1
|
Hao J, Jie Y, Lu Z, Ye T, Meng J, Liu C, Yan J, Zheng Y, Dong Z, Gu Z. Temporal changes in the transcriptome profile of Macrobrachium rosenbergii in response to decapod iridescent virus 1 infection. Front Immunol 2025; 16:1575476. [PMID: 40276510 PMCID: PMC12018387 DOI: 10.3389/fimmu.2025.1575476] [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: 02/12/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
The farming of Macrobrachium rosenbergii faces significant challenges due to infections caused by Decapod iridovirus 1 (DIV1). To gain deeper insights into the dynamic immune regulatory processes of M. rosenbergii in response to DIV1 infection, RNA sequencing (RNA-seq) was employed to profile the transcriptome in the hepatopancreas at 24, 48, 72, and 96 hours post-infection (hpi). Time-course analysis revealed 3,339 differentially expressed genes (DEGs), which exhibited distinct expression patterns across various stages of infection. At 24 hpi and 48 hpi, the top 20 enriched pathways included 3 immunity-related pathways (Lysosome, Phagosome, C-type lectin receptor signaling) and 7 metabolism-related pathways at 24 hpi, and 5 metabolism-related pathways at 48 hpi. In contrast, in the later stages of infection (72 hpi), 13 of the top 17 enriched pathways associated with DEGs were metabolism-related, including those involved in antioxidant defense, such as the Peroxisome, Cysteine and methionine metabolism, and Glutathione metabolism. At 96 hpi, pathways related to ECM-receptor interaction, Purine metabolism, and Lysosome were significantly enriched. Among the DEGs, a total of 16 genes were consistently identified across all time points, with 14 of these genes, including alpha-2-macroglobulin-like, alpha-amylase 1-like, putative aldolase class 2 protein PA3430, platelet-derived growth factor subunit B-like, serum amyloid A-5 protein-like, phenoloxidase-activating enzyme-like, pantetheinase-like, and perlucin-like protein, demonstrating sustained upregulation at all time points. In contrast, the gene encoding rhodanese domain-containing protein CG4456-like was consistantly downregulated. Additionally, weighted gene co-expression network analysis (WGCNA) indicated several hub genes that were tightly connected to intercellular communication, such as innexin shaking-B-like and innexin inx3-like, and endochitinase A1-like. The gene expression changes varied over time, exhibiting a dynamic, time-dependent pattern that underscores the complexity of host-pathogen interactions. These results provide new insights into the cellular mechanisms influenced by DIV1 throughout the infection process, offering valuable knowledge for developing virus control strategies in shrimp aquaculture.
Collapse
Affiliation(s)
- Jingwen Hao
- Xianghu Laboratory, Hangzhou, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Yukun Jie
- Xianghu Laboratory, Hangzhou, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Zhibin Lu
- Xianghu Laboratory, Hangzhou, China
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | | | | | - Cui Liu
- Xianghu Laboratory, Hangzhou, China
| | | | | | - Zaijie Dong
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | | |
Collapse
|
2
|
Qin X, Li J, Zhu F, Zhang J. Thioester-containing protein TEP15 promotes malaria parasite development in mosquitoes through negative regulation of melanization. Parasit Vectors 2025; 18:124. [PMID: 40170182 PMCID: PMC11963550 DOI: 10.1186/s13071-025-06772-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Thioester-containing proteins (TEPs) serve as crucial effectors and regulatory components within the innate immune system of mosquitoes. Despite their significance, the mechanisms by which TEPs exert negative regulation on the immune response in mosquitoes remain inadequately understood. This study aims to elucidate the role of TEPs in the negative regulation of melanization, thereby advancing our comprehension of their regulatory function in the immune response. METHODS We infected female Anopheles stephensi mosquitoes with Plasmodium yoelii by allowing them to feed on pre-infected female Kunming mice. Western blot, quantitative polymerase chain reaction, differential gene expression analyses, and gene silencing were then conducted. Student's t-test was used to analyze continuous variables, with statistical significance defined as p < 0.05. RESULTS A. stephensi TEP15 (AsTEP15) negatively regulated mosquitos' innate immunity and promoted Plasmodium development. AsTEP15 knockdown induced mosquito resistance to malaria parasite melanization during the oocyst stage and significantly reduced sporozoite numbers. Further analysis showed that AsTEP15 mainly negatively affects the TEP1 and immune deficiency (IMD) pathway, thereby inhibiting melanization. CONCLUSIONS We describe a mosquito TEP that negatively regulates immunity, further enriching the functional diversity of TEP family members. In addition, our results suggest that oocysts may exploit TEPs to escape or inhibit mosquito immunity, highlighting potential targets for blocking malaria transmission.
Collapse
Affiliation(s)
- Xin Qin
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| | - Jianyong Li
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| | - Feng Zhu
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China.
| | - Jian Zhang
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China.
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Dolan B, Correa Gaviria T, Dong Y, Cresswell P, Dimopoulos G, Chuang YM, Fikrig E. mosGILT antibodies interfere with Plasmodium sporogony in Anopheles gambiae. Nat Commun 2025; 16:592. [PMID: 39799117 PMCID: PMC11724845 DOI: 10.1038/s41467-025-55902-1] [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: 07/03/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025] Open
Abstract
Plasmodium, the causative agents of malaria, are obtained by mosquitoes from an infected human. Following Plasmodium acquisition by Anopheles gambiae, mosquito gamma-interferon-inducible lysosomal thiol reductase (mosGILT) plays a critical role in its subsequent sporogony in the mosquito. A critical location for this development is the midgut, a tissue we show expresses mosGILT. Using membrane-feeding and murine infection models, we demonstrate that antibodies against mosGILT reduce the number of P. falciparum and P. berghei oocysts in the midgut and the infection prevalence of both species in the mosquito. mosGILT antibodies act in the mosquito midgut, specifically impacting the Plasmodium oocyst stage. Targeting mosGILT can therefore interfere with the Plasmodium life cycle in the mosquito and potentially serve as a transmission-blocking vaccine.
Collapse
Affiliation(s)
- Brady Dolan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Tomás Correa Gaviria
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Peter Cresswell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yu-Min Chuang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
6
|
Xie MQ, Wang LJ, Xiao HM, Wei SJ. Regulatory networks of mRNAs and miRNAs involved in the immune response of diamondback moth, Plutella xylostella to fungal infection. BMC Genomics 2025; 26:15. [PMID: 39762741 PMCID: PMC11706182 DOI: 10.1186/s12864-024-11192-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND The entomopathogenic fungus, Isaria fumosorosea, shows promise as a biological control agent in managing the diamondback moth (DBM) Plutella xylostella, a highly destructive global pest of cruciferous vegetables. To date, the miRNA-mRNA regulatory networks underlying the immune response of DBM to I. fumosorosea infection are still poorly understood. Here, we characterize the expression profiles of miRNA and mRNA, and construct the miRNA-gene regulatory network in DBM infected with I. fumosorosea. RESULTS We identified 580 differentially expressed genes (DEGs) and 55 differentially expressed miRNAs (DEMs) in I. fumosorosea-infected DBM. Among these DEGs, we found 28 immunity-related genes, which mainly include pattern recognition receptors, signal modulators, and immune effectors. Integrated analysis discovered 87 negative correlation pairs between miRNA and mRNA, involving 40 DEMs and 62 DEGs in infected DBM. Additionally, 13 miRNAs and 10 corresponding mRNAs were identified as candidate miRNA-mRNA pairs for DBM immunity against fungal infection. Gene functional enrichment analysis indicated that these miRNAs could target genes associated with various pathways, such as the immune system, infectious diseases, digestive system, endocrine system, nervous system, and signal transduction. Finally, the regulatory relationships of six miRNA-mRNA pairs were validated using quantitative reverse transcription PCR. CONCLUSIONS For the first time, we present integrated miRNA and mRNA data to elucidate the immune response of the DBM to fungal infection. Our findings enhance the understanding of the immune response of the DBM to entomopathogenic fungi infection.
Collapse
Affiliation(s)
- Mei-Qiong Xie
- College of Life Sciences and Resources and Environment, Yichun University, Yichun, 336000, China
| | - Long-Jiang Wang
- College of Chemistry and Bioengineering, Yichun University, Yichun, 336000, China.
| | - Hua-Mei Xiao
- College of Life Sciences and Resources and Environment, Yichun University, Yichun, 336000, China
| | - Shu-Jun Wei
- Institute of Plant Protection, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China.
| |
Collapse
|
7
|
Belikoff EJ, Davis RJ, Williamson ME, Britt JW, Scott MJ. Identification of a gene promoter active in Lucilia sericata larval salivary glands using a rapid transient expression assay. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 173:104163. [PMID: 39068996 DOI: 10.1016/j.ibmb.2024.104163] [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: 03/08/2024] [Revised: 07/01/2024] [Accepted: 07/25/2024] [Indexed: 07/30/2024]
Abstract
Tissue-specific gene promoters are desired as they provide the specificity needed for control of gene expression in transgenic animals. Here we describe a relatively rapid two-component transient expression assay that was used to identify a gene promoter active in the larval salivary glands of the green blow fly, Lucilia sericata. Sterile L.sericata maggots are widely used for wound debridement. A larval salivary gland gene promoter could be used to make maggots that secrete factors for enhanced wound therapy. Embryos from a line that carry a tetracycline transactivator (tTA)-activated red fluorescent protein gene were injected with plasmid DNA with the tTA gene driven by a constitutive or tissue-specific gene promoter. The hatched larvae were reared on diet and then examined for red fluorescence. A promoter from the LsCG30371 gene was active in the larval salivary glands. The tissue-specificity of the promoter was subsequently confirmed with stable transgenic lines that carried the LsCG30371-tTA gene. The relatively rapid transient expression assay could potentially be used to determine the tissue-specificity of other gene promoters. Further, the stable LsCG30371-tTA lines could be used to make sterile maggots that secrete factors from the salivary glands for enhanced wound healing.
Collapse
Affiliation(s)
- Esther J Belikoff
- Department of Entomology, North Carolina State University, Campus Box 7613, Raleigh, NC, 27695-7613, USA.
| | - Rebecca J Davis
- Department of Entomology, North Carolina State University, Campus Box 7613, Raleigh, NC, 27695-7613, USA.
| | - Megan E Williamson
- Department of Entomology, North Carolina State University, Campus Box 7613, Raleigh, NC, 27695-7613, USA.
| | - John W Britt
- Department of Entomology, North Carolina State University, Campus Box 7613, Raleigh, NC, 27695-7613, USA.
| | - Maxwell J Scott
- Department of Entomology, North Carolina State University, Campus Box 7613, Raleigh, NC, 27695-7613, USA.
| |
Collapse
|
8
|
Liu T, Wang J, Li X, Yu S, Zheng D, Liu Z, Yang X, Wang Y. Human Defensin 5 Inhibits Plasmodium yoelii Development in Anopheles stephensi by Promoting Innate Immune Response. Trop Med Infect Dis 2024; 9:169. [PMID: 39195607 PMCID: PMC11360097 DOI: 10.3390/tropicalmed9080169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Malaria poses a serious threat to human health. Existing vector-based interventions have shortcomings, such as environmental pollution, strong resistance to chemical insecticides, and the slow effects of biological insecticides. Therefore, the need to develop novel strategies for controlling malaria, such as reducing mosquito vector competence, is escalating. Human defensin 5 (HD5) has broad-spectrum antimicrobial activity. To determine its effect on Plasmodium development in mosquitoes, HD5 was injected into Anopheles stephensi at various time points. The infection density of Plasmodium yoelii in An. stephensi was substantially reduced by HD5 treatment administered 24 h prior to infection or 6, 12, or 24 h post-infection (hpi). We found that HD5 treatment upregulated the expression of the innate immune effectors TEP1, MyD88, and Rel1 at 24 and 72 hpi. Furthermore, the RNA interference of MyD88, a key upstream molecule in the Toll signaling pathway, decreased the HD5-induced resistance of mosquitoes against Plasmodium infection. These results suggest that HD5 microinjection inhibits the development of malaria parasites in An. stephensi by activating the Toll signaling pathway.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Jing Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Xin Li
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Shasha Yu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Dan Zheng
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China
| | - Zhilong Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Xuesen Yang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Ying Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| |
Collapse
|
9
|
Kefi M, Cardoso-Jaime V, Saab SA, Dimopoulos G. Curing mosquitoes with genetic approaches for malaria control. Trends Parasitol 2024; 40:487-499. [PMID: 38760256 DOI: 10.1016/j.pt.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/19/2024]
Abstract
Malaria remains a persistent global public health challenge because of the limitations of current prevention tools. The use of transgenic mosquitoes incapable of transmitting malaria, in conjunction with existing methods, holds promise for achieving elimination of malaria and preventing its reintroduction. In this context, population modification involves the spread of engineered genetic elements through mosquito populations that render them incapable of malaria transmission. Significant progress has been made in this field over the past decade in revealing promising targets, optimizing genetic tools, and facilitating the transition from the laboratory to successful field deployments, which are subject to regulatory scrutiny. This review summarizes recent advances and ongoing challenges in 'curing' Anopheles vectors of the malaria parasite.
Collapse
Affiliation(s)
- Mary Kefi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Victor Cardoso-Jaime
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Sally A Saab
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - George Dimopoulos
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
10
|
Zmarlak NM, Lavazec C, Brito-Fravallo E, Genève C, Aliprandini E, Aguirre-Botero MC, Vernick KD, Mitri C. The Anopheles leucine-rich repeat protein APL1C is a pathogen binding factor recognizing Plasmodium ookinetes and sporozoites. PLoS Pathog 2024; 20:e1012008. [PMID: 38354186 PMCID: PMC10898737 DOI: 10.1371/journal.ppat.1012008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 02/27/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
Leucine-rich repeat (LRR) proteins are commonly involved in innate immunity of animals and plants, including for pattern recognition of pathogen-derived elicitors. The Anopheles secreted LRR proteins APL1C and LRIM1 are required for malaria ookinete killing in conjunction with the complement-like TEP1 protein. However, the mechanism of parasite immune recognition by the mosquito remains unclear, although it is known that TEP1 lacks inherent binding specificity. Here, we find that APL1C and LRIM1 bind specifically to Plasmodium berghei ookinetes, even after depletion of TEP1 transcript and protein, consistent with a role for the LRR proteins in pathogen recognition. Moreover, APL1C does not bind to ookinetes of the human malaria parasite Plasmodium falciparum, and is not required for killing of this parasite, which correlates LRR binding specificity and immune protection. Most of the live P. berghei ookinetes that migrated into the extracellular space exposed to mosquito hemolymph, and almost all dead ookinetes, are bound by APL1C, thus associating LRR protein binding with parasite killing. We also find that APL1C binds to the surface of P. berghei sporozoites released from oocysts into the mosquito hemocoel and forms a potent barrier limiting salivary gland invasion and mosquito infectivity. Pathogen binding by APL1C provides the first functional explanation for the long-known requirement of APL1C for P. berghei ookinete killing in the mosquito midgut. We propose that secreted mosquito LRR proteins are required for pathogen discrimination and orientation of immune effector activity, potentially as functional counterparts of the immunoglobulin-based receptors used by vertebrates for antigen recognition.
Collapse
Affiliation(s)
- Natalia Marta Zmarlak
- Institut Pasteur, Université de Paris, CNRS UMR2000, Unit of Genetics and Genomics of Insect Vectors, Department of Parasites and Insect Vectors, Paris, France
- Graduate School of Life Sciences ED515, Sorbonne Universities, UPMC Paris VI, Paris, France
| | - Catherine Lavazec
- Inserm U1016, CNRS UMR8104, Université de Paris, Institut Cochin, Paris, France
| | - Emma Brito-Fravallo
- Institut Pasteur, Université de Paris, CNRS UMR2000, Unit of Genetics and Genomics of Insect Vectors, Department of Parasites and Insect Vectors, Paris, France
| | - Corinne Genève
- Institut Pasteur, Université de Paris, CNRS UMR2000, Unit of Genetics and Genomics of Insect Vectors, Department of Parasites and Insect Vectors, Paris, France
| | - Eduardo Aliprandini
- Institut Pasteur, Université de Paris, Unit of Malaria Infection & Immunity, Department of Parasites and Insect Vectors, Paris, France
| | - Manuela Camille Aguirre-Botero
- Institut Pasteur, Université de Paris, Unit of Malaria Infection & Immunity, Department of Parasites and Insect Vectors, Paris, France
| | - Kenneth D. Vernick
- Institut Pasteur, Université de Paris, CNRS UMR2000, Unit of Genetics and Genomics of Insect Vectors, Department of Parasites and Insect Vectors, Paris, France
- Graduate School of Life Sciences ED515, Sorbonne Universities, UPMC Paris VI, Paris, France
| | - Christian Mitri
- Institut Pasteur, Université de Paris, CNRS UMR2000, Unit of Genetics and Genomics of Insect Vectors, Department of Parasites and Insect Vectors, Paris, France
| |
Collapse
|
11
|
Klug D, Blandin SA. Activation of complement-like antiparasitic responses in Anopheles mosquitoes. Curr Opin Microbiol 2023; 72:102280. [PMID: 36841199 DOI: 10.1016/j.mib.2023.102280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 02/25/2023]
Abstract
During their development in mosquitoes, malaria parasites undergo massive losses that are in part due to a potent antiparasitic response mounted by the vector. The most efficient and best-characterized response relies on a complement-like system particularly effective against parasites as they cross the mosquito midgut epithelium. While our vision of the molecular and cellular events that lead to parasite elimination is still partial, our understanding of the steps triggering complement activation at the surface of invading parasites has considerably progressed, not only through the identification of novel contributing genes, but also with the recent in-depth characterization of the different mosquito blood cell types, and the ability to track them in live mosquitoes. Here, we propose a simple model based on the time of invasion to explain how parasites may escape complement-like responses during midgut infection.
Collapse
Affiliation(s)
- Dennis Klug
- Université de Strasbourg, CNRS, Inserm, UPR9022/U1257, Mosquito Immune Responses (MIR), F-67000 Strasbourg, France
| | - Stephanie A Blandin
- Université de Strasbourg, CNRS, Inserm, UPR9022/U1257, Mosquito Immune Responses (MIR), F-67000 Strasbourg, France.
| |
Collapse
|
12
|
Bottino-Rojas V, James AA. Use of Insect Promoters in Genetic Engineering to Control Mosquito-Borne Diseases. Biomolecules 2022; 13:16. [PMID: 36671401 PMCID: PMC9855440 DOI: 10.3390/biom13010016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
Mosquito transgenesis and gene-drive technologies provide the basis for developing promising new tools for vector-borne disease prevention by either suppressing wild mosquito populations or reducing their capacity from transmitting pathogens. Many studies of the regulatory DNA and promoters of genes with robust sex-, tissue- and stage-specific expression profiles have supported the development of new tools and strategies that could bring mosquito-borne diseases under control. Although the list of regulatory elements available is significant, only a limited set of those can reliably drive spatial-temporal expression. Here, we review the advances in our ability to express beneficial and other genes in mosquitoes, and highlight the information needed for the development of new mosquito-control and anti-disease strategies.
Collapse
Affiliation(s)
- Vanessa Bottino-Rojas
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA 92697, USA
| | - Anthony A. James
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| |
Collapse
|
13
|
Hoermann A, Habtewold T, Selvaraj P, Del Corsano G, Capriotti P, Inghilterra MG, Kebede TM, Christophides GK, Windbichler N. Gene drive mosquitoes can aid malaria elimination by retarding Plasmodium sporogonic development. SCIENCE ADVANCES 2022; 8:eabo1733. [PMID: 36129981 PMCID: PMC9491717 DOI: 10.1126/sciadv.abo1733] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 08/04/2022] [Indexed: 05/12/2023]
Abstract
Gene drives hold promise for the genetic control of malaria vectors. The development of vector population modification strategies hinges on the availability of effector mechanisms impeding parasite development in transgenic mosquitoes. We augmented a midgut gene of the malaria mosquito Anopheles gambiae to secrete two exogenous antimicrobial peptides, magainin 2 and melittin. This small genetic modification, capable of efficient nonautonomous gene drive, hampers oocyst development in both Plasmodium falciparum and Plasmodium berghei. It delays the release of infectious sporozoites, while it simultaneously reduces the life span of homozygous female transgenic mosquitoes. Modeling the spread of this modification using a large-scale agent-based model of malaria epidemiology reveals that it can break the cycle of disease transmission across a range of transmission intensities.
Collapse
Affiliation(s)
- Astrid Hoermann
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Tibebu Habtewold
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Prashanth Selvaraj
- Institute for Disease Modeling, Bill and Melinda Gates Foundation, Seattle, WA 98109, USA
| | | | - Paolo Capriotti
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | | | - Temesgen M. Kebede
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | | | | |
Collapse
|
14
|
Onyango SA, Ochwedo KO, Machani MG, Olumeh JO, Debrah I, Omondi CJ, Ogolla SO, Lee MC, Zhou G, Kokwaro E, Kazura JW, Afrane YA, Githeko AK, Zhong D, Yan G. Molecular characterization and genotype distribution of thioester-containing protein 1 gene in Anopheles gambiae mosquitoes in western Kenya. Malar J 2022; 21:235. [PMID: 35948910 PMCID: PMC9364548 DOI: 10.1186/s12936-022-04256-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 08/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Evolutionary pressures lead to the selection of efficient malaria vectors either resistant or susceptible to Plasmodium parasites. These forces may favour the introduction of species genotypes that adapt to new breeding habitats, potentially having an impact on malaria transmission. Thioester-containing protein 1 (TEP1) of Anopheles gambiae complex plays an important role in innate immune defenses against parasites. This study aims to characterize the distribution pattern of TEP1 polymorphisms among populations of An. gambiae sensu lato (s.l.) in western Kenya. METHODS Anopheles gambiae adult and larvae were collected using pyrethrum spray catches (PSC) and plastic dippers respectively from Homa Bay, Kakamega, Bungoma, and Kisumu counties between 2017 and 2020. Collected adults and larvae reared to the adult stage were morphologically identified and then identified to sibling species by PCR. TEP1 alleles were determined in 627 anopheles mosquitoes using restriction fragment length polymorphisms-polymerase chain reaction (RFLP-PCR) and to validate the TEP1 genotyping results, a representative sample of the alleles was sequenced. RESULTS Two TEP1 alleles (TEP1*S1 and TEP1*R2) and three corresponding genotypes (*S1/S1, *R2/S1, and *R2/R2) were identified. TEP1*S1 and TEP1*R2 with their corresponding genotypes, homozygous *S1/S1 and heterozygous *R2/S1 were widely distributed across all sites with allele frequencies of approximately 80% and 20%, respectively both in Anopheles gambiae and Anopheles arabiensis. There was no significant difference detected among the populations and between the two mosquito species in TEP1 allele frequency and genotype frequency. The overall low levels in population structure (FST = 0.019) across all sites corresponded to an effective migration index (Nm = 12.571) and low Nei's genetic distance values (< 0.500) among the subpopulation. The comparative fixation index values revealed minimal genetic differentiation between species and high levels of gene flow among populations. CONCLUSION Genotyping TEP1 has identified two common TEP1 alleles (TEP1*S1 and TEP1*R2) and three corresponding genotypes (*S1/S1, *R2/S1, and *R2/R2) in An. gambiae s.l. The TEP1 allele genetic diversity and population structure are low in western Kenya.
Collapse
Affiliation(s)
- Shirley A. Onyango
- Department of Zoological Sciences, School of Science and Technology, Kenyatta University, Nairobi, Kenya
- Sub-Saharan Africa International Centre of Excellence for Malaria Research, Homa bay, Kenya
| | - Kevin O. Ochwedo
- Sub-Saharan Africa International Centre of Excellence for Malaria Research, Homa bay, Kenya
- Department of Biology, Faculty of Science and Technology, University of Nairobi, Nairobi, Kenya
| | - Maxwell G. Machani
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Julius O. Olumeh
- Sub-Saharan Africa International Centre of Excellence for Malaria Research, Homa bay, Kenya
| | - Isaiah Debrah
- Sub-Saharan Africa International Centre of Excellence for Malaria Research, Homa bay, Kenya
- Department of Biochemistry, Cell and Molecular Biology, West Africa Centre for Cell Biology of Infectious Pathogen, University of Ghana, Accra, Ghana
| | - Collince J. Omondi
- Sub-Saharan Africa International Centre of Excellence for Malaria Research, Homa bay, Kenya
- Department of Biology, Faculty of Science and Technology, University of Nairobi, Nairobi, Kenya
| | - Sidney O. Ogolla
- Department of Zoological Sciences, School of Science and Technology, Kenyatta University, Nairobi, Kenya
- Sub-Saharan Africa International Centre of Excellence for Malaria Research, Homa bay, Kenya
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
- Program in Public Health, College of Health Sciences, University of California at Irvine, Irvine, CA 92697 USA
- Department of Biology, Faculty of Science and Technology, University of Nairobi, Nairobi, Kenya
- Department of Medical Microbiology, Medical School, University of Ghana, University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, West Africa Centre for Cell Biology of Infectious Pathogen, University of Ghana, Accra, Ghana
- Center for Global Health and Diseases, Case Western Reserve University, LC 4983, Cleveland, OH 44106 USA
| | - Ming-Chieh Lee
- Program in Public Health, College of Health Sciences, University of California at Irvine, Irvine, CA 92697 USA
| | - Guofa Zhou
- Program in Public Health, College of Health Sciences, University of California at Irvine, Irvine, CA 92697 USA
| | - Elizabeth Kokwaro
- Department of Zoological Sciences, School of Science and Technology, Kenyatta University, Nairobi, Kenya
| | - James W. Kazura
- Center for Global Health and Diseases, Case Western Reserve University, LC 4983, Cleveland, OH 44106 USA
| | - Yaw A. Afrane
- Department of Medical Microbiology, Medical School, University of Ghana, University of Ghana, Accra, Ghana
| | - Andrew K. Githeko
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Daibin Zhong
- Program in Public Health, College of Health Sciences, University of California at Irvine, Irvine, CA 92697 USA
| | - Guiyun Yan
- Program in Public Health, College of Health Sciences, University of California at Irvine, Irvine, CA 92697 USA
| |
Collapse
|
15
|
Leung S, Windbichler N, Wenger EA, Bever CA, Selvaraj P. Population replacement gene drive characteristics for malaria elimination in a range of seasonal transmission settings: a modelling study. Malar J 2022; 21:226. [PMID: 35883100 PMCID: PMC9327287 DOI: 10.1186/s12936-022-04242-2] [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: 11/16/2021] [Accepted: 07/11/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gene drives are a genetic engineering method where a suite of genes is inherited at higher than Mendelian rates and has been proposed as a promising new vector control strategy to reinvigorate the fight against malaria in sub-Saharan Africa. METHODS Using an agent-based model of malaria transmission with vector genetics, the impacts of releasing population-replacement gene drive mosquitoes on malaria transmission are examined and the population replacement gene drive system parameters required to achieve local elimination within a spatially-resolved, seasonal Sahelian setting are quantified. The performance of two different gene drive systems-"classic" and "integral"-are evaluated. Various transmission regimes (low, moderate, and high-corresponding to annual entomological inoculation rates of 10, 30, and 80 infectious bites per person) and other simultaneous interventions, including deployment of insecticide-treated nets (ITNs) and passive healthcare-seeking, are also simulated. RESULTS Local elimination probabilities decreased with pre-existing population target site resistance frequency, increased with transmission-blocking effectiveness of the introduced antiparasitic gene and drive efficiency, and were context dependent with respect to fitness costs associated with the introduced gene. Of the four parameters, transmission-blocking effectiveness may be the most important to focus on for improvements to future gene drive strains because a single release of classic gene drive mosquitoes is likely to locally eliminate malaria in low to moderate transmission settings only when transmission-blocking effectiveness is very high (above ~ 80-90%). However, simultaneously deploying ITNs and releasing integral rather than classic gene drive mosquitoes significantly boosts elimination probabilities, such that elimination remains highly likely in low to moderate transmission regimes down to transmission-blocking effectiveness values as low as ~ 50% and in high transmission regimes with transmission-blocking effectiveness values above ~ 80-90%. CONCLUSION A single release of currently achievable population replacement gene drive mosquitoes, in combination with traditional forms of vector control, can likely locally eliminate malaria in low to moderate transmission regimes within the Sahel. In a high transmission regime, higher levels of transmission-blocking effectiveness than are currently available may be required.
Collapse
Affiliation(s)
- Shirley Leung
- Institute for Disease Modeling, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Nikolai Windbichler
- Department of Life Sciences, Imperial College London, South Kensington, London, UK
| | - Edward A Wenger
- Institute for Disease Modeling, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Caitlin A Bever
- Institute for Disease Modeling, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Prashanth Selvaraj
- Institute for Disease Modeling, Bill & Melinda Gates Foundation, Seattle, WA, USA.
| |
Collapse
|
16
|
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
|
17
|
Hun LV, Cheung KW, Brooks E, Zudekoff R, Luckhart S, Riehle MA. Increased insulin signaling in the Anopheles stephensi fat body regulates metabolism and enhances the host response to both bacterial challenge and Plasmodium falciparum infection. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 139:103669. [PMID: 34666189 PMCID: PMC8647039 DOI: 10.1016/j.ibmb.2021.103669] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/09/2021] [Accepted: 10/10/2021] [Indexed: 05/06/2023]
Abstract
In vertebrates and invertebrates, the insulin/insulin-like growth factor 1 (IGF1) signaling (IIS) cascade is highly conserved and plays a vital role in many different physiological processes. Among the many tissues that respond to IIS in mosquitoes, the fat body has a central role in metabolism, lifespan, reproduction, and innate immunity. We previously demonstrated that fat body specific expression of active Akt, a key IIS signaling molecule, in adult Anopheles stephensi and Aedes aegypti activated the IIS cascade and extended lifespan. Additionally, we found that transgenic females produced more vitellogenin (Vg) protein than non-transgenic mosquitoes, although this did not translate into increased fecundity. These results prompted us to further examine how IIS impacts immunity, metabolism, growth and development of these transgenic mosquitoes. We observed significant changes in glycogen, trehalose, triglycerides, glucose, and protein in young (3-5 d) transgenic mosquitoes relative to non-transgenic sibling controls, while only triglycerides were significantly changed in older (18 d) transgenic mosquitoes. More importantly, we demonstrated that enhanced fat body IIS decreased both the prevalence and intensity of Plasmodium falciparum infection in transgenic An. stephensi. Additionally, challenging transgenic An. stephensi with Gram-positive and Gram-negative bacteria altered the expression of several antimicrobial peptides (AMPs) and two anti-Plasmodium genes, nitric oxide synthase (NOS) and thioester complement-like protein (TEP1), relative to non-transgenic controls. Increased IIS in the fat body of adult female An. stephensi had little to no impact on body size, growth or development of progeny from transgenic mosquitoes relative to non-transgenic controls. This study both confirms and expands our understanding of the critical roles insulin signaling plays in regulating the diverse functions of the mosquito fat body.
Collapse
Affiliation(s)
- Lewis V Hun
- Department of Entomology, University of California Riverside, Riverside, CA, USA; Department of Entomology, University of Arizona, Tucson, AZ, USA
| | - Kong Wai Cheung
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Elizabeth Brooks
- Department of Entomology, University of Arizona, Tucson, AZ, USA
| | - Rissa Zudekoff
- Department of Entomology, University of Arizona, Tucson, AZ, USA
| | - Shirley Luckhart
- Departrment of Entomology, Plant Pathology and Nematology and Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - Michael A Riehle
- Department of Entomology, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
18
|
Rani J, Chauhan C, Das De T, Kumari S, Sharma P, Tevatiya S, Patel K, Mishra AK, Pandey KC, Singh N, Dixit R. Hemocyte RNA-Seq analysis of Indian malarial vectors Anopheles stephensi and Anopheles culicifacies: From similarities to differences. Gene 2021; 798:145810. [PMID: 34224830 DOI: 10.1016/j.gene.2021.145810] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 06/26/2021] [Accepted: 06/30/2021] [Indexed: 02/05/2023]
Abstract
Anopheles stephensi and Anopheles culicifacies are dominant malarial vectors in urban and rural India, respectively. Both species carry significant biological differences in their behavioral adaptation and immunity, but the genetic basis of these variations are still poorly understood. Here, we uncovered the genetic differences of immune blood cells, that influence several immune-physiological responses. We generated, analyzed and compared the hemocyte RNA-Seq database of both mosquitoes. A total of 5,837,223,769 assembled bases collapsed into 7,595 and 3,791 transcripts, originating from hemocytes of laboratory-reared 3-4 days old naïve (sugar-fed) mosquitoes, Anopheles stephensi and Anopheles culicifacies respectively. Comparative GO annotation analysis revealed that both mosquito hemocytes encode similar proteins. Furthermore, while An. stephensi hemocytes showed a higher percentage of immune transcripts encoding APHAG (Autophagy), IMD (Immune deficiency pathway), PRDX (Peroxiredoxin), SCR (Scavenger receptor), IAP (Inhibitor of apoptosis), GALE (galactoside binding lectins), BGBPs (1,3 beta D glucan binding proteins), CASPs (caspases) and SRRP (Small RNA regulatory pathway), An. culicifacies hemocytes yielded a relatively higher percentage of transcripts encoding CLIP (Clip domain serine protease), FREP (Fibrinogen related proteins), PPO (Prophenol oxidase), SRPN (Serpines), ML (Myeloid differentiation 2-related lipid recognition protein), Toll path and TEP (Thioester protein), family proteins. However, a detailed comparative Interproscan analysis showed An. stephensi mosquito hemocytes encode proteins with increased repeat numbers as compared to An. culicifacies. Notably, we observed an abundance of transcripts showing significant variability of encoded proteins with repeats such as LRR (Leucine rich repeat), WD40 (W-D dipeptide), Ankyrin, Annexin, Tetratricopeptide and Mitochondrial substrate carrier repeat-containing family proteins, which may have a direct influence on species-specific immune-physiological responses. Summarily, our deep sequencing analysis unraveled that An. stephensi evolved with an expansion of repeat sequences in hemocyte proteins as compared to An. culicifacies, possibly providing an advantage for better adaptation to diverse environments.
Collapse
Affiliation(s)
- Jyoti Rani
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria, Research, Dwarka, New Delhi 110077, India; Department of Bio and Nanotechnology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, India
| | - Charu Chauhan
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria, Research, Dwarka, New Delhi 110077, India
| | - Tanwee Das De
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria, Research, Dwarka, New Delhi 110077, India
| | - Seena Kumari
- 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
| | - Sanjay Tevatiya
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria, Research, Dwarka, New Delhi 110077, India
| | - Karan Patel
- DNA Xperts Private Limited, Sector 63, Noida, Uttar Pradesh 20130, India
| | - Ashwani K Mishra
- DNA Xperts Private Limited, Sector 63, Noida, Uttar Pradesh 20130, India
| | - Kailash C Pandey
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria, Research, Dwarka, New Delhi 110077, India
| | - Namita Singh
- Department of Bio and Nanotechnology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, India
| | - Rajnikant Dixit
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria, Research, Dwarka, New Delhi 110077, India.
| |
Collapse
|
19
|
Adelman ZN, Kojin BB. Malaria-Resistant Mosquitoes (Diptera: Culicidae); The Principle is Proven, But Will the Effectors Be Effective? JOURNAL OF MEDICAL ENTOMOLOGY 2021; 58:1997-2005. [PMID: 34018548 DOI: 10.1093/jme/tjab090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Indexed: 06/12/2023]
Abstract
Over the last few decades, a substantial number of anti-malarial effector genes have been evaluated for their ability to block parasite infection in the mosquito vector. While many of these approaches have yielded significant effects on either parasite intensity or prevalence of infection, just a few have been able to completely block transmission. Additionally, many approaches, while effective against the parasite, also disrupt or alter important aspects of mosquito physiology, leading to corresponding changes in lifespan, reproduction, and immunity. As the most promising approaches move towards field-based evaluation, questions of effector gene robustness and durability move to the forefront. In this forum piece, we critically evaluate past effector gene approaches with an eye towards developing a deeper pipeline to augment the current best candidates.
Collapse
Affiliation(s)
- Zach N Adelman
- Department of Entomology and AgriLife Research, Texas A&M University, College Station, TX, USA
| | - Bianca B Kojin
- Department of Entomology and AgriLife Research, Texas A&M University, College Station, TX, USA
| |
Collapse
|
20
|
Dong S, Dong Y, Simões ML, Dimopoulos G. Mosquito transgenesis for malaria control. Trends Parasitol 2021; 38:54-66. [PMID: 34483052 DOI: 10.1016/j.pt.2021.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022]
Abstract
Malaria is one of the deadliest diseases. Because of the ineffectiveness of current malaria-control methods, several novel mosquito vector-based control strategies have been proposed to supplement existing control strategies. Mosquito transgenesis and gene drive have emerged as promising tools for preventing the spread of malaria by either suppressing mosquito populations by self-destructing mosquitoes or replacing mosquito populations with disease-refractory populations. Here we review the development of mosquito transgenesis and its application for malaria control, highlighting the transgenic expression of antiparasitic effector genes, inactivation of host factor genes, and manipulation of miRNAs and lncRNAs. Overall, from a malaria-control perspective, mosquito transgenesis is not envisioned as a stand-alone approach; rather, its use is proposed as a complement to existing vector-control strategies.
Collapse
Affiliation(s)
- Shengzhang Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
21
|
Cerenius L, Söderhäll K. Immune properties of invertebrate phenoloxidases. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 122:104098. [PMID: 33857469 DOI: 10.1016/j.dci.2021.104098] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/12/2021] [Accepted: 04/04/2021] [Indexed: 06/12/2023]
Abstract
Melanin production from different types of phenoloxidases (POs) confers immunity from a variety of pathogens ranging from viruses and microorganisms to parasites. The arthropod proPO expresses a variety of activities including cytokine, opsonin and microbiocidal activities independent of and even without melanin production. Proteolytic processing of proPO and its activating enzyme gives rise to several peptide fragments with a variety of separate activities in a process reminiscent of vertebrate complement system activation although proPO bears no sequence similarity to vertebrate complement factors. Pathogens influence proPO activation and thereby what types of immune effects that will be produced. An increasing number of specialised pathogens - from parasites to viruses - have been identified who can synthesise compounds specifically aimed at the proPO-system. In invertebrates outside the arthropods phylogenetically unrelated POs are participating in melanization reactions obviously aimed at intruders and/or aberrant tissues.
Collapse
Affiliation(s)
- Lage Cerenius
- Department of Organismal Biology,Uppsala University, Norbyvägen 18A, 752 36 Uppsala, Sweden.
| | - Kenneth Söderhäll
- Department of Organismal Biology,Uppsala University, Norbyvägen 18A, 752 36 Uppsala, Sweden
| |
Collapse
|
22
|
Caragata EP, Dong S, Dong Y, Simões ML, Tikhe CV, Dimopoulos G. Prospects and Pitfalls: Next-Generation Tools to Control Mosquito-Transmitted Disease. Annu Rev Microbiol 2021; 74:455-475. [PMID: 32905752 DOI: 10.1146/annurev-micro-011320-025557] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mosquito-transmitted diseases, including malaria and dengue, are a major threat to human health around the globe, affecting millions each year. A diverse array of next-generation tools has been designed to eliminate mosquito populations or to replace them with mosquitoes that are less capable of transmitting key pathogens. Many of these new approaches have been built on recent advances in CRISPR/Cas9-based genome editing. These initiatives have driven the development of pathogen-resistant lines, new genetics-based sexing methods, and new methods of driving desirable genetic traits into mosquito populations. Many other emerging tools involve microorganisms, including two strategies involving Wolbachia that are achieving great success in the field. At the same time, other mosquito-associated bacteria, fungi, and even viruses represent untapped sources of new mosquitocidal or antipathogen compounds. Although there are still hurdles to be overcome, the prospect that such approaches will reduce the impact of these diseases is highly encouraging.
Collapse
Affiliation(s)
- E P Caragata
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - S Dong
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - Y Dong
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - M L Simões
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - C V Tikhe
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| | - G Dimopoulos
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA; , , , , ,
| |
Collapse
|
23
|
E Silva B, Matsena Zingoni Z, Koekemoer LL, Dahan-Moss YL. Microbiota identified from preserved Anopheles. Malar J 2021; 20:230. [PMID: 34022891 PMCID: PMC8141131 DOI: 10.1186/s12936-021-03754-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 05/08/2021] [Indexed: 11/23/2022] Open
Abstract
Background Mosquito species from the Anopheles gambiae complex and the Anopheles funestus group are dominant African malaria vectors. Mosquito microbiota play vital roles in physiology and vector competence. Recent research has focused on investigating the mosquito microbiota, especially in wild populations. Wild mosquitoes are preserved and transported to a laboratory for analyses. Thus far, microbial characterization post-preservation has been investigated in only Aedes vexans and Culex pipiens. Investigating the efficacy of cost-effective preservatives has also been limited to AllProtect reagent, ethanol and nucleic acid preservation buffer. This study characterized the microbiota of African Anopheles vectors: Anopheles arabiensis (member of the An. gambiae complex) and An. funestus (member of the An. funestus group), preserved on silica desiccant and RNAlater® solution. Methods Microbial composition and diversity were characterized using culture-dependent (midgut dissections, culturomics, MALDI-TOF MS) and culture-independent techniques (abdominal dissections, DNA extraction, next-generation sequencing) from laboratory (colonized) and field-collected mosquitoes. Colonized mosquitoes were either fresh (non-preserved) or preserved for 4 and 12 weeks on silica or in RNAlater®. Microbiota were also characterized from field-collected An. arabiensis preserved on silica for 8, 12 and 16 weeks. Results Elizabethkingia anophelis and Serratia oryzae were common between both vector species, while Enterobacter cloacae and Staphylococcus epidermidis were specific to females and males, respectively. Microbial diversity was not influenced by sex, condition (fresh or preserved), preservative, or preservation time-period; however, the type of bacterial identification technique affected all microbial diversity indices. Conclusions This study broadly characterized the microbiota of An. arabiensis and An. funestus. Silica- and RNAlater®-preservation were appropriate when paired with culture-dependent and culture-independent techniques, respectively. These results broaden the selection of cost-effective methods available for handling vector samples for downstream microbial analyses. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03754-7.
Collapse
Affiliation(s)
- Bianca E Silva
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Zvifadzo Matsena Zingoni
- Division of Epidemiology and Biostatistics, School of Public Health, University of the Witwatersrand, Parktown, South Africa
| | - Lizette L Koekemoer
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
| | - Yael L Dahan-Moss
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa. .,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa.
| |
Collapse
|
24
|
Nuss A, Sharma A, Gulia-Nuss M. Genetic Manipulation of Ticks: A Paradigm Shift in Tick and Tick-Borne Diseases Research. Front Cell Infect Microbiol 2021; 11:678037. [PMID: 34041045 PMCID: PMC8141593 DOI: 10.3389/fcimb.2021.678037] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/16/2021] [Indexed: 12/13/2022] Open
Abstract
Ticks are obligate hematophagous arthropods that are distributed worldwide and are one of the most important vectors of pathogens affecting humans and animals. Despite the growing burden of tick-borne diseases, research on ticks has lagged behind other arthropod vectors, such as mosquitoes. This is largely because of challenges in applying functional genomics and genetic tools to the idiosyncrasies unique to tick biology, particularly techniques for stable genetic transformations. CRISPR-Cas9 is transforming non-model organism research; however, successful germline editing has yet to be accomplished in ticks. Here, we review the ancillary methods needed for transgenic tick development and the use of CRISPR/Cas9, the most promising gene-editing approach, for tick genetic transformation.
Collapse
Affiliation(s)
- Andrew Nuss
- Department of Biochemistry and Molecular Biology, The University of Nevada, Reno, NV, United States
- Department of Agriculture, Veterinary, and Rangeland Sciences, The University of Nevada, Reno, NV, United States
| | - Arvind Sharma
- Department of Biochemistry and Molecular Biology, The University of Nevada, Reno, NV, United States
| | - Monika Gulia-Nuss
- Department of Biochemistry and Molecular Biology, The University of Nevada, Reno, NV, United States
| |
Collapse
|
25
|
Hoermann A, Tapanelli S, Capriotti P, Del Corsano G, Masters EK, Habtewold T, Christophides GK, Windbichler N. Converting endogenous genes of the malaria mosquito into simple non-autonomous gene drives for population replacement. eLife 2021; 10:58791. [PMID: 33845943 PMCID: PMC8043746 DOI: 10.7554/elife.58791] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 03/21/2021] [Indexed: 12/15/2022] Open
Abstract
Gene drives for mosquito population replacement are promising tools for malaria control. However, there is currently no clear pathway for safely testing such tools in endemic countries. The lack of well-characterized promoters for infection-relevant tissues and regulatory hurdles are further obstacles for their design and use. Here we explore how minimal genetic modifications of endogenous mosquito genes can convert them directly into non-autonomous gene drives without disrupting their expression. We co-opted the native regulatory sequences of three midgut-specific loci of the malaria vector Anopheles gambiae to host a prototypical antimalarial molecule and guide-RNAs encoded within artificial introns that support efficient gene drive. We assess the propensity of these modifications to interfere with the development of Plasmodium falciparum and their effect on fitness. Because of their inherent simplicity and passive mode of drive such traits could form part of an acceptable testing pathway of gene drives for malaria eradication.
Collapse
Affiliation(s)
- Astrid Hoermann
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Sofia Tapanelli
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Paolo Capriotti
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | | - Ellen Kg Masters
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Tibebu Habtewold
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | | - Nikolai Windbichler
- Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
26
|
Volohonsky G, Paul-Gilloteaux P, Štáfková J, Soichot J, Salamero J, Levashina EA. Kinetics of Plasmodium midgut invasion in Anopheles mosquitoes. PLoS Pathog 2020; 16:e1008739. [PMID: 32946522 PMCID: PMC7526910 DOI: 10.1371/journal.ppat.1008739] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 09/30/2020] [Accepted: 06/23/2020] [Indexed: 01/06/2023] Open
Abstract
Malaria-causing Plasmodium parasites traverse the mosquito midgut cells to establish infection at the basal side of the midgut. This dynamic process is a determinant of mosquito vector competence, yet the kinetics of the parasite migration is not well understood. Here we used transgenic mosquitoes of two Anopheles species and a Plasmodium berghei fluorescence reporter line to track parasite passage through the mosquito tissues at high spatial resolution. We provide new quantitative insight into malaria parasite invasion in African and Indian Anopheles species and propose that the mosquito complement-like system contributes to the species-specific dynamics of Plasmodium invasion. The traversal of the mosquito midgut cells is one of the critical stages in the life cycle of malaria parasites. Motile parasite forms, called ookinetes, traverse the midgut epithelium in a dynamic process which is not fully understood. Here, we harnessed transgenic reporters to track invasion of Plasmodium parasites in African and Indian mosquito species. We found important differences in parasite dynamics between the two Anopheles species and demonstrated a role of the mosquito complement-like system in regulation of parasite invasion of the midgut cells.
Collapse
Affiliation(s)
- Gloria Volohonsky
- INSERM U963, CNRS UPR9022, University of Strasbourg, Strasbourg, France
| | - Perrine Paul-Gilloteaux
- SERPICO Inria Team/CNRS UMR 144, Institut Curie, Paris, France.,National Biology and Health Infrastructure "France Bioimaging", Institut Curie, Paris, France.,Cell and Tissue Imaging Facility, IBiSA, Institut Curie, Paris, France
| | - Jitka Štáfková
- INSERM U963, CNRS UPR9022, University of Strasbourg, Strasbourg, France
| | - Julien Soichot
- INSERM U963, CNRS UPR9022, University of Strasbourg, Strasbourg, France
| | - Jean Salamero
- SERPICO Inria Team/CNRS UMR 144, Institut Curie, Paris, France.,National Biology and Health Infrastructure "France Bioimaging", Institut Curie, Paris, France.,Cell and Tissue Imaging Facility, IBiSA, Institut Curie, Paris, France
| | - Elena A Levashina
- INSERM U963, CNRS UPR9022, University of Strasbourg, Strasbourg, France.,Vector Biology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
27
|
Yu S, Wang P, Qin J, Zheng H, Wang J, Liu T, Yang X, Wang Y. Bacillus sphaericus exposure reduced vector competence of Anopheles dirus to Plasmodium yoelii by upregulating the Imd signaling pathway. Parasit Vectors 2020; 13:446. [PMID: 32891162 PMCID: PMC7487769 DOI: 10.1186/s13071-020-04321-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/30/2020] [Indexed: 12/05/2022] Open
Abstract
Background Vector control with Bacillus sphaericus (Bs) is an effective way to block the transmission of malaria. However, in practical application of Bs agents, a sublethal dose effect was often caused by insufficient dosing, and it is little known whether the Bs exposure would affect the surviving mosquitoes’ vector capacity to malaria. Methods A sublethal dose of the Bs 2362 strain was administrated to the early fourth-instar larvae of Anopheles dirus to simulate shortage use of Bs in field circumstance. To determine vector competence, mosquitoes were dissected and the oocysts in the midguts were examined on day 9–11 post-infection with Plasmodium yoelii. Meanwhile, a SYBR quantitative PCR assay was conducted to examine the transcriptional level of the key immune molecules of mosquitoes, and RNA interference was utilized to validate the role of key immune effector molecule TEP1. Results The sublethal dose of Bs treatment significantly reduced susceptibility of An. dirus to P. yoelii, with the decrease of P. yoelii infection intensity and rate. Although there existed a melanization response of adult An. dirus following challenge with P. yoelii, it was not involved in the decrease of vector competence as no significant difference of melanization rates and densities between the control and Bs groups was found. Further studies showed that Bs treatment significantly increased TEP1 expression in the fourth-instar larvae (L4), pupae (Pu), 48 h post-infection (hpi) and 72 hpi (P < 0.001). Further, gene-silencing of TEP1 resulted in disappearance of the Bs impact on vector competence of An. dirus to P. yoelii. Moreover, the transcriptional level of PGRP-LC and Rel2 were significantly elevated by Bs treatment with decreased expression of the negative regulator Caspar at 48 hpi, which implied that the Imd signaling pathway was upregulated by Bs exposure. Conclusions Bs exposure can reduce the vector competence of An. dirus to malaria parasites through upregulating Imd signaling pathway and enhancing the expression of TEP1. The data could not only help us to understand the impact and mechanism of Bs exposure on Anopheles’ vector competence to malaria but also provide us with novel clues for wiping out malaria using vector control.![]()
Collapse
Affiliation(s)
- Shasha Yu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Pan Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Jie Qin
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Jing Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Tingting Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Xuesen Yang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Ying Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
28
|
Nateghi Rostami M. CRISPR/Cas9 gene drive technology to control transmission of vector‐borne parasitic infections. Parasite Immunol 2020; 42:e12762. [DOI: 10.1111/pim.12762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Mahmoud Nateghi Rostami
- Laboratory of Biology of Host‐Parasite Interactions Department of Parasitology Pasteur Institute of Iran Tehran Iran
| |
Collapse
|
29
|
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
|
30
|
Epis S, Varotto-Boccazzi I, Crotti E, Damiani C, Giovati L, Mandrioli M, Biggiogera M, Gabrieli P, Genchi M, Polonelli L, Daffonchio D, Favia G, Bandi C. Chimeric symbionts expressing a Wolbachia protein stimulate mosquito immunity and inhibit filarial parasite development. Commun Biol 2020; 3:105. [PMID: 32144396 PMCID: PMC7060271 DOI: 10.1038/s42003-020-0835-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/18/2020] [Indexed: 12/28/2022] Open
Abstract
Wolbachia can reduce the capability of mosquitoes to transmit infectious diseases to humans and is currently exploited in campaigns for the control of arboviruses, like dengue and Zika. Under the assumption that Wolbachia-mediated activation of insect immunity plays a role in the reduction of mosquito vectorial capacity, we focused our attention on the Wolbachia surface protein (WSP), a potential inductor of innate immunity. We hypothesized that the heterologous expression of this protein in gut- and tissue-associated symbionts may reduce parasite transmission. We thus engineered the mosquito bacterial symbiont Asaia to express WSP (AsaiaWSP). AsaiaWSP induced activation of the host immune response in Aedes aegypti and Anopheles stephensi mosquitoes, and inhibited the development of the heartworm parasite Dirofilaria immitis in Ae. aegypti. These results consolidate previous evidence on the immune-stimulating property of WSP and make AsaiaWSP worth of further investigations as a potential tool for the control of mosquito-borne diseases. Epis and Varotto-Boccazzi et al. show that Wolbachia surface protein (WSP) activates host innate immunity in mosquitoes, inhibiting the development of the heartworm parasite in its insect host. This study suggests the possibility that the WSP-expressing symbiont may be harnessed to control mosquito-borne diseases.
Collapse
Affiliation(s)
- Sara Epis
- Department of Biosciences and Pediatric Clinical Research Center "Romeo and Enrica Invernizzi", University of Milan, Milan, Italy.,Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Milan, Italy
| | - Ilaria Varotto-Boccazzi
- Department of Biosciences and Pediatric Clinical Research Center "Romeo and Enrica Invernizzi", University of Milan, Milan, Italy.,Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Milan, Italy
| | - Elena Crotti
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Milan, Italy
| | - Claudia Damiani
- Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Milan, Italy.,School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Laura Giovati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Mauro Mandrioli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marco Biggiogera
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Paolo Gabrieli
- Department of Biosciences and Pediatric Clinical Research Center "Romeo and Enrica Invernizzi", University of Milan, Milan, Italy.,Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Milan, Italy
| | - Marco Genchi
- Department of Veterinary Sciences, University of Parma, Parma, Italy
| | - Luciano Polonelli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Daniele Daffonchio
- King Abdullah University of Science and Technology, Red Sea Research Center, Thuwal, Saudi Arabia
| | - Guido Favia
- Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Milan, Italy.,School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Claudio Bandi
- Department of Biosciences and Pediatric Clinical Research Center "Romeo and Enrica Invernizzi", University of Milan, Milan, Italy. .,Centro Interuniversitario di Ricerca sulla Malaria/Italian Malaria Network, Milan, Italy.
| |
Collapse
|
31
|
Yang J, Schleicher TR, Dong Y, Park HB, Lan J, Cresswell P, Crawford J, Dimopoulos G, Fikrig E. Disruption of mosGILT in Anopheles gambiae impairs ovarian development and Plasmodium infection. J Exp Med 2020; 217:e20190682. [PMID: 31658986 PMCID: PMC7037243 DOI: 10.1084/jem.20190682] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/06/2019] [Accepted: 10/25/2019] [Indexed: 11/04/2022] Open
Abstract
Plasmodium infection in Anopheles is influenced by mosquito-derived factors. We previously showed that a protein in saliva from infected Anopheles, mosquito gamma-interferon-inducible lysosomal thiol reductase (mosGILT), inhibits the ability of sporozoites to traverse cells and readily establish infection of the vertebrate host. To determine whether mosGILT influences Plasmodium within the mosquito, we generated Anopheles gambiae mosquitoes carrying mosaic mutations in the mosGILT gene using CRISPR/CRISPR associated protein 9 (Cas9). Here, we show that female mosaic mosGILT mutant mosquitoes display defects in ovarian development and refractoriness to Plasmodium. Following infection by either Plasmodium berghei or Plasmodium falciparum, mutant mosquitoes have significantly reduced oocyst numbers as a result of increased thioester-containing protein 1 (TEP1)-dependent parasite killing. Expression of vitellogenin (Vg), the major yolk protein that can reduce the parasite-killing efficiency of TEP1, is severely impaired in mutant mosquitoes. MosGILT is a mosquito factor that is essential for ovarian development and indirectly protects both human and rodent Plasmodium species from mosquito immunity.
Collapse
Affiliation(s)
- Jing Yang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Tyler R. Schleicher
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Yuemei Dong
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Hyun Bong Park
- Department of Chemistry, Yale University, New Haven, CT
- Chemical Biology Institute, Yale University, West Haven, CT
| | - Jiangfeng Lan
- College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Peter Cresswell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Jason Crawford
- Department of Chemistry, Yale University, New Haven, CT
- Chemical Biology Institute, Yale University, West Haven, CT
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT
| | - George Dimopoulos
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Chevy Chase, MD
| |
Collapse
|
32
|
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
|
33
|
Reyes Ruiz VM, Sousa GL, Sneed SD, Farrant KV, Christophides GK, Povelones M. Stimulation of a protease targeting the LRIM1/APL1C complex reveals specificity in complement-like pathway activation in Anopheles gambiae. PLoS One 2019; 14:e0214753. [PMID: 30958840 PMCID: PMC6453449 DOI: 10.1371/journal.pone.0214753] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/19/2019] [Indexed: 12/15/2022] Open
Abstract
The complement-like pathway of the African malaria mosquito Anopheles gambiae provides protection against infection by diverse pathogens. A functional requirement for a core set of proteins during infections by rodent and human malaria parasites, bacteria, and fungi suggests a similar mechanism operates against different pathogens. However, the extent to which the molecular mechanisms are conserved is unknown. In this study we probed the biochemical responses of complement-like pathway to challenge by the Gram-positive bacterium Staphyloccocus aureus. Western blot analysis of the hemolymph revealed that S. aureus challenge activates a TEP1 convertase-like activity and promotes the depletion of the protein SPCLIP1. S. aureus challenge did not lead to an apparent change in the abundance of the LRIM1/APL1C complex compared to challenge by the Gram-negative bacterium, Escherichia coli. Following up on this observation using a panel of LRIM1 and APL1C antibodies, we found that E. coli challenge, but not S. aureus, specifically activates a protease that cleaves the C-terminus of APL1C. Inhibitor studies in vivo and in vitro protease assays suggest that a serine protease is responsible for APL1C cleavage. This study reveals that despite different challenges converging on activation of a TEP1 convertase-like activity, the mosquito complement-like pathway also includes pathogen-specific reactions.
Collapse
Affiliation(s)
- Valeria M. Reyes Ruiz
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gregory L. Sousa
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sarah D. Sneed
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Katie V. Farrant
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | | - Michael Povelones
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
34
|
Yan Y, Hillyer JF. Complement-like proteins TEP1, TEP3 and TEP4 are positive regulators of periostial hemocyte aggregation in the mosquito Anopheles gambiae. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 107:1-9. [PMID: 30690067 DOI: 10.1016/j.ibmb.2019.01.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/16/2019] [Accepted: 01/18/2019] [Indexed: 06/09/2023]
Abstract
The mosquito immune and circulatory systems are functionally integrated. During an infection, hemocytes aggregate around the ostia (valves) of the dorsal vessel - areas of the heart called the periostial regions - where they phagocytose live and melanized pathogens. Although periostial hemocyte aggregation is an immune response that occurs following infection with bacteria and malaria parasites, the molecular basis of this process remains poorly understood. Here, we show that the thioester-containing proteins, TEP1, TEP3 and TEP4 are positive regulators of periostial hemocyte aggregation in the African malaria mosquito, Anopheles gambiae. RNAi-based knockdown of TEP1, TEP3 and TEP4 resulted in fewer periostial hemocytes following Escherichia coli infection, without affecting the adjacent population of non-periostial, sessile hemocytes. Moreover, knockdown of TEP1, TEP3 and TEP4 expression resulted in reduced bacterial accumulation and melanin deposition at the periostial regions. Finally, this study confirmed the role that TEP1 plays in reducing infection intensity in the hemocoel. Overall, this research shows that the complement-like proteins, TEP1, TEP3 and TEP4, are positive regulators of the functional integration between the immune and circulatory systems of mosquitoes.
Collapse
Affiliation(s)
- Yan Yan
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Julián F Hillyer
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
35
|
Pinaud S, Portet A, Allienne JF, Belmudes L, Saint-Beat C, Arancibia N, Galinier R, Du Pasquier L, Duval D, Gourbal B. Molecular characterisation of immunological memory following homologous or heterologous challenges in the schistosomiasis vector snail, Biomphalaria glabrata. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 92:238-252. [PMID: 30529491 DOI: 10.1016/j.dci.2018.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/30/2018] [Accepted: 12/01/2018] [Indexed: 05/16/2023]
Abstract
Invertebrate immune response may be primed by a current infection in a sustained manner, leading to the failure of a secondary infection with the same pathogen. The present study focuses on the Schistosomiasis vector snail Biomphalaria glabrata, in which a specific genotype-dependent immunological memory was demonstrated as a shift from a cellular to a humoral immune response. Herein, we investigate the complex molecular bases associated with this genotype-dependant immunological memory response. We demonstrate that Biomphalaria regulates a polymorphic set of immune recognition molecules and immune effector repertoires to respond to different strains of Schistosoma parasites. These results suggest a combinatorial usage of pathogen recognition receptors (PRRs) that distinguish different strains of parasites during the acquisition of immunological memory. Immunizations also show that snails become resistant after exposure to parasite extracts. Hemolymph transfer and a label-free proteomic analysis proved that circulating hemolymph compounds can be produced and released to more efficiently kill the newly encountered parasite of the same genetic lineage.
Collapse
Affiliation(s)
- Silvain Pinaud
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environments UMR 5244, CNRS, IFREMER, Univ. Montpellier, F-66860, Perpignan, France.
| | - Anaïs Portet
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environments UMR 5244, CNRS, IFREMER, Univ. Montpellier, F-66860, Perpignan, France.
| | - Jean-François Allienne
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environments UMR 5244, CNRS, IFREMER, Univ. Montpellier, F-66860, Perpignan, France.
| | - Lucid Belmudes
- CEA-Grenoble, Exploring the Dynamics of Proteomes (EDyP), F-38054, Grenoble, Cedex 9, France.
| | - Cécile Saint-Beat
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environments UMR 5244, CNRS, IFREMER, Univ. Montpellier, F-66860, Perpignan, France.
| | - Nathalie Arancibia
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environments UMR 5244, CNRS, IFREMER, Univ. Montpellier, F-66860, Perpignan, France.
| | - Richard Galinier
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environments UMR 5244, CNRS, IFREMER, Univ. Montpellier, F-66860, Perpignan, France.
| | - Louis Du Pasquier
- University of Basel, Zoological Institute, Department of Zoology and Evolutionary Biology Vesalgasse 1, Basel, Switzerland.
| | - David Duval
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environments UMR 5244, CNRS, IFREMER, Univ. Montpellier, F-66860, Perpignan, France.
| | - Benjamin Gourbal
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environments UMR 5244, CNRS, IFREMER, Univ. Montpellier, F-66860, Perpignan, France.
| |
Collapse
|
36
|
Abstract
Vector control programs based on population reduction by matings with mass-released sterile insects require the release of only male mosquitoes, as the release of females, even if sterile, would increase the number of biting and potentially disease-transmitting individuals. While small-scale releases demonstrated the applicability of sterile males releases to control the yellow fever mosquito Aedes aegypti, large-scale programs for mosquitoes are currently prevented by the lack of efficient sexing systems in any of the vector species.Different approaches of sexing are pursued, including classical genetic and mechanical methods of sex separation. Another strategy is the development of transgenic sexing systems. Such systems already exist in other insect pests. Genome modification tools could be used to apply similar strategies to mosquitoes. Three major tools to modify mosquito genomes are currently used: transposable elements, site-specific recombination systems, and genome editing via TALEN or CRISPR/Cas. All three can serve the purpose of developing sexing systems and vector control strains in mosquitoes in two ways: first, via their use in basic research. A better understanding of mosquito biology, including the sex-determining pathways and the involved genes can greatly facilitate the development of sexing strains. Moreover, basic research can help to identify other regulatory elements and genes potentially useful for the construction of transgenic sexing systems. Second, these genome modification tools can be used to apply the gained knowledge to build and test mosquito sexing strains for vector control.
Collapse
Affiliation(s)
- Irina Häcker
- Institute for Insect Biotechnology, Justus-Liebig-University Giessen, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany.
| | - Marc F Schetelig
- Institute for Insect Biotechnology, Justus-Liebig-University Giessen, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany
| |
Collapse
|
37
|
Adolfi A, Lycett GJ. Opening the toolkit for genetic analysis and control of Anopheles mosquito vectors. CURRENT OPINION IN INSECT SCIENCE 2018; 30:8-18. [PMID: 30553490 DOI: 10.1016/j.cois.2018.07.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 07/24/2018] [Indexed: 06/09/2023]
Abstract
Anopheles is the only genus of mosquitoes that transmit human malaria and consequently the focus of large scale genome and transcriptome-wide association studies. Genetic tools to define the function of the candidate genes arising from these analyses are vital. Moreover, genome editing offers the potential to modify Anopheles population structure at local and global scale to provide complementary tools towards the ultimate goal of malaria elimination. Major breakthroughs in Anopheles genetic analysis came with the development of germline transformation and RNA interference technology. Yet, the field has been revolutionised again by precise genome editing now possible through site-specific nucleases. Here we review the components of the current genetic toolkit available to study Anopheles, focusing particularly on how these technical advances are used to gain insight into malaria transmission and the design of genetic methods to control Anopheles vectors.
Collapse
Affiliation(s)
- Adriana Adolfi
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, CA 92697-4500, USA
| | - Gareth John Lycett
- Vector Biology Department, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| |
Collapse
|
38
|
Zumaya-Estrada FA, Rodríguez MC, Rodríguez MH. Pathogen-insect interaction candidate molecules for transmission-blocking control strategies of vector borne diseases. SALUD PUBLICA DE MEXICO 2018; 60:77-85. [PMID: 29689660 DOI: 10.21149/8140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 02/24/2017] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To analyze the current knowledge of pathogen-insect interactions amenable for the design of molecular-based control strategies of vector-borne diseases. MATERIALS AND METHODS We examined malaria, dengue, and Chagas disease pathogens and insect molecules that participate in interactions during their vectors infection. RESULTS Pathogen molecules that participate in the insect intestine invasion and induced vector immune molecules are presented, and their inclusion in transmission blocking vaccines (TBV) and in genetically modify insect (GMI) vectors or symbiotic bacteria are discussed. CONCLUSIONS Disruption of processes by blocking vector-pathogen interactions provides several candidates for molecular control strategies, but TBV and GMI efficacies are still limited and other secondary effects of GMI (improving transmission of other pathogens, affectation of other organisms) should be discarded.
Collapse
Affiliation(s)
- Federico Alonso Zumaya-Estrada
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Cuernavaca, Morelos, México
| | - María Carmen Rodríguez
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Cuernavaca, Morelos, México
| | - Mario Henry Rodríguez
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Cuernavaca, Morelos, México
| |
Collapse
|
39
|
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
|
40
|
Adolfi A, Pondeville E, Lynd A, Bourgouin C, Lycett GJ. Multi-tissue GAL4-mediated gene expression in all Anopheles gambiae life stages using an endogenous polyubiquitin promoter. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2018; 96:1-9. [PMID: 29578046 DOI: 10.1016/j.ibmb.2018.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/12/2018] [Accepted: 03/19/2018] [Indexed: 06/08/2023]
Abstract
The ability to manipulate the Anopheles gambiae genome and alter gene expression effectively and reproducibly is a prerequisite for functional genetic analysis and for the development of novel control strategies in this important disease vector. However, in vivo transgenic analysis in mosquitoes is limited by the lack of promoters active ubiquitously. To address this, we used the GAL4/UAS system to investigate the promoter of the An. gambiae Polyubiquitin-c (PUBc) gene and demonstrated its ability to drive expression in mosquito cell culture before incorporation into An. gambiae transgenic driver lines. To generate such lines, piggyBac-mediated insertion was used to identify genomic regions able to sustain widespread expression and to create φC31 docking lines at these permissive sites. Patterns of expression induced by PUBc-GAL4 drivers carrying single intergenic insertions were assessed by crossing with a novel responder UAS-mCD8:mCherry line that was created by φC31-mediated integration. Amongst the drivers created at single, unique chromosomal integration loci, two were isolated that induced differential expression levels in a similar multiple-tissue spatial pattern throughout the mosquito life cycle. This work expands the tools available for An. gambiae functional analysis by providing a novel promoter for investigating phenotypes resulting from widespread multi-tissue expression, as well as identifying and tagging genomic sites that sustain broad transcriptional activity.
Collapse
Affiliation(s)
- Adriana Adolfi
- Liverpool School of Tropical Medicine, Vector Biology Department, Liverpool, UK.
| | - Emilie Pondeville
- Institut Pasteur, Genetics and Genomics of Insect Vectors, CNRS Unit URA3012, Paris, France.
| | - Amy Lynd
- Liverpool School of Tropical Medicine, Vector Biology Department, Liverpool, UK
| | - Catherine Bourgouin
- Institut Pasteur, Genetics and Genomics of Insect Vectors, CNRS Unit URA3012, Paris, France
| | - Gareth J Lycett
- Liverpool School of Tropical Medicine, Vector Biology Department, Liverpool, UK.
| |
Collapse
|
41
|
Liu PW, Xu JB, Dong YQ, Chen XG, Gu JB. Use of a Recombinant Mosquito Densovirus As a Gene Delivery Vector for the Functional Analysis of Genes in Mosquito Larvae. J Vis Exp 2017. [PMID: 29053694 DOI: 10.3791/56121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
In vivo microinjection is the most commonly used gene transfer technique for analyzing the gene functions in individual mosquitoes. However, this method requires a more technically demanding operation and involves complicated procedures, especially when used in larvae due to their small size, relatively thin and fragile cuticle, and high mortality, which limit its application. In contrast, viral vectors for gene delivery have been developed to surmount extracellular and intracellular barriers. These systems have the advantages of easy manipulation, high gene transduction efficiency, long-term maintenance of gene expression, and the ability to produce persistent effects in vivo. Mosquito densoviruses (MDVs) are mosquito-specific, small single-stranded DNA viruses that can effectively deliver foreign nucleic acids into mosquito cells; however, the replacement or insertion of foreign genes to create recombinant viruses typically causes a loss of packaging and/or replication abilities, which is a barrier to the development of these viruses as delivery vectors. Herein, we report using an artificial intronic small-RNA expression strategy to develop a non-defective recombinant Aedes aegypti densovirus (AaeDV) in vivo delivery system. Detailed procedures for the construction, packaging and quantitative analysis of the rAaeDV vectors, and for larval infection are described. This study demonstrates, for the first time, the feasibility of developing a non-defective recombinant MDV micro RNA (miRNA) expression system, and thus providing a powerful tool for the functional analysis of genes in mosquito and establishing a basis for the application of viral paratransgenesis for controlling mosquito-borne diseases. We demonstrated that Aedes albopictus 1st instar larvae could be easily and effectively infected by introducing the virus into the water body of the larvae breeding site and that the developed rAaeDVs could be used to overexpress or knock down the expression of a specific target gene in larvae, providing a tool for the functional analysis of mosquito genes.
Collapse
Affiliation(s)
- Pei-Wen Liu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University
| | - Jia-Bao Xu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University
| | - Yun-Qiao Dong
- Reproductive Medical Centre of Guangdong Women and Children's Hospital
| | - Xiao-Guang Chen
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University
| | - Jin-Bao Gu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University;
| |
Collapse
|
42
|
Nakhleh J, Christophides GK, Osta MA. The serine protease homolog CLIPA14 modulates the intensity of the immune response in the mosquito Anopheles gambiae. J Biol Chem 2017; 292:18217-18226. [PMID: 28928218 PMCID: PMC5672044 DOI: 10.1074/jbc.m117.797787] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/13/2017] [Indexed: 11/06/2022] Open
Abstract
Clip domain serine protease homologs (SPHs) are positive and negative regulators of Anopheles gambiae immune responses mediated by the complement-like protein TEP1 against Plasmodium malaria parasites and other microbial infections. We have previously reported that the SPH CLIPA2 is a negative regulator of the TEP1-mediated response by showing that CLIPA2 knockdown (kd) enhances mosquito resistance to infections with fungi, bacteria, and Plasmodium parasites. Here, we identify another SPH, CLIPA14, as a novel regulator of mosquito immunity. We found that CLIPA14 is a hemolymph protein that is rapidly cleaved following a systemic infection. CLIPA14 kd mosquitoes elicited a potent melanization response against Plasmodium berghei ookinetes and exhibited significantly increased resistance to Plasmodium infections as well as to systemic and oral bacterial infections. The activity of the enzyme phenoloxidase, which initiates melanin biosynthesis, dramatically increased in the hemolymph of CLIPA14 kd mosquitoes in response to systemic bacterial infections. Ookinete melanization and hemolymph phenoloxidase activity were further increased after cosilencing CLIPA14 and CLIPA2, suggesting that these two SPHs act in concert to control the melanization response. Interestingly, CLIPA14 RNAi phenotypes and its infection-induced cleavage were abolished in a TEP1 loss-of-function background. Our results suggest that a complex network of SPHs functions downstream of TEP1 to regulate the melanization reaction.
Collapse
Affiliation(s)
- Johnny Nakhleh
- From the Department of Biology, American University of Beirut, Beirut 1107 2020, Lebanon and
| | | | - Mike A Osta
- From the Department of Biology, American University of Beirut, Beirut 1107 2020, Lebanon and
| |
Collapse
|
43
|
Liew JW, Fong MY, Lau YL. Quantitative real-time PCR analysis of Anopheles dirus TEP1 and NOS during Plasmodium berghei infection, using three reference genes. PeerJ 2017; 5:e3577. [PMID: 28761783 PMCID: PMC5533154 DOI: 10.7717/peerj.3577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/23/2017] [Indexed: 01/12/2023] Open
Abstract
Quantitative reverse transcription PCR (qRT-PCR) has been an integral part of characterizing the immunity of Anopheles mosquitoes towards Plasmodium invasion. Two anti-Plasmodium factors of Anopheles, thioester-containing protein 1 (TEP1) and nitric oxide synthase (NOS), play a role in the refractoriness of Anopheles towards Plasmodium infection and are generally expressed during infection. However, these are less studied in Anopheles dirus, a dominant malaria vector in Southeast Asia. Furthermore, most studies used a single reference gene for normalization during gene expression analysis without proper validation. This may lead to erroneous quantification of expression levels. Therefore, the present study characterized and investigated the expression profiles of TEP1 and NOS of Anopheles dirus during P. berghei infection. Prior to that, the elongation factor 1-alpha (EF1), actin 1 (Act) and ribosomal protein S7 (S7) genes were validated for their suitability as a set of reference genes. TEP1 and NOS expressions in An. dirus were found to be significantly induced after P. berghei infection.
Collapse
Affiliation(s)
- Jonathan W.K. Liew
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mun Yik Fong
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yee Ling Lau
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|