1
|
Seck F, Diop MF, Mané K, Diallo A, Dieng I, Namountougou M, Diabate A, Amambua-Ngwa A, Dia I, Assogba BS. Reduced Genetic Diversity of Key Fertility and Vector Competency Related Genes in Anopheles gambiae s.l. Across Sub-Saharan Africa. Genes (Basel) 2025; 16:543. [PMID: 40428366 PMCID: PMC12111087 DOI: 10.3390/genes16050543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Insecticide resistance challenges the vector control efforts towards malaria elimination and proving the development of complementary tools. Targeting the genes that are involved in mosquito fertility and susceptibility to Plasmodium with small molecule inhibitors has been a promising alternative to curb the vector population and drive the transmission down. However, such an approach would require a comprehensive knowledge of the genetic diversity of the targeted genes to ensure the broad efficacy of new tools across the natural vector populations. METHODS Four fertility and parasite susceptibility genes were identified from a systematic review of the literature. The Single Nucleotide Polymorphisms (SNPs) found within the regions spanned by these four genes, genotyped across 2784 wild-caught Anopheles gambiae s.l. from 19 sub-Saharan African (SSA) countries, were extracted from the whole genome SNP data of the Ag1000G project (Ag3.0). The population genetic analysis on gene-specific data included the determination of the population structure, estimation of the differentiation level between the populations, evaluation of the linkage between the non-synonymous SNPs (nsSNPs), and a few statistical tests. RESULTS As potential targets for small molecule inhibitors to reduce malaria transmission, our set of four genes associated with Anopheles fertility and their susceptibility to Plasmodium comprises the mating-induced stimulator of oogenesis protein (MISO, AGAP002620), Vitellogenin (Vg, AGAP004203), Lipophorin (Lp, AGAP001826), and Haem-peroxidase 15 (HPX15, AGAP013327). The analyses performed on these potential targets of small inhibitor molecules revealed that the genes are conserved within SSA populations of An. gambiae s.l. The overall low Fst values and low clustering of principal component analysis between species indicated low genetic differentiation at all the genes (MISO, Vg, Lp and HPX15). The low nucleotide diversity (>0.10), negative Tajima's D values, and heterozygosity analysis provided ecological insights into the purifying selection that acts to remove deleterious mutations, maintaining genetic diversity at low levels within the populations. None of MISO nsSNPs were identified in linkage disequilibrium, whereas a few weakly linked nsSNPs with ambiguous haplotyping were detected at other genes. CONCLUSIONS This integrated finding on the genetic features of major malaria vectors' biological factors across natural populations offer new insights for developing sustainable malaria control tools. These loci were reasonably conserved, allowing for the design of effective targeting with small molecule inhibitors towards controlling vector populations and lowering global malaria transmission.
Collapse
Affiliation(s)
- Fatoumata Seck
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul P.O. Box 273, The Gambia; (F.S.); (M.F.D.); (K.M.); (A.A.-N.)
- Institut Pasteur de Dakar, Dakar 220, Senegal (I.D.); (I.D.)
| | - Mouhamadou Fadel Diop
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul P.O. Box 273, The Gambia; (F.S.); (M.F.D.); (K.M.); (A.A.-N.)
| | - Karim Mané
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul P.O. Box 273, The Gambia; (F.S.); (M.F.D.); (K.M.); (A.A.-N.)
| | - Amadou Diallo
- Institut Pasteur de Dakar, Dakar 220, Senegal (I.D.); (I.D.)
| | - Idrissa Dieng
- Institut Pasteur de Dakar, Dakar 220, Senegal (I.D.); (I.D.)
| | - Moussa Namountougou
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso 01 BP 545, Burkina Faso; (M.N.); (A.D.)
| | - Abdoulaye Diabate
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso 01 BP 545, Burkina Faso; (M.N.); (A.D.)
| | - Alfred Amambua-Ngwa
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul P.O. Box 273, The Gambia; (F.S.); (M.F.D.); (K.M.); (A.A.-N.)
| | - Ibrahima Dia
- Institut Pasteur de Dakar, Dakar 220, Senegal (I.D.); (I.D.)
| | - Benoit Sessinou Assogba
- Medical Research Council Unit, The Gambia at the London School of Hygiene and Tropical Medicine, Banjul P.O. Box 273, The Gambia; (F.S.); (M.F.D.); (K.M.); (A.A.-N.)
| |
Collapse
|
2
|
Barletta ABF, Barillas-Mury C, Molina-Cruz A. Mosquito immune responses to Plasmodium parasites that limit malaria transmission. Cell Mol Life Sci 2025; 82:143. [PMID: 40192851 PMCID: PMC11977068 DOI: 10.1007/s00018-025-05667-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 12/05/2024] [Accepted: 03/17/2025] [Indexed: 04/10/2025]
Abstract
The mosquito immune system is a major barrier that malaria parasites must overcome for their successful development and disease transmission. At each developmental stage in the vector, Plasmodium parasites can be potentially targeted by the mosquito innate immune responses, which involve epithelial, humoral, and cellular components. The immune response to Plasmodium ookinetes can be powerful and some of the underlying effector mechanisms are well characterized. However, the defense responses to oocysts and sporozoites appear to be less effective and are less well understood. Plasmodium parasites are under constant pressure to avoid elimination by evading and/or manipulating the mosquito immune system. Understanding the intricate interaction between Plasmodium parasites and the mosquito immune system is fundamental to understand the epidemiology of malaria transmission and to devise innovative control strategies.
Collapse
Affiliation(s)
- Ana Beatriz F Barletta
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA.
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Alvaro Molina-Cruz
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA.
| |
Collapse
|
3
|
Qin X, Li J, Zhu F, Zhang J. Thioester-containing protein TEP15 promotes malaria parasite development in mosquitoes through negative regulation of melanization. Parasit Vectors 2025; 18:124. [PMID: 40170182 PMCID: PMC11963550 DOI: 10.1186/s13071-025-06772-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Thioester-containing proteins (TEPs) serve as crucial effectors and regulatory components within the innate immune system of mosquitoes. Despite their significance, the mechanisms by which TEPs exert negative regulation on the immune response in mosquitoes remain inadequately understood. This study aims to elucidate the role of TEPs in the negative regulation of melanization, thereby advancing our comprehension of their regulatory function in the immune response. METHODS We infected female Anopheles stephensi mosquitoes with Plasmodium yoelii by allowing them to feed on pre-infected female Kunming mice. Western blot, quantitative polymerase chain reaction, differential gene expression analyses, and gene silencing were then conducted. Student's t-test was used to analyze continuous variables, with statistical significance defined as p < 0.05. RESULTS A. stephensi TEP15 (AsTEP15) negatively regulated mosquitos' innate immunity and promoted Plasmodium development. AsTEP15 knockdown induced mosquito resistance to malaria parasite melanization during the oocyst stage and significantly reduced sporozoite numbers. Further analysis showed that AsTEP15 mainly negatively affects the TEP1 and immune deficiency (IMD) pathway, thereby inhibiting melanization. CONCLUSIONS We describe a mosquito TEP that negatively regulates immunity, further enriching the functional diversity of TEP family members. In addition, our results suggest that oocysts may exploit TEPs to escape or inhibit mosquito immunity, highlighting potential targets for blocking malaria transmission.
Collapse
Affiliation(s)
- Xin Qin
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| | - Jianyong Li
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| | - Feng Zhu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China.
| | - Jian Zhang
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China.
| |
Collapse
|
4
|
Rhodes VL, Waterhouse RM, Michel K. The molecular toll pathway repertoire in anopheline mosquitoes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105287. [PMID: 39522894 PMCID: PMC11717629 DOI: 10.1016/j.dci.2024.105287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Innate immunity in mosquitoes has received much attention due to its potential impact on vector competence for vector-borne disease pathogens, including malaria parasites. The nuclear factor (NF)-κB-dependent Toll pathway is a major regulator of innate immunity in insects. In mosquitoes, this pathway controls transcription of the majority of the known canonical humoral immune effectors, mediates anti-bacterial, anti-fungal and anti-viral immune responses, and contributes to malaria parasite killing. However, besides initial gene annotation of putative Toll pathway members and genetic analysis of the contribution of few key components to immunity, the molecular make-up and function of the Toll pathway in mosquitoes is largely unexplored. To facilitate functional analyses of the Toll pathway in mosquitoes, we report here manually annotated and refined gene models of Toll-like receptors and all putative components of the intracellular signal transduction cascade across 19 anopheline genomes, and in two culicine genomes. In addition, based on phylogenetic analyses, we identified differing levels of evolutionary constraint across the intracellular Toll pathway members, and identified a recent radiation of TOLL1/5 within the Anopheles gambiae complex. Together, this study provides insight into the evolution of TLRs and the putative members of the intracellular signal transduction cascade within the genus Anopheles.
Collapse
Affiliation(s)
- Victoria L Rhodes
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA; Biology Department, Missouri Southern University, Joplin, MO 64801, USA
| | | | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
5
|
Jalovecka M, Malandrin L, Urbanova V, Mahmood S, Snebergerova P, Peklanska M, Pavlasova V, Sima R, Kopacek P, Perner J, Hajdusek O. Activation of the tick Toll pathway to control infection of Ixodes ricinus by the apicomplexan parasite Babesia microti. PLoS Pathog 2024; 20:e1012743. [PMID: 39680508 DOI: 10.1371/journal.ppat.1012743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
The vector competence of blood-feeding arthropods is influenced by the interaction between pathogens and the immune system of the vector. The Toll and IMD (immune deficiency) signaling pathways play a key role in the regulation of innate immunity in both the Drosophila model and blood-feeding insects. However, in ticks (chelicerates), immune determination for pathogen acquisition and transmission has not yet been fully explored. Here, we have mapped homologs of insect Toll and IMD pathways in the European tick Ixodes ricinus, an important vector of human and animal diseases. We show that most genes of the Toll pathway are well conserved, whereas the IMD pathway has been greatly reduced. We therefore investigated the functions of the individual components of the tick Toll pathway and found that, unlike in Drosophila, it was specifically activated by Gram-negative bacteria. The activation of pathway induced the expression of defensin (defIR), the first identified downstream effector gene of the tick Toll pathway. Borrelia, an atypical bacterium and causative agent of Lyme borreliosis, bypassed Toll-mediated recognition in I. ricinus and also resisted systemic effector molecules when the Toll pathway was activated by silencing its repressor cactus via RNA interference. Babesia, an apicomplexan parasite, also avoided Toll-mediated recognition. Strikingly, unlike Borrelia, the number of Babesia parasites reaching the salivary glands during tick infection was significantly reduced by knocking down cactus. The simultaneous silencing of cactus and dorsal resulted in greater infections and underscored the importance of tick immunity in regulating parasite infections in these important disease vectors.
Collapse
Affiliation(s)
- Marie Jalovecka
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Laurence Malandrin
- Nantes-Atlantic National College of Veterinary Medicine (ONIRIS), UMR1300 BiOEPAR, National Research Institute for Agriculture, Food and the Environment (INRAE), Nantes, France
| | - Veronika Urbanova
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Sazzad Mahmood
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Pavla Snebergerova
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Miriama Peklanska
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Veronika Pavlasova
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Radek Sima
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Petr Kopacek
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Jan Perner
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| | - Ondrej Hajdusek
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czech Republic
| |
Collapse
|
6
|
Rhodes VL, Waterhouse RM, Michel K. The Molecular Toll Pathway Repertoire in Anopheline Mosquitoes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612760. [PMID: 39345384 PMCID: PMC11429875 DOI: 10.1101/2024.09.12.612760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Innate immunity in mosquitoes has received much attention due to its potential impact on vector competence for vector-borne disease pathogens, including malaria parasites. The nuclear factor (NF)-κB-dependent Toll pathway is a major regulator of innate immunity in insects. In mosquitoes, this pathway controls transcription of the majority of the known canonical humoral immune effectors, mediates anti-bacterial, anti-fungal and anti-viral immune responses, and contributes to malaria parasite killing. However, besides initial gene annotation of putative Toll pathway members and genetic analysis of the contribution of few key components to immunity, the molecular make-up and function of the Toll pathway in mosquitoes is largely unexplored. To facilitate functional analyses of the Toll pathway in mosquitoes, we report here manually annotated and refined gene models of Toll-like receptors and all putative components of the intracellular signal transduction cascade across 19 anopheline genomes, and in two culicine genomes. In addition, based on phylogenetic analyses, we identified differing levels of evolutionary constraint across the intracellular Toll pathway members, and identified a recent radiation of TOLL1/5 within the An. gambiae complex. Together, this study provides insight into the evolution of TLRs and the putative members of the intracellular signal transduction cascade within the genus Anopheles.
Collapse
Affiliation(s)
- Victoria L. Rhodes
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
- Biology Department, Missouri Southern University, Joplin, MO 64801, USA
| | | | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
7
|
Mousavi Shafi ZS, Firouz ZM, Pirahmadi S. Gene expression analysis of Anopheles Meigen, 1818 (Diptera: Culicidae) innate immunity after Plasmodium Marchiafava & Celli, 1885 (Apicomplexa) infection: Toward developing new malaria control strategies. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 123:105650. [PMID: 39089500 DOI: 10.1016/j.meegid.2024.105650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/27/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Despite the critical role of the Anopheles innate immune system in defending against Plasmodium infection, there is still limited information about the key immune mechanisms in Anopheles. This review assesses recent findings on the expression characteristics of immune-related genes in Anopheles following exposure to Plasmodium. A literature review, unrestricted by publication date, was conducted to evaluate immune-related gene expression in different organs of Anopheles after Plasmodium infection. Mosquito immune responses in the midgut are essential for reducing parasite populations. Additionally, innate immune responses in the salivary glands and hemocytes circulating in the hemocoel play key roles in defense against the parasite. Transcriptomic analysis of the mosquito's innate immune response to Plasmodium infection provides valuable insights into key immune mechanisms in mosquito defense. A deeper understanding of immune mechanisms in different organs of Anopheles following Plasmodium infection will aid in discovering critical targets for designing novel control strategies.
Collapse
Affiliation(s)
- Zahra Sadat Mousavi Shafi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Zeinab Mohammadi Firouz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
8
|
Ratcliffe NA, Mello CB, Castro HC, Dyson P, Figueiredo M. Immune Reactions of Vector Insects to Parasites and Pathogens. Microorganisms 2024; 12:568. [PMID: 38543619 PMCID: PMC10974449 DOI: 10.3390/microorganisms12030568] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 11/12/2024] Open
Abstract
This overview initially describes insect immune reactions and then brings together present knowledge of the interactions of vector insects with their invading parasites and pathogens. It is a way of introducing this Special Issue with subsequent papers presenting the latest details of these interactions in each particular group of vectors. Hopefully, this paper will fill a void in the literature since brief descriptions of vector immunity have now been brought together in one publication and could form a starting point for those interested and new to this important area. Descriptions are given on the immune reactions of mosquitoes, blackflies, sandflies, tsetse flies, lice, fleas and triatomine bugs. Cellular and humoral defences are described separately but emphasis is made on the co-operation of these processes in the completed immune response. The paper also emphasises the need for great care in extracting haemocytes for subsequent study as appreciation of their fragile nature is often overlooked with the non-sterile media, smearing techniques and excessive centrifugation sometimes used. The potential vital role of eicosanoids in the instigation of many of the immune reactions described is also discussed. Finally, the priming of the immune system, mainly in mosquitoes, is considered and one possible mechanism is presented.
Collapse
Affiliation(s)
- Norman Arthur Ratcliffe
- Department of Biosciences, Swansea University, Singleton Park, Swansea SA28PP, UK
- Biology Institute, Universidade Federal Fluminense, Niterói 24210-130, RJ, Brazil; (C.B.M.); (H.C.C.)
| | - Cicero Brasileiro Mello
- Biology Institute, Universidade Federal Fluminense, Niterói 24210-130, RJ, Brazil; (C.B.M.); (H.C.C.)
| | - Helena Carla Castro
- Biology Institute, Universidade Federal Fluminense, Niterói 24210-130, RJ, Brazil; (C.B.M.); (H.C.C.)
| | - Paul Dyson
- Institute of Life Science, Medical School, Swansea University, Singleton Park, Swansea SA28PP, UK; (P.D.); (M.F.)
| | - Marcela Figueiredo
- Institute of Life Science, Medical School, Swansea University, Singleton Park, Swansea SA28PP, UK; (P.D.); (M.F.)
| |
Collapse
|
9
|
Li M, Zhou Y, Cheng J, Wang Y, Lan C, Shen Y. Response of the mosquito immune system and symbiotic bacteria to pathogen infection. Parasit Vectors 2024; 17:69. [PMID: 38368353 PMCID: PMC10874582 DOI: 10.1186/s13071-024-06161-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/24/2024] [Indexed: 02/19/2024] Open
Abstract
Mosquitoes are the deadliest animal in the word, transmitting a variety of insect-borne infectious diseases, such as malaria, dengue fever, yellow fever, and Zika, causing more deaths than any other vector-borne pathogen. Moreover, in the absence of effective drugs and vaccines to prevent and treat insect-borne diseases, mosquito control is particularly important as the primary measure. In recent decades, due to the gradual increase in mosquito resistance, increasing attention has fallen on the mechanisms and effects associated with pathogen infection. This review provides an overview of mosquito innate immune mechanisms in terms of physical and physiological barriers, pattern recognition receptors, signalling pathways, and cellular and humoral immunity, as well as the antipathogenic effects of mosquito symbiotic bacteria. This review contributes to an in-depth understanding of the interaction process between mosquitoes and pathogens and provides a theoretical basis for biological defence strategies against mosquito-borne infectious diseases.
Collapse
Affiliation(s)
- Manjin Li
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yang Zhou
- Nanjing Medical University, Nanjing, 211166, China
| | - Jin Cheng
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yiqing Wang
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Cejie Lan
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
| | - Yuan Shen
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
- Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
10
|
Garrigós M, Ylla G, Martínez-de la Puente J, Figuerola J, Ruiz-López MJ. Two avian Plasmodium species trigger different transcriptional responses on their vector Culex pipiens. Mol Ecol 2023:e17240. [PMID: 38108558 DOI: 10.1111/mec.17240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 11/01/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
Malaria is a mosquito-borne disease caused by protozoans of the genus Plasmodium that affects both humans and wildlife. The fitness consequences of infections by avian malaria are well known in birds, however, little information exists on its impact on mosquitoes. Here we study how Culex pipiens mosquitoes transcriptionally respond to infection by two different Plasmodium species, P. relictum and P. cathemerium, differing in their virulence (mortality rate) and transmissibility (parasite presence in exposed mosquitoes' saliva). We studied the mosquito response to the infection at three critical stages of parasite development: the formation of ookinetes at 24 h post-infection (hpi), the release of sporozoites into the hemocoel at 10 days post-infection (dpi), and the storage of sporozoites in the salivary glands at 21 dpi. For each time point, we characterized the gene expression of mosquitoes infected with each P. relictum and P. cathemerium and mosquitoes fed on an uninfected bird and, subsequently, compared their transcriptomic responses. Differential gene expression analysis showed that most transcriptomic changes occurred during the early infection stage (24 hpi), especially when comparing P. relictum and P. cathemerium-infected mosquitoes. Differentially expressed genes in mosquitoes infected with each species were related mainly to the metabolism of the immune response, trypsin, and other serine-proteases. We conclude that these differences in response may partly play a role in the differential virulence and transmissibility previously observed between P. relictum and P. cathemerium in Cx. pipiens.
Collapse
Affiliation(s)
- Marta Garrigós
- Department of Parasitology, University of Granada, Granada, Spain
| | - Guillem Ylla
- Bioinformatics and Genome Biology Lab, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Josué Martínez-de la Puente
- Department of Parasitology, University of Granada, Granada, Spain
- CIBER Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
| | - Jordi Figuerola
- CIBER Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
- Department of Wetland Ecology, Estación Biológica de Doñana, CSIC, Sevilla, Spain
| | - María José Ruiz-López
- CIBER Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
- Department of Wetland Ecology, Estación Biológica de Doñana, CSIC, Sevilla, Spain
| |
Collapse
|
11
|
Alves e Silva TL, Canepa GE, Sweeney B, Hessab Alvarenga P, Zhao M, Vega-Rodríguez J, Molina-Cruz A, Barillas-Mury C. The heat shock protein Hsc70-3 mediates an anti-apoptotic response critical for Plasmodium evasion of Anopheles gambiae immunity. Microbiol Spectr 2023; 11:e0094023. [PMID: 37982627 PMCID: PMC10715144 DOI: 10.1128/spectrum.00940-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 10/06/2023] [Indexed: 11/21/2023] Open
Abstract
IMPORTANCE Malaria transmission by Anopheles gambiae mosquitoes is very effective, in part because the parasite expresses a surface protein called Pfs47 that allows it to evade the mosquito immune system. Here we investigate how this protein changes the response of mosquito midgut epithelial cells to invasion by the parasite. Pfs47 is known to interact with P47Rec, a mosquito midgut receptor. We found that Pf47Rec inhibits caspase-mediated apoptosis by interacting with the Hsc70-3. This disrupts nitration of midgut epithelial cells invaded by the parasite and the release of hemocyte-derived microvesicles, which are critical for effective activation of the mosquito complement system that eliminates the parasite.
Collapse
Affiliation(s)
- Thiago Luiz Alves e Silva
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Gaspar E. Canepa
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Brendan Sweeney
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Patricia Hessab Alvarenga
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Ming Zhao
- Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Alvaro Molina-Cruz
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
12
|
Hernandez-Caballero I, Hellgren O, Garcia-Longoria Batanete L. Genomic advances in the study of the mosquito vector during avian malaria infection. Parasitology 2023; 150:1330-1339. [PMID: 37614176 PMCID: PMC10941221 DOI: 10.1017/s0031182023000756] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023]
Abstract
Invertebrate host–parasite associations are one of the keystones in order to understand vector-borne diseases. The study of these specific interactions provides information not only about how the vector is affected by the parasite at the gene-expression level, but might also reveal mosquito strategies for blocking the transmission of the parasites. A very well-known vector for human malaria is Anopheles gambiae. This mosquito species has been the main focus for genomics studies determining essential key genes and pathways over the course of a malaria infection. However, to-date there is an important knowledge gap concerning other non-mammophilic mosquito species, for example some species from the Culex genera which may transmit avian malaria but also zoonotic pathogens such as West Nile virus. From an evolutionary perspective, these 2 mosquito genera diverged 170 million years ago, hence allowing studies in both species determining evolutionary conserved genes essential during malaria infections, which in turn might help to find key genes for blocking malaria cycle inside the mosquito. Here, we extensively review the current knowledge on key genes and pathways expressed in Anopheles over the course of malaria infections and highlight the importance of conducting genomic investigations for detecting pathways in Culex mosquitoes linked to infection of avian malaria. By pooling this information, we underline the need to increase genomic studies in mosquito–parasite associations, such as the one in Culex–Plasmodium, that can provide a better understanding of the infection dynamics in wildlife and reduce the negative impact on ecosystems.
Collapse
Affiliation(s)
- Irene Hernandez-Caballero
- Department of Anatomy, Cellular Biology and Zoology, University of Extremadura, E-06071 Badajoz, Spain
| | - Olof Hellgren
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Sölvegatan 37, SE-22362, Sweden
| | | |
Collapse
|
13
|
Mehmood N, Hassan A, Zhong X, Zhu Y, Ouyang G, Huang Q. Entomopathogenic fungal infection following immune gene silencing decreased behavioral and physiological fitness in Aedes aegypti mosquitoes. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 195:105535. [PMID: 37666588 DOI: 10.1016/j.pestbp.2023.105535] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 09/06/2023]
Abstract
Entomopathogenic fungi are a promising category of biocontrol agents with mosquitocidal properties. Prior studies have proved their potential to reduce fecundity, human biting and vector competence, all of them together determine vectorial capacity of the mosquitoes. Unfortunately, conventional vector control strategies are inadequate with growing problem of insecticide resistance and environmental deterioration. Therefore, alternate vector control measures are immediately needed and to accomplish that, an improved understanding of behavioral and physiological defense mechanisms of the mosquitoes against fungal infection is essential. In this study, fitness was considered with respect to different behavioral (self-grooming and flight), physiological (antifungal activity and antimicrobial peptides) parameters and survival rates as compared to the control group. We found a significant upregulation in CLSP2, TEP22, Rel1 and Rel2 genes at multiple time periods of fungal infection, which indicates the successful fungal infection and activation of Toll and IMD pathways in mosquitoes. RNAi-mediated silencing of Rel1 and Rel2 genes (transcription factors of Toll and IMD pathways, respectively) significantly reduced the survival, self-grooming frequencies and durations, and flight locomotor activity among adult Ae. aegypti female mosquitoes. Moreover, Rel1 and Rel2 knockdown significantly decreased antifungal activity and antimicrobial peptides expression levels in target mosquitoes. These results indicate an overall decrease in fitness of the mosquitoes after fungal challenge following Rel1 and Rel2 silencing. These findings provide an improved understanding of behavioral and physiological responses in mosquitoes with altered immunity against entomopathogenic fungal infections which can guide us towards the development of novel biocontrol strategies against mosquitoes.
Collapse
Affiliation(s)
- Nasir Mehmood
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Ali Hassan
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Xueshan Zhong
- Yuexiu Center for Disease Control and Prevention, Guangzhou 510055, Guangdong Province, China
| | - Yongzhang Zhu
- Guangzhou Yongliang Environmental Protection Technology Service CO., LTD, Guangzhou 510405, Guangdong Province, China
| | - Guang Ouyang
- Guangzhou Yongliang Environmental Protection Technology Service CO., LTD, Guangzhou 510405, Guangdong Province, China
| | - Qiuying Huang
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
14
|
Ramelow J, Keleta Y, Niu G, Wang X, Li J. Plasmodium parasitophorous vacuole membrane protein Pfs16 promotes malaria transmission by silencing mosquito immunity. J Biol Chem 2023:104824. [PMID: 37196765 DOI: 10.1016/j.jbc.2023.104824] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/07/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
With rising cases for the first time in years, malaria remains a significant public health burden. The sexual stage of the malaria parasite infects mosquitoes to transmit malaria from host to host. Hence, an infected mosquito plays an essential role in malaria transmission. Plasmodium falciparum is the most dominant and dangerous malaria pathogen. Previous studies identified a sexual stage-specific protein 16 (Pfs16) localized to the parasitophorous vacuole membrane (PVM). Here we elucidate the function of Pfs16 during malaria transmission. Our structural analysis identified Pfs16 as an alpha-helical integral membrane protein with one transmembrane domain connecting to two regions across PVM. ELISA assays showed that insect cell-expressed recombinant Pfs16 (rPfs16) interacted with An. gambiae midguts, and microscopy found that rPfs16 bound to midgut epithelial cells. Transmission-blocking assays demonstrated that polyclonal antibodies against Pfs16 significantly reduced the number of oocysts in mosquito midguts. However, on the contrary, feeding rPfs16 increased the number of oocysts. Further analysis revealed that Pfs16 reduced the activity of mosquito midgut caspase 3/7, a key enzyme in the mosquito Jun-N-terminal kinase (JNK) immune pathway. We conclude that Pfs16 facilitates parasites to invade mosquito midguts by actively silencing the mosquito's innate immunity through its interaction with the midgut epithelial cells. Therefore, Pfs16 is a potential target to control malaria transmission.
Collapse
Affiliation(s)
- Julian Ramelow
- Biomedical Sciences Graduate Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Yacob Keleta
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Guodong Niu
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Xiaohong Wang
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Jun Li
- Biomedical Sciences Graduate Program, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; Department of Biological Sciences, Florida International University, Miami, FL 33199, USA; Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
15
|
Morejon B, Michel K. A zone-of-inhibition assay to screen for humoral antimicrobial activity in mosquito hemolymph. Front Cell Infect Microbiol 2023; 13:891577. [PMID: 36779191 PMCID: PMC9908765 DOI: 10.3389/fcimb.2023.891577] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
In insects, antibacterial immunity largely depends on the activation of downstream signaling and effector responses, leading to the synthesis and secretion of soluble effector molecules, such as antimicrobial peptides (AMPs). AMPs are acute infection response peptides secreted into the hemolymph upon bacterial stimulation. The transcription of innate immunity genes encoding for AMPs is highly dependent on several signaling cascade pathways, such as the Toll pathway. In the African malaria mosquito, Anopheles gambiae, AMPs hold a special interest as their upregulation have been shown to limit the growth of malaria parasites, bacteria, and fungi. Most of the current knowledge on the regulation of insect AMPs in microbial infection have been obtained from Drosophila. However, largely due to the lack of convenient assays, the regulation of antimicrobial activity in mosquito hemolymph is still not completely understood. In this study, we report a zone of inhibition assay to identify the contribution of AMPs and components of the Toll pathway to the antimicrobial activity of A. gambiae hemolymph. As a proof of principle, we demonstrate that Micrococcus luteus challenge induces antimicrobial activity in the adult female mosquito hemolymph, which is largely dependent on defensin 1. Moreover, by using RNAi to silence Cactus, REL1, and MyD88, we showed that Cactus kd induces antimicrobial activity in the mosquito hemolymph, whereas the antimicrobial activity in REL1 kd and MyD88 kd is reduced after challenge. Finally, while injection itself is not sufficient to induce antimicrobial activity, our results show that it primes the response to bacterial challenge. Our study provides information that increases our knowledge of the regulation of antimicrobial activity in response to microbial infections in mosquitoes. Furthermore, this assay represents an ex vivo medium throughput assay that can be used to determine the upstream regulatory elements of antimicrobial activity in A. gambiae hemolymph.
Collapse
Affiliation(s)
- Bianca Morejon
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | | |
Collapse
|
16
|
Ramirez JL, Hampton KJ, Rosales AM, Muturi EJ. Multiple mosquito AMPs are needed to potentiate their antifungal effect against entomopathogenic fungi. Front Microbiol 2023; 13:1062383. [PMID: 36687607 PMCID: PMC9852974 DOI: 10.3389/fmicb.2022.1062383] [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: 10/05/2022] [Accepted: 12/12/2022] [Indexed: 01/09/2023] Open
Abstract
Mosquito resistance to microbial infections, including fungal entomopathogens that are selected for mosquito control, depend on a range of antimicrobial effectors, among them antimicrobial peptides (AMPs). These short peptides, along the antimicrobial effector lysozyme, act by disrupting the microbial cell membrane or by interfering with microbial physiological processes. While the induction of AMPs and lysozyme during fungal entomopathogenic infections have been reported, their contribution to the mosquito antifungal response has not been evaluated. In this study, we assessed the induction of Ae. aegypti AMPs and lysozyme genes at two points of infection and against distinct entomopathogenic fungi. Our results indicate that fungal infection elicits the expression of cecropin, defensin, diptericin, holotricin, and lysozyme, but do not affect those of attacin or gambicin. We further evaluated the role of these antimicrobial effectors via RNAi-based depletion of select AMPs during challenges with two entomopathogenic fungi. Our results reveal that AMPs and lysozyme are critical to the antifungal response, acting in concert, rather than individually, to potentiate their antimicrobial effect against entomopathogenic fungi. This study further contributes to a better understanding of the mechanisms that confer resistance to entomopathogenic fungi in an important mosquito vector.
Collapse
Affiliation(s)
- José L. Ramirez
- Crop Bioprotection Research Unit, United States Department of Agriculture, Agricultural Research Service, National Center for Agricultural Utilization Research, Peoria, IL, United States,*Correspondence: José L. Ramirez, ,
| | - Kylie J. Hampton
- Crop Bioprotection Research Unit, United States Department of Agriculture, Agricultural Research Service, National Center for Agricultural Utilization Research, Peoria, IL, United States
| | | | - Ephantus J. Muturi
- Crop Bioprotection Research Unit, United States Department of Agriculture, Agricultural Research Service, National Center for Agricultural Utilization Research, Peoria, IL, United States
| |
Collapse
|
17
|
Sneed SD, Dwivedi SB, DiGate C, Denecke S, Povelones M. Aedes aegypti Malpighian tubules are immunologically activated following systemic Toll activation. Parasit Vectors 2022; 15:469. [PMID: 36522779 PMCID: PMC9753289 DOI: 10.1186/s13071-022-05567-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/02/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Canine heartworm is a widespread and potentially fatal mosquito-borne disease caused by infections with the parasitic nematode, Dirofilaria immitis. We have previously shown that systemic activation of the Toll immune pathway via silencing of the negative regulator Cactus in Aedes aegypti blocks parasite development in the Malpighian tubules (MT), the mosquito renal organ. However, it was not established whether the MT were directly responding to Toll activation or were alternatively responding to upregulated proteins or other changes to the hemolymph driven by other tissues. Distinguishing these possibilities is crucial for developing more precise strategies to block D. immitis while potentially avoiding the fitness cost to the mosquito associated with Cactus silencing. METHODS This study defines the transcriptional response of the MT and changes to the hemolymph proteome of Ae. aegypti after systemic Toll activation via intra-thoracic injection of double-stranded Cactus (dsCactus) RNA. RESULTS Malpighian tubules significantly increased expression of the Toll pathway target genes that significantly overlapped expression changes occurring in whole mosquitoes. A significant overlap between the transcriptional response of the MT and proteins upregulated in the hemolymph was also observed. CONCLUSIONS Our data show that MT are capable of RNA interference-mediated gene silencing and directly respond to dsCactus treatment by upregulating targets of the canonical Toll pathway. Although not definitive, the strong correspondence between the MT transcriptional response and the hemolymph proteomic responses provides evidence that the MT may contribute to mosquito humoral immunity.
Collapse
Affiliation(s)
- Sarah D. Sneed
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Sutopa B. Dwivedi
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Cameron DiGate
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Shane Denecke
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Michael Povelones
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
18
|
Yan Y, Sigle LT, Rinker DC, Estévez-Lao TY, Capra JA, Hillyer JF. The immune deficiency and c-Jun N-terminal kinase pathways drive the functional integration of the immune and circulatory systems of mosquitoes. Open Biol 2022; 12:220111. [PMID: 36069078 PMCID: PMC9449813 DOI: 10.1098/rsob.220111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The immune and circulatory systems of animals are functionally integrated. In mammals, the spleen and lymph nodes filter and destroy microbes circulating in the blood and lymph, respectively. In insects, immune cells that surround the heart valves (ostia), called periostial haemocytes, destroy pathogens in the areas of the body that experience the swiftest haemolymph (blood) flow. An infection recruits additional periostial haemocytes, amplifying heart-associated immune responses. Although the structural mechanics of periostial haemocyte aggregation have been defined, the genetic factors that regulate this process remain less understood. Here, we conducted RNA sequencing in the African malaria mosquito, Anopheles gambiae, and discovered that an infection upregulates multiple components of the immune deficiency (IMD) and c-Jun N-terminal kinase (JNK) pathways in the heart with periostial haemocytes. This upregulation is greater in the heart with periostial haemocytes than in the circulating haemocytes or the entire abdomen. RNA interference-based knockdown then showed that the IMD and JNK pathways drive periostial haemocyte aggregation and alter phagocytosis and melanization on the heart, thereby demonstrating that these pathways regulate the functional integration between the immune and circulatory systems. Understanding how insects fight infection lays the foundation for novel strategies that could protect beneficial insects and harm detrimental ones.
Collapse
Affiliation(s)
- Yan Yan
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Leah T. Sigle
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - David C. Rinker
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | | | - John A. Capra
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA,Bakar Computational Health Sciences Institute and Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Julián F. Hillyer
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
19
|
Zhu F, Zheng H, Chen S, Zhang K, Qin X, Zhang J, Liu T, Fan Y, Wang L, Li X, Zhang J, Xu W. Malaria oocysts require circumsporozoite protein to evade mosquito immunity. Nat Commun 2022; 13:3208. [PMID: 35680915 PMCID: PMC9184642 DOI: 10.1038/s41467-022-30988-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 05/25/2022] [Indexed: 11/08/2022] Open
Abstract
Malaria parasites are less vulnerable to mosquito immune responses once ookinetes transform into oocysts, facilitating parasite development in the mosquito. However, the underlying mechanisms of oocyst resistance to mosquito defenses remain unclear. Here, we show that circumsporozoite protein (CSP) is required for rodent malaria oocysts to avoid mosquito defenses. Mosquito infection with CSPmut parasites (mutation in the CSP pexel I/II domains) induces nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 5 (NOX5)-mediated hemocyte nitration, thus activating Toll pathway and melanization of mature oocysts, upregulating hemocyte TEP1 expression, and causing defects in the release of sporozoites from oocysts. The pre-infection of mosquitoes with the CSPmut parasites reduces the burden of infection when re-challenged with CSPwt parasites by inducing hemocyte nitration. Thus, we demonstrate why oocysts are invisible to mosquito immunity and reveal an unknown role of CSP in the immune evasion of oocysts, indicating it as a potential target to block malaria transmission.
Collapse
Affiliation(s)
- Feng Zhu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| | - Hong Zheng
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Suilin Chen
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| | - Kun Zhang
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| | - Xin Qin
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| | - Jingru Zhang
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| | - Taiping Liu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| | - Yongling Fan
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| | - Liting Wang
- Biomedical Analysis Center, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Xiaoxu Li
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
- Department of High Altitude Physiology and Pathology, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jian Zhang
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China.
| | - Wenyue Xu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China.
| |
Collapse
|
20
|
Maya-Maldonado K, Cardoso-Jaime V, González-Olvera G, Osorio B, Recio-Tótoro B, Manrique-Saide P, Rodríguez-Sánchez IP, Lanz-Mendoza H, Missirlis F, Hernández-Hernández FDLC. Mosquito metallomics reveal copper and iron as critical factors for Plasmodium infection. PLoS Negl Trop Dis 2021; 15:e0009509. [PMID: 34161336 PMCID: PMC8221525 DOI: 10.1371/journal.pntd.0009509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 05/26/2021] [Indexed: 11/18/2022] Open
Abstract
Iron and copper chelation restricts Plasmodium growth in vitro and in mammalian hosts. The parasite alters metal homeostasis in red blood cells to its favor, for example metabolizing hemoglobin to hemozoin. Metal interactions with the mosquito have not, however, been studied. Here, we describe the metallomes of Anopheles albimanus and Aedes aegypti throughout their life cycle and following a blood meal. Consistent with previous reports, we found evidence of maternal iron deposition in embryos of Ae. aegypti, but less so in An. albimanus. Sodium, potassium, iron, and copper are present at higher concentrations during larval developmental stages. Two An. albimanus phenotypes that differ in their susceptibility to Plasmodium berghei infection were studied. The susceptible white stripe (ws) phenotype was named after a dorsal white stripe apparent during larval stages 3, 4, and pupae. During larval stage 3, ws larvae accumulate more iron and copper than the resistant brown stripe (bs) phenotype counterparts. A similar increase in copper and iron accumulation was also observed in the susceptible ws, but not in the resistant bs phenotype following P. berghei infection. Feeding ws mosquitoes with extracellular iron and copper chelators before and after receiving Plasmodium-infected blood protected from infection and simultaneously affected follicular development in the case of iron chelation. Unexpectedly, the application of the iron chelator to the bs strain reverted resistance to infection. Besides a drop in iron, iron-chelated bs mosquitoes experienced a concomitant loss of copper. Thus, the effect of metal chelation on P. berghei infectivity was strain-specific.
Collapse
Affiliation(s)
- Krystal Maya-Maldonado
- Departamento de Infectómica y Patogénesis Molecular, Cinvestav, Ciudad de México, México
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, México
| | - Victor Cardoso-Jaime
- Departamento de Infectómica y Patogénesis Molecular, Cinvestav, Ciudad de México, México
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, México
| | - Gabriela González-Olvera
- Unidad Colaborativa para Bioensayos Entomológicos, Campus de Ciencias Biológicas y Agropecuarias, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Beatriz Osorio
- Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav, Ciudad de México, México
| | - Benito Recio-Tótoro
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, México
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Pablo Manrique-Saide
- Unidad Colaborativa para Bioensayos Entomológicos, Campus de Ciencias Biológicas y Agropecuarias, Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Iram Pablo Rodríguez-Sánchez
- Laboratorio de Fisiología Molecular y Estructural, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Humberto Lanz-Mendoza
- Centro de Investigaciones sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, México
| | - Fanis Missirlis
- Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav, Ciudad de México, México
| | | |
Collapse
|
21
|
Nattoh G, Bargul JL, Magoma G, Mbaisi L, Butungi H, Mararo E, Teal E, Herren JK. The fungus Leptosphaerulina persists in Anopheles gambiae and induces melanization. PLoS One 2021; 16:e0246452. [PMID: 33617536 PMCID: PMC7899377 DOI: 10.1371/journal.pone.0246452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Anopheles mosquitoes are colonized by diverse microorganisms that may impact on host biology and vectorial capacity. Eukaryotic symbionts such as fungi have been isolated from Anopheles, but whether they are stably associated with mosquitoes and transmitted transstadially across mosquito life stages or to subsequent generations remains largely unexplored. Here, we show that a Leptosphaerulina sp. fungus isolated from the midgut of An. gambiae can be stably associated with An. gambiae host and that it imposes low fitness cost when re-introduced through co-feeding. This fungus is transstadially transmitted across An. gambiae developmental stages and to their progeny. It is present in field-caught larvae and adult mosquitoes at moderate levels across geographical regions. We observed that Leptosphaerulina sp. induces a distinctive melanotic phenotype across the developmental stages of mosquito. As a eukaryotic symbiont that is stably associated with An. gambiae Leptosphaerulina sp. can be explored for paratransgenesis.
Collapse
Affiliation(s)
- Godfrey Nattoh
- International Centre of Insect Physiology and Ecology, Nairobi, Kenya
- Pan African University Institute for Basic Sciences Technology and Innovation, Nairobi, Kenya
- * E-mail:
| | - Joel L. Bargul
- International Centre of Insect Physiology and Ecology, Nairobi, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Science and Technology, Nairobi, Kenya
| | - Gabriel Magoma
- Pan African University Institute for Basic Sciences Technology and Innovation, Nairobi, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Science and Technology, Nairobi, Kenya
| | - Lilian Mbaisi
- International Centre of Insect Physiology and Ecology, Nairobi, Kenya
| | - Hellen Butungi
- International Centre of Insect Physiology and Ecology, Nairobi, Kenya
- Wits Research Institute for Malaria, University of the Witwatersrand, Johannesburg, South Africa
| | - Enock Mararo
- International Centre of Insect Physiology and Ecology, Nairobi, Kenya
| | - Evan Teal
- International Centre of Insect Physiology and Ecology, Nairobi, Kenya
| | | |
Collapse
|
22
|
Rolandelli A, Nascimento AEC, Silva LS, Rivera-Pomar R, Guarneri AA. Modulation of IMD, Toll, and Jak/STAT Immune Pathways Genes in the Fat Body of Rhodnius prolixus During Trypanosoma rangeli Infection. Front Cell Infect Microbiol 2021; 10:598526. [PMID: 33537241 PMCID: PMC7848085 DOI: 10.3389/fcimb.2020.598526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/30/2020] [Indexed: 12/31/2022] Open
Abstract
Trypanosoma rangeli is the second most common American trypanosome that infects man. It is vectored by triatomines from the genus Rhodnius, in which it invades the hemolymph and infects the salivary glands, avoiding the bug immune responses. In insects, these responses are initiated by well conserved pathways, mainly the IMD, Toll, and Jak/STAT. We hypothesize that long-term infection with T. rangeli in the gut or hemolymph of Rhodnius prolixus triggers different systemic immune responses, which influence the number of parasites that survive inside the vector. Thus, we investigated groups of insects with infections in the gut and/or hemolymph, and evaluated the parasite load and the expression in the fat body of transcription factors (Rp-Relish, Rp-Dorsal, and Rp-STAT) and inhibitors (Rp-Cactus and Rp-Caspar) of the IMD, Toll, and Jak/STAT pathways. We detected lower parasite counts in the gut of insects without hemolymph infection, compared to hemolymph-infected groups. Besides, we measured higher parasite numbers in the gut of bugs that were first inoculated with T. rangeli and then fed on infected mice, compared with control insects, indicating that hemolymph infection increases parasite numbers in the gut. Interestingly, we observed that genes from the three immune pathways where differentially modulated, depending on the region parasites were present, as we found (1) Rp-Relish downregulated in gut-and/or-hemolymph-infected insects, compared with controls; (2) Rp-Cactus upregulated in gut-infected insect, compared with controls and gut-and-hemolymph-infected groups; and (3) Rp-STAT downregulated in all groups of hemolymph-infected insects. Finally, we uncovered negative correlations between parasite loads in the gut and Rp-Relish and Rp-Cactus expression, and between parasite counts in the hemolymph and Rp-Relish levels, suggesting an association between parasite numbers and the IMD and Toll pathways. Overall, our findings reveal new players in R. prolixus-T. rangeli interactions that could be key for the capacity of the bug to transmit the pathogen.
Collapse
Affiliation(s)
- Agustín Rolandelli
- Centro de Bioinvestigaciones (CeBio), Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT-NOBA), Universidad Nacional del Noroeste de la Provincia de Buenos Aires (UNNOBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Pergamino, Argentina
| | - Adeisa E C Nascimento
- Vector Behaviour and Pathogen Interaction Group, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| | - Leticia S Silva
- Vector Behaviour and Pathogen Interaction Group, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| | - Rolando Rivera-Pomar
- Centro de Bioinvestigaciones (CeBio), Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT-NOBA), Universidad Nacional del Noroeste de la Provincia de Buenos Aires (UNNOBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Pergamino, Argentina
| | - Alessandra A Guarneri
- Vector Behaviour and Pathogen Interaction Group, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| |
Collapse
|
23
|
Zhang X, Li M, El Moussawi L, Saab S, Zhang S, Osta MA, Michel K. CLIPB10 is a Terminal Protease in the Regulatory Network That Controls Melanization in the African Malaria Mosquito Anopheles gambiae. Front Cell Infect Microbiol 2021; 10:585986. [PMID: 33520733 PMCID: PMC7843523 DOI: 10.3389/fcimb.2020.585986] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022] Open
Abstract
Humoral immune responses in animals are often tightly controlled by regulated proteolysis. This proteolysis is exerted by extracellular protease cascades, whose activation culminates in the proteolytic cleavage of key immune proteins and enzymes. A model for such immune system regulation is the melanization reaction in insects, where the activation of prophenoxidase (proPO) leads to the rapid formation of eumelanin on the surface of foreign entities such as parasites, bacteria and fungi. ProPO activation is tightly regulated by a network of so-called clip domain serine proteases, their proteolytically inactive homologs, and their serpin inhibitors. In Anopheles gambiae, the major malaria vector in sub-Saharan Africa, manipulation of this protease network affects resistance to a wide range of microorganisms, as well as host survival. However, thus far, our understanding of the molecular make-up and regulation of the protease network in mosquitoes is limited. Here, we report the function of the clip domain serine protease CLIPB10 in this network, using a combination of genetic and biochemical assays. CLIPB10 knockdown partially reversed melanotic tumor formation induced by Serpin 2 silencing in the absence of infection. CLIPB10 was also partially required for the melanization of ookinete stages of the rodent malaria parasite Plasmodium berghei in a refractory mosquito genetic background. Recombinant serpin 2 protein, a key inhibitor of the proPO activation cascade in An. gambiae, formed a SDS-stable protein complex with activated recombinant CLIPB10, and efficiently inhibited CLIPB10 activity in vitro at a stoichiometry of 1.89:1. Recombinant activated CLIPB10 increased PO activity in Manduca sexta hemolymph ex vivo, and directly activated purified M. sexta proPO in vitro. Taken together, these data identify CLIPB10 as the second protease with prophenoloxidase-activating function in An. gambiae, in addition to the previously described CLIPB9, suggesting functional redundancy in the protease network that controls melanization. In addition, our data suggest that tissue melanization and humoral melanization of parasites are at least partially mediated by the same proteases.
Collapse
Affiliation(s)
- Xin Zhang
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Miao Li
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Layla El Moussawi
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Sally Saab
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Shasha Zhang
- Division of Biology, Kansas State University, Manhattan, KS, United States.,Department of Entomology, China Agricultural University, Beijing, China
| | - Mike A Osta
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
24
|
Hajkazemian M, Bossé C, Mozūraitis R, Emami SN. Battleground midgut: The cost to the mosquito for hosting the malaria parasite. Biol Cell 2020; 113:79-94. [PMID: 33125724 DOI: 10.1111/boc.202000039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/31/2020] [Accepted: 10/12/2020] [Indexed: 12/27/2022]
Abstract
In eco-evolutionary studies of parasite-host interactions, virulence is defined as a reduction in host fitness as a result of infection relative to an uninfected host. Pathogen virulence may either promote parasite transmission, when correlated with higher parasite replication rate, or decrease the transmission rate if the pathogen quickly kills the host. This evolutionary mechanism, referred to as 'trade-off' theory, proposes that pathogen virulence evolves towards a level that most benefits the transmission. It has been generally predicted that pathogens evolve towards low virulence in their insect vectors, mainly due to the high dependence of parasite transmission on their vector survival. Therefore, the degree of virulence which malaria parasites impose on mosquito vectors may depend on several external and internal factors. Here, we review briefly (i) the role of mosquito in parasite development, with a particular focus on mosquito midgut as the battleground between Plasmodium and the mosquito host. We aim to point out (ii) the histology of the mosquito midgut epithelium and its role in host defence against parasite's countermeasures in the three main battle sites, namely (a) the lumen (microbiota and biochemical environment), (b) the peritrophic membrane (physical barrier) and (c) the tubular epithelium including the basal membrane (physical and biochemical barrier). Lastly, (iii) we describe the impact which malaria parasite and its virulence factors have on mosquito fitness.
Collapse
Affiliation(s)
- Melika Hajkazemian
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Clément Bossé
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.,François Rabelais University, Tours, France
| | - Raimondas Mozūraitis
- Laboratory of Chemical and Behavioural Ecology, Institute of Ecology, Nature Research Centre, Vilnius, Lithuania.,Department of Zoology, Stockholm University, Stockholm, Sweden
| | - S Noushin Emami
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.,Molecular Attraction AB, Hägersten, Stockholm, Sweden.,Natural Resources Institute, FES, University of Greenwich, London, UK
| |
Collapse
|
25
|
Volohonsky G, Paul-Gilloteaux P, Štáfková J, Soichot J, Salamero J, Levashina EA. Kinetics of Plasmodium midgut invasion in Anopheles mosquitoes. PLoS Pathog 2020; 16:e1008739. [PMID: 32946522 PMCID: PMC7526910 DOI: 10.1371/journal.ppat.1008739] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 09/30/2020] [Accepted: 06/23/2020] [Indexed: 01/06/2023] Open
Abstract
Malaria-causing Plasmodium parasites traverse the mosquito midgut cells to establish infection at the basal side of the midgut. This dynamic process is a determinant of mosquito vector competence, yet the kinetics of the parasite migration is not well understood. Here we used transgenic mosquitoes of two Anopheles species and a Plasmodium berghei fluorescence reporter line to track parasite passage through the mosquito tissues at high spatial resolution. We provide new quantitative insight into malaria parasite invasion in African and Indian Anopheles species and propose that the mosquito complement-like system contributes to the species-specific dynamics of Plasmodium invasion. The traversal of the mosquito midgut cells is one of the critical stages in the life cycle of malaria parasites. Motile parasite forms, called ookinetes, traverse the midgut epithelium in a dynamic process which is not fully understood. Here, we harnessed transgenic reporters to track invasion of Plasmodium parasites in African and Indian mosquito species. We found important differences in parasite dynamics between the two Anopheles species and demonstrated a role of the mosquito complement-like system in regulation of parasite invasion of the midgut cells.
Collapse
Affiliation(s)
- Gloria Volohonsky
- INSERM U963, CNRS UPR9022, University of Strasbourg, Strasbourg, France
| | - Perrine Paul-Gilloteaux
- SERPICO Inria Team/CNRS UMR 144, Institut Curie, Paris, France.,National Biology and Health Infrastructure "France Bioimaging", Institut Curie, Paris, France.,Cell and Tissue Imaging Facility, IBiSA, Institut Curie, Paris, France
| | - Jitka Štáfková
- INSERM U963, CNRS UPR9022, University of Strasbourg, Strasbourg, France
| | - Julien Soichot
- INSERM U963, CNRS UPR9022, University of Strasbourg, Strasbourg, France
| | - Jean Salamero
- SERPICO Inria Team/CNRS UMR 144, Institut Curie, Paris, France.,National Biology and Health Infrastructure "France Bioimaging", Institut Curie, Paris, France.,Cell and Tissue Imaging Facility, IBiSA, Institut Curie, Paris, France
| | - Elena A Levashina
- INSERM U963, CNRS UPR9022, University of Strasbourg, Strasbourg, France.,Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
26
|
Yu S, Wang P, Qin J, Zheng H, Wang J, Liu T, Yang X, Wang Y. Bacillus sphaericus exposure reduced vector competence of Anopheles dirus to Plasmodium yoelii by upregulating the Imd signaling pathway. Parasit Vectors 2020; 13:446. [PMID: 32891162 PMCID: PMC7487769 DOI: 10.1186/s13071-020-04321-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/30/2020] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Vector control with Bacillus sphaericus (Bs) is an effective way to block the transmission of malaria. However, in practical application of Bs agents, a sublethal dose effect was often caused by insufficient dosing, and it is little known whether the Bs exposure would affect the surviving mosquitoes' vector capacity to malaria. METHODS A sublethal dose of the Bs 2362 strain was administrated to the early fourth-instar larvae of Anopheles dirus to simulate shortage use of Bs in field circumstance. To determine vector competence, mosquitoes were dissected and the oocysts in the midguts were examined on day 9-11 post-infection with Plasmodium yoelii. Meanwhile, a SYBR quantitative PCR assay was conducted to examine the transcriptional level of the key immune molecules of mosquitoes, and RNA interference was utilized to validate the role of key immune effector molecule TEP1. RESULTS The sublethal dose of Bs treatment significantly reduced susceptibility of An. dirus to P. yoelii, with the decrease of P. yoelii infection intensity and rate. Although there existed a melanization response of adult An. dirus following challenge with P. yoelii, it was not involved in the decrease of vector competence as no significant difference of melanization rates and densities between the control and Bs groups was found. Further studies showed that Bs treatment significantly increased TEP1 expression in the fourth-instar larvae (L4), pupae (Pu), 48 h post-infection (hpi) and 72 hpi (P < 0.001). Further, gene-silencing of TEP1 resulted in disappearance of the Bs impact on vector competence of An. dirus to P. yoelii. Moreover, the transcriptional level of PGRP-LC and Rel2 were significantly elevated by Bs treatment with decreased expression of the negative regulator Caspar at 48 hpi, which implied that the Imd signaling pathway was upregulated by Bs exposure. CONCLUSIONS Bs exposure can reduce the vector competence of An. dirus to malaria parasites through upregulating Imd signaling pathway and enhancing the expression of TEP1. The data could not only help us to understand the impact and mechanism of Bs exposure on Anopheles' vector competence to malaria but also provide us with novel clues for wiping out malaria using vector control.
Collapse
Affiliation(s)
- Shasha Yu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038 China
| | - Pan Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038 China
| | - Jie Qin
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038 China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037 China
| | - Jing Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038 China
| | - Tingting Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038 China
| | - Xuesen Yang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038 China
| | - Ying Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038 China
| |
Collapse
|
27
|
Dong Y, Simões ML, Dimopoulos G. Versatile transgenic multistage effector-gene combinations for Plasmodium falciparum suppression in Anopheles. SCIENCE ADVANCES 2020; 6:eaay5898. [PMID: 32426491 PMCID: PMC7220273 DOI: 10.1126/sciadv.aay5898] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/27/2020] [Indexed: 05/14/2023]
Abstract
The malaria parasite's complex journey through the Anopheles mosquito vector provides multiple opportunities for targeting Plasmodium with recombinant effectors at different developmental stages and different host tissues. We have designed and expressed transgenes that efficiently suppress Plasmodium infection by targeting the parasite with multiple independent endogenous and exogenous effectors at multiple infection stages to potentiate suppression and minimize the probability for development of resistance to develop. We have also addressed the fitness impact of transgene expression on the mosquito. We show that highly potent suppression can be achieved by targeting both pre-oocyst stages by transgenically overexpressing either the endogenous immune deficiency immune pathway transcription factor Rel2 or a polycistronic mRNA encoding multiple antiparasitic effectors and simultaneously targeting the sporozoite stages with an anti-sporozoite single-chain antibody fused to the antiparasitic protein Scorpine. Expression of the selected endogenous effector systems appears to pose a lower fitness cost than does the use of foreign genes.
Collapse
|
28
|
PIMMS43 is required for malaria parasite immune evasion and sporogonic development in the mosquito vector. Proc Natl Acad Sci U S A 2020; 117:7363-7373. [PMID: 32165544 PMCID: PMC7132314 DOI: 10.1073/pnas.1919709117] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Malaria is transmitted among humans through mosquito bites. Here, we characterize a protein found on the surface of mosquito stages of malaria parasites and reveal that it serves to evade the mosquito immune system and ensure disease transmission. Neutralization of PIMMS43 (Plasmodium Infection of the Mosquito Midgut Screen 43), either by eliminating it from the parasite genome or by preincubating parasites with antibodies that bind to the PIMMS43 protein, inhibits mosquito infection with malaria parasites. Differences in PIMMS43 detected between African malaria parasite populations suggest that these populations have adapted for transmission by different mosquito vectors that are also differentially distributed across the continent. We conclude that targeting PIMMS43 can block malaria parasites inside mosquitoes before they can infect humans. After being ingested by a female Anopheles mosquito during a bloodmeal on an infected host, and before they can reach the mosquito salivary glands to be transmitted to a new host, Plasmodium parasites must establish an infection of the mosquito midgut in the form of oocysts. To achieve this, they must first survive a series of robust innate immune responses that take place prior to, during, and immediately after ookinete traversal of the midgut epithelium. Understanding how parasites may evade these responses could highlight new ways to block malaria transmission. We show that an ookinete and sporozoite surface protein designated as PIMMS43 (Plasmodium Infection of the Mosquito Midgut Screen 43) is required for parasite evasion of the Anopheles coluzzii complement-like response. Disruption of PIMMS43 in the rodent malaria parasite Plasmodium berghei triggers robust complement activation and ookinete elimination upon mosquito midgut traversal. Silencing components of the complement-like system through RNAi largely restores ookinete-to-oocyst transition but oocysts remain small in size and produce a very small number of sporozoites that additionally are not infectious, indicating that PIMMS43 is also essential for sporogonic development in the oocyst. Antibodies that bind PIMMS43 interfere with parasite immune evasion when ingested with the infectious blood meal and significantly reduce the prevalence and intensity of infection. PIMMS43 genetic structure across African Plasmodium falciparum populations indicates allelic adaptation to sympatric vector populations. These data add to our understanding of mosquito–parasite interactions and identify PIMMS43 as a target of malaria transmission blocking.
Collapse
|
29
|
Mitri C, Bischoff E, Belda Cuesta E, Volant S, Ghozlane A, Eiglmeier K, Holm I, Dieme C, Brito-Fravallo E, Guelbeogo WM, Sagnon N, Riehle MM, Vernick KD. Leucine-Rich Immune Factor APL1 Is Associated With Specific Modulation of Enteric Microbiome Taxa in the Asian Malaria Mosquito Anopheles stephensi. Front Microbiol 2020; 11:306. [PMID: 32174902 PMCID: PMC7054466 DOI: 10.3389/fmicb.2020.00306] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/11/2020] [Indexed: 01/14/2023] Open
Abstract
The commensal gut microbiome is contained by the enteric epithelial barrier, but little is known about the degree of specificity of host immune barrier interactions for particular bacterial taxa. Here, we show that depletion of leucine-rich repeat immune factor APL1 in the Asian malaria mosquito Anopheles stephensi is associated with higher midgut abundance of just the family Enterobacteraceae, and not generalized dysbiosis of the microbiome. The effect is explained by the response of a narrow clade containing two main taxa related to Klebsiella and Cedecea. Analysis of field samples indicate that these two taxa are recurrent members of the wild Anopheles microbiome. Triangulation using sequence and functional data incriminated relatives of C. neteri and Cedecea NFIX57 as candidates for the Cedecea component, and K. michiganensis, K. oxytoca, and K.sp. LTGPAF-6F as candidates for the Klebsiella component. APL1 presence is associated with host ability to specifically constrain the abundance of a narrow microbiome clade of the Enterobacteraceae, and the immune factor may promote homeostasis of this clade in the enteric microbiome for host benefit.
Collapse
Affiliation(s)
- Christian Mitri
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Emmanuel Bischoff
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Eugeni Belda Cuesta
- Integromics Unit, Institute of Cardiometabolism and Nutrition, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France
| | - Stevenn Volant
- Bioinformatics and Biostatistics Hub, Department of Computational Biology, Institut Pasteur, Paris, France
- CNRS USR 3756, Institut Pasteur, Paris, France
| | - Amine Ghozlane
- Bioinformatics and Biostatistics Hub, Department of Computational Biology, Institut Pasteur, Paris, France
- CNRS USR 3756, Institut Pasteur, Paris, France
| | - Karin Eiglmeier
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Inge Holm
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Constentin Dieme
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Emma Brito-Fravallo
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Wamdaogo M. Guelbeogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - N’Fale Sagnon
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso
| | - Michelle M. Riehle
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kenneth D. Vernick
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| |
Collapse
|
30
|
Edgerton EB, McCrea AR, Berry CT, Kwok JY, Thompson LK, Watson B, Fuller EM, Nolan TJ, Lok JB, Povelones M. Activation of mosquito immunity blocks the development of transmission-stage filarial nematodes. Proc Natl Acad Sci U S A 2020; 117:3711-3717. [PMID: 32015105 PMCID: PMC7035481 DOI: 10.1073/pnas.1909369117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mosquito-borne helminth infections are responsible for a significant worldwide disease burden in both humans and animals. Accordingly, development of novel strategies to reduce disease transmission by targeting these pathogens in the vector are of paramount importance. We found that a strain of Aedes aegypti that is refractory to infection by Dirofilaria immitis, the agent of canine heartworm disease, mounts a stronger immune response during infection than does a susceptible strain. Moreover, activation of the Toll immune signaling pathway in the susceptible strain arrests larval development of the parasite, thereby decreasing the number of transmission-stage larvae. Notably, this strategy also blocks transmission-stage Brugia malayi, an agent of human lymphatic filariasis. Our data show that mosquito immunity can play a pivotal role in restricting filarial nematode development and suggest that genetically engineering mosquitoes with enhanced immunity will help reduce pathogen transmission.
Collapse
Affiliation(s)
- Elizabeth B Edgerton
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Abigail R McCrea
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Corbett T Berry
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Jenny Y Kwok
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Letitia K Thompson
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Brittany Watson
- Department of Clinical Sciences & Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | | | - Thomas J Nolan
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - James B Lok
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Michael Povelones
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104;
| |
Collapse
|
31
|
King JG. Developmental and comparative perspectives on mosquito immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 103:103458. [PMID: 31377103 DOI: 10.1016/j.dci.2019.103458] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 06/10/2023]
Abstract
Diseases spread by mosquitoes have killed more people than those spread by any other group of arthropod vectors and remain an important factor in determining global health and economic stability. The mosquito innate immune system can act to either modulate infection with human pathogens or fight off entomopathogens and increase the fitness and longevity of infected mosquitoes. While work remains towards understanding the larval immune system and the development of the mosquito immune system, it has recently become clearer that environmental factors heavily shape the developing mosquito immune system and continue to influence the adult immune system as well. The adult immune system has been well-studied and is known to involve multiple tissues and diverse molecular mechanisms. This review summarizes and synthesizes what is currently understood about the development of the mosquito immune system and includes comparisons of immune components unique to mosquitoes among the blood-feeding arthropods as well as important distinguishing factors between the anopheline and culicine mosquitoes. An explanation is included for how mosquito immunity factors into vector competence and vectorial capacity is presented along with a model for the interrelationships between nutrition, microbiome, pathogen interactions and behavior as they relate to mosquito development, immune status, adult female fitness and ultimately, vectorial capacity. Novel discoveries in the fields of mosquito ecoimmunology, neuroimmunology, and intracellular antiviral responses are highlighted.
Collapse
Affiliation(s)
- Jonas G King
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, 32 Creelman Street, Dorman 402, Mississippi State, MS 39762, USA.
| |
Collapse
|
32
|
Mitri C, Bischoff E, Eiglmeier K, Holm I, Dieme C, Brito-Fravallo E, Raz A, Zakeri S, Nejad MIK, Djadid ND, Vernick KD, Riehle MM. Gene copy number and function of the APL1 immune factor changed during Anopheles evolution. Parasit Vectors 2020; 13:18. [PMID: 31931885 PMCID: PMC6958605 DOI: 10.1186/s13071-019-3868-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/19/2019] [Indexed: 12/02/2022] Open
Abstract
Background The recent reference genome assembly and annotation of the Asian malaria vector Anopheles stephensi detected only one gene encoding the leucine-rich repeat immune factor APL1, while in the Anopheles gambiae and sibling Anopheles coluzzii, APL1 factors are encoded by a family of three paralogs. The phylogeny and biological function of the unique APL1 gene in An. stephensi have not yet been specifically examined. Methods The APL1 locus was manually annotated to confirm the computationally predicted single APL1 gene in An. stephensi. APL1 evolution within Anopheles was explored by phylogenomic analysis. The single or paralogous APL1 genes were silenced in An. stephensi and An. coluzzii, respectively, followed by mosquito survival analysis, experimental infection with Plasmodium and expression analysis. Results APL1 is present as a single ancestral gene in most Anopheles including An. stephensi but has expanded to three paralogs in an African lineage that includes only the Anopheles gambiae species complex and Anopheles christyi. Silencing of the unique APL1 copy in An. stephensi results in significant mosquito mortality. Elevated mortality of APL1-depleted An. stephensi is rescued by antibiotic treatment, suggesting that pathology due to bacteria is the cause of mortality, and indicating that the unique APL1 gene is essential for host survival. Successful Plasmodium development in An. stephensi depends upon APL1 activity for protection from high host mortality due to bacteria. In contrast, silencing of all three APL1 paralogs in An. coluzzii does not result in elevated mortality, either with or without Plasmodium infection. Expression of the single An. stephensi APL1 gene is regulated by both the Imd and Toll immune pathways, while the two signaling pathways regulate different APL1 paralogs in the expanded APL1 locus. Conclusions APL1 underwent loss and gain of functions concomitant with expansion from a single ancestral gene to three paralogs in one lineage of African Anopheles. We infer that activity of the unique APL1 gene promotes longevity in An. stephensi by conferring protection from or tolerance to an effect of bacterial pathology. The evolution of an expanded APL1 gene family could be a factor contributing to the exceptional levels of malaria transmission mediated by human-feeding members of the An. gambiae species complex in Africa.![]()
Collapse
Affiliation(s)
- Christian Mitri
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.,CNRS Unit of Evolutionary Genomics, Modeling and Health (UMR2000), Institut Pasteur, Paris, France
| | - Emmanuel Bischoff
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.,CNRS Unit of Evolutionary Genomics, Modeling and Health (UMR2000), Institut Pasteur, Paris, France
| | - Karin Eiglmeier
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.,CNRS Unit of Evolutionary Genomics, Modeling and Health (UMR2000), Institut Pasteur, Paris, France
| | - Inge Holm
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.,CNRS Unit of Evolutionary Genomics, Modeling and Health (UMR2000), Institut Pasteur, Paris, France
| | - Constentin Dieme
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.,CNRS Unit of Evolutionary Genomics, Modeling and Health (UMR2000), Institut Pasteur, Paris, France.,Wadsworth Center, New York State Department of Health, Slingerlands, NY, USA
| | - Emma Brito-Fravallo
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.,CNRS Unit of Evolutionary Genomics, Modeling and Health (UMR2000), Institut Pasteur, Paris, France
| | - Abbasali Raz
- Malaria and Vector Research Group, Biotechnology Research Center, Institut Pasteur of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group, Biotechnology Research Center, Institut Pasteur of Iran, Tehran, Iran
| | - Mahdokht I K Nejad
- Malaria and Vector Research Group, Biotechnology Research Center, Institut Pasteur of Iran, Tehran, Iran
| | - Navid D Djadid
- Malaria and Vector Research Group, Biotechnology Research Center, Institut Pasteur of Iran, Tehran, Iran
| | - Kenneth D Vernick
- Unit of Insect Vector Genetics and Genomics, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France. .,CNRS Unit of Evolutionary Genomics, Modeling and Health (UMR2000), Institut Pasteur, Paris, France.
| | - Michelle M Riehle
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
33
|
Abstract
Insects possess powerful immune systems that have evolved to defend against wounding and environmental pathogens such as bacteria, fungi, protozoans, and parasitoids. This surprising sophistication is accomplished through the activation of multiple immune pathways comprised of a large array of components, many of which have been identified and studied in detail using both genetic manipulations and traditional biochemical techniques. Recent advances indicate that certain pathways activate arrays of proteins that interact to form large functional complexes. Here we discuss three examples from multiple insects that exemplify such processes, including pathogen recognition, melanization, and coagulation. The functionality of each depends on integrating recognition with the recruitment of immune effectors capable of healing wounds and destroying pathogens. In both melanization and coagulation, protein interactions also appear to be essential for enzymatic activities tied to the formation of melanin and for the recruitment of hemocytes. The importance of these immune complexes is highlighted by the evolution of mechanisms in pathogens to disrupt their formation, an example of which is provided. While technically difficult to study, and not always readily amenable to dissection through genetics, modern mass spectrometry has become an indispensable tool in the study of these higher-order protein interactions. The formation of immune complexes should be viewed as an essential and emerging frontier in the study of insect immunity.
Collapse
|
34
|
Cappelli A, Damiani C, Mancini MV, Valzano M, Rossi P, Serrao A, Ricci I, Favia G. Asaia Activates Immune Genes in Mosquito Eliciting an Anti- Plasmodium Response: Implications in Malaria Control. Front Genet 2019; 10:836. [PMID: 31608103 PMCID: PMC6774264 DOI: 10.3389/fgene.2019.00836] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/13/2019] [Indexed: 01/04/2023] Open
Abstract
In mosquitoes, the discovery of the numerous interactions between components of the microbiota and the host immune response opens up the attractive possibility of the development of novel control strategies against mosquito borne diseases. We have focused our attention to Asaia, a symbiont of several mosquito vectors who has been proposed as one of the most potential tool for paratransgenic applications; although being extensively characterized, its interactions with the mosquito immune system has never been investigated. Here we report a study aimed at describing the interactions between Asaia and the immune system of two vectors of malaria, Anophelesstephensi and An. gambiae. The introduction of Asaia isolates induced the activation of the basal level of mosquito immunity and lower the development of malaria parasite in An. stephensi. These findings confirm and expand the potential of Asaia in mosquito borne diseases control, not only through paratransgenesis, but also as a natural effector for mosquito immune priming.
Collapse
Affiliation(s)
- Alessia Cappelli
- School of Biosciences & Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Claudia Damiani
- School of Biosciences & Veterinary Medicine, University of Camerino, Camerino, Italy
| | | | - Matteo Valzano
- School of Biosciences & Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Paolo Rossi
- School of Biosciences & Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Aurelio Serrao
- School of Biosciences & Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Irene Ricci
- School of Biosciences & Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Guido Favia
- School of Biosciences & Veterinary Medicine, University of Camerino, Camerino, Italy
| |
Collapse
|
35
|
Tawidian P, Rhodes VL, Michel K. Mosquito-fungus interactions and antifungal immunity. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 111:103182. [PMID: 31265904 PMCID: PMC6639037 DOI: 10.1016/j.ibmb.2019.103182] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 06/28/2019] [Indexed: 05/14/2023]
Abstract
The mosquito immune system has evolved in the presence of continuous encounters with fungi that range from food to foes. Herein, we review the field of mosquito-fungal interactions, providing an overview of current knowledge and topics of interest. Mosquitoes encounter fungi in their aquatic and terrestrial habitats. Mosquito larvae are exposed to fungi on plant detritus, within the water column, and at the water surface. Adult mosquitoes are exposed to fungi during indoor and outdoor resting, blood and sugar feeding, mating, and oviposition. Fungi enter the mosquito body through different routes, including ingestion and through active or passive breaches in the cuticle. Oral uptake of fungi can be beneficial to mosquitoes, as yeasts hold nutritional value and support larval development. However, ingestion of or surface contact with fungal entomopathogens leads to colonization of the mosquito with often lethal consequences to the host. The mosquito immune system recognizes fungi and mounts cellular and humoral immune responses in the hemocoel, and possibly epithelial immune responses in the gut. These responses are regulated transcriptionally through multiple signal transduction pathways. Proteolytic protease cascades provide additional regulation of antifungal immunity. Together, these immune responses provide an efficient barrier to fungal infections, which need to be overcome by entomopathogens. Therefore, fungi constitute an excellent tool to examine the molecular underpinnings of mosquito immunity and to identify novel antifungal peptides. In addition, recent advances in mycobiome analyses can now be used to examine the contribution of fungi to various mosquito traits, including vector competence.
Collapse
Affiliation(s)
- P Tawidian
- Division of Biology, Kansas State University, 267 Chalmers Hall, Manhattan, KS, 66506, USA
| | - V L Rhodes
- Missouri Southern State University, Biology Department, Reynolds Hall 220, 3950 E. Newman Rd., Joplin, MO, 64801-1595, USA
| | - K Michel
- Division of Biology, Kansas State University, 267 Chalmers Hall, Manhattan, KS, 66506, USA.
| |
Collapse
|
36
|
Interaction of RNA viruses of the natural virome with the African malaria vector, Anopheles coluzzii. Sci Rep 2019; 9:6319. [PMID: 31004099 PMCID: PMC6474895 DOI: 10.1038/s41598-019-42825-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 04/10/2019] [Indexed: 11/08/2022] Open
Abstract
Mosquitoes are colonized by a little-studied natural virome. Like the bacterial microbiome, the virome also probably influences the biology and immunity of mosquito vector populations, but tractable experimental models are lacking. We recently discovered two novel viruses in the virome of wild Anopheles and in colonies of the malaria vector Anopheles coluzzii: Anopheles C virus and Anopheles cypovirus. Here, we describe biological interactions between these two viruses and An. coluzzii mosquitoes. Viral abundance varies reproducibly during mosquito development. DNA forms of these viruses were not detected, and thus viral persistence is likely based on vertical transmission of RNA genomes. At least Anopheles C virus is vertically transmitted by an intraembryonic route. Relative abundance of the two viruses is inversely correlated in individual mosquitoes. One possible mechanism for this could be interactions with host immunity, and functional genomic analysis indicated differential influence of at least the Toll and JAK/STAT immune signaling pathways upon the viruses. The nonrandom distributions and interactions with host immunity suggest that these and other members of the natural virome may constitute a source of unrecognized heterogeneity in mosquito vector populations.
Collapse
|
37
|
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
|
38
|
Werling K, Shaw WR, Itoe MA, Westervelt KA, Marcenac P, Paton DG, Peng D, Singh N, Smidler AL, South A, Deik AA, Mancio-Silva L, Demas AR, March S, Calvo E, Bhatia SN, Clish CB, Catteruccia F. Steroid Hormone Function Controls Non-competitive Plasmodium Development in Anopheles. Cell 2019; 177:315-325.e14. [PMID: 30929905 DOI: 10.1016/j.cell.2019.02.036] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/14/2018] [Accepted: 02/20/2019] [Indexed: 12/26/2022]
Abstract
Transmission of malaria parasites occurs when a female Anopheles mosquito feeds on an infected host to acquire nutrients for egg development. How parasites are affected by oogenetic processes, principally orchestrated by the steroid hormone 20-hydroxyecdysone (20E), remains largely unknown. Here we show that Plasmodium falciparum development is intimately but not competitively linked to processes shaping Anopheles gambiae reproduction. We unveil a 20E-mediated positive correlation between egg and oocyst numbers; impairing oogenesis by multiple 20E manipulations decreases parasite intensities. These manipulations, however, accelerate Plasmodium growth rates, allowing sporozoites to become infectious sooner. Parasites exploit mosquito lipids for faster growth, but they do so without further affecting egg development. These results suggest that P. falciparum has adopted a non-competitive evolutionary strategy of resource exploitation to optimize transmission while minimizing fitness costs to its mosquito vector. Our findings have profound implications for currently proposed control strategies aimed at suppressing mosquito populations.
Collapse
Affiliation(s)
- Kristine Werling
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - W Robert Shaw
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Maurice A Itoe
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Kathleen A Westervelt
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Perrine Marcenac
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Douglas G Paton
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Duo Peng
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Naresh Singh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Andrea L Smidler
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA
| | - Adam South
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Amy A Deik
- Broad Institute, Cambridge, MA 02142, USA
| | - Liliana Mancio-Silva
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Allison R Demas
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20852, USA
| | - Sangeeta N Bhatia
- Broad Institute, Cambridge, MA 02142, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
39
|
Guo Z, Qin J, Zhou X, Zhang Y. Insect Transcription Factors: A Landscape of Their Structures and Biological Functions in Drosophila and beyond. Int J Mol Sci 2018; 19:ijms19113691. [PMID: 30469390 PMCID: PMC6274879 DOI: 10.3390/ijms19113691] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/17/2022] Open
Abstract
Transcription factors (TFs) play essential roles in the transcriptional regulation of functional genes, and are involved in diverse physiological processes in living organisms. The fruit fly Drosophila melanogaster, a simple and easily manipulated organismal model, has been extensively applied to study the biological functions of TFs and their related transcriptional regulation mechanisms. It is noteworthy that with the development of genetic tools such as CRISPR/Cas9 and the next-generation genome sequencing techniques in recent years, identification and dissection the complex genetic regulatory networks of TFs have also made great progress in other insects beyond Drosophila. However, unfortunately, there is no comprehensive review that systematically summarizes the structures and biological functions of TFs in both model and non-model insects. Here, we spend extensive effort in collecting vast related studies, and attempt to provide an impartial overview of the progress of the structure and biological functions of current documented TFs in insects, as well as the classical and emerging research methods for studying their regulatory functions. Consequently, considering the importance of versatile TFs in orchestrating diverse insect physiological processes, this review will assist a growing number of entomologists to interrogate this understudied field, and to propel the progress of their contributions to pest control and even human health.
Collapse
Affiliation(s)
- Zhaojiang Guo
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Jianying Qin
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
- Longping Branch, Graduate School of Hunan University, Changsha 410125, China.
| | - Xiaomao Zhou
- Longping Branch, Graduate School of Hunan University, Changsha 410125, China.
| | - Youjun Zhang
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
40
|
Shaw WR, Catteruccia F. Vector biology meets disease control: using basic research to fight vector-borne diseases. Nat Microbiol 2018; 4:20-34. [PMID: 30150735 DOI: 10.1038/s41564-018-0214-7] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 06/29/2018] [Indexed: 12/11/2022]
Abstract
Human pathogens that are transmitted by insects are a global problem, particularly those vectored by mosquitoes; for example, malaria parasites transmitted by Anopheles species, and viruses such as dengue, Zika and chikungunya that are carried by Aedes mosquitoes. Over the past 15 years, the prevalence of malaria has been substantially reduced and virus outbreaks have been contained by controlling mosquito vectors using insecticide-based approaches. However, disease control is now threatened by alarming rates of insecticide resistance in insect populations, prompting the need to develop a new generation of specific strategies that can reduce vector-mediated transmission. Here, we review how increased knowledge in insect biology and insect-pathogen interactions is stimulating new concepts and tools for vector control. We focus on strategies that either interfere with the development of pathogens within their vectors or directly impact insect survival, including enhancement of vector-mediated immune control, manipulation of the insect microbiome, or use of powerful new genetic tools such as CRISPR-Cas systems to edit vector genomes. Finally, we offer a perspective on the implementation hurdles as well as the knowledge gaps that must be filled in the coming years to safely realize the potential of these novel strategies to eliminate the scourge of vector-borne disease.
Collapse
Affiliation(s)
- W Robert Shaw
- Harvard T. H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA, USA.
| | - Flaminia Catteruccia
- Harvard T. H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA, USA.
| |
Collapse
|
41
|
Kumar A, Srivastava P, Sirisena P, Dubey SK, Kumar R, Shrinet J, Sunil S. Mosquito Innate Immunity. INSECTS 2018; 9:insects9030095. [PMID: 30096752 PMCID: PMC6165528 DOI: 10.3390/insects9030095] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 06/17/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022]
Abstract
Mosquitoes live under the endless threat of infections from different kinds of pathogens such as bacteria, parasites, and viruses. The mosquito defends itself by employing both physical and physiological barriers that resist the entry of the pathogen and the subsequent establishment of the pathogen within the mosquito. However, if the pathogen does gain entry into the insect, the insect mounts a vigorous innate cellular and humoral immune response against the pathogen, thereby limiting the pathogen's propagation to nonpathogenic levels. This happens through three major mechanisms: phagocytosis, melanization, and lysis. During these processes, various signaling pathways that engage intense mosquito⁻pathogen interactions are activated. A critical overview of the mosquito immune system and latest information about the interaction between mosquitoes and pathogens are provided in this review. The conserved, innate immune pathways and specific anti-pathogenic strategies in mosquito midgut, hemolymph, salivary gland, and neural tissues for the control of pathogen propagation are discussed in detail.
Collapse
Affiliation(s)
- Ankit Kumar
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Priyanshu Srivastava
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Pdnn Sirisena
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Sunil Kumar Dubey
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Ramesh Kumar
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Jatin Shrinet
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Sujatha Sunil
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| |
Collapse
|
42
|
Rhodes VL, Thomas MB, Michel K. The interplay between dose and immune system activation determines fungal infection outcome in the African malaria mosquito, Anopheles gambiae. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 85:125-133. [PMID: 29649553 PMCID: PMC5935592 DOI: 10.1016/j.dci.2018.04.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 05/27/2023]
Abstract
The Toll pathway is a central regulator of antifungal immunity in insects. In mosquitoes, the Toll pathway affects infections with the fungal entomopathogen, Beauveria bassiana, which is considered a potential mosquito biopesticide. We report here the use of B. bassiana strain I93-825 in Anopheles gambiae to analyze the impact of Toll pathway modulation on mosquito survival. Exposure to a narrow dose range of conidia by direct contact decreased mosquito longevity and median survival. In addition, fungal exposure dose correlated positively and linearly with hazard ratio. Increased Toll signaling by knockdown of its inhibitor, cactus, decreased survivorship of uninfected females, increased mosquito survival after low dose B. bassiana exposure, but had little effect following exposure to higher doses. This observed trade-off could have implications for development of B. bassiana as a prospective vector control tool. On the one hand, selection for small increases in mosquito immune signaling across a narrow dose range could impair efficacy of B. bassiana. On the other hand, costs of immunity and the capacity for higher doses of fungus to overwhelm immune responses could limit evolution of resistance.
Collapse
Affiliation(s)
- Victoria L Rhodes
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Matthew B Thomas
- Department of Entomology and Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
43
|
Meng E, Qiao T, Tang B, Hou Y, Yu W, Chen Z. Effects of ovarian fluid, venom and egg surface characteristics of Tetrastichus brontispae (Hymenoptera: Eulophidae) on the immune response of Octodonta nipae (Coleoptera: Chrysomelidae). JOURNAL OF INSECT PHYSIOLOGY 2018; 109:125-137. [PMID: 30025717 DOI: 10.1016/j.jinsphys.2018.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/16/2018] [Accepted: 07/16/2018] [Indexed: 06/08/2023]
Abstract
Although the importance of parasitoids as biocontrol agents has long been recognized, systematic studies of the physiological mechanisms are scarce, especially in those parasitoids that are able to successfully invade their hosts by activating host immune responses. This study explored this phenomenon by investigating the effects of ovarian fluid, venom and egg surface characteristics of Tetrastichus brontispae (Hymenoptera: Eulophidae) on host immunity. The results showed that the injection of venom alone induced higher phenoloxidase activity, while a mixture of ovarian plus venom fluids provoked higher granulocyte and plasmatocyte spreading ratios, highlighting the role that egg surface characteristics may play in successful parasitism. After thorough investigation, the presence of a hemomucin homologue was documented on the egg surface (which was named Tetrastichus brontispae adipocyte plasma membrane associated protein-like, TbAPMAP-like), while the absence of polydnaviruses, fibrous layers and virus-like filaments was confirmed. The higher encapsulation index of eggs incubated with TbAPMAP-like polyclonal antibody demonstrated the protection of the protein against encapsulation. These results contribute to our understanding of the mechanisms used by endoparasitoids to evade encapsulation during the early parasitism stage while enriching our knowledge of local active regulatory mechanisms. It is likely that this is the first study to determine the egg protective properties of TbAPMAP-like in host-parasite systems.
Collapse
Affiliation(s)
- E Meng
- State Key Laboratory of Ecological Pest Control of Fujian-Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Ting Qiao
- State Key Laboratory of Ecological Pest Control of Fujian-Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Baozhen Tang
- State Key Laboratory of Ecological Pest Control of Fujian-Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Youming Hou
- State Key Laboratory of Ecological Pest Control of Fujian-Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| | - Weizhen Yu
- State Key Laboratory of Ecological Pest Control of Fujian-Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Fujian Provincial Key Laboratory of Insect Ecology, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Zhiming Chen
- Fuzhou Entry-Exit Inspection & Quarantine Bureau of P.R.C, Fuzhou, 350002, China
| |
Collapse
|
44
|
Unbiased classification of mosquito blood cells by single-cell genomics and high-content imaging. Proc Natl Acad Sci U S A 2018; 115:E7568-E7577. [PMID: 30038005 PMCID: PMC6094101 DOI: 10.1073/pnas.1803062115] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Mosquito blood cells are immune cells that help control infection by vector-borne pathogens. Despite their importance, little is known about mosquito blood cell biology beyond morphological and functional criteria used for their classification. Here, we combined the power of single-cell RNA sequencing, high-content imaging flow cytometry, and single-molecule RNA hybridization to analyze a subset of blood cells of the malaria mosquito Anopheles gambiae By demonstrating that blood cells express nearly half of the mosquito transcriptome, our dataset represents an unprecedented view into their transcriptional program. Analyses of differentially expressed genes identified transcriptional signatures of two cell types and provide insights into the current classification of these cells. We further demonstrate the active transfer of a cellular marker between blood cells that may confound their identification. We propose that cell-to-cell exchange may contribute to cellular diversity and functional plasticity seen across biological systems.
Collapse
|
45
|
Wang J, Song X, Wang M. Peptidoglycan recognition proteins in hematophagous arthropods. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 83:89-95. [PMID: 29269264 PMCID: PMC5889321 DOI: 10.1016/j.dci.2017.12.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/11/2017] [Accepted: 12/18/2017] [Indexed: 05/24/2023]
Abstract
Hematophagous arthropods are medically important disease vectors that transmit a variety of pathogens. Unlike mammals that employ both innate and adaptive immunity to clear invading pathogens, these vectors rely mainly on an innate immune system to combat pathogens. Peptidoglycan recognition proteins (PGRPs) are important components of innate immune signaling pathways and are responsible for recognizing microbe-associated molecular patterns (MAMPs), thus regulating host immune interactions with both harmful and helpful microbes. Here we review a number of recent studies in different vectors that address the function of PGRPs in immune regulation. Further, we discuss the variation of PGRPs between vectors and Drosophila.
Collapse
Affiliation(s)
- Jingwen Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, PR China.
| | - Xiumei Song
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, PR China
| | - Mengfei Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, PR China
| |
Collapse
|
46
|
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
|
47
|
Gleason-Rodríguez G, Castillo-Méndez M, Maya K, Ramos-Castañeda J, Valverde-Garduño V. Dengue virus infection induces chromatin remodeling at locus AAEL006536 in the midgut of Aedes aegypti. SALUD PUBLICA DE MEXICO 2018; 60:41-47. [PMID: 29689655 DOI: 10.21149/8471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 08/14/2017] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE To identify and characterize Aedes aegypti's AAEL006536 gene proximal upstream cis-regulatory sequences activated by dengue virus infection. MATERIALS AND METHODS A. aegypti Rockefeller strain mosquitoes were blood fed or infected with dengue virus 2. Open chromatinprofiling was then carried out in pools of midguts from each group of mosquitoes. RESULTS The proximal upstream region does not contain open chromatin sites in the midguts of blood-fed mosquitoes as detected by FAIRE-qPCR. In contrast, two cis-regulatory sites were identified in the same upstream region of dengue virus-infected mosquito midguts. The distal sequence contains STAT-, REL- and C/EBP-type transcription factor binding sites. CONCLUSIONS The activation of two proximal cis-regulatory sequences, induced by dengue virus infection, is mediated by chromatin remodeling mechanisms. Binding sites suggest a dengue virus infectioninduced participation of immunity transcription factors in the up-regulation of this gene. This suggests the participation of the AAEL006536 gene in the mosquito's antiviral innate immune response.
Collapse
Affiliation(s)
- Graciela Gleason-Rodríguez
- Departamento de Infección e Inmunidad, Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. México.,Escuela de Salud Pública de México, Instituto Nacional de Salud Pública. México
| | - Manuel Castillo-Méndez
- Departamento de Infección e Inmunidad, Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. México.,Escuela de Salud Pública de México, Instituto Nacional de Salud Pública. México
| | - Krystal Maya
- Departamento de Infección e Inmunidad, Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. México
| | - José Ramos-Castañeda
- Departamento de Infección e Inmunidad, Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. México
| | - Verónica Valverde-Garduño
- Departamento de Infección e Inmunidad, Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. México.,Escuela de Salud Pública de México, Instituto Nacional de Salud Pública. México
| |
Collapse
|
48
|
Pan X, Pike A, Joshi D, Bian G, McFadden MJ, Lu P, Liang X, Zhang F, Raikhel AS, Xi Z. The bacterium Wolbachia exploits host innate immunity to establish a symbiotic relationship with the dengue vector mosquito Aedes aegypti. ISME JOURNAL 2017; 12:277-288. [PMID: 29099491 PMCID: PMC5739022 DOI: 10.1038/ismej.2017.174] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/30/2017] [Accepted: 09/06/2017] [Indexed: 01/15/2023]
Abstract
A host's immune system plays a central role in shaping the composition of the microbiota and, in return, resident microbes influence immune responses. Symbiotic associations of the maternally transmitted bacterium Wolbachia occur with a wide range of arthropods. It is, however, absent from the dengue and Zika vector mosquito Aedes aegypti in nature. When Wolbachia is artificially forced to form symbiosis with this new mosquito host, it boosts the basal immune response and enhances the mosquito's resistance to pathogens, including dengue, Zika virus and malaria parasites. The mechanisms involved in establishing a symbiotic relationship between Wolbachia and A. aegypti, and the long-term outcomes of this interaction, are not well understood. Here, we have demonstrated that both the immune deficiency (IMD) and Toll pathways are activated by the Wolbachia strain wAlbB upon its introduction into A. aegypti. Silencing the Toll and IMD pathways via RNA interference reduces the wAlbB load. Notably, wAlbB induces peptidoglycan recognition protein (PGRP)-LE expression in the carcass of A. aegypti, and its silencing results in a reduction of symbiont load. Using transgenic mosquitoes with stage-specific induction of the IMD and Toll pathways, we have shown that elevated wAlbB infection in these mosquitoes is maintained via maternal transmission. These results indicate that host innate immunity is utilized to establish and promote host-microbial symbiosis. Our results will facilitate a long-term projection of the stability of the Wolbachia-A. aegypti mosquito system that is being developed to control dengue and Zika virus transmission to humans.
Collapse
Affiliation(s)
- Xiaoling Pan
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.,School of Medicine, Hunan Normal University, Changsha, Hunan 410013, China
| | - Andrew Pike
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Deepak Joshi
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Guowu Bian
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Michael J McFadden
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Peng Lu
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Xiao Liang
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Fengrui Zhang
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Alexander S Raikhel
- Department of Entomology and Institute for Integrative Molecular Biology, University of California, Riverside, CA 92521, USA
| | - Zhiyong Xi
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.,Sun Yat-sen University-Michigan State University Joint Center of Vector Control for Tropical Diseases, Guangzhou, Guangdong 510080, China
| |
Collapse
|
49
|
Kumar M, Mohanty AK, Sreenivasamurthy SK, Dey G, Advani J, Pinto SM, Kumar A, Prasad TSK. Response to Blood Meal in the Fat Body of Anopheles stephensi Using Quantitative Proteomics: Toward New Vector Control Strategies Against Malaria. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2017; 21:520-530. [PMID: 28873011 DOI: 10.1089/omi.2017.0092] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Malaria remains a grand challenge for disruptive innovation in global health therapeutics and diagnostics. Anopheles stephensi is one of the major vectors of malaria in Asia. Vector and transmission control are key focus areas in the fight against malaria, a field of postgenomics research where proteomics can play a substantive role. Moreover, to identify novel strategies to control the vector population, it is necessary to understand the vector life processes at a global and molecular scale. In this context, fat body is a vital organ required for vitellogenesis, vector immunity, vector physiology, and vector-parasite interaction. Given its central role in energy metabolism, vitellogenesis, and immune function, the proteome profile of the fat body and the impact of blood meal (BM) ingestion on the protein abundances of this vital organ have not been investigated so far. Therefore, using a proteomics approach, we identified the proteins expressed in the fat body of An. stephensi and their differential expression in response to BM ingestion. In all, we identified 3,218 proteins in the fat body using high-resolution mass spectrometry, of which 483 were found to be differentially expressed in response to the BM ingestion. Bioinformatics analysis of these proteins underscored their role in amino acid metabolism, vitellogenesis, lipid transport, signal peptide processing, mosquito immunity, and oxidation-reduction processes. Interestingly, we identified five novel genes, which were found to be differentially expressed upon BM ingestion. Proteins that exhibited altered expression in the present study are potential targets for vector control strategies and development of transmission blocking vaccines in the fight against malaria.
Collapse
Affiliation(s)
- Manish Kumar
- 1 Institute of Bioinformatics , Bangalore, India .,2 Manipal University , Manipal, India
| | | | | | - Gourav Dey
- 1 Institute of Bioinformatics , Bangalore, India .,2 Manipal University , Manipal, India
| | - Jayshree Advani
- 1 Institute of Bioinformatics , Bangalore, India .,2 Manipal University , Manipal, India
| | - Sneha M Pinto
- 4 YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University , Mangalore, India
| | - Ashwani Kumar
- 3 National Institute of Malaria Research (ICMR) , Panjim, India
| | - Thottethodi Subrahmanya Keshava Prasad
- 1 Institute of Bioinformatics , Bangalore, India .,4 YU-IOB Center for Systems Biology and Molecular Medicine, Yenepoya University , Mangalore, India .,5 NIMHANS-IOB Proteomics and Bioinformatics Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neuro Sciences , Bangalore, India
| |
Collapse
|
50
|
Short SM, Mongodin EF, MacLeod HJ, Talyuli OAC, Dimopoulos G. Amino acid metabolic signaling influences Aedes aegypti midgut microbiome variability. PLoS Negl Trop Dis 2017; 11:e0005677. [PMID: 28753661 PMCID: PMC5549995 DOI: 10.1371/journal.pntd.0005677] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 08/09/2017] [Accepted: 06/02/2017] [Indexed: 12/26/2022] Open
Abstract
The mosquito midgut microbiota has been shown to influence vector competence for multiple human pathogens. The microbiota is highly variable in the field, and the sources of this variability are not well understood, which limits our ability to understand or predict its effects on pathogen transmission. In this work, we report significant variation in female adult midgut bacterial load between strains of A. aegypti which vary in their susceptibility to dengue virus. Composition of the midgut microbiome was similar overall between the strains, with 81-92% of reads coming from the same five bacterial families, though we did detect differences in the presence of some bacterial families including Flavobacteriaceae and Entobacteriaceae. We conducted transcriptomic analysis on the two mosquito strains that showed the greatest difference in bacterial load, and found that they differ in transcript abundance of many genes implicated in amino acid metabolism, in particular the branched chain amino acid degradation pathway. We then silenced this pathway by targeting multiple genes using RNA interference, which resulted in strain-specific bacterial proliferation, thereby eliminating the difference in midgut bacterial load between the strains. This suggests that the branched chain amino acid (BCAA) degradation pathway controls midgut bacterial load, though the mechanism underlying this remains unclear. Overall, our results indicate that amino acid metabolism can act to influence the midgut microbiota. Moreover, they suggest that genetic or physiological variation in BCAA degradation pathway activity may in part explain midgut microbiota variation in the field.
Collapse
Affiliation(s)
- Sarah M. Short
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Emmanuel F. Mongodin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Hannah J. MacLeod
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Octavio A. C. Talyuli
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|