, the parasite causing malaria, and mosquito-borne viruses, such as dengue virus and Zika virus, within their mosquito vectors, and we discuss the similarities and differences between Plasmodium and viral systems, looking toward the elucidation of new targets for disease control.
Collapse
Affiliation(s)
- Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; These authors contributed equally
| | - Eric P Caragata
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; These authors contributed equally
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
31
|
Thiévent K, Hofer L, Rapp E, Tambwe MM, Moore S, Koella JC. Malaria infection in mosquitoes decreases the personal protection offered by permethrin-treated bednets. Parasit Vectors 2018; 11:284. [PMID: 29728155 PMCID: PMC5936035 DOI: 10.1186/s13071-018-2846-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/12/2018] [Indexed: 12/19/2022] Open
Abstract
Background Insecticides targeting adult mosquitoes are the main way of controlling malaria. They work not only by killing mosquitoes, but also by repelling and irritating them. Indeed their repellent action gives valuable personal protection against biting mosquitoes. In the context of malaria control this personal protection is especially relevant when mosquitoes are infectious, whereas to protect the community we would prefer that the mosquitoes that are not yet infectious are killed (so, not repelled) by the insecticide. As the infectious stage of malaria parasites increases the motivation of mosquitoes to bite, we predicted that it would also change their behavioural response to insecticides. Results With two systems, a laboratory isolate of the rodent malaria Plasmodium berghei infecting Anopheles gambiae and several isolates of P. falciparum obtained from schoolchildren in Tanzania that infected Anopheles arabiensis, we found that mosquitoes harbouring the infectious stage (the sporozoites) of the parasite were less repelled by permethrin-treated nets than uninfected ones. Conclusions Our results suggest that, at least in the laboratory, malaria infection decreases the personal protection offered by insecticide-treated nets at the stage where the personal protection is most valuable. Further studies must investigate whether these results hold true in the field and whether the less effective personal protection can be balanced by increased community protection. Electronic supplementary material The online version of this article (10.1186/s13071-018-2846-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kevin Thiévent
- Institute of Biology, University of Neuchâtel, Rue Emile-Argand 11, 2000, Neuchâtel, Switzerland.
| | - Lorenz Hofer
- Institute of Biology, University of Neuchâtel, Rue Emile-Argand 11, 2000, Neuchâtel, Switzerland
| | - Elise Rapp
- Institute of Biology, University of Neuchâtel, Rue Emile-Argand 11, 2000, Neuchâtel, Switzerland
| | - Mgeni Mohamed Tambwe
- Ifakara Health Institute, Intervention and Environmental Health and Ecological Sciences, P.O. Box 74, Bagamoyo, Tanzania
| | - Sarah Moore
- Ifakara Health Institute, Intervention and Environmental Health and Ecological Sciences, P.O. Box 74, Bagamoyo, Tanzania.,Swiss Tropical & Public Health Institute, Socinstrasse 57, 4002, Basel, Switzerland
| | - Jacob C Koella
- Institute of Biology, University of Neuchâtel, Rue Emile-Argand 11, 2000, Neuchâtel, Switzerland
| |
Collapse
|
32
|
Dong Y, Simões ML, Marois E, Dimopoulos G. CRISPR/Cas9 -mediated gene knockout of Anopheles gambiae FREP1 suppresses malaria parasite infection. PLoS Pathog 2018. [PMID: 29518156 PMCID: PMC5843335 DOI: 10.1371/journal.ppat.1006898] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Plasmodium relies on numerous agonists during its journey through the mosquito vector, and these agonists represent potent targets for transmission-blocking by either inhibiting or interfering with them pre- or post-transcriptionally. The recently developed CRISPR/Cas9-based genome editing tools for Anopheles mosquitoes provide new and promising opportunities for the study of agonist function and for developing malaria control strategies through gene deletion to achieve complete agonist inactivation. Here we have established a modified CRISPR/Cas9 gene editing procedure for the malaria vector Anopheles gambiae, and studied the effect of inactivating the fibrinogen-related protein 1 (FREP1) gene on the mosquito’s susceptibility to Plasmodium and on mosquito fitness. FREP1 knockout mutants developed into adult mosquitoes that showed profound suppression of infection with both human and rodent malaria parasites at the oocyst and sporozoite stages. FREP1 inactivation, however, resulted in fitness costs including a significantly lower blood-feeding propensity, fecundity and egg hatching rate, a retarded pupation time, and reduced longevity after a blood meal. The causative agent of malaria, Plasmodium, has to complete a complex infection cycle in the Anopheles gambiae mosquito vector in order to reach the salivary gland from where it can be transmitted to a human host. The parasite’s development in the mosquito relies on numerous host factors (agonists), and their inhibition or inactivation can thereby result in suppression of infection and consequently malaria transmission. The recently developed CRISPR/Cas9-based genome editing tools for Anopheles mosquitoes provide new and promising opportunities to delete (inactivate) Plasmodium agonists to better understand their function and for blocking malaria transmission. Here we have established a modified CRISPR/Cas9 genome editing technique for malaria vector A. gambiae mosquitoes. Through this approach we have inactivated the fibrinogen-related protein 1 (FREP1) gene, via CRISPR/Cas9 genome editing, and the impact of this manipulation on the mosquito’s susceptibility to Plasmodium and on mosquito fitness. FREP1 knockout mutants showed a profound suppression of infection with both human and rodent malaria parasites, while it also resulted in fitness costs: a significantly lower blood-feeding propensity, fecundity and egg hatching rate, and a retarded larval development and pupation time, and reduced longevity after a blood meal.
Collapse
Affiliation(s)
- Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Maria L. Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Eric Marois
- Inserm, CNRS, Université de Strasbourg, Strasbourg, France
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
33
|
Mitchell SN, Catteruccia F. Anopheline Reproductive Biology: Impacts on Vectorial Capacity and Potential Avenues for Malaria Control. Cold Spring Harb Perspect Med 2017; 7:a025593. [PMID: 28389513 PMCID: PMC5710097 DOI: 10.1101/cshperspect.a025593] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Vectorial capacity is a mathematical approximation of the efficiency of vector-borne disease transmission, measured as the number of new infections disseminated per case per day by an insect vector. Multiple elements of mosquito biology govern their vectorial capacity, including survival, population densities, feeding preferences, and vector competence. Intriguingly, biological pathways essential to mosquito reproductive fitness directly or indirectly influence a number of these elements. Here, we explore this complex interaction, focusing on how the interplay between mating and blood feeding in female Anopheles not only shapes their reproductive success but also influences their ability to sustain Plasmodium parasite development. Central to malaria transmission, mosquito reproductive biology has recently become the focus of research strategies aimed at malaria control, and we discuss promising new methods based on the manipulation of key reproductive steps. In light of widespread resistance to all public health-approved insecticides targeting mosquito reproduction may prove crucial to the success of malaria-eradication campaigns.
Collapse
Affiliation(s)
- Sara N Mitchell
- Harvard T.H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, Massachusetts 02115
| | - Flaminia Catteruccia
- Harvard T.H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, Massachusetts 02115
| |
Collapse
|
34
|
Nguyen PL, Vantaux A, Hien DF, Dabiré KR, Yameogo BK, Gouagna LC, Fontenille D, Renaud F, Simard F, Costantini C, Thomas F, Cohuet A, Lefèvre T. No evidence for manipulation of Anopheles gambiae, An. coluzzii and An. arabiensis host preference by Plasmodium falciparum. Sci Rep 2017; 7:9415. [PMID: 28842622 PMCID: PMC5572726 DOI: 10.1038/s41598-017-09821-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/21/2017] [Indexed: 01/08/2023] Open
Abstract
Whether malaria parasites can manipulate mosquito host choice in ways that enhance parasite transmission toward suitable hosts and/or reduce mosquito attraction to unsuitable hosts (i.e. specific manipulation) is unknown. To address this question, we experimentally infected three species of mosquito vectors with wild isolates of the human malaria parasite Plasmodium falciparum, and examined the effects of immature and mature infections on mosquito behavioural responses to combinations of calf odour, human odour and outdoor air using a dual-port olfactometer. Regardless of parasite developmental stage and mosquito species, P. falciparum infection did not alter mosquito activation rate or their choice for human odours. The overall expression pattern of host choice of all three mosquito species was consistent with a high degree of anthropophily, with infected and uninfected individuals showing higher attraction toward human odour over calf odour, human odour over outdoor air, and outdoor air over calf odour. Our results suggest that, in this system, the parasite may not be able to manipulate the early long-range behavioural steps involved in the mosquito host-feeding process. Future studies are required to test whether malaria parasites can modify their mosquito host choice at a shorter range to enhance transmission.
Collapse
Affiliation(s)
- Phuong L Nguyen
- MIVEGEC (Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle), UMR IRD 224-CNRS, 5290-UM, Montpellier, France
- Institut de Biologie, École Normale Supérieur, 46 rue d'Ulm, 75012, Paris, France
| | - Amélie Vantaux
- MIVEGEC (Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle), UMR IRD 224-CNRS, 5290-UM, Montpellier, France.
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso, Burkina Faso.
- Malaria Molecular Epidemiology Unit, Institut Pasteur of Cambodia, 5 Bd Monivong, PO Box 983, Phnom Penh, 12 201, Cambodia.
| | - Domonbabele FdS Hien
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso, Burkina Faso
| | - Kounbobr R Dabiré
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso, Burkina Faso
- Laboratoire mixte international sur les vecteurs (LAMIVECT), Bobo Dioulasso, Burkina Faso
| | - Bienvenue K Yameogo
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso, Burkina Faso
| | - Louis-Clément Gouagna
- MIVEGEC (Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle), UMR IRD 224-CNRS, 5290-UM, Montpellier, France
| | - Didier Fontenille
- MIVEGEC (Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle), UMR IRD 224-CNRS, 5290-UM, Montpellier, France
- Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - François Renaud
- MIVEGEC (Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle), UMR IRD 224-CNRS, 5290-UM, Montpellier, France
| | - Frédéric Simard
- MIVEGEC (Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle), UMR IRD 224-CNRS, 5290-UM, Montpellier, France
| | - Carlo Costantini
- MIVEGEC (Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle), UMR IRD 224-CNRS, 5290-UM, Montpellier, France
| | - Fréderic Thomas
- MIVEGEC (Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle), UMR IRD 224-CNRS, 5290-UM, Montpellier, France
| | - Anna Cohuet
- MIVEGEC (Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle), UMR IRD 224-CNRS, 5290-UM, Montpellier, France
| | - Thierry Lefèvre
- MIVEGEC (Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle), UMR IRD 224-CNRS, 5290-UM, Montpellier, France
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso, Burkina Faso
- Laboratoire mixte international sur les vecteurs (LAMIVECT), Bobo Dioulasso, Burkina Faso
| |
Collapse
|
35
|
Sandeu MM, Bayibéki AN, Tchioffo MT, Abate L, Gimonneau G, Awono-Ambéné PH, Nsango SE, Diallo D, Berry A, Texier G, Morlais I. Do the venous blood samples replicate malaria parasite densities found in capillary blood? A field study performed in naturally-infected asymptomatic children in Cameroon. Malar J 2017; 16:345. [PMID: 28818084 PMCID: PMC5561596 DOI: 10.1186/s12936-017-1978-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 08/07/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The measure of new drug- or vaccine-based approaches for malaria control is based on direct membrane feeding assays (DMFAs) where gametocyte-infected blood samples are offered to mosquitoes through an artificial feeder system. Gametocyte donors are identified by the microscopic detection and quantification of malaria blood stages on blood films prepared using either capillary or venous blood. However, parasites are known to sequester in the microvasculature and this phenomenon may alter accurate detection of parasites in blood films. The blood source may then impact the success of mosquito feeding experiments and investigations are needed for the implementation of DMFAs under natural conditions. METHODS Thick blood smears were prepared from blood obtained from asymptomatic children attending primary schools in the vicinity of Mfou (Cameroon) over four transmission seasons. Parasite densities were determined microscopically from capillary and venous blood for 137 naturally-infected gametocyte carriers. The effect of the blood source on gametocyte and asexual stage densities was then assessed by fitting cumulative link mixed models (CLMM). DMFAs were performed to compare the infectiousness of gametocytes from the different blood sources to mosquitoes. RESULTS Prevalence of Plasmodium falciparum asexual stages among asymptomatic children aged from 4 to 15 years was 51.8% (2116/4087). The overall prevalence of P. falciparum gametocyte carriage was 8.9% and varied from one school to another. No difference in the density of gametocyte and asexual stages was found between capillary and venous blood. Attempts to perform DMFAs with capillary blood failed. CONCLUSIONS Plasmodium falciparum malaria parasite densities do not differ between capillary and venous blood in asymptomatic subjects for both gametocyte and trophozoite stages. This finding suggests that the blood source should not interfere with transmission efficiency in DMFAs.
Collapse
Affiliation(s)
- Maurice M. Sandeu
- Laboratoire de Recherche sur le Paludisme, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, BP288, Yaoundé, Cameroon
| | - Albert N. Bayibéki
- Laboratoire de Recherche sur le Paludisme, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, BP288, Yaoundé, Cameroon
| | - Majoline T. Tchioffo
- Laboratoire de Recherche sur le Paludisme, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, BP288, Yaoundé, Cameroon
- UMR MIVEGEC, Institut de Recherche pour le Développement, 911 Avenue Agropolis, BP64501, 34394 Montpellier Cedex, France
| | - Luc Abate
- UMR MIVEGEC, Institut de Recherche pour le Développement, 911 Avenue Agropolis, BP64501, 34394 Montpellier Cedex, France
| | - Geoffrey Gimonneau
- UMR MIVEGEC, Institut de Recherche pour le Développement, 911 Avenue Agropolis, BP64501, 34394 Montpellier Cedex, France
| | - Parfait H. Awono-Ambéné
- Laboratoire de Recherche sur le Paludisme, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, BP288, Yaoundé, Cameroon
| | - Sandrine E. Nsango
- Laboratoire de Recherche sur le Paludisme, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, BP288, Yaoundé, Cameroon
- Université de Douala, Faculté de Médecine et des Sciences Pharmaceutiques, BP2701 Douala, Cameroon
| | - Diadier Diallo
- PATH Malaria Vaccine Initiative, Washington, DC 20001 USA
| | - Antoine Berry
- Centre de Physiopathologie de Toulouse Purpan, INSERM U1043, CNRS, UMR5282, Université de Toulouse III, BP 3028, 31024 Toulouse Cedex 03, France
- Centre Hospitalier Universitaire de Toulouse, TSA 40031, 31059 Toulouse, France
| | - Gaétan Texier
- Centre d’épidémiologie et de santé publique des armées, 111 avenue de la Corse, BP40026, 13568 Marseille Cedex 02, France
- UMR 912-SESSTIM-INSERM/IRD, Université Aix-Marseille, 27 bd Jean Moulin, 13385 Marseille Cedex 05, France
| | - Isabelle Morlais
- Laboratoire de Recherche sur le Paludisme, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, BP288, Yaoundé, Cameroon
- UMR MIVEGEC, Institut de Recherche pour le Développement, 911 Avenue Agropolis, BP64501, 34394 Montpellier Cedex, France
| |
Collapse
|
36
|
Dhawan R, Gupta K, Kajla M, Kakani P, Choudhury TP, Kumar S, Kumar V, Gupta L. Apolipophorin-III Acts as a Positive Regulator of Plasmodium Development in Anopheles stephensi. Front Physiol 2017; 8:185. [PMID: 28439240 PMCID: PMC5383653 DOI: 10.3389/fphys.2017.00185] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/10/2017] [Indexed: 01/30/2023] Open
Abstract
Apolipophorin III (ApoLp-III) is a well-known hemolymph protein having a functional role in lipid transport and immune responses of insects. Here we report the molecular and functional characterization of Anopheles stephensi Apolipophorin-III (AsApoLp-III) gene. This gene consists of 679 nucleotides arranged into two exons of 45 and 540 bp that give an ORF encoding 194 amino acid residues. Excluding a putative signal peptide of the first 19 amino acid residues, the 175-residues in mature AsApoLp-III protein has a calculated molecular mass of 22 kDa. Phylogenetic analysis revealed the divergence of mosquitoes (Order Diptera) ApoLp-III from their counterparts in moths (Order: Lepidoptera). Also, it revealed a close relatedness of AsApoLp-III to ApoLp-III of An. gambiae. AsApoLp-III mRNA expression is strongly induced in Plasmodium berghei infected mosquito midguts suggesting its crucial role in parasite development. AsApoLp-III silencing decreased P. berghei oocysts numbers by 7.7 fold against controls. These effects might be due to the interruption of AsApoLp-III mediated lipid delivery to the developing oocysts. In addition, nitric oxide synthase (NOS), an antiplasmodial gene, is also highly induced in AsApoLp-III silenced midguts suggesting that this gene acts like an agonist and protects Plasmodium against the mosquito immunity.
Collapse
Affiliation(s)
- Rini Dhawan
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and SciencePilani, India
| | - Kuldeep Gupta
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and SciencePilani, India
| | - Mithilesh Kajla
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and SciencePilani, India
| | - Parik Kakani
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and SciencePilani, India
| | - Tania P Choudhury
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and SciencePilani, India
| | - Sanjeev Kumar
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and SciencePilani, India.,Department of Biotechnology, Chaudhary Bansi Lal UniversityBhiwani, India
| | - Vikas Kumar
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and SciencePilani, India
| | - Lalita Gupta
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and SciencePilani, India.,Department of Zoology, Chaudhary Bansi Lal UniversityBhiwani, India
| |
Collapse
|
37
|
Barik TK, Suzuki Y, Rasgon JL. Factors influencing infection and transmission of Anopheles gambiae densovirus (AgDNV) in mosquitoes. PeerJ 2016; 4:e2691. [PMID: 27867767 PMCID: PMC5111888 DOI: 10.7717/peerj.2691] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/15/2016] [Indexed: 11/20/2022] Open
Abstract
Anopheles gambiae densovirus (AgDNV) is a potential microbial agent for paratransgenesis and gene transduction in An. gambiae, the major vector of human malaria in sub-Saharan Africa. Understanding the interaction between AgDNV and An. gambiae is critical for using AgDNV in a basic and applied manner for Anopheles gene manipulation. Here, we tested the effects of mosquito age, sex, blood feeding status, and potential for horizontal transmission using an enhanced green fluorescent protein (EGFP) reporter AgDNV system. Neither mosquito age at infection nor feeding regime affected viral titers. Female mosquitoes were more permissive to viral infection than males. Despite low viral titers, infected males were able to venereally transmit virus to females during mating, where the virus was localized with the transferred sperm in the spermathecae. These findings will be useful for designing AgDNV-based strategies to manipulate Anopheles gambiae.
Collapse
Affiliation(s)
- Tapan K Barik
- Applied Entomology Laboratory, Post Graduate Department of Zoology, Berhampur University, Berhampur, Odisha, India.,Department of Entomology, Pennsylvania State University, University Park, PA, United States
| | - Yasutsugu Suzuki
- Department of Entomology, Pennsylvania State University, University Park, PA, United States.,Department of Virology, Institute Pasteur, Paris, France
| | - Jason L Rasgon
- Department of Entomology, Pennsylvania State University, University Park, PA, United States.,Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA, United States.,The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
38
|
Saraiva RG, Kang S, Simões ML, Angleró-Rodríguez YI, Dimopoulos G. Mosquito gut antiparasitic and antiviral immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 64:53-64. [PMID: 26827888 DOI: 10.1016/j.dci.2016.01.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/16/2016] [Accepted: 01/26/2016] [Indexed: 06/05/2023]
Abstract
Mosquitoes are responsible for the transmission of diseases with a serious impact on global human health, such as malaria and dengue. All mosquito-transmitted pathogens complete part of their life cycle in the insect gut, where they are exposed to mosquito-encoded barriers and active factors that can limit their development. Here we present the current understanding of mosquito gut immunity against malaria parasites, filarial worms, and viruses such as dengue, Chikungunya, and West Nile. The most recently proposed immune mediators involved in intestinal defenses are discussed, as well as the synergies identified between the recognition of gut microbiota and the mounting of the immune response.
Collapse
Affiliation(s)
- Raúl G Saraiva
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Seokyoung Kang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Yesseinia I Angleró-Rodríguez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
39
|
Salgueiro P, Lopes AS, Mendes C, Charlwood JD, Arez AP, Pinto J, Silveira H. Molecular evolution and population genetics of a Gram-negative binding protein gene in the malaria vector Anopheles gambiae (sensu lato). Parasit Vectors 2016; 9:515. [PMID: 27658383 PMCID: PMC5034674 DOI: 10.1186/s13071-016-1800-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/14/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Clarifying the role of the innate immune system of the malaria vector Anopheles gambiae is a potential way to block the development of the Plasmodium parasites. Pathogen recognition is the first step of innate immune response, where pattern recognition proteins like GNBPs play a central role. RESULTS We analysed 70 sequences of the protein coding gene GNBPB2 from two species, Anopheles gambiae (s.s.) and An. coluzzii, collected in six African countries. We detected 135 segregating sites defining 63 distinct haplotypes and 30 proteins. Mean nucleotide diversity (π) was 0.014 for both species. We found no significant genetic differentiation between species, but a significant positive correlation between genetic differentiation and geographical distance among populations. CONCLUSIONS Species status seems to contribute less for the molecular differentiation in GNBPB2 than geographical region in the African continent (West and East). Purifying selection was found to be the most common form of selection, as in many other immunity-related genes. Diversifying selection may be also operating in the GNBPB2 gene.
Collapse
Affiliation(s)
- Patrícia Salgueiro
- Global Health and Tropical Medicine Centre (GHTM), Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Ana Sofia Lopes
- Global Health and Tropical Medicine Centre (GHTM), Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Cristina Mendes
- Global Health and Tropical Medicine Centre (GHTM), Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Jacques Derek Charlwood
- Global Health and Tropical Medicine Centre (GHTM), Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Lisboa, Portugal
- London School of Hygiene and Tropical Medicine, London, UK
| | - Ana Paula Arez
- Global Health and Tropical Medicine Centre (GHTM), Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Lisboa, Portugal
| | - João Pinto
- Global Health and Tropical Medicine Centre (GHTM), Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Lisboa, Portugal
| | - Henrique Silveira
- Global Health and Tropical Medicine Centre (GHTM), Unidade de Parasitologia Médica, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Lisboa, Portugal
| |
Collapse
|
40
|
Shaw WR, Marcenac P, Childs LM, Buckee CO, Baldini F, Sawadogo SP, Dabiré RK, Diabaté A, Catteruccia F. Wolbachia infections in natural Anopheles populations affect egg laying and negatively correlate with Plasmodium development. Nat Commun 2016; 7:11772. [PMID: 27243367 PMCID: PMC4895022 DOI: 10.1038/ncomms11772] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/27/2016] [Indexed: 11/21/2022] Open
Abstract
The maternally inherited alpha-proteobacterium Wolbachia has been proposed as a tool to block transmission of devastating mosquito-borne infectious diseases like dengue and malaria. Here we study the reproductive manipulations induced by a recently identified Wolbachia strain that stably infects natural mosquito populations of a major malaria vector, Anopheles coluzzii, in Burkina Faso. We determine that these infections significantly accelerate egg laying but do not induce cytoplasmic incompatibility or sex-ratio distortion, two parasitic reproductive phenotypes that facilitate the spread of other Wolbachia strains within insect hosts. Analysis of 221 blood-fed A. coluzzii females collected from houses shows a negative correlation between the presence of Plasmodium parasites and Wolbachia infection. A mathematical model incorporating these results predicts that infection with these endosymbionts may reduce malaria prevalence in human populations. These data suggest that Wolbachia may be an important player in malaria transmission dynamics in Sub-Saharan Africa. Wolbachia bacteria infect insects and could potentially be used to control populations of malaria-transmitting mosquitoes. Here, the authors provide evidence that natural Wolbachia infections affect the rate of egg laying and are associated with reduced presence of malaria parasites in Anopheles mosquitoes.
Collapse
Affiliation(s)
- W Robert Shaw
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Perrine Marcenac
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Lauren M Childs
- Center for Communicable Disease Dynamics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Caroline O Buckee
- Center for Communicable Disease Dynamics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Francesco Baldini
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow G12 8QQ, UK
| | - Simon P Sawadogo
- Institut de Recherche en Sciences de la Santé/Centre Muraz, O1 BP 390 Bobo-Dioulasso 01, Burkina Faso
| | - Roch K Dabiré
- Institut de Recherche en Sciences de la Santé/Centre Muraz, O1 BP 390 Bobo-Dioulasso 01, Burkina Faso
| | - Abdoulaye Diabaté
- Institut de Recherche en Sciences de la Santé/Centre Muraz, O1 BP 390 Bobo-Dioulasso 01, Burkina Faso
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| |
Collapse
|
41
|
Kamareddine L, Nakhleh J, Osta MA. Functional Interaction between Apolipophorins and Complement Regulate the Mosquito Immune Response to Systemic Infections. J Innate Immun 2016; 8:314-26. [PMID: 26950600 DOI: 10.1159/000443883] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/09/2016] [Indexed: 01/14/2023] Open
Abstract
The complement-like protein thioester-containing protein 1 (TEP1) is the hallmark effector molecule against Plasmodium ookinetes in the malaria vector Anopheles gambiae. We have previously shown that the knockdown of the noncatalytic clip domain serine protease CLIPA2 increased TEP1-mediated killing rendering mosquitoes more resistant to Plasmodium, bacterial and fungal infections. Here, CLIPA2 coimmunoprecipitation from the hemolymph of Beauveria bassiana-infected mosquitoes followed by mass spectrometry and functional genetic analysis led to the identification of the Apolipophorin-II/I gene, encoding the two lipid carrier proteins Apo-I and II, as a novel negative regulator of TEP1-mediated immune response during mosquito systemic infections. Apo-II/I exhibits a similar RNAi phenotype as CLIPA2 in mosquito bioassays characterized by increased resistance to B. bassiana and Escherichia coli infections. We provide evidence that this enhanced resistance to systemic infections is TEP1 dependent. Interestingly, silencing Apo-II/I but not CLIPA2 upregulated the expression of TEP1 following systemic infections with E. coli and B. bassiana in a c-Jun N-terminal kinase pathway-dependent manner. Our results suggest that mosquito Apo-II/I plays an important immune regulatory role during systemic infections and provide novel insight into the functional interplay between lipid metabolism and immune gene regulation.
Collapse
Affiliation(s)
- Layla Kamareddine
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | | | | |
Collapse
|
42
|
Lawniczak MK. Connecting genotypes to medically relevant phenotypes in major vector mosquitoes. CURRENT OPINION IN INSECT SCIENCE 2015; 10:59-64. [PMID: 29588015 DOI: 10.1016/j.cois.2015.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 04/16/2015] [Indexed: 06/08/2023]
Abstract
Transmission of mosquito-borne human disease relies on vectors maintaining strong human host preference and continued susceptibility to disease-causing pathogens or parasites. These traits are affected by the genetics and the environments of all involved organisms, and genotypic interactions are common between parasite and vector, and between virus and vector. A recent study on Aedes host preference has exploited natural genetic variation to make great progress. Here I review our current understanding of the genetic basis of transmission-relevant traits in Anopheles and Aedes, highlighting additional research areas that would benefit from the integration of natural genetic variation.
Collapse
Affiliation(s)
- Mara Kn Lawniczak
- Wellcome Trust Sanger Institute, Malaria Programme, Hinxton CB10 1SA, United Kingdom; Imperial College London, Department of Life Sciences, London SW7 2AZ, United Kingdom.
| |
Collapse
|
43
|
Morlais I, Nsango SE, Toussile W, Abate L, Annan Z, Tchioffo MT, Cohuet A, Awono-Ambene PH, Fontenille D, Rousset F, Berry A. Plasmodium falciparum mating patterns and mosquito infectivity of natural isolates of gametocytes. PLoS One 2015; 10:e0123777. [PMID: 25875840 PMCID: PMC4397039 DOI: 10.1371/journal.pone.0123777] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 02/26/2015] [Indexed: 11/19/2022] Open
Abstract
Plasmodium falciparum infections in malaria endemic areas often harbor multiple clones of parasites. However, the transmission success of the different genotypes within the mosquito vector has remained elusive so far. The genetic diversity of malaria parasites was measured by using microsatellite markers in gametocyte isolates from 125 asymptomatic carriers. For a subset of 49 carriers, the dynamics of co-infecting genotypes was followed until their development within salivary glands. Also, individual oocysts from midguts infected with blood from 9 donors were genotyped to assess mating patterns. Multiplicity of infection (MOI) was high both in gametocyte isolates and sporozoite populations, reaching up to 10 genotypes. Gametocyte isolates with multiple genotypes gave rise to lower infection prevalence and intensity. Fluctuations of genotype number occurred during the development within the mosquito and sub-patent genotypes, not detected in gametocyte isolates, were identified in the vector salivary glands. The inbreeding coefficient Fis was positively correlated to the oocyst loads, suggesting that P. falciparum parasites use different reproductive strategies according to the genotypes present in the gametocyte isolate. The number of parasite clones within an infection affects the transmission success and the mosquito has an important role in maintaining P. falciparum genetic diversity. Our results emphasize the crucial importance of discriminating between the different genotypes within an infection when studying the A. gambiae natural resistance to P. falciparum, and the need to monitor parasite diversity in areas where malaria control interventions are implemented.
Collapse
Affiliation(s)
- Isabelle Morlais
- Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, Yaoundé, Cameroon
- Institut de Recherche pour le Développement, Montpellier, France
- * E-mail:
| | - Sandrine E. Nsango
- Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, Yaoundé, Cameroon
- Institut de Recherche pour le Développement, Montpellier, France
- Faculté de Médecine et des Sciences Pharmaceutiques, Douala, Cameroon
| | | | - Luc Abate
- Institut de Recherche pour le Développement, Montpellier, France
| | - Zeinab Annan
- Institut de Recherche pour le Développement, Montpellier, France
| | | | - Anna Cohuet
- Institut de Recherche pour le Développement, Montpellier, France
| | - Parfait H. Awono-Ambene
- Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, Yaoundé, Cameroon
| | | | | | - Antoine Berry
- Centre Hospitalier Universitaire de Toulouse, Toulouse, France
- Centre de Physiopathologie de Toulouse Purpan, Toulouse, France
| |
Collapse
|
44
|
Taszłow P, Wojda I. Changes in the hemolymph protein profiles in Galleria mellonella infected with Bacillus thuringiensis involve apolipophorin III. The effect of heat shock. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2015; 88:123-143. [PMID: 25308190 DOI: 10.1002/arch.21208] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This report concerns the effect of heat shock on host-pathogen interaction in Galleria mellonella infected with Bacillus thuringiensis. We show enhanced activity against Gram-positive bacteria in the hemolymph of larvae pre-exposed to heat shock before infection with B. thuringiensis. Heat shock influenced the protein pattern in the hemolymph of infected larvae: more peptides with a molecular weight below 10 kDa were detected in comparison with nonshocked animals. Additionally, we noticed that the amount of apolipophorin III (apoLp-III) in the hemolymph decreased transiently following infection, which was considerably higher in larvae pre-exposed to heat shock. On the other hand, its expression in the fat body showed a consequent infection-induced decline, observed equally in shocked and nonshocked animals. This suggests that the amount of apoLp-III in the hemolymph of G. mellonella larvae is regulated at multiple levels. We also report that this protein is more resistant to degradation in the hemolymph of larvae pre-exposed to heat shock in comparison to nonshocked larvae. Two-dimensional analysis revealed the presence of three isoforms of apoLp-III, all susceptible to proteolytic degradation. However, one of them was the most abundant, both in the protease-treated and untreated hemolymph. Taking into consideration that, in general, apoLp-III has a stimulative effect on different immune-related hemolymph proteins and peptides, the reported findings bring us closer to understanding the effect of heat shock on the resistance of G. mellonella to infection.
Collapse
Affiliation(s)
- Paulina Taszłow
- Department of Immunobiology, Faculty of Biology and Biotechnology, Institute of Biology and Biochemistry, Maria Curie-Sklodowska University, Lublin, Poland
| | | |
Collapse
|
45
|
Da DF, Churcher TS, Yerbanga RS, Yaméogo B, Sangaré I, Ouedraogo JB, Sinden RE, Blagborough AM, Cohuet A. Experimental study of the relationship between Plasmodium gametocyte density and infection success in mosquitoes; implications for the evaluation of malaria transmission-reducing interventions. Exp Parasitol 2014; 149:74-83. [PMID: 25541384 DOI: 10.1016/j.exppara.2014.12.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 12/10/2014] [Accepted: 12/15/2014] [Indexed: 11/29/2022]
Abstract
The evaluation of transmission reducing interventions (TRI) to control malaria widely uses membrane feeding assays. In such assays, the intensity of Plasmodium infection in the vector might affect the measured efficacy of the candidates to block transmission. Gametocyte density in the host blood is a determinant of the infection success in the mosquito, however, uncertain estimates of parasite densities and intrinsic characteristics of the infected blood can induce variability. To reduce this variation, a feasible method is to dilute infectious blood samples. We describe the effect of diluting samples of Plasmodium-containing blood samples to allow accurate relative measures of gametocyte densities and their impact on mosquito infectivity and TRI efficacy. Natural Plasmodium falciparum samples were diluted to generate a wide range of parasite densities, and fed to Anopheles coluzzii mosquitoes. This was compared with parallel dilutions conducted on Plasmodium berghei infections. We examined how blood dilution influences the observed blocking activity of anti-Pbs28 monoclonal antibody using the P. berghei/Anopheles stephensi system. In the natural species combination P. falciparum/An. coluzzii, blood dilution using heat-inactivated, infected blood as diluents, revealed positive near linear relationships, between gametocyte densities and oocyst loads in the range tested. A similar relationship was observed in the P. berghei/An. stephensi system when using a similar dilution method. In contrast, diluting infected mice blood with fresh uninfected blood dramatically increases the infectiousness. This suggests that highly infected mice blood contains inhibitory factors or reduced blood moieties, which impede infection and may in turn, lead to misinterpretation when comparing individual TRI evaluation assays. In the lab system, the transmission blocking activity of an antibody specific for Pbs28 was confirmed to be density-dependent. This highlights the need to carefully interpret evaluations of TRI candidates, regarding gametocyte densities in the P. berghei/An. stephensi system.
Collapse
Affiliation(s)
- Dari F Da
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso; Institut de Recherche pour le Développement, unité MIVEGEC (UM1-UM2-CNRS 5290-IRD 224), 911 avenue Agropolis, Montpellier Cedex 5 34394, France
| | - Thomas S Churcher
- Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Rakiswendé S Yerbanga
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso
| | - Bienvenue Yaméogo
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso
| | - Ibrahim Sangaré
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso; Institut de Recherche pour le Développement, unité MIVEGEC (UM1-UM2-CNRS 5290-IRD 224), 911 avenue Agropolis, Montpellier Cedex 5 34394, France
| | - Jean Bosco Ouedraogo
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso
| | - Robert E Sinden
- Division of Cell and Molecular Biology, Imperial College London, London, United Kingdom; The Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Andrew M Blagborough
- Division of Cell and Molecular Biology, Imperial College London, London, United Kingdom
| | - Anna Cohuet
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso; Institut de Recherche pour le Développement, unité MIVEGEC (UM1-UM2-CNRS 5290-IRD 224), 911 avenue Agropolis, Montpellier Cedex 5 34394, France.
| |
Collapse
|
46
|
Marie A, Holzmuller P, Tchioffo MT, Rossignol M, Demettre E, Seveno M, Corbel V, Awono-Ambéné P, Morlais I, Remoue F, Cornelie S. Anopheles gambiae salivary protein expression modulated by wild Plasmodium falciparum infection: highlighting of new antigenic peptides as candidates of An. gambiae bites. Parasit Vectors 2014; 7:599. [PMID: 25526764 PMCID: PMC4287575 DOI: 10.1186/s13071-014-0599-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 12/10/2014] [Indexed: 12/24/2022] Open
Abstract
Background Malaria is the major parasitic disease worldwide caused by Plasmodium infection. The objective of integrated malaria control programs is to decrease malaria transmission, which needs specific tools to be accurately assessed. In areas where the transmission is low or has been substantially reduced, new complementary tools have to be developed to improve surveillance. A recent approach, based on the human antibody response to Anopheles salivary proteins, has been shown to be efficient in evaluating human exposure to Anopheles bites. The aim of the present study was to identify new An. gambiae salivary proteins as potential candidate biomarkers of human exposure to P. falciparum-infective bites. Methods Experimental infections of An. gambiae by wild P. falciparum were carried out in semi-field conditions. Then a proteomic approach, combining 2D-DIGE and mass spectrometry, was used to identify the overexpressed salivary proteins in infected salivary glands compared to uninfected An. gambiae controls. Subsequently, a peptide design of each potential candidate was performed in silico and their antigenicity was tested by an epitope-mapping technique using blood from individuals exposed to Anopheles bites. Results Five salivary proteins (gSG6, gSG1b, TRIO, SG5 and long form D7) were overexpressed in the infected salivary glands. Eighteen peptides were designed from these proteins and were found antigenic in children exposed to the Anopheles bites. Moreover, the results showed that the presence of wild P. falciparum in salivary glands modulates the expression of several salivary proteins and also appeared to induce post-translational modifications. Conclusions This study is, to our knowledge, the first that compares the sialome of An. gambiae both infected and not infected by wild P. falciparum, making it possible to mimic the natural conditions of infection. This is a first step toward a better understanding of the close interactions between the parasite and the salivary gland of mosquitoes. In addition, these results open the way to define biomarkers of infective bites of Anopheles, which could, in the future, improve the estimation of malaria transmission and the evaluation of malaria vector control tools. Electronic supplementary material The online version of this article (doi:10.1186/s13071-014-0599-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexandra Marie
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France.
| | - Philippe Holzmuller
- CIRAD Département Systèmes Biologiques BIOS UMR 15 CMAEE "Contrôle des Maladies Exotiques et Emergentes", Campus International de Baillarguet, TA A-15/G, Montpellier cedex 5, 34398, France.
| | - Majoline T Tchioffo
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France.
| | - Marie Rossignol
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France.
| | - Edith Demettre
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, INSERM U661, UM1, UM2, Plate-forme de Protéomique Fonctionnelle CNRS UMS BioCampus 3426, Montpellier, 34094, France.
| | - Martial Seveno
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, INSERM U661, UM1, UM2, Plate-forme de Protéomique Fonctionnelle CNRS UMS BioCampus 3426, Montpellier, 34094, France.
| | - Vincent Corbel
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France. .,Department of Entomology, Faculty of Agriculture, Kasetsart University, 50 Ngam Wong Wan Rd, Ladyaow Chatuchak, Bangkok, 10900, Thailand.
| | - Parfait Awono-Ambéné
- Laboratoire de Recherche sur le Paludisme, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale (OCEAC), Yaoundé, BP 288, Cameroun.
| | - Isabelle Morlais
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France. .,Laboratoire de Recherche sur le Paludisme, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale (OCEAC), Yaoundé, BP 288, Cameroun.
| | - Franck Remoue
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France.
| | - Sylvie Cornelie
- MIVEGEC (UMR IRD224 CNRS 5290 UM1-UM2), Institut de Recherche pour le développement (IRD), 911 avenue Agropolis, Montpellier cedex 5, 34394, France. .,MIVEGEC- IRD- CREC, Cotonou, 01 BP4414 RP, Bénin.
| |
Collapse
|
47
|
Schlegelmilch T, Vlachou D. Cell biological analysis of mosquito midgut invasion: the defensive role of the actin-based ookinete hood. Pathog Glob Health 2014; 107:480-92. [PMID: 24428832 PMCID: PMC4073529 DOI: 10.1179/2047772413z.000000000180] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Successful completion of the Plasmodium lifecycle in the mosquito vector is critical for malaria transmission. It has been documented that the fate of Plasmodium in the mosquito ultimately depends on a fine interplay of molecular mosquito factors that act as parasite agonists and antagonists. Here we investigate whether the cellular responses of the invaded midgut epithelium can also determine the parasite fate and development. We show that the parasite hood, an actin-rich structure formed around the ookinete as it exits the epithelium, is a local epithelial defence reaction observed around 60% of invading parasites. The hood co-localizes with WASP, a promoter of actin filament nucleation, suggesting that it is an active reaction of the invaded cell against invading parasites. Importantly, depletion of WASP by RNAi leads to a significant reduction in hood formation, which is consistent with the previously documented role of this gene as a potent parasite antagonist. Indeed, in mosquitoes that are either genetically selected or manipulated by RNAi to be refractory to Plasmodium, most dead parasites exhibit an actin hood. In these mosquitoes, invading ookinetes are killed by lysis or melanization while exiting the midgut epithelium. Silencing WASP in these mosquitoes inhibits the formation of the hood and allows many parasites to develop to oocysts. These data in conjunction with fine microscopic observations suggest that the presence of the hood is linked to ookinete killing through lysis.
Collapse
|
48
|
Severo MS, Levashina EA. Mosquito defenses against Plasmodium parasites. CURRENT OPINION IN INSECT SCIENCE 2014; 3:30-36. [PMID: 32846668 DOI: 10.1016/j.cois.2014.07.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/28/2014] [Accepted: 07/30/2014] [Indexed: 06/11/2023]
Abstract
Malaria, the human infectious disease caused by Plasmodium parasites, is transmitted by the bite of the mosquito Anopheles gambiae. Mosquitoes actively detect Plasmodium and mount efficient responses that eliminate the majority of invading parasites. Such responses include hemocyte-mediated defenses, activation of the complement-like system, melanization, and immune signaling cascades. This review aims to summarize our current knowledge of the mosquito immune responses to Plasmodium and to highlight the remaining gaps in our understanding of these events.
Collapse
Affiliation(s)
- Maiara S Severo
- Vector Biology Unit, Max-Planck-Institut für Infektionsbiologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Elena A Levashina
- Vector Biology Unit, Max-Planck-Institut für Infektionsbiologie, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
49
|
Suzuki Y, Niu G, Hughes GL, Rasgon JL. A viral over-expression system for the major malaria mosquito Anopheles gambiae. Sci Rep 2014; 4:5127. [PMID: 24875042 PMCID: PMC4038844 DOI: 10.1038/srep05127] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/14/2014] [Indexed: 11/08/2022] Open
Abstract
Understanding pathogen/mosquito interactions is essential for developing novel strategies to control mosquito-borne diseases. Technical advances in reverse-genetics, such as RNA interference (RNAi), have facilitated elucidation of components of the mosquito immune system that are antagonistic to pathogen development, and host proteins essential for parasite development. Forward genetic approaches, however, are limited to generation of transgenic insects, and while powerful, mosquito transgenesis is a resource- and time-intensive technique that is not broadly available to most laboratories. The ability to easily "over-express" genes would enhance molecular studies in vector biology and expedite elucidation of pathogen-refractory genes without the need to make transgenic insects. We developed and characterized an efficient Anopheles gambiae densovirus (AgDNV) over-expression system for the major malaria vector Anopheles gambiae. High-levels of gene expression were detected at 3 days post-infection and increased over time, suggesting this is an effective system for gene induction. Strong expression was observed in the fat body and ovaries. We validated multiple short promoters for gene induction studies. Finally, we developed a polycistronic system to simultaneously express multiple genes of interest. This AgDNV-based toolset allows for consistent transduction of genes of interest and will be a powerful molecular tool for research in Anopheles gambiae mosquitoes.
Collapse
Affiliation(s)
- Yasutsugu Suzuki
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| | - Guodong Niu
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
- Current address: Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, 73019, United States of America
| | - Grant L. Hughes
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| | - Jason L. Rasgon
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| |
Collapse
|
50
|
Alout H, Yameogo B, Djogbénou LS, Chandre F, Dabiré RK, Corbel V, Cohuet A. Interplay between Plasmodium infection and resistance to insecticides in vector mosquitoes. J Infect Dis 2014; 210:1464-70. [PMID: 24829465 DOI: 10.1093/infdis/jiu276] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Despite its epidemiological importance, the impact of insecticide resistance on vector-parasite interactions and malaria transmission is poorly understood. Here, we explored the impact of Plasmodium infection on the level of insecticide resistance to dichlorodiphenyltrichloroethane (DDT) in field-caught Anopheles gambiae sensu stricto homozygous for the kdr mutation. Results showed that kdr homozygous mosquitoes that fed on infectious blood were more susceptible to DDT than mosquitoes that fed on noninfectious blood during both ookinete development (day 1 after the blood meal) and oocyst maturation (day 7 after the blood meal) but not during sporozoite invasion of the salivary glands. Plasmodium falciparum infection seemed to impose a fitness cost on mosquitoes by reducing the ability of kdr homozygous A. gambiae sensu stricto to survive exposure to DDT. These results suggest an interaction between Plasmodium infection and the insecticide susceptibility of mosquitoes carrying insecticide-resistant alleles. We discuss this finding in relation to vector control efficacy.
Collapse
Affiliation(s)
- Haoues Alout
- Institut de Recherche pour le Développement, Maladies Infectieuses et Vecteurs, Ecologie, Génétique, Evolution et Contrôle, UM1-UM2-CNRS5290-IRD 224, Montpellier, France Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Bienvenue Yameogo
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | | | - Fabrice Chandre
- Institut de Recherche pour le Développement, Maladies Infectieuses et Vecteurs, Ecologie, Génétique, Evolution et Contrôle, UM1-UM2-CNRS5290-IRD 224, Montpellier, France
| | | | - Vincent Corbel
- Institut de Recherche pour le Développement, Maladies Infectieuses et Vecteurs, Ecologie, Génétique, Evolution et Contrôle, UM1-UM2-CNRS5290-IRD 224, Montpellier, France Department of Entomology, Faculty of Agriculture, Kasetsart University, Bangkok, Thailand
| | - Anna Cohuet
- Institut de Recherche pour le Développement, Maladies Infectieuses et Vecteurs, Ecologie, Génétique, Evolution et Contrôle, UM1-UM2-CNRS5290-IRD 224, Montpellier, France Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| |
Collapse
|