1
|
Samantsidis GR, Karim S, Smith RC. Influence of blood feeding and infection on arthropod hemocytes. CURRENT OPINION IN INSECT SCIENCE 2025; 69:101341. [PMID: 39938680 DOI: 10.1016/j.cois.2025.101341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/14/2025]
Abstract
Blood feeding provides essential nutrients for development and reproduction in hematophagous arthropods yet also initiates significant other physiological alterations in immune function. Immune cells, or hemocytes, are integral components of the arthropod innate immune system with notable roles in defining vector competence. Evidence suggests that both blood feeding and infection drive substantial changes in hemocyte phenotypes, including proliferation, immune activation, and differentiation, which directly and indirectly influence pathogen infection outcomes. These dynamics have fueled extensive research into hemocyte biology in recent years, which aided by emerging single-cell technologies and methods of phagocyte depletion, have provided novel molecular insights into hemocyte populations and additional support for their important contributions to parasite, virus, and bacterial infections. Despite this progress, many aspects of arthropod immune cell biology remain unclear. Focusing on mosquitoes and ticks as two of the most prominent and well-studied arthropod vectors, this review summarizes the effects of blood feeding and infection on mosquito and tick hemocytes, highlighting hemocyte classifications, and the known mechanisms by which hemocytes can have positive or negative impacts on vector-borne pathogen infection.
Collapse
Affiliation(s)
| | - Shahid Karim
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, USA
| | - Ryan C Smith
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA, USA.
| |
Collapse
|
2
|
Kwon H, Smith RC. Protocol to chemically deplete phagocytic hemocytes in Anopheles gambiae using clodronate liposomes. STAR Protoc 2025; 6:103819. [PMID: 40381199 DOI: 10.1016/j.xpro.2025.103819] [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: 02/21/2025] [Revised: 04/02/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025] Open
Abstract
Understanding the roles of phagocytic hemocytes in mosquito innate immunity has been significantly limited due to the lack of genetic tools. Here, we present a protocol for depleting phagocytic hemocytes in Anopheles gambiae mosquitoes using clodronate liposomes. We describe steps for mosquito injection, as well as validation by microscopy, quantitative real-time PCR (real-time qPCR), and flow cytometry analysis. This protocol allows for the delineation of phagocytic hemocyte function in mosquito immunity, which can be more broadly applied to other arthropod systems. For complete details on the use and execution of this protocol, please refer to Kwon et al.1.
Collapse
Affiliation(s)
- Hyeogsun Kwon
- Department of Plant Pathology, Entomology, and Microbiology, Iowa State University, Ames, IA 50011, USA.
| | - Ryan C Smith
- Department of Plant Pathology, Entomology, and Microbiology, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
3
|
Camacho E, Dong Y, Chrissian C, Cordero RJ, Saraiva RG, Anglero-Rodriguez Y, Smith DF, Jacobs E, Hartshorn I, Patiño-Medina JA, DePasquale M, Dziedzic A, Jedlicka A, Smith B, Mlambo G, Tripathi A, Broderick NA, Stark RE, Dimopoulos G, Casadevall A. Dietary L-3,4-dihydroxyphenylalanine (L-DOPA) augments cuticular melanization in Anopheles mosquitos while reducing their lifespan and malaria parasite burden. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.30.615839. [PMID: 40166253 PMCID: PMC11956902 DOI: 10.1101/2024.09.30.615839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
L-3,4-dihydroxyphenylalanine (L-DOPA), a naturally occurring tyrosine derivative, is prevalent in environments that include mosquito habitats, potentially serving as part of their diet. Given its role as a precursor for melanin synthesis we investigate the effect of dietary L-DOPA on mosquito physiology and immunity to Plasmodium falciparum and Cryptococcus neoformans infection. Dietary L-DOPA is incorporated into mosquito melanin via a non-canonical pathway and has a profound transcriptional effect associated with enhanced immunity, increased pigmentation, and reduced lifespan. Increased melanization results in an enhanced capacity to absorb electromagnetic radiation that affects mosquito temperatures. Bacteria in the mosquito microbiome act as sources of dopamine, a substrate for melanization. Our results illustrate how an environmentally abundant amino acid analogue can affect mosquito physiology and suggest its potential usefulness as an environmentally friendly vector control agent to reduce malaria transmission, warranting further research and field studies.
Collapse
|
4
|
Saab SA, Cardoso-Jaime V, Kefi M, Dimopoulos G. Advances in the dissection of Anopheles-Plasmodium interactions. PLoS Pathog 2025; 21:e1012965. [PMID: 40163471 PMCID: PMC11957333 DOI: 10.1371/journal.ppat.1012965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Malaria is a life-threatening mosquito-borne disease caused by the Plasmodium parasite, responsible for more than half a million deaths annually and principally involving children. The successful transmission of malaria by Anopheles mosquitoes relies on complex successive interactions between the parasite and various mosquito organs, host factors, and restriction factors. This review summarizes our current understanding of the mechanisms regulating Plasmodium infection of the mosquito vector at successive plasmodial developmental stages and highlights potential transmission-blocking targets and strategies.
Collapse
Affiliation(s)
- Sally A. Saab
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| | - Victor Cardoso-Jaime
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| | - Mary Kefi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| |
Collapse
|
5
|
Cardoso-Jaime V, Dimopoulos G. Anopheles gambiae phagocytic hemocytes promote Plasmodium falciparum infection by regulating midgut epithelial integrity. Nat Commun 2025; 16:1465. [PMID: 39920122 PMCID: PMC11805967 DOI: 10.1038/s41467-025-56313-y] [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: 05/25/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025] Open
Abstract
For successful transmission, the malaria parasite must traverse tissue epithelia and survive attack from the insect's innate immune system. Hemocytes play a multitude of roles in mosquitoes, including defense against invading pathogens. Here, we show that hemocytes of the major malaria vector Anopheles gambiae promote Plasmodium falciparum infection by maintaining midgut epithelial integrity by controlling cell proliferation upon blood feeding. The mosquito's hemocytes also control the midgut microbiota and immune gene expression. Our study unveils novel hemocyte functions that are exploited by the human malaria parasite to evade the mosquito's immune system.
Collapse
Affiliation(s)
- Victor Cardoso-Jaime
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - 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
|
6
|
Zdybicka-Barabas A, Stączek S, Kunat-Budzyńska M, Cytryńska M. Innate Immunity in Insects: The Lights and Shadows of Phenoloxidase System Activation. Int J Mol Sci 2025; 26:1320. [PMID: 39941087 PMCID: PMC11818254 DOI: 10.3390/ijms26031320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Melanogenesis and melanin deposition are processes essential for the effective immune response of insects to various invaders. Phenoloxidase (PO), produced in specialized cells as an inactive precursor prophenoloxidase (proPO), is the key enzyme for melanin formation. The precursor is activated via limited proteolysis by a dedicated serine proteinase, which is the final element in the cascade of serine proteinases (SPs) that make up the PO system. Melanogenesis provides different cytotoxic molecules active in fighting infections, as well as melanin, which is important for sequestration of invaders. However, since the cytotoxic reactive compounds generated during melanization also pose a threat to host cells, strict control of the PO system is necessary for host self-protection. Different pathogens and parasites influence the PO system and melanization through various strategies, which allow them to survive and develop in the host insect body. In this review, we characterize "the lights and shadows" of PO system activation, indicating, on one hand, its advantages as an efficient and effective mechanism of the insect immune response and, on the other hand, the dangers for the insect host associated with the improper functioning of this system and selected strategies for regulating its activity by entomopathogenic organisms.
Collapse
Affiliation(s)
| | | | | | - Małgorzata Cytryńska
- Department of Immunobiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19 St., 20-033 Lublin, Poland; (A.Z.-B.); (S.S.); (M.K.-B.)
| |
Collapse
|
7
|
Vieira CS, Bisogno S, Salvemini M, Loza Telleria E, Volf P. Azadirachtin disrupts ecdysone signaling and alters sand fly immunity. Parasit Vectors 2024; 17:526. [PMID: 39707409 DOI: 10.1186/s13071-024-06589-8] [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: 09/03/2024] [Accepted: 11/17/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Leishmaniasis is a group of neglected vector-borne diseases transmitted by phlebotomine sand flies. Leishmania parasites must overcome various defenses in the sand fly midgut, including the insects's immune response. Insect immunity is regulated by the ecdysone hormone, which binds to its nuclear receptor (EcR) and activates the transcription of genes involved in insect immunity. However, the role of ecdysone in sand fly immunity has never been studied. Phlebotomus perniciosus is a natural vector of Leishmania infantum; here, we manipulated its neuroendocrine system using azadirachtin (Aza), a natural compound known to affect ecdysone synthesis. METHODS Phlebotomus perniciosus larvae and adult females were fed on food containing either Aza alone or Aza plus ecdysone, and the effects on mortality and ecdysis were evaluated. Genes related to ecdysone signaling and immunity were identified in P. perniciosus, and the expression of antimicrobial peptides (AMPs), EcR, the ecdysone-induced genes Eip74EF and Eip75B, and the transcription factor serpent were analyzed using quantitative polymerase chain reaction (PCR). RESULTS Aza treatment inhibited molting of first-instar (L1) larvae to L2, with only 10% of larvae molting compared to 95% in the control group. Serpent and Eip74EF, attacin, defensin 1, and defensin 2 genes were downregulated by Aza treatment in larvae. Similarly, Aza-treated adult females also presented suppression of ecdysone signaling-related genes and the AMPs attacin and defensin 2. Notably, all gene repression caused by Aza was reversed by adding ecdysone concomitantly with Aza to the larval or female food, indicating that these genes are effective markers for ecdysone repression. CONCLUSIONS These results highlight the critical role of ecdysone in regulating the development and immunity of P. perniciosus, which potentially could interfere with Leishmania infection.
Collapse
Affiliation(s)
- Cecilia Stahl Vieira
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic.
| | - Sara Bisogno
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Marco Salvemini
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Erich Loza Telleria
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
8
|
Samantsidis GR, Smith RC. Exploring new dimensions of immune cell biology in Anopheles gambiae through genetic immunophenotyping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619690. [PMID: 39484609 PMCID: PMC11526922 DOI: 10.1101/2024.10.22.619690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Mosquito immune cells, or hemocytes, are integral components of the innate immune responses that define vector competence. However, the lack of genetic resources has limited their characterization and our understanding of their functional roles in immune signaling. To overcome these challenges, we engineered transgenic Anopheles gambiae that express fluorescent proteins under the control of candidate hemocyte promoters. Following the characterization of five transgenic constructs through gene expression and microscopy-based approaches, we examine mosquito immune cell populations by leveraging advanced spectral imaging flow cytometry. Our results comprehensively map the composition of mosquito hemocytes, classifying them into twelve distinct populations based on size, granularity, ploidy, phagocytic capacity, and the expression of PPO6, SPARC, and LRIM15 genetic markers. Together, our novel use of morphological properties and genetic markers provides increased resolution into our understanding of mosquito hemocytes, highlighting the complexity and plasticity of these immune cell populations, while providing the foundation for deeper investigations into their roles in immunity and pathogen transmission.
Collapse
Affiliation(s)
| | - Ryan C Smith
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, Iowa, USA
| |
Collapse
|
9
|
Camacho E, Dong Y, Chrissian C, Cordero RJ, Saravia RG, Anglero-Rodriguez Y, Smith DF, Jacobs E, Hartshorn I, Patiño-Medina JA, DePasquale M, Dziedzic A, Jedlicka A, Smith B, Mlambo G, Tripathi A, Broderick NA, Stark RE, Dimopoulos G, Casadevall A. Dietary L-3,4-dihydroxyphenylalanine (L-DOPA) augments cuticular melanization in Anopheles mosquitos while reducing their lifespan and malaria parasite burden. RESEARCH SQUARE 2024:rs.3.rs-5167892. [PMID: 39483913 PMCID: PMC11527263 DOI: 10.21203/rs.3.rs-5167892/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
L-3,4-dihydroxyphenylalanine (L-DOPA), a naturally occurring tyrosine derivative, is prevalent in environments that include mosquito habitats, potentially serving as part of their diet. Given its role as a precursor for melanin synthesis we investigated the effect of dietary L-DOPA on mosquito physiology and immunity to Plasmodium falciparum and Cryptococcus neoformans infection. Dietary L-DOPA was incorporated into mosquito melanin via a non-canonical pathway and had profound transcriptional effects that were associated with enhanced immunity, increased pigmentation, and reduced lifespan. Increased melanization resulted in an enhanced capacity to absorb electromagnetic radiation that affected mosquito temperatures. Bacteria in the mosquito microbiome were sources of dopamine, which is a substrate for melanization. Our results illustrate how an environmentally abundant amino acid analogue can affect mosquito physiology and suggest its potential usefulness as an environmentally friendly vector control agent to reduce malaria transmission, warranting further research and field studies.
Collapse
|
10
|
Ratcliffe NA. Back to the future: Forgotten protocols for optimizing the isolation of arthropod haemocytes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 159:105223. [PMID: 38960294 DOI: 10.1016/j.dci.2024.105223] [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: 05/13/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024]
Abstract
Consideration is given to previous and more recent protocols for harvesting arthropod haemocytes from Galleria, Drosophila, mosquitoes, Limulus and crustaceans. The optimal harvesting of these cells is essential for meaningful studies of invertebrate immunity in vitro. The results of such experiments, however, have often been flawed due to a lack of understanding of the fragile nature of arthropod haemocytes on exposure to bacterial lipopolysaccharides, resulting in the aggregation and loss of cell types during haemolymph clotting. This article emphasizes that although there are similarities between mammalian neutrophils and arthropod haemocytes, the protocols required for the successful harvesting of these cells vary significantly. The various stages for the successful harvesting of arthropod haemocytes are described in detail and should provide invaluable advice to those requiring both high cell viability and recovery of the different cell types for subsequent experimentation.
Collapse
Affiliation(s)
- Norman A Ratcliffe
- Biology Institute, Universidade Federal Fluminense, Niterói, RJ, 24210-130, Brazil; Department of Biosciences, Swansea University, Singleton Park, Swansea, SA28PP, UK.
| |
Collapse
|
11
|
Mameli E, Samantsidis GR, Viswanatha R, Kwon H, Hall DR, Butnaru M, Hu Y, Mohr SE, Perrimon N, Smith RC. A genome-wide CRISPR screen in Anopheles mosquito cells identifies essential genes and required components of clodronate liposome function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614595. [PMID: 39386635 PMCID: PMC11463579 DOI: 10.1101/2024.09.24.614595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Anopheles mosquitoes are the sole vector of human malaria, the most burdensome vector-borne disease worldwide. Strategies aimed at reducing mosquito populations and limiting their ability to transmit disease show the most promise for disease control. Therefore, gaining an improved understanding of mosquito biology, and specifically that of the immune response, can aid efforts to develop new approaches that limit malaria transmission. Here, we use a genome-wide CRISPR screening approach for the first time in mosquito cells to identify essential genes in Anopheles and identify genes for which knockout confers resistance to clodronate liposomes, which have been widely used in mammals and arthropods to ablate immune cells. In the essential gene screen, we identified a set of 1280 Anopheles genes that are highly enriched for genes involved in fundamental cell processes. For the clodronate liposome screen, we identified several candidate resistance factors and confirm their roles in the uptake and processing of clodronate liposomes through in vivo validation in Anopheles gambiae, providing new mechanistic detail of phagolysosome formation and clodronate liposome function. In summary, we demonstrate the application of a genome-wide CRISPR knockout platform in a major malaria vector and the identification of genes that are important for fitness and immune-related processes.
Collapse
Affiliation(s)
- Enzo Mameli
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - George-Rafael Samantsidis
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA 50011, USA
| | - Raghuvir Viswanatha
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Hyeogsun Kwon
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA 50011, USA
| | - David R. Hall
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA 50011, USA
| | - Matthew Butnaru
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Stephanie E. Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
- HHMI, Harvard Medical School, Boston, MA, 02115, USA
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
12
|
Amaya Romero JE, Chenal C, Ben Chehida Y, Miles A, Clarkson CS, Pedergnana V, Wertheim B, Fontaine MC. Mitochondrial Variation in Anopheles gambiae and Anopheles coluzzii: Phylogeographic Legacy and Mitonuclear Associations With Metabolic Resistance to Pathogens and Insecticides. Genome Biol Evol 2024; 16:evae172. [PMID: 39226386 PMCID: PMC11370803 DOI: 10.1093/gbe/evae172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 09/05/2024] Open
Abstract
Mitochondrial DNA has been a popular marker in phylogeography, phylogeny, and molecular ecology, but its complex evolution is increasingly recognized. Here, we investigated mitochondrial DNA variation in Anopheles gambiae and Anopheles coluzzii, in relation to other species in the Anopheles gambiae complex, by assembling the mitogenomes of 1,219 mosquitoes across Africa. The mitochondrial DNA phylogeny of the Anopheles gambiae complex was consistent with previously reported highly reticulated evolutionary history, revealing important discordances with the species tree. The three most widespread species (An. gambiae, An. coluzzii, and Anopheles arabiensis), known for extensive historical introgression, could not be discriminated based on mitogenomes. Furthermore, a monophyletic clustering of the three saltwater-tolerant species (Anopheles merus, Anopheles melas, and Anopheles bwambae) in the Anopheles gambiae complex also suggested that introgression and possibly selection shaped mitochondrial DNA evolution. Mitochondrial DNA variation in An. gambiae and An. coluzzii across Africa revealed significant partitioning among populations and species. A peculiar mitochondrial DNA lineage found predominantly in An. coluzzii and in the hybrid taxon of the African "far-west" exhibited divergence comparable to the interspecies divergence in the Anopheles gambiae complex, with a geographic distribution matching closely An. coluzzii's geographic range. This phylogeographic relict of the An. coluzzii and An. gambiae split was associated with population and species structure, but not with the rare Wolbachia occurrence. The lineage was significantly associated with single nucleotide polymorphisms in the nuclear genome, particularly in genes associated with pathogen and insecticide resistance. These findings underline potential mitonuclear coevolution history and the role played by mitochondria in shaping metabolic responses to pathogens and insecticides in Anopheles.
Collapse
Affiliation(s)
- Jorge E Amaya Romero
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen 9747 AG, Netherlands
- MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
| | - Clothilde Chenal
- MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
- Institut des Science de l’Évolution de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Yacine Ben Chehida
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen 9747 AG, Netherlands
- Ecology and Evolutionary Biology, School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Alistair Miles
- Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | | | | | - Bregje Wertheim
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen 9747 AG, Netherlands
| | - Michael C Fontaine
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen 9747 AG, Netherlands
- MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
| |
Collapse
|
13
|
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
|
14
|
Schinkel M, Bousema T, van Rij RP. Tripartite interactions between viruses, parasites, and mosquitoes. CURRENT OPINION IN INSECT SCIENCE 2024; 64:101222. [PMID: 38908822 DOI: 10.1016/j.cois.2024.101222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
Mosquito-borne diseases have a major impact on global human health. Biological agents that colonize the mosquito vector are increasingly explored as an intervention strategy to prevent vector-borne disease transmission. For instance, the release of mosquitoes carrying the endosymbiotic bacterium Wolbachia effectively reduced dengue virus incidence and disease. Insect-specific viruses are likewise considered as biocontrol agents against vector-borne diseases. While most studies focused on insect-specific viruses as an intervention against arthropod-borne viruses, we here consider whether mosquito-specific viruses may affect the transmission of the malaria-causing Plasmodium parasite by Anopheles mosquitoes. Although there is no direct experimental evidence addressing this question, we found that viral infections in dipteran insects activate some of the immune pathways that are antiparasitic in Anopheles. These findings suggest that indirect virus-parasite interactions could occur and that insect-specific viruses may modulate malaria transmission. Tripartite interactions between viruses, parasites, and Anopheles mosquitoes thus merit further investigation.
Collapse
Affiliation(s)
- Michelle Schinkel
- Department of Medical Microbiology, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, the Netherlands.
| |
Collapse
|
15
|
Samantsidis GR, Kwon H, Wendland M, Fonder C, Smith RC. TNF signaling mediates cellular immune function and promotes malaria parasite killing in the mosquito Anopheles gambiae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592209. [PMID: 38746363 PMCID: PMC11092648 DOI: 10.1101/2024.05.02.592209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Tumor Necrosis Factor-α (TNF-α) is a proinflammatory cytokine and a master regulator of immune cell function in vertebrates. While previous studies have implicated TNF signaling in invertebrate immunity, the roles of TNF in mosquito innate immunity and vector competence have yet to be explored. Herein, we confirm the identification of a conserved TNF-α pathway in Anopheles gambiae consisting of the TNF-α ligand, Eiger, and its cognate receptors Wengen and Grindelwald. Through gene expression analysis, RNAi, and in vivo injection of recombinant TNF-α, we provide direct evidence for the requirement of TNF signaling in regulating mosquito immune cell function by promoting granulocyte midgut attachment, increased granulocyte abundance, and oenocytoid rupture. Moreover, our data demonstrate that TNF signaling is an integral component of anti-Plasmodium immunity that limits malaria parasite survival. Together, our data support the existence of a highly conserved TNF signaling pathway in mosquitoes that mediates cellular immunity and influences Plasmodium infection outcomes, offering potential new approaches to interfere with malaria transmission by targeting the mosquito host.
Collapse
Affiliation(s)
| | - Hyeogsun Kwon
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA, USA
| | - Megan Wendland
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA, USA
| | - Catherine Fonder
- Molecular, Cellular and Developmental Biology Interdepartmental Graduate Program, Iowa State University, Ames, IA, USA
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, IA, USA
| |
Collapse
|
16
|
Hall DR, Johnson RM, Kwon H, Ferdous Z, Laredo-Tiscareño SV, Blitvich BJ, Brackney DE, Smith RC. Mosquito immune cells enhance dengue and Zika virus dissemination in Aedes aegypti. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587950. [PMID: 38617257 PMCID: PMC11014501 DOI: 10.1101/2024.04.03.587950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Mosquito-borne viruses cause more than 400 million annual infections and place over half of the world's population at risk. Despite this importance, the mechanisms by which arboviruses infect the mosquito host and disseminate to tissues required for transmission are not well understood. Here, we provide evidence that mosquito immune cells, known as hemocytes, play an integral role in the dissemination of dengue virus (DENV) and Zika virus (ZIKV) in the mosquito Aedes aegypti. We establish that phagocytic hemocytes are a focal point for virus infection and demonstrate that these immune cell populations facilitate virus dissemination to the ovaries and salivary glands. Additional transfer experiments confirm that virus-infected hemocytes confer a virus infection to non-infected mosquitoes more efficiently than free virus in acellular hemolymph, revealing that hemocytes are an important tropism to enhance virus dissemination in the mosquito host. These data support a "trojan horse" model of virus dissemination where infected hemocytes transport virus through the hemolymph to deliver virus to mosquito tissues required for transmission and parallels vertebrate systems where immune cell populations promote virus dissemination to secondary sites of infection. In summary, this study significantly advances our understanding of virus infection dynamics in mosquitoes and highlights conserved roles of immune cells in virus dissemination across vertebrate and invertebrate systems.
Collapse
Affiliation(s)
- David R. Hall
- Interdepartmental Program in Genetics and Genomics, Iowa State University, Ames, Iowa
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, Iowa
| | - Rebecca M. Johnson
- Center for Vector-Borne and Zoonotic Diseases, Department of Entomology, The Connecticut Agricultural Experiment Station, New Haven, Connecticut
| | - Hyeogsun Kwon
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, Iowa
| | - Zannatul Ferdous
- Center for Vector-Borne and Zoonotic Diseases, Department of Entomology, The Connecticut Agricultural Experiment Station, New Haven, Connecticut
| | | | - Bradley J. Blitvich
- Department of Veterinary Microbiology and Preventative Medicine, Iowa State University, Ames, Iowa
| | - Doug E. Brackney
- Center for Vector-Borne and Zoonotic Diseases, Department of Entomology, The Connecticut Agricultural Experiment Station, New Haven, Connecticut
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, Iowa
| |
Collapse
|
17
|
Adegoke A, Ribeiro JMC, Brown S, Smith RC, Karim S. Rickettsia parkeri hijacks tick hemocytes to manipulate cellular and humoral transcriptional responses. Front Immunol 2023; 14:1094326. [PMID: 36845157 PMCID: PMC9950277 DOI: 10.3389/fimmu.2023.1094326] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/16/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Blood-feeding arthropods rely on robust cellular and humoral immunity to control pathogen invasion and replication. Tick hemocytes produce factors that can facilitate or suppress microbial infection and pathogenesis. Despite the importance of hemocytes in regulating microbial infection, understanding of their basic biology and molecular mechanisms remains limited. Methods Here we combined histomorphology and functional analysis to identify five distinct phagocytic and non-phagocytic hemocyte populations circulating within the Gulf Coast tick Amblyomma maculatum. Results and discussion Depletion of phagocytic hemocytes using clodronate liposomes revealed their function in eliminating bacterial infection. We provide the first direct evidence that an intracellular tick-borne pathogen, Rickettsia parkeri, infects phagocytic hemocytes in Am. maculatum to modify tick cellular immune responses. A hemocyte-specific RNA-seq dataset generated from hemocytes isolated from uninfected and R. parkeri-infected partially blood-fed ticks generated ~40,000 differentially regulated transcripts, >11,000 of which were immune genes. Silencing two differentially regulated phagocytic immune marker genes (nimrod B2 and eater-two Drosophila homologs), significantly reduced hemocyte phagocytosis. Conclusion Together, these findings represent a significant step forward in understanding how hemocytes regulate microbial homeostasis and vector competence.
Collapse
Affiliation(s)
- Abdulsalam Adegoke
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Jose M. C. Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Sidney Brown
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology, and Microbiology, Iowa State University, Ames, IA, United States
| | - Shahid Karim
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| |
Collapse
|
18
|
Mishra R, Hua G, Bagal UR, Champagne DE, Adang MJ. Anopheles gambiae strain (Ag55) cultured cells originated from Anopheles coluzzii and are phagocytic with hemocyte-like gene expression. INSECT SCIENCE 2022; 29:1346-1360. [PMID: 35358364 DOI: 10.1111/1744-7917.13036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 02/21/2022] [Accepted: 03/07/2022] [Indexed: 06/14/2023]
Abstract
Anopheles gambiae and Anopheles coluzzii are closely related species that are predominant vectors of malaria in Africa. Recently, A. gambiae form M was renamed A. coluzzii and we now conclude on the basis of a diagnostic PCR-restriction fragment length polymorphism assay that Ag55 cells were derived from A. coluzzii. We established an Ag55 cell transcriptome, and KEGG pathway analysis showed that Ag55 cells are enriched in phagosome pathway transcripts. The Ag55 transcriptome has an abundance of specific transcripts characteristic of mosquito hemocytes. Functional E. coli bioparticle uptake experiments visualized by fluorescence microscopy and confocal microscopy and quantified by flow cytometry establish the phagocytic competence of Ag55 cells. Results from this investigation of Ag55 cell properties will guide researchers in the use and engineering of the Ag55 cell line to better enable investigations of Plasmodium, other microbes, and insecticidal toxins.
Collapse
Affiliation(s)
- Ruchir Mishra
- Department of Entomology, College of Agricultural and Environmental Sciences, University of Georgia, Athens, Georgia, USA
- Department of Entomology and Nematology, University of Florida, Gainesville, Florida, USA
| | - Gang Hua
- Department of Entomology, College of Agricultural and Environmental Sciences, University of Georgia, Athens, Georgia, USA
| | - Ujwal R Bagal
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, USA
| | - Donald E Champagne
- Department of Entomology, College of Agricultural and Environmental Sciences, University of Georgia, Athens, Georgia, USA
| | - Michael J Adang
- Department of Entomology, College of Agricultural and Environmental Sciences, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
19
|
Bruno D, Montali A, Gariboldi M, Wrońska AK, Kaczmarek A, Mohamed A, Tian L, Casartelli M, Tettamanti G. Morphofunctional characterization of hemocytes in black soldier fly larvae. INSECT SCIENCE 2022. [PMID: 36065570 DOI: 10.1111/1744-7917.13111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/03/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
In insects, the cell-mediated immune response involves an active role of hemocytes in phagocytosis, nodulation, and encapsulation. Although these processes have been well documented in multiple species belonging to different insect orders, information concerning the immune response, particularly the hemocyte types and their specific function in the black soldier fly Hermetia illucens, is still limited. This is a serious gap in knowledge given the high economic relevance of H. illucens larvae in waste management strategies and considering that the saprophagous feeding habits of this dipteran species have likely shaped its immune system to efficiently respond to infections. The present study represents the first detailed characterization of black soldier fly hemocytes and provides new insights into the cell-mediated immune response of this insect. In particular, in addition to prohemocytes, we identified five hemocyte types that mount the immune response in the larva, and analyzed their behavior, role, and morphofunctional changes in response to bacterial infection and injection of chromatographic beads. Our results demonstrate that the circulating phagocytes in black soldier fly larvae are plasmatocytes. These cells also take part in nodulation and encapsulation with granulocytes and lamellocyte-like cells, developing a starting core for nodule/capsule formation to remove/encapsulate large bacterial aggregates/pathogens from the hemolymph, respectively. These processes are supported by the release of melanin precursors from crystal cells and likely by mobilizing nutrient reserves in newly circulating adipohemocytes, which could thus trophically support other hemocytes during the immune response. Finally, the regulation of the cell-mediated immune response by eicosanoids was investigated.
Collapse
Affiliation(s)
- Daniele Bruno
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Aurora Montali
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Marzia Gariboldi
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Anna Katarzyna Wrońska
- Host Parasites Molecular Interaction Research Unit, Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
| | - Agata Kaczmarek
- Host Parasites Molecular Interaction Research Unit, Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
| | - Amr Mohamed
- Department of Entomology, Faculty of Science, Cairo University, Giza, Egypt
| | - Ling Tian
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Morena Casartelli
- Department of Biosciences, University of Milano, Milano, Italy
- Interuniversity Center for Studies on Bioinspired Agro-environmental Technology (BAT Center), University of Napoli Federico II, Portici, Italy
| | - Gianluca Tettamanti
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Interuniversity Center for Studies on Bioinspired Agro-environmental Technology (BAT Center), University of Napoli Federico II, Portici, Italy
| |
Collapse
|
20
|
Polenogova OV, Noskov YA, Artemchenko AS, Zhangissina S, Klementeva TN, Yaroslavtseva ON, Khodyrev VP, Kruykova NA, Glupov VV. Citrobacter freundii, a natural associate of the Colorado potato beetle, increases larval susceptibility to Bacillus thuringiensis. PEST MANAGEMENT SCIENCE 2022; 78:3823-3835. [PMID: 35238478 DOI: 10.1002/ps.6856] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND We assume that certain representatives of gut microflora mediate immune changes during dysbiosis, accelerating septicemia caused by Bacillus thuringiensis. RESULTS Co-introduction of Citrobacter freundii with Bacillus thuringiensis var. tenebrionis (morrisoni) (Bt) led to an increase in Colorado potato beetle (CPB) larval mortality to 69.0% (1.3-5×) and a synergistic effect was observed from day 1 to day 6. Ultrathin sections of the CPB midgut showed autophagosome formation and partial destruction of gut microvilli under the influence of Bt, which was accompanied by pronounced hypersecretion of the endoplasmic reticulum with apocrine vesicle formation and oncotic changes in cells under the action of C. freundii. The destruction of gut tissues was accompanied by suppression of detoxification processes under the action of the bacteria and a decrease (2.8-3.5×) in the concentration of lipid oxidation products during Bt infection. In the first hours post combined treatment, we registered a slight increase in the total hemocyte count (THC) especially a predomination (1.4×) of immune-competent plasmatocytes. Oral administration of symbiotic and entomopathogenic bacteria to the CPB larvae significantly decreased the THC (1.4×) after 24 h and increased (1.1-1.5×) the detoxifying enzymes level in the lymph. These changes are likely to be associated with the destruction of hemocytes and the need to remove the toxic products of reactive oxygen species. CONCLUSION The obtained results indicate that feeding of C. freundii and B. thuringiensis to the CPB larvae is accompanied by tissue changes that significantly affect the cellular and humoral immunity of the insect, increasing its susceptibility to Bt. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Olga V Polenogova
- Institute of Systematics and Ecology of Animals, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Yury A Noskov
- Institute of Systematics and Ecology of Animals, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
- National Research Tomsk State University, Tomsk, Russia
| | - Anna S Artemchenko
- Institute of Systematics and Ecology of Animals, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Saule Zhangissina
- Institute of Systematics and Ecology of Animals, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Tatyana N Klementeva
- Institute of Systematics and Ecology of Animals, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Olga N Yaroslavtseva
- Institute of Systematics and Ecology of Animals, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Viktor P Khodyrev
- Institute of Systematics and Ecology of Animals, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Natalya A Kruykova
- Institute of Systematics and Ecology of Animals, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Viktor V Glupov
- Institute of Systematics and Ecology of Animals, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
21
|
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: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [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. Circumsporozoite protein (CSP), the major surface protein of Plasmodium sporozoites, is important for parasite targeting to mosquito salivary glands and the mammalian liver. Here, Zhu et al. show that CSP is required for rodent malaria oocysts to evade mosquito immunity by inducing hemocyte nitration and causing subsequent defects in sporozoite-release from oocysts.
Collapse
|
22
|
Kwon H, Mohammed M, Franzén O, Ankarklev J, Smith RC. Single-cell analysis of mosquito hemocytes identifies signatures of immune cell subtypes and cell differentiation. eLife 2021; 10:66192. [PMID: 34318744 PMCID: PMC8376254 DOI: 10.7554/elife.66192] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
Mosquito immune cells, known as hemocytes, are integral to cellular and humoral responses that limit pathogen survival and mediate immune priming. However, without reliable cell markers and genetic tools, studies of mosquito immune cells have been limited to morphological observations, leaving several aspects of their biology uncharacterized. Here, we use single-cell RNA sequencing (scRNA-seq) to characterize mosquito immune cells, demonstrating an increased complexity to previously defined prohemocyte, oenocytoid, and granulocyte subtypes. Through functional assays relying on phagocytosis, phagocyte depletion, and RNA-FISH experiments, we define markers to accurately distinguish immune cell subtypes and provide evidence for immune cell maturation and differentiation. In addition, gene-silencing experiments demonstrate the importance of lozenge in defining the mosquito oenocytoid cell fate. Together, our scRNA-seq analysis provides an important foundation for future studies of mosquito immune cell biology and a valuable resource for comparative invertebrate immunology.
Collapse
Affiliation(s)
- Hyeogsun Kwon
- Department of Entomology, Iowa State University, Ames, United States
| | - Mubasher Mohammed
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Oscar Franzén
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Novum, Huddinge, Sweden
| | - Johan Ankarklev
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.,Microbial Single Cell Genomics facility, SciLifeLab, Biomedical Center (BMC) Uppsala University, Uppsala, Sweden
| | - Ryan C Smith
- Department of Entomology, Iowa State University, Ames, United States
| |
Collapse
|
23
|
Kumari S, Chauhan C, Tevatiya S, Singla D, De TD, Sharma P, Thomas T, Rani J, Savargaonkar D, Pandey KC, Pande V, Dixit R. Genetic changes of Plasmodium vivax tempers host tissue-specific responses in Anopheles stephensi. CURRENT RESEARCH IN IMMUNOLOGY 2021; 2:12-22. [PMID: 35492403 PMCID: PMC9040150 DOI: 10.1016/j.crimmu.2021.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/13/2021] [Accepted: 02/14/2021] [Indexed: 02/02/2023] Open
Abstract
Recently, we showed how an early restriction of gut flora proliferation by Plasmodium vivax favors immune-suppression and Plasmodium survival in the gut lumen (Sharma et al., 2020). Here, we asked post gut invasion how P. vivax interacts with individual tissues such as the midgut, hemocyte, and salivary glands, and manages its survival in the mosquito host. Our data from tissue-specific comparative RNA-Seq analysis and extensive temporal/spatial expression profiling of selected mosquito transcripts in the uninfected and P. vivax infected mosquito's tissues indicated that (i) a transient suppression of gut metabolic machinery by early oocysts; (ii) enriched expression of nutritional responsive proteins and immune proteins against late oocysts, together may ensure optimal parasite development and gut homeostasis restoration; (iii) pre-immune activation of hemocyte by early gut-oocysts infection via REL induction (p < 0.003); and altered expression of hemocyte-encoded immune proteins may cause rapid removal of free circulating sporozoites from hemolymph; (iv) while a strong suppression of salivary metabolic activities, and elevated expression of salivary specific secretory, as well as immune proteins together, may favor the long-term storage and survival of invaded sporozoites. Finally, our RNA-Seq-based discovery of 4449 transcripts of Plasmodium vivax origin, and their developmental stage-specific expression modulation in the corresponding infected mosquito tissues, predicts a possible mechanism of mosquito responses evasion by P. vivax. Conclusively, our system-wide RNA-Seq analysis provides the first genetic evidence of direct mosquito-Plasmodium interaction and establishes a functional correlation.
Collapse
Affiliation(s)
- Seena Kumari
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Charu Chauhan
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Sanjay Tevatiya
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Deepak Singla
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
- School of Agricultural Biotechnology, Punjab Agricultural University, Ludhiana, Punjab, India
| | - Tanwee Das De
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Punita Sharma
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Tina Thomas
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Jyoti Rani
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
- Bio and Nanotechnology Department, Guru Jambheshwar University of Science and Technology, Haryana, India
| | - Deepali Savargaonkar
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Kailash C. Pandey
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Veena Pande
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, India
| | - Rajnikant Dixit
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| |
Collapse
|
24
|
Ramesh Kumar J, Smith JP, Kwon H, Smith RC. Use of Clodronate Liposomes to Deplete Phagocytic Immune Cells in Drosophila melanogaster and Aedes aegypti. Front Cell Dev Biol 2021; 9:627976. [PMID: 33604338 PMCID: PMC7884637 DOI: 10.3389/fcell.2021.627976] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
The innate immune system is the primary defense response to limit invading pathogens for all invertebrate species. In insects, immune cells are central to both cellular and humoral immune responses, however few genetic resources exist beyond Drosophila to study immune cell function. Therefore, the development of innovative tools that can be widely applied to a variety of insect systems is of importance to advance the study of insect immunity. Here, we have adapted the use of clodronate liposomes (CLD) to deplete phagocytic immune cells in the vinegar fly, Drosophila melanogaster, and the yellow fever mosquito, Aedes aegypti. Through microscopy and molecular techniques, we validate the depletion of phagocytic cell populations in both insect species and demonstrate the integral role of phagocytes in combating bacterial pathogens. Together, these data demonstrate the wide utility of CLD in insect systems to advance the study of phagocyte function in insect innate immunity.
Collapse
Affiliation(s)
- Jyothsna Ramesh Kumar
- Interdepartmental Graduate Program in Immunobiology, Iowa State University, Ames, IA, United States.,Department of Entomology, Iowa State University, Ames, IA, United States
| | - Jessica P Smith
- Department of Entomology, Iowa State University, Ames, IA, United States
| | - Hyeogsun Kwon
- Department of Entomology, Iowa State University, Ames, IA, United States
| | - Ryan C Smith
- Department of Entomology, Iowa State University, Ames, IA, United States
| |
Collapse
|
25
|
Kumari S, Chauhan C, Tevatiya S, Singla D, De TD, Sharma P, Thomas T, Rani J, Savargaonkar D, Pandey KC, Pande V, Dixit R. Genetic changes of Plasmodium vivax tempers host tissue-specific responses in Anopheles stephensi. CURRENT RESEARCH IN IMMUNOLOGY 2021. [DOI: https:/doi.org/10.1016/j.crimmu.2021.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
26
|
Kumari S, Chauhan C, Tevatiya S, Singla D, De TD, Sharma P, Thomas T, Rani J, Savargaonkar D, Pandey KC, Pande V, Dixit R. Genetic changes of Plasmodium vivax tempers host tissue-specific responses in Anopheles stephensi. CURRENT RESEARCH IN IMMUNOLOGY 2021. [DOI: https://doi.org/10.1016/j.crimmu.2021.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
27
|
Gioseffi A, Hamerly T, Van K, Zhang N, Dinglasan RR, Yates PA, Kima PE. Leishmania-infected macrophages release extracellular vesicles that can promote lesion development. Life Sci Alliance 2020; 3:3/12/e202000742. [PMID: 33122174 PMCID: PMC7652379 DOI: 10.26508/lsa.202000742] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 12/12/2022] Open
Abstract
Macrophages infected with Leishmania donovani release extracellular vesicles that are composed of parasite and host-derived molecules that have the potential to induce vascular changes in tissues. Leishmania donovani infection of macrophages results in quantitative and qualitative changes in the protein profile of extracellular vesicles (EVs) released by the infected host cells. We confirmed mass spectrometry results orthogonally by performing Western blots for several Leishmania-infected macrophage-enriched EVs (LieEVs) molecules. Several host cell proteins in LieEVs have been implicated in promoting vascular changes in other systems. We also identified 59 parasite-derived proteins in LieEVs, including a putative L. donovani homolog of mammalian vasohibins (LdVash), which in mammals promotes angiogenesis. We developed a transgenic parasite that expressed an endogenously tagged LdVash/mNeonGreen (mNG) and confirmed that LdVash/mNG is indeed expressed in infected macrophages and in LieEVs. We further observed that LieEVs induce endothelial cells to release angiogenesis promoting mediators including IL-8, G-CSF/CSF-3, and VEGF-A. In addition, LieEVs induce epithelial cell migration and tube formation by endothelial cells in surrogate angiogenesis assays. Taken together, these studies show that Leishmania infection alters the composition of EVs from infected cells and suggest that LieEVs may play a role in the promotion of vascularization of Leishmania infections.
Collapse
Affiliation(s)
- Anna Gioseffi
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Tim Hamerly
- Emerging Pathogens Institute and Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, USA
| | - Kha Van
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Naixin Zhang
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Rhoel R Dinglasan
- Emerging Pathogens Institute and Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, USA
| | - Phillip A Yates
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR, USA
| | - Peter E Kima
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| |
Collapse
|
28
|
Prediction of malaria transmission drivers in Anopheles mosquitoes using artificial intelligence coupled to MALDI-TOF mass spectrometry. Sci Rep 2020; 10:11379. [PMID: 32647135 PMCID: PMC7347643 DOI: 10.1038/s41598-020-68272-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/16/2020] [Indexed: 11/21/2022] Open
Abstract
Vector control programmes are a strategic priority in the fight against malaria. However, vector control interventions require rigorous monitoring. Entomological tools for characterizing malaria transmission drivers are limited and are difficult to establish in the field. To predict Anopheles drivers of malaria transmission, such as mosquito age, blood feeding and Plasmodium infection, we evaluated artificial neural networks (ANNs) coupled to matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry (MS) and analysed the impact on the proteome of laboratory-reared Anopheles stephensi mosquitoes. ANNs were sensitive to Anopheles proteome changes and specifically recognized spectral patterns associated with mosquito age (0–10 days, 11–20 days and 21–28 days), blood feeding and P. berghei infection, with best prediction accuracies of 73%, 89% and 78%, respectively. This study illustrates that MALDI-TOF MS coupled to ANNs can be used to predict entomological drivers of malaria transmission, providing potential new tools for vector control. Future studies must assess the field validity of this new approach in wild-caught adult Anopheles. A similar approach could be envisaged for the identification of blood meal source and the detection of insecticide resistance in Anopheles and to other arthropods and pathogens.
Collapse
|
29
|
Potts R, King JG, Pietri JE. Ex vivo characterization of the circulating hemocytes of bed bugs and their responses to bacterial exposure. J Invertebr Pathol 2020; 174:107422. [PMID: 32526226 PMCID: PMC9254597 DOI: 10.1016/j.jip.2020.107422] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 11/17/2022]
Abstract
Bed bugs (Cimex spp.) are urban pests of global importance. Knowledge of the immune system of bed bugs has implications for understanding their susceptibility to biological control agents, their potential to transmit human pathogens, and the basic comparative immunology of insects. Nonetheless, the immunological repertoire of the family Cimicidae remains poorly characterized. Here, we use microscopy, flow cytometry, and RNA sequencing to provide a basal characterization of the circulating hemocytes of the common bed bug, Cimex lectularius. We also examine the responses of these specialized cells to E. coli exposure using the same techniques. Our results show that circulating hemocytes are comprised of at least four morphologically distinct cell types that are capable of phagocytosis, undergo degranulation, and exhibit additional markers of activation following stimulation, including size shift and DNA replication. Furthermore, transcriptomic profiling reveals expression of predicted Toll/IMD signaling pathway components, antimicrobial effectors and other potentially immunoresponsive genes in these cells. Together, our data demonstrate the conservation of several canonical cellular immune responses in the common bed bug and provide a foundation for additional mechanistic immunological studies with specific pathogens of interest.
Collapse
Affiliation(s)
- Rashaun Potts
- University of South Dakota, Sanford School of Medicine, Division of Basic Biomedical Sciences, Vermillion, SD, United States
| | - Jonas G King
- Mississippi State University, Department of Biochemistry, Molecular Biology, Entomology & Plant Pathology, Starkville, MS, United States
| | - Jose E Pietri
- University of South Dakota, Sanford School of Medicine, Division of Basic Biomedical Sciences, Vermillion, SD, United States.
| |
Collapse
|
30
|
Powers JC, Turangan R, Joosse BA, Hillyer JF. Adult Mosquitoes Infected with Bacteria Early in Life Have Stronger Antimicrobial Responses and More Hemocytes after Reinfection Later in Life. INSECTS 2020; 11:insects11060331. [PMID: 32481519 PMCID: PMC7349202 DOI: 10.3390/insects11060331] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022]
Abstract
The immunological strategies employed by insects to overcome infection vary with the type of infection and may change with experience. We investigated how a bacterial infection in the hemocoel of the African malaria mosquito, Anopheles gambiae, prepares the immune system to face a subsequent bacterial infection. For this, adult female mosquitoes were separated into three groups—unmanipulated, injured, or infected with Escherichia coli—and five days later all the mosquitoes were infected with a different strain of E. coli. We found that an injury or a bacterial infection early in life enhances the ability of mosquitoes to kill bacteria later in life. This protection results in higher mosquito survival and is associated with an increased hemocyte density, altered phagocytic activity by individual hemocytes, and the increased expression of nitric oxide synthase and perhaps prophenoloxidase 6. Protection from a second infection likely occurs because of heightened immune awareness due to an already existing infection instead of memory arising from an earlier, cured infection. This study highlights the dynamic nature of the mosquito immune response and how one infection prepares mosquitoes to survive a subsequent infection.
Collapse
|
31
|
Pondeville E, Puchot N, Parvy JP, Carissimo G, Poidevin M, Waterhouse RM, Marois E, Bourgouin C. Hemocyte-targeted gene expression in the female malaria mosquito using the hemolectin promoter from Drosophila. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2020; 120:103339. [PMID: 32105779 PMCID: PMC7181189 DOI: 10.1016/j.ibmb.2020.103339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 06/10/2023]
Abstract
Hemocytes, the immune cells in mosquitoes, participate in immune defenses against pathogens including malaria parasites. Mosquito hemocytes can also be infected by arthropod-borne viruses but the pro- or anti-viral nature of this interaction is unknown. Although there has been progress on hemocyte characterization during pathogen infection in mosquitoes, the specific contribution of hemocytes to immune responses and the hemocyte-specific functions of immune genes and pathways remain unresolved due to the lack of genetic tools to manipulate gene expression in these cells specifically. Here, we used the Gal4-UAS system to characterize the activity of the Drosophila hemocyte-specific hemolectin promoter in the adults of Anopheles gambiae, the malaria mosquito. We established an hml-Gal4 driver line that we further crossed to a fluorescent UAS responder line, and examined the expression pattern in the adult progeny driven by the hml promoter. We show that the hml regulatory region drives hemocyte-specific transgene expression in a subset of hemocytes, and that transgene expression is triggered after a blood meal. The hml promoter drives transgene expression in differentiating prohemocytes as well as in differentiated granulocytes. Analysis of different immune markers in hemocytes in which the hml promoter drives transgene expression revealed that this regulatory region could be used to study phagocytosis as well as melanization. Finally, the hml promoter drives transgene expression in hemocytes in which o'nyong-nyong virus replicates. Altogether, the Drosophila hml promoter constitutes a good tool to drive transgene expression in hemocyte only and to analyze the function of these cells and the genes they express during pathogen infection in Anopheles gambiae.
Collapse
Affiliation(s)
- Emilie Pondeville
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France.
| | - Nicolas Puchot
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | | | - Guillaume Carissimo
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France
| | - Mickael Poidevin
- Centre de Génétique Moléculaire, CNRS UPR 2167, Gif-sur-Yvette, France
| | - Robert M Waterhouse
- Department of Ecology and Evolution, Swiss Institute of Bioinformatics, University of Lausanne, 1015, Lausanne, Switzerland
| | - Eric Marois
- CNRS UPR9022, INSERM U1257, Université de Strasbourg, Strasbourg, France
| | - Catherine Bourgouin
- CNRS Unit of Evolutionary Genomics, Modeling, and Health (UMR2000), Institut Pasteur, Paris, France.
| |
Collapse
|
32
|
20-Hydroxyecdysone Primes Innate Immune Responses That Limit Bacterial and Malarial Parasite Survival in Anopheles gambiae. mSphere 2020; 5:5/2/e00983-19. [PMID: 32295874 PMCID: PMC7160685 DOI: 10.1128/msphere.00983-19] [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: 01/09/2023] Open
Abstract
Blood feeding is an integral behavior of mosquitoes to acquire nutritional resources needed for reproduction. This requirement also enables mosquitoes to serve as efficient vectors to acquire and potentially transmit a multitude of mosquito-borne diseases, most notably malaria. Recent studies suggest that mosquito immunity is stimulated following a blood meal, independent of infection status. Since blood feeding promotes production of the hormone 20-hydroxyecdysone (20E), we hypothesized that 20E plays an important role in priming the immune response for pathogen challenge. Here, we examine the immunological effects of priming Anopheles gambiae with 20E prior to pathogen infection, demonstrating a significant reduction in bacteria and Plasmodium berghei survival in the mosquito host. Transcriptome sequencing (RNA-seq) analysis following 20E treatment identifies several known 20E-regulated genes, as well as several immune genes with previously reported function in antipathogen defense. Together, these data demonstrate that 20E influences cellular immune function and antipathogen immunity following mosquito blood feeding, arguing the importance of hormones in the regulation of mosquito innate immune function.IMPORTANCE Blood feeding is required to provide nutrients for mosquito egg production and serves as a mechanism to acquire and transmit pathogens. Shortly after a blood meal is taken, there is a peak in the production of 20-hydroxyecdysone (20E), a mosquito hormone that initiates physiological changes, including yolk protein production and mating refractoriness. Here, we examine additional roles of 20E in the regulation of mosquito immunity, demonstrating that priming the immune system with 20E increases mosquito resistance to pathogens. We identify differentially expressed genes in response to 20E treatment, including several involved in innate immune function as well as lipid metabolism and transport. Together, these data argue that 20E stimulates mosquito cellular immune function and innate immunity shortly after blood feeding.
Collapse
|
33
|
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
|
34
|
Chemical depletion of phagocytic immune cells in Anopheles gambiae reveals dual roles of mosquito hemocytes in anti- Plasmodium immunity. Proc Natl Acad Sci U S A 2019; 116:14119-14128. [PMID: 31235594 DOI: 10.1073/pnas.1900147116] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mosquito immunity is composed of both cellular and humoral factors that provide protection from invading pathogens. Immune cells known as hemocytes, have been intricately associated with phagocytosis and innate immune signaling. However, the lack of genetic tools has limited hemocyte study despite their importance in mosquito anti-Plasmodium immunity. To address these limitations, we employ the use of a chemical-based treatment to deplete phagocytic immune cells in Anopheles gambiae, demonstrating the role of phagocytes in complement recognition and prophenoloxidase production that limit the ookinete and oocyst stages of malaria parasite development, respectively. Through these experiments, we also define specific subtypes of phagocytic immune cells in An. gambiae, providing insights beyond the morphological characteristics that traditionally define mosquito hemocyte populations. Together, this study represents a significant advancement in our understanding of the roles of mosquito phagocytes in mosquito vector competence and demonstrates the utility of clodronate liposomes as an important tool in the study of invertebrate immunity.
Collapse
|
35
|
Brown LD, Shapiro LLM, Thompson GA, Estévez‐Lao TY, Hillyer JF. Transstadial immune activation in a mosquito: Adults that emerge from infected larvae have stronger antibacterial activity in their hemocoel yet increased susceptibility to malaria infection. Ecol Evol 2019; 9:6082-6095. [PMID: 31161020 PMCID: PMC6540708 DOI: 10.1002/ece3.5192] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 01/24/2023] Open
Abstract
Larval and adult mosquitoes mount immune responses against pathogens that invade their hemocoel. Although it has been suggested that a correlation exists between immune processes across insect life stages, the influence that an infection in the hemocoel of a larva has on the immune system of the eclosed adult remains unknown. Here, we used Anopheles gambiae to test whether a larval infection influences the adult response to a subsequent bacterial or malaria parasite infection. We found that for both female and male mosquitoes, a larval infection enhances the efficiency of bacterial clearance following a secondary infection in the hemocoel of adults. The adults that emerge from infected larvae have more hemocytes than adults that emerge from naive or injured larvae, and individual hemocytes have greater phagocytic activity. Furthermore, mRNA abundance of immune genes-such as cecropin A, Lysozyme C1, Stat-A, and Tep1-is higher in adults that emerge from infected larvae. A larval infection, however, does not have a meaningful effect on the probability that female adults will survive a systemic bacterial infection, and increases the susceptibility of females to Plasmodium yoelii, as measured by oocyst prevalence and intensity in the midgut. Finally, immune proficiency varies by sex; females exhibit increased bacterial killing, have twice as many hemocytes, and more highly express immune genes. Together, these results show that a larval hemocoelic infection induces transstadial immune activation-possibly via transstadial immune priming-but that it confers both costs and benefits to the emerged adults.
Collapse
Affiliation(s)
- Lisa D. Brown
- Department of Biological SciencesVanderbilt UniversityNashvilleTennessee
- Present address:
Department of BiologyGeorgia Southern UniversityStatesboroGeorgia
| | | | | | | | - Julián F. Hillyer
- Department of Biological SciencesVanderbilt UniversityNashvilleTennessee
| |
Collapse
|
36
|
Hugo RLE, Birrell GW. Proteomics of Anopheles Vectors of Malaria. Trends Parasitol 2018; 34:961-981. [DOI: 10.1016/j.pt.2018.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 12/12/2022]
|
37
|
Sigle LT, Hillyer JF. Mosquito Hemocytes Associate With Circulatory Structures That Support Intracardiac Retrograde Hemolymph Flow. Front Physiol 2018; 9:1187. [PMID: 30210361 PMCID: PMC6121077 DOI: 10.3389/fphys.2018.01187] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/07/2018] [Indexed: 01/28/2023] Open
Abstract
A powerful immune system protects mosquitoes from pathogens and influences their ability to transmit disease. The mosquito's immune and circulatory systems are functionally integrated, whereby intense immune processes occur in areas of high hemolymph flow. The primary circulatory organ of mosquitoes is the dorsal vessel, which consists of a thoracic aorta and an abdominal heart. In adults of the African malaria mosquito, Anopheles gambiae, the heart periodically alternates contraction direction, resulting in intracardiac hemolymph flowing toward the head (anterograde) and toward the posterior of the abdomen (retrograde). During anterograde contractions, hemolymph enters the dorsal vessel through ostia located in abdominal segments 2-7, and exits through an excurrent opening located in the head. During retrograde contractions, hemolymph enters the dorsal vessel through ostia located at the thoraco-abdominal junction, and exits through posterior excurrent openings located in the eighth abdominal segment. The ostia in abdominal segments 2 to 7-which function in anterograde intracardiac flow-are sites of intense immune activity, as a subset of hemocytes, called periostial hemocytes, respond to infection by aggregating, phagocytosing, and killing pathogens. Here, we assessed whether hemocytes are present and active at two sites important for retrograde intracardiac hemolymph flow: the thoraco-abdominal ostia and the posterior excurrent openings of the heart. We detected sessile hemocytes around both of these structures, and these hemocytes readily engage in phagocytosis. However, they are few in number and a bacterial infection does not induce the aggregation of additional hemocytes at these locations. Finally, we describe the process of hemocyte attachment and detachment to regions of the dorsal vessel involved in intracardiac retrograde flow.
Collapse
Affiliation(s)
| | - Julián F. Hillyer
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
38
|
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
|
39
|
Koiwai K, Kondo H, Hirono I. RNA-seq identifies integrin alpha of kuruma shrimp Marsupenaeus japonicus as a candidate molecular marker for phagocytic hemocytes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 81:271-278. [PMID: 29258750 DOI: 10.1016/j.dci.2017.12.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/13/2017] [Accepted: 12/13/2017] [Indexed: 06/07/2023]
Abstract
Phagocytosis is main cellular immunity, however, it is still unknown or debated upon which types of hemocyte contributes phagocytosis in penaeid shrimps. The hemocyte characterization in kuruma shrimp have been mainly performed based on its morphology by microscopic observation. Therefore, establishment of molecular markers to distinguish phagocytic hemocytes is required. In this study, using magnetic fluorescent beads, we enriched phagocytic hemocytes and conducted RNA-seq analysis between total and enriched phagocytic hemocytes. The data demonstrated functional difference between total and phagocytic hemocytes. In addition, a transcript homologous to integrin-alpha was highly expressed in phagocytic hemocytes, and named Mj-Intgα. Using anti-serum against Mj-Intgα revealed that around 60% of total hemocytes and more than 90% of phagocytic hemocytes showed positive for Mj-Intgα. This study presents Mj-Intgα as a candidate molecular marker for future functional characterization of hemocytes.
Collapse
Affiliation(s)
- Keiichiro Koiwai
- Laboratory of Genome Science, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato, Tokyo, 108-8477, Japan
| | - Hidehiro Kondo
- Laboratory of Genome Science, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato, Tokyo, 108-8477, Japan
| | - Ikuo Hirono
- Laboratory of Genome Science, Tokyo University of Marine Science and Technology, 4-5-7 Konan, Minato, Tokyo, 108-8477, Japan.
| |
Collapse
|
40
|
Immune Response and Evasion Mechanisms of Plasmodium falciparum Parasites. J Immunol Res 2018; 2018:6529681. [PMID: 29765991 PMCID: PMC5889876 DOI: 10.1155/2018/6529681] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/17/2018] [Accepted: 02/21/2018] [Indexed: 02/07/2023] Open
Abstract
Malaria causes approximately 212 million cases and 429 thousand deaths annually. Plasmodium falciparum is responsible for the vast majority of deaths (99%) than others. The virulence of P. falciparum is mostly associated with immune response-evading ability. It has different mechanisms to evade both Anopheles mosquito and human host immune responses. Immune-evading mechanisms in mosquito depend mainly on the Pfs47 gene that inhibits Janus kinase-mediated activation. Host complement factor also protects human complement immune attack of extracellular gametes in Anopheles mosquito midgut. In the human host, evasion largely results from antigenic variation, polymorphism, and sequestration. They also induce Kupffer cell apoptosis at the preerythrocytic stage and interfere with phagocytic functions of macrophage by hemozoin in the erythrocytic stage. Lack of major histocompatibility complex-I molecule expression on the surface red blood cells also avoids recognition by CD8+ T cells. Complement proteins could allow for the entry of parasite into the red blood cell. Intracellular survival also assists the escape of malarial parasite. Invading, evading, and immune response mechanisms both in malaria vector and human host are critical to design appropriate vaccine. As a result, the receptors and ligands involved in different stages of malaria parasites should be elucidated.
Collapse
|
41
|
Zhang J, Huang J, Zhu F, Zhang J. Differential gene expression in Anopheles stephensi following infection with drug-resistant Plasmodium yoelii. Parasit Vectors 2017; 10:401. [PMID: 28851458 PMCID: PMC5576267 DOI: 10.1186/s13071-017-2326-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 08/08/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The transmission of drug-resistant parasites by the mosquito may be influenced by the altered biological fitness of drug-resistant parasites and different immune reactions or metabolic change in the mosquito. At this point, little is known about the variations in mosquito immunity and metabolism when mosquitoes are infected with drug-resistant parasites. To understand the differential gene expression in Anopheles following infection with drug-resistant Plasmodium, we conducted a genome-wide transcriptomic profiling analysis of Anopheles stephensi following feeding on mice with drug-resistant or drug-sensitive P. yoelii, observed changes in gene expression profiles and identified transcripts affected by the drug-resistant parasite. RESULTS To study the impact of drug-resistant Plasmodium infections on An. stephensi gene transcription, we analyzed the three major transition stages of Plasmodium infections: at 24 h and 13 and 19 days after blood-feeding. Six cDNA libraries (R-As24h, R-As13d, R-As19d,S-As24h, S-As13dand S-As19d) were constructed, and RNA sequencing was subsequently performed. In total, approximately 50.1 million raw reads, 47.9 million clean reads and 7.18G clean bases were obtained. Following differentially expressed gene (DEG) analysis, GO enrichment analysis of DEGs, and functional classification by KEGG, we showed that the variations in gene expression in An. stephensi infected by the drug-resistant P. yoelii NSM occurred mainly at 13 days after blood meal during sporozoite migration through the hemolymph. The differentially expressed genes included those functioning in some important immune reaction and iron metabolism pathways, such as pattern recognition receptors, regulators of the JNK pathway, components of the phagosome pathway, regulators of the melanization response, activators of complement reactions, insulin signaling cascade members, oxidative stress and detoxification proteins. CONCLUSIONS Our study shows that drug-resistant P. yoelii NSM has an impact on the transcript abundance levels of An.stephensi mostly at 13 days after blood meal during sporozoite migration through the hemolymph and that most differentially expressed genes were downregulated. Our results highlight the need for a better understanding of selective pressures from these differentially expressed genes of the drug-resistant Plasmodium in the mosquito and the different transmission patterns of drug-resistant Plasmodium by Anopheles.
Collapse
Affiliation(s)
- Jingru Zhang
- Department of Pathobiology, the Third Military Medical University, Chongqing, People's Republic of China
| | - Jiacheng Huang
- Students brigade 5, The Third Military Medical University, Chongqing, People's Republic of China
| | - Feng Zhu
- Department of Pathobiology, the Third Military Medical University, Chongqing, People's Republic of China
| | - Jian Zhang
- Department of Pathobiology, the Third Military Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
42
|
Kwon H, Arends BR, Smith RC. Late-phase immune responses limiting oocyst survival are independent of TEP1 function yet display strain specific differences in Anopheles gambiae. Parasit Vectors 2017; 10:369. [PMID: 28764765 PMCID: PMC5540282 DOI: 10.1186/s13071-017-2308-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/25/2017] [Indexed: 11/18/2022] Open
Abstract
Background There is emerging evidence that mosquito anti-Plasmodium immunity is multimodal with distinct mechanisms for killing malaria parasites at either the ookinete or oocyst stages. Early-phase responses targeting the ookinete require complement-like components circulating in the mosquito hemolymph that result in TEP1-mediated lysis or melanization. Additional responses mediated by the LL3 and STAT pathways limit oocyst survival through unknown mechanisms that require mosquito hemocyte function. While previous experiments argue that these mechanisms of parasite killing are independent, the transient nature of gene-silencing has rendered these experiments inconclusive. To address this issue, we outline experiments using a TALEN-derived TEP1 mutant line to examine the role of TEP1 in the Anopheles gambiae late-phase immune response. Results Despite higher early oocyst numbers in the TEP1 mutant line, no differences in oocyst survival were observed when compared to control mosquitoes, suggesting that TEP1 function is independent of the late-phase immune response. To further validate this phenotype in the TEP1 mutant, oocyst survival was evaluated in the TEP1 mutant background by silencing either LL3 or STAT-A. Surprisingly, only STAT-A silenced mosquitoes were able to reconstitute the late-phase immune phenotype increasing oocyst survival in the TEP1 mutant line. Additional experiments highlight significant differences in LL3 expression in the M/S hybrid genetic background of the TEP1 mutant line compared to that of the Keele strain (M form) of An. gambiae, and demonstrate that LL3 is not required for granulocyte differentiation in the M/S hybrid G3 genetic background in response to malaria parasite infection. Conclusions Through the combination of genetic experiments utilizing genetic mutants and reverse genetic approaches, new information has emerged regarding the mechanisms of mosquito late-phase immunity. When combined with previously published experiments, the body of evidence argues that Plasmodium oocyst survival is TEP1 independent, thus establishing that the mechanisms of early- and late-phase immunity are distinct. Moreover, we identify that the known components that mediate oocyst survival are susceptible to strain-specific differences depending on their genetic background and provide further evidence that the signals that promote hemocyte differentiation are required to limit oocyst survival. Together, this study provides new insights into the mechanisms of oocyst killing and the importance of genetics in shaping mosquito vector competence. Electronic supplementary material The online version of this article (doi:10.1186/s13071-017-2308-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyeogsun Kwon
- Department of Entomology, Iowa State University, Ames, Iowa, 50011, USA
| | - Benjamin R Arends
- Department of Entomology, Iowa State University, Ames, Iowa, 50011, USA
| | - Ryan C Smith
- Department of Entomology, Iowa State University, Ames, Iowa, 50011, USA.
| |
Collapse
|
43
|
League GP, Estévez-Lao TY, Yan Y, Garcia-Lopez VA, Hillyer JF. Anopheles gambiae larvae mount stronger immune responses against bacterial infection than adults: evidence of adaptive decoupling in mosquitoes. Parasit Vectors 2017; 10:367. [PMID: 28764812 PMCID: PMC5539753 DOI: 10.1186/s13071-017-2302-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/20/2017] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The immune system of adult mosquitoes has received significant attention because of the ability of females to vector disease-causing pathogens while ingesting blood meals. However, few studies have focused on the immune system of larvae, which, we hypothesize, is highly robust due to the high density and diversity of microorganisms that larvae encounter in their aquatic environments and the strong selection pressures at work in the larval stage to ensure survival to reproductive maturity. Here, we surveyed a broad range of cellular and humoral immune parameters in larvae of the malaria mosquito, Anopheles gambiae, and compared their potency to that of newly-emerged adults and older adults. RESULTS We found that larvae kill bacteria in their hemocoel with equal or greater efficiency compared to newly-emerged adults, and that antibacterial ability declines further with adult age, indicative of senescence. This phenotype correlates with more circulating hemocytes and a differing spatial arrangement of sessile hemocytes in larvae relative to adults, as well as with the individual hemocytes of adults carrying a greater phagocytic burden. The hemolymph of larvae also possesses markedly stronger antibacterial lytic and melanization activity than the hemolymph of adults. Finally, infection induces a stronger transcriptional upregulation of immunity genes in larvae than in adults, including differences in the immunity genes that are regulated. CONCLUSIONS These results demonstrate that immunity is strongest in larvae and declines after metamorphosis and with adult age, and suggest that adaptive decoupling, or the independent evolution of larval and adult traits made possible by metamorphosis, has occurred in the mosquito lineage.
Collapse
Affiliation(s)
- Garrett P. League
- Department of Biological Sciences, Vanderbilt University, Nashville, TN USA
| | | | - Yan Yan
- Department of Biological Sciences, Vanderbilt University, Nashville, TN USA
| | | | - Julián F. Hillyer
- Department of Biological Sciences, Vanderbilt University, Nashville, TN USA
| |
Collapse
|
44
|
Whitten MMA, Coates CJ. Re-evaluation of insect melanogenesis research: Views from the dark side. Pigment Cell Melanoma Res 2017; 30:386-401. [PMID: 28378380 DOI: 10.1111/pcmr.12590] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 03/29/2017] [Indexed: 12/16/2022]
Abstract
Melanins (eumelanin and pheomelanin) are synthesized in insects for several purposes including cuticle sclerotization and color patterning, clot formation, organogenesis, and innate immunity. Traditional views of insect immunity detail the storage of pro-phenoloxidases inside specialized blood cells (hemocytes) and their release upon recognition of foreign bodies. Activated phenoloxidases convert monophenols into reactive quinones in a two-step enzymatic reaction, and until recently, the mechanism of tyrosine hydroxylation remained a mystery. Herein, we present our interpretations of these enzyme-substrate complexes. The resultant melanins are deposited onto the surface of microbes to immobilize, agglutinate, and suffocate them. Phenoloxidase activity and melanin production are not limited to the blood (hemolymph) or cuticle, as recent evidence points to more diverse, sophisticated interactions in the gut and with the resident symbionts. This review offers insight into the somewhat neglected areas of insect melanogenesis research, particularly in innate immunity, its role in beneficial insects such as pollinators, the functional versatility of phenoloxidases, and the limitations of common experimental approaches that may impede progress inadvertently.
Collapse
|