1
|
Omokungbe B, Centurión A, Stiehler S, Morr A, Vilcinskas A, Steinbrink A, Hardes K. Gene silencing in the aedine cell lines C6/36 and U4.4 using long double-stranded RNA. Parasit Vectors 2024; 17:255. [PMID: 38863029 PMCID: PMC11167938 DOI: 10.1186/s13071-024-06340-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/30/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND RNA interference (RNAi) is a target-specific gene silencing method that can be used to determine gene functions and investigate host-pathogen interactions, as well as facilitating the development of ecofriendly pesticides. Commercially available transfection reagents (TRs) can improve the efficacy of RNAi. However, we currently lack a product and protocol for the transfection of insect cell lines with long double-stranded RNA (dsRNA). METHODS We used agarose gel electrophoresis to determine the capacity of eight TRs to form complexes with long dsRNA. A CellTiter-Glo assay was then used to assess the cytotoxicity of the resulting lipoplexes. We also measured the cellular uptake of dsRNA by fluorescence microscopy using the fluorophore Cy3 as a label. Finally, we analyzed the TRs based on their transfection efficacy and compared the RNAi responses of Aedes albopictus C6/36 and U4.4 cells by knocking down an mCherry reporter Semliki Forest virus in both cell lines. RESULTS The TRs from Biontex (K4, Metafectene Pro, and Metafectene SI+) showed the best complexing capacity and the lowest dsRNA:TR ratio needed for complete complex formation. Only HiPerFect was unable to complex the dsRNA completely, even at a ratio of 1:9. Most of the complexes containing mCherry-dsRNA were nontoxic at 2 ng/µL, but Lipofectamine 2000 was toxic at 1 ng/µL in U4.4 cells and at 2 ng/µL in C6/36 cells. The transfection of U4.4 cells with mCherry-dsRNA/TR complexes achieved significant knockdown of the virus reporter. Comparison of the RNAi response in C6/36 and U4.4 cells suggested that C6/36 cells lack the antiviral RNAi response because there was no significant knockdown of the virus reporter in any of the treatments. CONCLUSIONS C6/36 cells have an impaired RNAi response as previously reported. This investigation provides valuable information for future RNAi experiments by showing how to mitigate the adverse effects attributed to TRs. This will facilitate the judicious selection of TRs and transfection conditions conducive to RNAi research in mosquitoes.
Collapse
Affiliation(s)
- Bodunrin Omokungbe
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE TBG), Senckenberganlage 25, 60325, Frankfurt Am Main, Germany
- Institute for Insect Biotechnology, Justus-Liebig University, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany
| | - Alejandra Centurión
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE TBG), Senckenberganlage 25, 60325, Frankfurt Am Main, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch of Bioresources, Ohlebergsweg 12, 35392, Giessen, Germany
| | - Sabrina Stiehler
- Institute for Insect Biotechnology, Justus-Liebig University, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany
| | - Antonia Morr
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch of Bioresources, Ohlebergsweg 12, 35392, Giessen, Germany
| | - Andreas Vilcinskas
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE TBG), Senckenberganlage 25, 60325, Frankfurt Am Main, Germany
- Institute for Insect Biotechnology, Justus-Liebig University, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch of Bioresources, Ohlebergsweg 12, 35392, Giessen, Germany
| | - Antje Steinbrink
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE TBG), Senckenberganlage 25, 60325, Frankfurt Am Main, Germany
- Institute for Insect Biotechnology, Justus-Liebig University, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany
| | - Kornelia Hardes
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE TBG), Senckenberganlage 25, 60325, Frankfurt Am Main, Germany.
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch of Bioresources, Ohlebergsweg 12, 35392, Giessen, Germany.
- BMBF Junior Research Group in Infection Research "ASCRIBE", Ohlebergsweg 12, 35392, Giessen, Germany.
| |
Collapse
|
2
|
Li M, Zhou Y, Cheng J, Wang Y, Lan C, Shen Y. Response of the mosquito immune system and symbiotic bacteria to pathogen infection. Parasit Vectors 2024; 17:69. [PMID: 38368353 PMCID: PMC10874582 DOI: 10.1186/s13071-024-06161-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/24/2024] [Indexed: 02/19/2024] Open
Abstract
Mosquitoes are the deadliest animal in the word, transmitting a variety of insect-borne infectious diseases, such as malaria, dengue fever, yellow fever, and Zika, causing more deaths than any other vector-borne pathogen. Moreover, in the absence of effective drugs and vaccines to prevent and treat insect-borne diseases, mosquito control is particularly important as the primary measure. In recent decades, due to the gradual increase in mosquito resistance, increasing attention has fallen on the mechanisms and effects associated with pathogen infection. This review provides an overview of mosquito innate immune mechanisms in terms of physical and physiological barriers, pattern recognition receptors, signalling pathways, and cellular and humoral immunity, as well as the antipathogenic effects of mosquito symbiotic bacteria. This review contributes to an in-depth understanding of the interaction process between mosquitoes and pathogens and provides a theoretical basis for biological defence strategies against mosquito-borne infectious diseases.
Collapse
Affiliation(s)
- Manjin Li
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yang Zhou
- Nanjing Medical University, Nanjing, 211166, China
| | - Jin Cheng
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yiqing Wang
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Cejie Lan
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
| | - Yuan Shen
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
- Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
3
|
Svobodová K, Krištůfek V, Kubásek J, Krejčí A. Alcohol extract of the gypsy mushroom (Cortinarius caperatus) inhibits the development of Deformed wing virus infection in western honey bee (Apis mellifera). JOURNAL OF INSECT PHYSIOLOGY 2024; 152:104583. [PMID: 37979771 DOI: 10.1016/j.jinsphys.2023.104583] [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: 03/14/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/20/2023]
Abstract
Deformed wing virus (DWV) transmitted by the parasitic mite Varroa destructor is one of the most significant factors contributing to massive losses of managed colonies of western honey bee (Apis mellifera) subspecies of European origin reported worldwide in recent decades. Despite this fact, no antiviral treatment against honey bee viruses is currently available for practical applications and the level of viral infection can only be controlled indirectly by reducing the number of Varroa mites in honey bee colonies. In this study, we investigated the antiviral potential of the gypsy mushroom (Cortinarius caperatus) to reduce DWV infection in honey bees. Our results indicate that the alcohol extract of C. caperatus prevented the development of DWV infection in cage experiments as well as after direct application to honey bee colonies in a field experiment. The applied doses did not shorten the lifespan of honey bees. The reduced levels of DWV in C. caperatus-treated honey bees in cage experiments were accompanied by significant changes in the gene expression of Tep7, Bap1, and Vago. The C. caperatus treatment was not effective against the trypanosomatid Lotmaria passim. No residues of C.caperatus were found in honey harvested in the spring from colonies supplemented with the mushroom extract for their winter feeding. These findings suggest that C. caperatus alcohol extract could be a potential natural remedy to treat DWV infection in honey bees.
Collapse
Affiliation(s)
- Karolína Svobodová
- University of South Bohemia, Faculty of Science, Ceske Budejovice, Czech Republic.
| | - Václav Krištůfek
- Czech Academy of Sciences, Biology Centre, Institute of Soil Biology, Ceske Budejovice, Czech Republic
| | - Jiří Kubásek
- University of South Bohemia, Faculty of Science, Ceske Budejovice, Czech Republic
| | - Alena Krejčí
- University of South Bohemia, Faculty of Science, Ceske Budejovice, Czech Republic; Czech Academy of Sciences, Biology Centre, Institute of Entomology, Ceske Budejovice, Czech Republic.
| |
Collapse
|
4
|
Prince BC, Walsh E, Torres TZB, Rückert C. Recognition of Arboviruses by the Mosquito Immune System. Biomolecules 2023; 13:1159. [PMID: 37509194 PMCID: PMC10376960 DOI: 10.3390/biom13071159] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Arthropod-borne viruses (arboviruses) pose a significant threat to both human and animal health worldwide. These viruses are transmitted through the bites of mosquitoes, ticks, sandflies, or biting midges to humans or animals. In humans, arbovirus infection often results in mild flu-like symptoms, but severe disease and death also occur. There are few vaccines available, so control efforts focus on the mosquito population and virus transmission control. One area of research that may enable the development of new strategies to control arbovirus transmission is the field of vector immunology. Arthropod vectors, such as mosquitoes, have coevolved with arboviruses, resulting in a balance of virus replication and vector immune responses. If this balance were disrupted, virus transmission would likely be reduced, either through reduced replication, or even through enhanced replication, resulting in mosquito mortality. The first step in mounting any immune response is to recognize the presence of an invading pathogen. Recent research advances have been made to tease apart the mechanisms of arbovirus detection by mosquitoes. Here, we summarize what is known about arbovirus recognition by the mosquito immune system, try to generate a comprehensive picture, and highlight where there are still gaps in our current understanding.
Collapse
Affiliation(s)
- Brian C Prince
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Elizabeth Walsh
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Tran Zen B Torres
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Claudia Rückert
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
5
|
Sri-In C, Thontiravong A, Bartholomay LC, Wechtaisong W, Thongmeesee K, Riana E, Tiawsirisup S. 34-kDa salivary protein enhances duck Tembusu virus infectivity in the salivary glands of Aedes albopictus by modulating the innate immune response. Sci Rep 2023; 13:9098. [PMID: 37277542 DOI: 10.1038/s41598-023-35914-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 05/25/2023] [Indexed: 06/07/2023] Open
Abstract
Duck Tembusu virus (DTMUV) is an important flavivirus that can be transmitted to poultry via Aedes albopictus bites. Furthermore, humans residing in the DTMUV epidemic area display activated antiviral immune responses to local DTMUV isolates during the pathogenic invasion, thereby raising the primary concern that this flavivirus may be transmitted to humans via mosquito bites. Therefore, we identified the gene AALF004421, which is a homolog of the 34-kDa salivary protein (34 kDa) of Ae. albopictus and studied the salivary protein-mediated enhancement of DTMUV infection in Ae. albopictus salivary glands. We observed that double-stranded RNA-mediated silencing of the 34 kDa in mosquito salivary glands demonstrated that the silenced 34 kDa impaired DTMUV infectivity, similar to inhibition through serine protease. This impairment occurred as a consequence of triggering the innate immune response function of a macroglobulin complement-related factor (MCR). 34-kDa in the salivary gland which had similar activity as a serine protease, results in the abrogation of antimicrobial peptides production and strong enhance DTMUV replication and transmission. Although the function of the 34 kDa in Ae. albopictus is currently unknown; in the present study, we showed that it may have a major role in DTMUV infection in mosquito salivary glands through the suppression of the antiviral immune response in the earliest stages of infection. This finding provides the first identification of a prominently expressed 34 kDa protein in Ae. albopictus saliva that could serve as a target for controlling DTMUV replication in mosquito vectors.
Collapse
Affiliation(s)
- Chalida Sri-In
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Aunyaratana Thontiravong
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Lyric C Bartholomay
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Wisconsin, USA
| | - Wittawat Wechtaisong
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kritsada Thongmeesee
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Elizabeth Riana
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sonthaya Tiawsirisup
- Animal Vector-Borne Disease Research Unit, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
6
|
Cottis S, Blisnick AA, Failloux AB, Vernick KD. Determinants of Chikungunya and O'nyong-Nyong Virus Specificity for Infection of Aedes and Anopheles Mosquito Vectors. Viruses 2023; 15:589. [PMID: 36992298 PMCID: PMC10051923 DOI: 10.3390/v15030589] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/02/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
Mosquito-borne diseases caused by viruses and parasites are responsible for more than 700 million infections each year. Anopheles and Aedes are the two major vectors for, respectively, malaria and arboviruses. Anopheles mosquitoes are the primary vector of just one known arbovirus, the alphavirus o'nyong-nyong virus (ONNV), which is closely related to the chikungunya virus (CHIKV), vectored by Aedes mosquitoes. However, Anopheles harbor a complex natural virome of RNA viruses, and a number of pathogenic arboviruses have been isolated from Anopheles mosquitoes in nature. CHIKV and ONNV are in the same antigenic group, the Semliki Forest virus complex, are difficult to distinguish via immunodiagnostic assay, and symptomatically cause essentially the same human disease. The major difference between the arboviruses appears to be their differential use of mosquito vectors. The mechanisms governing this vector specificity are poorly understood. Here, we summarize intrinsic and extrinsic factors that could be associated with vector specificity by these viruses. We highlight the complexity and multifactorial aspect of vectorial specificity of the two alphaviruses, and evaluate the level of risk of vector shift by ONNV or CHIKV.
Collapse
Affiliation(s)
- Solène Cottis
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris Cité, CNRS UMR2000, F-75015 Paris, France
- Graduate School of Life Sciences ED515, Sorbonne Université UPMC Paris VI, 75252 Paris, France
| | - Adrien A. Blisnick
- Arboviruses and Insect Vectors Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, F-75015 Paris, France
| | - Anna-Bella Failloux
- Arboviruses and Insect Vectors Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, F-75015 Paris, France
| | - Kenneth D. Vernick
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris Cité, CNRS UMR2000, F-75015 Paris, France
- Graduate School of Life Sciences ED515, Sorbonne Université UPMC Paris VI, 75252 Paris, France
| |
Collapse
|
7
|
Zhao BR, Wang XX, Liu PP, Wang XW. Complement-related proteins in crustacean immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 139:104577. [PMID: 36265592 DOI: 10.1016/j.dci.2022.104577] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/02/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
As an important part of innate immune system, complement system is widely involved in defense response and immune regulation, and plays an important biological role. The complement system has been deeply studied. More than 30 complement-related molecules and three major complement-activation pathways have been identified in vertebrates. Crustacean animals do not have complement system. There are only some complement-related proteins in crustaceans which are important for host defense. In this review, we summarize the current knowledge about complement-related proteins in crustaceans, and their functions in crustacean immunity. We also make a comparation of the crustacean pro-phenoloxidase activating system and the mammalian complement system. This review provides a better understanding of the evolution and function of complement-related proteins in crustaceans.
Collapse
Affiliation(s)
- Bao-Rui Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, And State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, 266237, China
| | - Xin-Xin Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, And State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, 266237, China
| | - Ping-Ping Liu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, And State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, 266237, China
| | - Xian-Wei Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, And State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, 266237, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, 266237, China.
| |
Collapse
|
8
|
Cardoso-Jaime V, Tikhe CV, Dong S, Dimopoulos G. The Role of Mosquito Hemocytes in Viral Infections. Viruses 2022; 14:v14102088. [PMID: 36298644 PMCID: PMC9608948 DOI: 10.3390/v14102088] [Citation(s) in RCA: 8] [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: 08/16/2022] [Revised: 09/03/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Insect hemocytes are the only immune cells that can mount a humoral and cellular immune response. Despite the critical involvement of hemocytes in immune responses against bacteria, fungi, and parasites in mosquitoes, our understanding of their antiviral potential is still limited. It has been shown that hemocytes express humoral factors such as TEP1, PPO, and certain antimicrobial peptides that are known to restrict viral infections. Insect hemocytes also harbor the major immune pathways, such as JAK/STAT, TOLL, IMD, and RNAi, which are critical for the control of viral infection. Recent research has indicated a role for hemocytes in the regulation of viral infection through RNA interference and autophagy; however, the specific mechanism by which this regulation occurs remains uncharacterized. Conversely, some studies have suggested that hemocytes act as agonists of arboviral infection because they lack basal lamina and circulate throughout the whole mosquito, likely facilitating viral dissemination to other tissues such as salivary glands. In addition, hemocytes produce arbovirus agonist factors such as lectins, which enhance viral infection. Here, we summarize our current understanding of hemocytes’ involvement in viral infections.
Collapse
|
9
|
Marquez J, Dinguirard N, Gonzalez A, Kane A, Joffe N, Yoshino T, Castillo M. Molecular characterization of thioester-containing proteins in Biomphalaria glabrata and their differential gene expression upon Schistosoma mansoni exposure. Front Immunol 2022; 13:903158. [PMID: 35967434 PMCID: PMC9363628 DOI: 10.3389/fimmu.2022.903158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/27/2022] [Indexed: 11/30/2022] Open
Abstract
Schistosomiasis is a disease caused by trematode parasites of the genus Schistosoma that affects approximately 200 million people worldwide. Schistosomiasis has been a persistent problem in endemic areas as there is no vaccine available, currently used anti-helmintic medications do not prevent reinfection, and most concerning, drug resistance has been documented in laboratory and field isolates. Thus, alternative approaches to curtail this human disease are warranted. Understanding the immunobiology of the obligate intermediate host of these parasites, which include the freshwater snail Biomphalaria glabrata, may facilitate the development of novel methods to stop or reduce transmission to humans. Molecules from the thioester-containing protein (TEP) superfamily have been shown to be involved in immunological functions in many animals including corals and humans. In this study we identified, characterized, and compared TEP transcripts and their expression upon S. mansoni exposure in resistant and susceptible strains of B. glabrata snails. Results showed the expression of 11 unique TEPs in B. glabrata snails. These transcripts present high sequence identity at the nucleotide and putative amino acid levels between susceptible and resistant strains. Further analysis revealed differences in several TEPs’ constitutive expression levels between resistant and susceptible snail strains, with C3-1, C3-3, and CD109 having higher constitutive expression levels in the resistant (BS90) strain, whereas C3-2 and TEP-1 showed higher constitutive expression levels in the susceptible (NMRI) strain. Furthermore, TEP-specific response to S. mansoni miracidia exposure reiterated their differential expression, with resistant snails upregulating the expression of both TEP-4 and TEP-3 at 2 h and 48 h post-exposure, respectively. Further understanding the diverse TEP genes and their functions in invertebrate animal vectors will not only expand our knowledge in regard to this ancient family of immune proteins, but also offer the opportunity to identify novel molecular targets that could aid in the efforts to develop control methods to reduce schistosomiasis transmission.
Collapse
Affiliation(s)
- J. Marquez
- Department of Biology, New Mexico State University, Las Cruces, NM, United States
| | - N. Dinguirard
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - A. Gonzalez
- Department of Biology, New Mexico State University, Las Cruces, NM, United States
| | - A.E. Kane
- Department of Biology, New Mexico State University, Las Cruces, NM, United States
| | - N.R. Joffe
- Department of Biology, New Mexico State University, Las Cruces, NM, United States
| | - T.P. Yoshino
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - M.G. Castillo
- Department of Biology, New Mexico State University, Las Cruces, NM, United States
- *Correspondence: M.G. Castillo,
| |
Collapse
|
10
|
Viglietta M, Bellone R, Blisnick AA, Failloux AB. Vector Specificity of Arbovirus Transmission. Front Microbiol 2021; 12:773211. [PMID: 34956136 PMCID: PMC8696169 DOI: 10.3389/fmicb.2021.773211] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022] Open
Abstract
More than 25% of human infectious diseases are vector-borne diseases (VBDs). These diseases, caused by pathogens shared between animals and humans, are a growing threat to global health with more than 2.5 million annual deaths. Mosquitoes and ticks are the main vectors of arboviruses including flaviviruses, which greatly affect humans. However, all tick or mosquito species are not able to transmit all viruses, suggesting important molecular mechanisms regulating viral infection, dissemination, and transmission by vectors. Despite the large distribution of arthropods (mosquitoes and ticks) and arboviruses, only a few pairings of arthropods (family, genus, and population) and viruses (family, genus, and genotype) successfully transmit. Here, we review the factors that might limit pathogen transmission: internal (vector genetics, immune responses, microbiome including insect-specific viruses, and coinfections) and external, either biotic (adult and larvae nutrition) or abiotic (temperature, chemicals, and altitude). This review will demonstrate the dynamic nature and complexity of virus–vector interactions to help in designing appropriate practices in surveillance and prevention to reduce VBD threats.
Collapse
Affiliation(s)
- Marine Viglietta
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| | - Rachel Bellone
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| | - Adrien Albert Blisnick
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| | - Anna-Bella Failloux
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| |
Collapse
|
11
|
Silva JMF, Nagata T, Melo FL, Elena SF. Heterogeneity in the Response of Different Subtypes of Drosophila melanogaster Midgut Cells to Viral Infections. Viruses 2021; 13:2284. [PMID: 34835089 PMCID: PMC8623525 DOI: 10.3390/v13112284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/05/2021] [Accepted: 11/13/2021] [Indexed: 11/29/2022] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) offers the possibility to monitor both host and pathogens transcriptomes at the cellular level. Here, public scRNA-seq datasets from Drosophila melanogaster midgut cells were used to compare the differences in replication strategy and cellular responses between two fly picorna-like viruses, Thika virus (TV) and D. melanogaster Nora virus (DMelNV). TV exhibited lower levels of viral RNA accumulation but infected a higher number of cells compared to DMelNV. In both cases, viral RNA accumulation varied according to cell subtype. The cellular heat shock response to TV and DMelNV infection was cell-subtype- and virus-specific. Disruption of bottleneck genes at later stages of infection in the systemic response, as well as of translation-related genes in the cellular response to DMelNV in two cell subtypes, may affect the virus replication.
Collapse
Affiliation(s)
- João M. F. Silva
- Departamento de Biologia Celular, Universidade de Brasília, Brasília 70910-900, Brazil; (J.M.F.S.); (T.N.); (F.L.M.)
- Instituto de Biología Integrativa de Sistemas (I2SysBio), CSIC-Universitat de València, 46980 Paterna, València, Spain
| | - Tatsuya Nagata
- Departamento de Biologia Celular, Universidade de Brasília, Brasília 70910-900, Brazil; (J.M.F.S.); (T.N.); (F.L.M.)
| | - Fernando L. Melo
- Departamento de Biologia Celular, Universidade de Brasília, Brasília 70910-900, Brazil; (J.M.F.S.); (T.N.); (F.L.M.)
| | - Santiago F. Elena
- Instituto de Biología Integrativa de Sistemas (I2SysBio), CSIC-Universitat de València, 46980 Paterna, València, Spain
- The Santa Fe Institute, Santa Fe, NM 87501, USA
| |
Collapse
|
12
|
McMenamin AJ, Brutscher LM, Daughenbaugh KF, Flenniken ML. The Honey Bee Gene Bee Antiviral Protein-1 Is a Taxonomically Restricted Antiviral Immune Gene. FRONTIERS IN INSECT SCIENCE 2021; 1:749781. [PMID: 38468887 PMCID: PMC10926557 DOI: 10.3389/finsc.2021.749781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/20/2021] [Indexed: 03/13/2024]
Abstract
Insects have evolved a wide range of strategies to combat invading pathogens, including viruses. Genes that encode proteins involved in immune responses often evolve under positive selection due to their co-evolution with pathogens. Insect antiviral defense includes the RNA interference (RNAi) mechanism, which is triggered by recognition of non-self, virally produced, double-stranded RNAs. Indeed, insect RNAi genes (e.g., dicer and argonaute-2) are under high selective pressure. Honey bees (Apis mellifera) are eusocial insects that respond to viral infections via both sequence specific RNAi and a non-sequence specific dsRNA triggered pathway, which is less well-characterized. A transcriptome-level study of virus-infected and/or dsRNA-treated honey bees revealed increased expression of a novel antiviral gene, GenBank: MF116383, and in vivo experiments confirmed its antiviral function. Due to in silico annotation and sequence similarity, MF116383 was originally annotated as a probable cyclin-dependent serine/threonine-protein kinase. In this study, we confirmed that MF116383 limits virus infection, and carried out further bioinformatic and phylogenetic analyses to better characterize this important gene-which we renamed bee antiviral protein-1 (bap1). Phylogenetic analysis revealed that bap1 is taxonomically restricted to Hymenoptera and Blatella germanica (the German cockroach) and that the majority of bap1 amino acids are evolving under neutral selection. This is in-line with the results from structural prediction tools that indicate Bap1 is a highly disordered protein, which likely has relaxed structural constraints. Assessment of honey bee gene expression using a weighted gene correlation network analysis revealed that bap1 expression was highly correlated with several immune genes-most notably argonaute-2. The coexpression of bap1 and argonaute-2 was confirmed in an independent dataset that accounted for the effect of virus abundance. Together, these data demonstrate that bap1 is a taxonomically restricted, rapidly evolving antiviral immune gene. Future work will determine the role of bap1 in limiting replication of other viruses and examine the signal cascade responsible for regulating the expression of bap1 and other honey bee antiviral defense genes, including coexpressed ago-2, and determine whether the virus limiting function of bap1 acts in parallel or in tandem with RNAi.
Collapse
Affiliation(s)
- Alexander J. McMenamin
- Department of Plant Sciences and Plant Pathology, Montana State University, Bozeman, MT, United States
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
- Pollinator Health Center, Montana State University, Bozeman, MT, United States
| | - Laura M. Brutscher
- Pollinator Health Center, Montana State University, Bozeman, MT, United States
| | - Katie F. Daughenbaugh
- Department of Plant Sciences and Plant Pathology, Montana State University, Bozeman, MT, United States
- Pollinator Health Center, Montana State University, Bozeman, MT, United States
| | - Michelle L. Flenniken
- Department of Plant Sciences and Plant Pathology, Montana State University, Bozeman, MT, United States
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
- Pollinator Health Center, Montana State University, Bozeman, MT, United States
| |
Collapse
|
13
|
McFarlane M, Laureti M, Levée T, Terry S, Kohl A, Pondeville E. Improved transient silencing of gene expression in the mosquito female Aedes aegypti. INSECT MOLECULAR BIOLOGY 2021; 30:355-365. [PMID: 33715239 DOI: 10.1111/imb.12700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/30/2020] [Accepted: 03/26/2021] [Indexed: 06/12/2023]
Abstract
Gene silencing using RNA interference (RNAi) has become a widely used genetic technique to study gene function in many organisms. In insects, this technique is often applied through the delivery of dsRNA. In the adult female Aedes aegypti, a main vector of human-infecting arboviruses, efficiency of gene silencing following dsRNA injection varies greatly according to targeted genes. Difficult knockdowns using dsRNA can thus hamper gene function analysis. Here, by analysing silencing of three different genes in female Ae. aegypti (p400, ago2 and E75), we show that gene silencing can indeed be dsRNA sequence dependent but different efficiencies do not correlate with dsRNA length. Our findings suggest that silencing is likely also gene dependent, probably due to gene-specific tissue expression and/or feedback mechanisms. We demonstrate that use of high doses of dsRNA can improve knockdown efficiency, and injection of a transfection reagent along with dsRNA reduces the variability in efficiency between replicates. Finally, we show that gene silencing cannot be achieved using siRNA injection in Ae. aegypti adult females. Overall, this work should help future gene function analyses using RNAi in adult females Ae. aegypti, leading toward a better understanding of physiological and infectious processes.
Collapse
Affiliation(s)
- M McFarlane
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - M Laureti
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - T Levée
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - S Terry
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - A Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - E Pondeville
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| |
Collapse
|
14
|
Weng SC, Li HH, Li JC, Liu WL, Chen CH, Shiao SH. A Thioester-Containing Protein Controls Dengue Virus Infection in Aedes aegypti Through Modulating Immune Response. Front Immunol 2021; 12:670122. [PMID: 34054842 PMCID: PMC8155531 DOI: 10.3389/fimmu.2021.670122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/28/2021] [Indexed: 01/13/2023] Open
Abstract
Complement-like proteins in arthropods defend against invading pathogens in the early phases of infection. Thioester-containing proteins (TEPs), which exhibit high similarity to mammalian complement C3, are thought to play a key role in the innate immunity of arthropods. We identified and characterized anti-dengue virus (DENV) host factors, in particular complement-like proteins, in the mosquito Aedes aegypti. Our results indicate that TEP1 limits DENV infection in Ae. aegypti. We showed that TEP1 transcription is highly induced in mosquitoes following DENV infection. Silencing TEP1 resulted in the up-regulation of viral RNA and proteins. In addition, the production of infectious virus particles increased in the absence of TEP1. We generated a transgenic mosquito line with a TEP1 loss-of-function phenotype under a blood meal-inducible promoter. We showed that viral protein and titers increased in transgenic mosquitoes after an infectious blood meal. Interestingly, expression of transcription factor Rel2 and certain anti-microbial peptides (AMPs) were inhibited in transgenic mosquitoes. Overall, our results suggest that TEP1 regulates the immune response and consequently controls the replication of dengue virus in mosquitoes. This finding provides new insight into the molecular mechanisms of mosquito host factors in the regulation of DENV replication.
Collapse
Affiliation(s)
- Shih-Che Weng
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsing-Han Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Jian-Chiuan Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Wei-Liang Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan.,National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Shin-Hong Shiao
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
15
|
Chowdhury A, Modahl CM, Tan ST, Wong Wei Xiang B, Missé D, Vial T, Kini RM, Pompon JF. JNK pathway restricts DENV2, ZIKV and CHIKV infection by activating complement and apoptosis in mosquito salivary glands. PLoS Pathog 2020; 16:e1008754. [PMID: 32776975 PMCID: PMC7444518 DOI: 10.1371/journal.ppat.1008754] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 08/20/2020] [Accepted: 06/26/2020] [Indexed: 11/18/2022] Open
Abstract
Arbovirus infection of Aedes aegypti salivary glands (SGs) determines transmission. However, there is a dearth of knowledge on SG immunity. Here, we characterized SG immune response to dengue, Zika and chikungunya viruses using high-throughput transcriptomics. We also describe a transcriptomic response associated to apoptosis, blood-feeding and lipid metabolism. The three viruses differentially regulate components of Toll, Immune deficiency (IMD) and c-Jun N- terminal Kinase (JNK) pathways. However, silencing of the Toll and IMD pathway components showed variable effects on SG infection by each virus. In contrast, regulation of the JNK pathway produced consistent responses in both SGs and midgut. Infection by the three viruses increased with depletion of the activator Kayak and decreased with depletion of the negative regulator Puckered. Virus-induced JNK pathway regulates the complement factor, Thioester containing protein-20 (TEP20), and the apoptosis activator, Dronc, in SGs. Individual and co-silencing of these genes demonstrate their antiviral effects and that both may function together. Co-silencing either TEP20 or Dronc with Puckered annihilates JNK pathway antiviral effect. Upon infection in SGs, TEP20 induces antimicrobial peptides (AMPs), while Dronc is required for apoptosis independently of TEP20. In conclusion, we revealed the broad antiviral function of JNK pathway in SGs and showed that it is mediated by a TEP20 complement and Dronc-induced apoptosis response. These results expand our understanding of the immune arsenal that blocks arbovirus transmission. Arboviral diseases caused by dengue (DENV), Zika (ZIKV) and chikungunya (CHIKV) viruses are responsible for large number of death and debilitation around the world. These viruses are transmitted to humans by the mosquito vector, Aedes aegypti. During the bites, infected salivary glands (SGs) release saliva containing viruses, which initiate human infection. As the tissue where transmitted viruses are produced, SG infection is a key determinant of transmission. To bridge the knowledge gap in vector-virus molecular interactions in SGs, we describe the transcriptome after DENV, ZIKV and CHIKV infection using RNA-sequencing and characterized the immune response in this tissue. Our study reveals the broad antiviral function of c-Jun N-terminal kinase (JNK) pathway against DENV, ZIKV and CHIKV in SGs. We further show that it is mediated by the complement system and apoptosis, identifying the mechanism. Our study adds the JNK pathway to the immune arsenal that can be harnessed to engineer refractory vectors.
Collapse
Affiliation(s)
- Avisha Chowdhury
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Cassandra M. Modahl
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Siok Thing Tan
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | - Dorothée Missé
- MIVEGEC, IRD, CNRS, Univ. Montpellier, Montpellier, France
| | - Thomas Vial
- Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - R. Manjunatha Kini
- Department of Biological Sciences, National University of Singapore, Singapore
- * E-mail: (RMK); (JFP)
| | - Julien Francis Pompon
- Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- MIVEGEC, IRD, CNRS, Univ. Montpellier, Montpellier, France
- * E-mail: (RMK); (JFP)
| |
Collapse
|
16
|
A New Assessment of Thioester-Containing Proteins Diversity of the Freshwater Snail Biomphalaria glabrata. Genes (Basel) 2020; 11:genes11010069. [PMID: 31936127 PMCID: PMC7016707 DOI: 10.3390/genes11010069] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 12/16/2022] Open
Abstract
Thioester-containing proteins (TEPs) superfamily is known to play important innate immune functions in a wide range of animal phyla. TEPs are involved in recognition, and in the direct or mediated killing of several invading organisms or pathogens. While several TEPs have been identified in many invertebrates, only one TEP (named BgTEP) has been previously characterized in the freshwater snail, Biomphalaria glabrata. As the presence of a single member of that family is particularly intriguing, transcriptomic data and the recently published genome were used to explore the presence of other BgTEP related genes in B. glabrata. Ten other TEP members have been reported and classified into different subfamilies: Three complement-like factors (BgC3-1 to BgC3-3), one α-2-macroblobulin (BgA2M), two macroglobulin complement-related proteins (BgMCR1, BgMCR2), one CD109 (BgCD109), and three insect TEP (BgTEP2 to BgTEP4) in addition to the previously characterized BgTEP that we renamed BgTEP1. This is the first report on such a level of TEP diversity and of the presence of macroglobulin complement-related proteins (MCR) in mollusks. Gene structure analysis revealed alternative splicing in the highly variable region of three members (BgA2M, BgCD109, and BgTEP2) with a particularly unexpected diversity for BgTEP2. Finally, different gene expression profiles tend to indicate specific functions for such novel family members.
Collapse
|
17
|
The Mosquito Immune System and the Life of Dengue Virus: What We Know and Do Not Know. Pathogens 2019; 8:pathogens8020077. [PMID: 31200426 PMCID: PMC6631187 DOI: 10.3390/pathogens8020077] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 01/10/2023] Open
Abstract
Flaviviruses are largely transmitted to humans by their arthropod vectors such as mosquitoes or ticks. The dengue virus (DENV) is one of the members of the family Flaviviridae and is the causative agent of dengue fever. In the mosquito vector, DENV enters through viremic blood meal and replicates in the mid-gut. Newly formed virion particles circulate to various mosquito organs and get transmitted to the next host in subsequent bites. Aedes aegypti and Aedes albopictus have intricate immune control to allow DENV production at a sub-pathogenic level. In the mosquito, antimicrobial peptides (AMP) and RNA inference (RNAi) are the two main antiviral strategies used against DENV. Apart from innate immunity, mosquito resident microbes play a significant role in modulating DENV replication. In this review, we discuss different immune mechanisms and preventive strategies that act against DENV in two of its vectors: Aedes aegypti and Aedes albopictus.
Collapse
|
18
|
Homologs of Human Dengue-Resistance Genes, FKBP1B and ATCAY, Confer Antiviral Resistance in Aedes aegypti Mosquitoes. INSECTS 2019; 10:insects10020046. [PMID: 30717390 PMCID: PMC6409984 DOI: 10.3390/insects10020046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 01/26/2019] [Accepted: 01/29/2019] [Indexed: 02/06/2023]
Abstract
Dengue virus (DENV) is transmitted by mosquitoes and is a major public health concern. The study of innate mosquito defense mechanisms against DENV have revealed crucial roles for the Toll, Imd, JAK-STAT, and RNAi pathways in mediating DENV in the mosquito. Often overlooked in such studies is the role of intrinsic cellular defense mechanisms that we hypothesize to work in concert with the classical immune pathways to affect organismal defense. Our understanding of the molecular interaction of DENV with mosquito host cells is limited, and we propose to expand upon the recent results from a genome-scale, small interfering RNA (siRNA)-based study that identified mammalian host proteins associated with resistance to dengue/West Nile virus (DENV/WNV) infection. The study identified 22 human DENV/WNV resistance genes (DVR), and we hypothesized that a subset would be functionally conserved in Aedes aegypti mosquitoes, imparting cellular defense against flaviviruses in this species. We identified 12 homologs of 22 human DVR genes in the Ae. aegypti genome. To evaluate their possible role in cellular resistance/antiviral defense against DENV, we used siRNA silencing targeted against each of the 12 homologs in an Ae. aegypti cell line (Aag2) infected with DENV2 and identified that silencing of the two candidates, AeFKBP1 and AeATCAY, homologs of human FKBP1B and ATCAY, were associated with a viral increase. We then used dsRNA to silence each of the two genes in adult mosquitoes to validate the observed antiviral functions in vivo. Depletion of AeFKBP1 or AeATCAY increased viral dissemination through the mosquito at 14 days post-infection. Our results demonstrated that AeFKBP1 and AeATCAY mediate resistance to DENV akin to what has been described for their homologs in humans. AeFKBP1 and AeATCAY provide a rare opportunity to elucidate a DENV-resistance mechanism that may be evolutionarily conserved between humans and Ae. aegypti.
Collapse
|
19
|
Kumar A, Srivastava P, Sirisena P, Dubey SK, Kumar R, Shrinet J, Sunil S. Mosquito Innate Immunity. INSECTS 2018; 9:insects9030095. [PMID: 30096752 PMCID: PMC6165528 DOI: 10.3390/insects9030095] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 06/17/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022]
Abstract
Mosquitoes live under the endless threat of infections from different kinds of pathogens such as bacteria, parasites, and viruses. The mosquito defends itself by employing both physical and physiological barriers that resist the entry of the pathogen and the subsequent establishment of the pathogen within the mosquito. However, if the pathogen does gain entry into the insect, the insect mounts a vigorous innate cellular and humoral immune response against the pathogen, thereby limiting the pathogen's propagation to nonpathogenic levels. This happens through three major mechanisms: phagocytosis, melanization, and lysis. During these processes, various signaling pathways that engage intense mosquito⁻pathogen interactions are activated. A critical overview of the mosquito immune system and latest information about the interaction between mosquitoes and pathogens are provided in this review. The conserved, innate immune pathways and specific anti-pathogenic strategies in mosquito midgut, hemolymph, salivary gland, and neural tissues for the control of pathogen propagation are discussed in detail.
Collapse
Affiliation(s)
- Ankit Kumar
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Priyanshu Srivastava
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Pdnn Sirisena
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Sunil Kumar Dubey
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Ramesh Kumar
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Jatin Shrinet
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| | - Sujatha Sunil
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi-110067, India.
| |
Collapse
|
20
|
Mills MK, Michel K, Pfannenstiel RS, Ruder MG, Veronesi E, Nayduch D. Culicoides-virus interactions: infection barriers and possible factors underlying vector competence. CURRENT OPINION IN INSECT SCIENCE 2017; 22:7-15. [PMID: 28805641 DOI: 10.1016/j.cois.2017.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/01/2017] [Indexed: 06/07/2023]
Abstract
In the United States, Culicoides midges vector arboviruses of economic importance such as Bluetongue Virus and Epizootic Hemorrhagic Disease Virus. A limited number of studies have demonstrated the complexities of midge-virus interactions, including dynamic changes in virus titer and prevalence over the infection time course. These dynamics are, in part, dictated by mesenteron infection and escape barriers. This review summarizes the overarching trends in viral titer and prevalence throughout the course of infection. Essential barriers to infection and dissemination in the midge are highlighted, along with heritable and extrinsic factors that likely contribute to these barriers. Next generation molecular tools and techniques, now available for Culicoides midges, give researchers the opportunity to test how these factors contribute to vector competence.
Collapse
Affiliation(s)
- Mary K Mills
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Robert S Pfannenstiel
- United States Department of Agriculture, Agricultural Research Service, Arthropod Borne Animal Diseases Research Unit, Manhattan, KS 66502, USA
| | - Mark G Ruder
- Southeastern Cooperative Wildlife Disease Study, Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Eva Veronesi
- Vector-borne Viral Diseases Programme, The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF, United Kingdom
| | - Dana Nayduch
- United States Department of Agriculture, Agricultural Research Service, Arthropod Borne Animal Diseases Research Unit, Manhattan, KS 66502, USA.
| |
Collapse
|
21
|
Brutscher LM, Daughenbaugh KF, Flenniken ML. Virus and dsRNA-triggered transcriptional responses reveal key components of honey bee antiviral defense. Sci Rep 2017; 7:6448. [PMID: 28743868 PMCID: PMC5526946 DOI: 10.1038/s41598-017-06623-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/04/2017] [Indexed: 12/22/2022] Open
Abstract
Recent high annual losses of honey bee colonies are associated with many factors, including RNA virus infections. Honey bee antiviral responses include RNA interference and immune pathway activation, but their relative roles in antiviral defense are not well understood. To better characterize the mechanism(s) of honey bee antiviral defense, bees were infected with a model virus in the presence or absence of dsRNA, a virus associated molecular pattern. Regardless of sequence specificity, dsRNA reduced virus abundance. We utilized next generation sequencing to examine transcriptional responses triggered by virus and dsRNA at three time-points post-infection. Hundreds of genes exhibited differential expression in response to co-treatment of dsRNA and virus. Virus-infected bees had greater expression of genes involved in RNAi, Toll, Imd, and JAK-STAT pathways, but the majority of differentially expressed genes are not well characterized. To confirm the virus limiting role of two genes, including the well-characterized gene, dicer, and a probable uncharacterized cyclin dependent kinase in honey bees, we utilized RNAi to reduce their expression in vivo and determined that virus abundance increased, supporting their involvement in antiviral defense. Together, these results further our understanding of honey bee antiviral defense, particularly the role of a non-sequence specific dsRNA-mediated antiviral pathway.
Collapse
Affiliation(s)
- Laura M Brutscher
- Department of Plant Sciences and Plant Pathology, Montana State University, Bozeman, MT, USA.,Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA.,Pollinator Health Center, Montana State University, Bozeman, MT, USA
| | - Katie F Daughenbaugh
- Department of Plant Sciences and Plant Pathology, Montana State University, Bozeman, MT, USA.,Pollinator Health Center, Montana State University, Bozeman, MT, USA
| | - Michelle L Flenniken
- Department of Plant Sciences and Plant Pathology, Montana State University, Bozeman, MT, USA. .,Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA. .,Pollinator Health Center, Montana State University, Bozeman, MT, USA.
| |
Collapse
|
22
|
Shokal U, Eleftherianos I. Evolution and Function of Thioester-Containing Proteins and the Complement System in the Innate Immune Response. Front Immunol 2017; 8:759. [PMID: 28706521 PMCID: PMC5489563 DOI: 10.3389/fimmu.2017.00759] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/16/2017] [Indexed: 01/09/2023] Open
Abstract
The innate immune response is evolutionary conserved among organisms. The complement system forms an important and efficient immune defense mechanism. It consists of plasma proteins that participate in microbial detection, which ultimately results in the production of various molecules with antimicrobial activity. Thioester-containing proteins (TEPs) are a superfamily of secreted effector proteins. In vertebrates, certain TEPs act in the innate immune response by promoting recruitment of immune cells, phagocytosis, and direct lysis of microbial invaders. Insects are excellent models for dissecting the molecular basis of innate immune recognition and response to a wide range of microbial infections. Impressive progress in recent years has generated crucial information on the role of TEPs in the antibacterial and antiparasite response of the tractable model insect Drosophila melanogaster and the mosquito malaria vector Anopheles gambiae. This knowledge is critical for better understanding the evolution of TEPs and their involvement in the regulation of the host innate immune system.
Collapse
Affiliation(s)
- Upasana Shokal
- Department of Biological Sciences, The George Washington University, Washington, DC, United States
| | - Ioannis Eleftherianos
- Department of Biological Sciences, The George Washington University, Washington, DC, United States
| |
Collapse
|
23
|
Biological Control Strategies for Mosquito Vectors of Arboviruses. INSECTS 2017; 8:insects8010021. [PMID: 28208639 PMCID: PMC5371949 DOI: 10.3390/insects8010021] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/21/2017] [Indexed: 12/16/2022]
Abstract
Historically, biological control utilizes predatory species and pathogenic microorganisms to reduce the population of mosquitoes as disease vectors. This is particularly important for the control of mosquito-borne arboviruses, which normally do not have specific antiviral therapies available. Although development of resistance is likely, the advantages of biological control are that the resources used are typically biodegradable and ecologically friendly. Over the past decade, the advancement of molecular biology has enabled optimization by the manipulation of genetic materials associated with biological control agents. Two significant advancements are the discovery of cytoplasmic incompatibility induced by Wolbachia bacteria, which has enhanced replacement programs, and the introduction of dominant lethal genes into local mosquito populations through the release of genetically modified mosquitoes. As various arboviruses continue to be significant public health threats, biological control strategies have evolved to be more diverse and become critical tools to reduce the disease burden of arboviruses.
Collapse
|
24
|
Baxter RHG, Contet A, Krueger K. Arthropod Innate Immune Systems and Vector-Borne Diseases. Biochemistry 2017; 56:907-918. [PMID: 28072517 DOI: 10.1021/acs.biochem.6b00870] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Arthropods, especially ticks and mosquitoes, are the vectors for a number of parasitic and viral human diseases, including malaria, sleeping sickness, Dengue, and Zika, yet arthropods show tremendous individual variation in their capacity to transmit disease. A key factor in this capacity is the group of genetically encoded immune factors that counteract infection by the pathogen. Arthropod-specific pattern recognition receptors and protease cascades detect and respond to infection. Proteins such as antimicrobial peptides, thioester-containing proteins, and transglutaminases effect responses such as lysis, phagocytosis, melanization, and agglutination. Effector responses are initiated by damage signals such as reactive oxygen species signaling from epithelial cells and recognized by cell surface receptors on hemocytes. Antiviral immunity is primarily mediated by siRNA pathways but coupled with interferon-like signaling, antimicrobial peptides, and thioester-containing proteins. Molecular mechanisms of immunity are closely linked to related traits of longevity and fertility, and arthropods have the capacity for innate immunological memory. Advances in understanding vector immunity can be leveraged to develop novel control strategies for reducing the rate of transmission of both ancient and emerging threats to global health.
Collapse
Affiliation(s)
- Richard H G Baxter
- Department of Chemistry and Molecular Biophysics & Biochemistry, Yale University , New Haven, Connecticut 06511, United States
| | - Alicia Contet
- Department of Chemistry and Molecular Biophysics & Biochemistry, Yale University , New Haven, Connecticut 06511, United States
| | - Kathryn Krueger
- Department of Chemistry and Molecular Biophysics & Biochemistry, Yale University , New Haven, Connecticut 06511, United States
| |
Collapse
|
25
|
Krzywinska E, Dennison NJ, Lycett GJ, Krzywinski J. A maleness gene in the malaria mosquito Anopheles gambiae. Science 2016; 353:67-9. [PMID: 27365445 DOI: 10.1126/science.aaf5605] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 06/01/2016] [Indexed: 12/22/2022]
Abstract
The molecular pathways controlling gender are highly variable and have been identified in only a few nonmammalian model species. In many insects, maleness is conferred by a Y chromosome-linked M factor of unknown nature. We have isolated and characterized a gene, Yob, for the M factor in the malaria mosquito Anopheles gambiae Yob, activated at the beginning of zygotic transcription and expressed throughout a male's life, controls male-specific splicing of the doublesex gene. Silencing embryonic Yob expression is male-lethal, whereas ectopic embryonic delivery of Yob transcripts yields male-only broods. This female-killing property may be an invaluable tool for creation of conditional male-only transgenic Anopheles strains for malaria control programs.
Collapse
Affiliation(s)
| | - Nathan J Dennison
- Vector Biology Department, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK. Department of Life Sciences, Imperial College, South Kensington Campus, London SW7 2AZ, UK
| | - Gareth J Lycett
- Vector Biology Department, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Jaroslaw Krzywinski
- The Pirbright Institute, Ash Road, Pirbright, Surrey, GU24 0NF, UK. Vector Biology Department, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK.
| |
Collapse
|
26
|
Liu YX, Li FX, Liu ZZ, Jia ZR, Zhou YH, Zhang H, Yan H, Zhou XQ, Chen XG. Integrated analysis of miRNAs and transcriptomes in Aedes albopictus midgut reveals the differential expression profiles of immune-related genes during dengue virus serotype-2 infection. INSECT SCIENCE 2016; 23:377-385. [PMID: 27029517 DOI: 10.1111/1744-7917.12339] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 03/12/2016] [Accepted: 03/21/2016] [Indexed: 06/05/2023]
Abstract
Mosquito microRNAs (miRNAs) are involved in host-virus interaction, and have been reported to be altered by dengue virus (DENV) infection in Aedes albopictus (Diptera: Culicidae). However, little is known about the molecular mechanisms of Aedes albopictus midgut-the first organ to interact with DENV-involved in its resistance to DENV. Here we used high-throughput sequencing to characterize miRNA and messenger RNA (mRNA) expression patterns in Aedes albopictus midgut in response to dengue virus serotype 2. A total of three miRNAs and 777 mRNAs were identified to be differentially expressed upon DENV infection. For the mRNAs, we identified 198 immune-related genes and 31 of them were differentially expressed. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses also showed that the differentially expressed immune-related genes were involved in immune response. Then the differential expression patterns of six immune-related genes and three miRNAs were confirmed by real-time reverse transcription polymerase chain reaction. Furthermore, seven known miRNA-mRNA interaction pairs were identified by aligning our two datasets. These analyses of miRNA and mRNA transcriptomes provide valuable information for uncovering the DENV response genes and provide a basis for future study of the resistance mechanisms in Aedes albopictus midgut.
Collapse
Affiliation(s)
- Yan-Xia Liu
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
- Laboratory of Emerging Infectious Diseases and Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Fen-Xiang Li
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Zhuan-Zhuan Liu
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Zhi-Rong Jia
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Yan-He Zhou
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Hao Zhang
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | - Hui Yan
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| | | | - Xiao-Guang Chen
- Key Laboratory of Prevention and Control of Emerging Infectious Diseases of Guangdong Higher Education Institutes, Department of Pathogen Biology, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Troupin A, Londono-Renteria B, Conway MJ, Cloherty E, Jameson S, Higgs S, Vanlandingham DL, Fikrig E, Colpitts TM. A novel mosquito ubiquitin targets viral envelope protein for degradation and reduces virion production during dengue virus infection. Biochim Biophys Acta Gen Subj 2016; 1860:1898-909. [PMID: 27241849 PMCID: PMC4949077 DOI: 10.1016/j.bbagen.2016.05.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 04/27/2016] [Accepted: 05/26/2016] [Indexed: 11/17/2022]
Abstract
Background Dengue virus (DENV) is a mosquito-borne flavivirus that causes significant human disease and mortality in the tropics and subtropics. By examining the effects of virus infection on gene expression, and interactions between virus and vector, new targets for prevention of infection and novel treatments may be identified in mosquitoes. We previously performed a microarray analysis of the Aedes aegypti transcriptome during infection with DENV and found that mosquito ubiquitin protein Ub3881 (AAEL003881) was specifically and highly down-regulated. Ubiquitin proteins have multiple functions in insects, including marking proteins for proteasomal degradation, regulating apoptosis and mediating innate immune signaling. Methods We used qRT-PCR to quantify gene expression and infection, and RNAi to reduce Ub3881 expression. Mosquitoes were infected with DENV through blood feeding. We transfected DENV protein expression constructs to examine the effect of Ub3881 on protein degradation. We used site-directed mutagenesis and transfection to determine what amino acids are involved in Ub3881-mediated protein degradation. Immunofluorescence, Co-immunoprecipitation and Western blotting were used to examine protein interactions and co-localization. Results The overexpression of Ub3881, but not related ubiquitin proteins, decreased DENV infection in mosquito cells and live Ae. aegypti. The Ub3881 protein was demonstrated to be involved in DENV envelope protein degradation and reduce the number of infectious virions released. Conclusions We conclude that Ub3881 has several antiviral functions in the mosquito, including specific viral protein degradation. General significance Our data highlights Ub3881 as a target for future DENV prevention strategies in the mosquito transmission vector. A novel mosquito ubiquitin, Ub3881, is identified in Aedes aegypti. Ub3881 is shown to have antiviral functions during dengue virus infection of the mosquito. Ub3881 targets a dengue viral protein for degradation during infection. Future dengue virus prevention strategies could incorporate Ub3881 as a target to prevent mosquito infection.
Collapse
Affiliation(s)
- Andrea Troupin
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States
| | - Berlin Londono-Renteria
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States
| | - Michael J Conway
- Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, MI 48859, United States
| | - Erin Cloherty
- Department of Tropical Medicine, Tulane University School of Public Health, New Orleans, LA 70112, United States
| | - Samuel Jameson
- Department of Tropical Medicine, Tulane University School of Public Health, New Orleans, LA 70112, United States
| | - Stephen Higgs
- Biosecurity Research Institute, Kansas State University, Manhattan, KS 66506, United States; Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Dana L Vanlandingham
- Biosecurity Research Institute, Kansas State University, Manhattan, KS 66506, United States; Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, United States; Howard Hughes Medical Institute, Chevy Chase, MD 20815, United States
| | - Tonya M Colpitts
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States.
| |
Collapse
|
28
|
Londono-Renteria B, Grippin C, Cardenas JC, Troupin A, Colpitts TM. Human C5a Protein Participates in the Mosquito Immune Response Against Dengue Virus. JOURNAL OF MEDICAL ENTOMOLOGY 2016; 53:505-512. [PMID: 26843451 PMCID: PMC4892811 DOI: 10.1093/jme/tjw003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/11/2016] [Indexed: 06/05/2023]
Abstract
Dengue virus (DENV) is transmitted by Aedes spp mosquitoes during a bloodmeal uptake. The bloodmeal consists of host cells, immune factors, and possibly blood-borne pathogens, such as arboviruses. Human cells and immune-related factors, like the complement system, can remain active in the bloodmeal and may be able to interact with pathogens in the mosquito. Previous studies have shown that active complement proteins impact Plasmodium parasite viability in the Anopheles midgut. Thus, we investigated the effects of the human complement on DENV infection in the midgut of Aedes aegypti. Our findings indicate that mosquitoes receiving DENV mixed with normal non-inactivated human serum showed significantly lower viremia than those fed with heat-inactivated serum. This implies that human complement may act to limit DENV infection in the mosquito midgut. In addition, we found that human complement C5a protein was able to directly communicate with mosquito cells, affecting the cell antiviral response against DENV. Our results also show that human C5a protein is able to interact with several membrane-bound mosquito proteins. Together these results suggest an important role of human complement protein in DENV transmission.
Collapse
Affiliation(s)
- Berlin Londono-Renteria
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29209 (; ),
| | - Crystal Grippin
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, 70130 , and
| | - Jenny C Cardenas
- Microbiology and Clinical Laboratory, Hospital San Juan de Dios, Los Patios - Norte de Santander, Colombia
| | - Andrea Troupin
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29209 (; )
| | - Tonya M Colpitts
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, 29209 (; )
| |
Collapse
|
29
|
Cheng G, Liu Y, Wang P, Xiao X. Mosquito Defense Strategies against Viral Infection. Trends Parasitol 2016; 32:177-186. [PMID: 26626596 PMCID: PMC4767563 DOI: 10.1016/j.pt.2015.09.009] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/25/2015] [Accepted: 09/28/2015] [Indexed: 01/23/2023]
Abstract
Mosquito-borne viral diseases are a major concern of global health and result in significant economic losses in many countries. As natural vectors, mosquitoes are very permissive to and allow systemic and persistent arbovirus infection. Intriguingly, persistent viral propagation in mosquito tissues neither results in dramatic pathological sequelae nor impairs the vectorial behavior or lifespan, indicating that mosquitoes have evolved mechanisms to tolerate persistent infection and developed efficient antiviral strategies to restrict viral replication to nonpathogenic levels. Here we provide an overview of recent progress in understanding mosquito antiviral immunity and advances in the strategies by which mosquitoes control viral infection in specific tissues.
Collapse
Affiliation(s)
- Gong Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University, Beijing 100084, PR China.
| | - Yang Liu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University, Beijing 100084, PR China; School of Life Science, Tsinghua University, Beijing 100084, PR China
| | - Penghua Wang
- Department of Microbiology and Immunology, School of Medicine, New York Medical College, Valhalla, NY 10595, USA
| | - Xiaoping Xiao
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Tsinghua University, Beijing 100084, PR China
| |
Collapse
|
30
|
Londono-Renteria B, Troupin A, Conway MJ, Vesely D, Ledizet M, Roundy CM, Cloherty E, Jameson S, Vanlandingham D, Higgs S, Fikrig E, Colpitts TM. Dengue Virus Infection of Aedes aegypti Requires a Putative Cysteine Rich Venom Protein. PLoS Pathog 2015; 11:e1005202. [PMID: 26491875 PMCID: PMC4619585 DOI: 10.1371/journal.ppat.1005202] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/11/2015] [Indexed: 12/20/2022] Open
Abstract
Dengue virus (DENV) is a mosquito-borne flavivirus that causes serious human disease and mortality worldwide. There is no specific antiviral therapy or vaccine for DENV infection. Alterations in gene expression during DENV infection of the mosquito and the impact of these changes on virus infection are important events to investigate in hopes of creating new treatments and vaccines. We previously identified 203 genes that were ≥5-fold differentially upregulated during flavivirus infection of the mosquito. Here, we examined the impact of silencing 100 of the most highly upregulated gene targets on DENV infection in its mosquito vector. We identified 20 genes that reduced DENV infection by at least 60% when silenced. We focused on one gene, a putative cysteine rich venom protein (SeqID AAEL000379; CRVP379), whose silencing significantly reduced DENV infection in Aedes aegypti cells. Here, we examine the requirement for CRVP379 during DENV infection of the mosquito and investigate the mechanisms surrounding this phenomenon. We also show that blocking CRVP379 protein with either RNAi or specific antisera inhibits DENV infection in Aedes aegypti. This work identifies a novel mosquito gene target for controlling DENV infection in mosquitoes that may also be used to develop broad preventative and therapeutic measures for multiple flaviviruses.
Collapse
Affiliation(s)
- Berlin Londono-Renteria
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Andrea Troupin
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Michael J Conway
- Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, Michigan, United States of America
| | - Diana Vesely
- L2 Diagnostics, New Haven, Connecticut, United States of America
| | - Michael Ledizet
- L2 Diagnostics, New Haven, Connecticut, United States of America
| | - Christopher M. Roundy
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Erin Cloherty
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Samuel Jameson
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Dana Vanlandingham
- Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, United States of America
- Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, United States of America
| | - Stephen Higgs
- Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, United States of America
- Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, United States of America
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Tonya M. Colpitts
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
31
|
Williams M, Baxter R. The structure and function of thioester-containing proteins in arthropods. Biophys Rev 2014; 6:261-272. [PMID: 28510031 DOI: 10.1007/s12551-014-0142-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 06/19/2014] [Indexed: 10/25/2022] Open
Abstract
Thioester-containing proteins (TEPs) form an ancient and diverse family of secreted proteins that play central roles in the innate immune response. Two families of TEPs, complement factors and α2-macroglobulins, have been known and studied in vertebrates for many years, but only in the last decade have crystal structures become available. In the same period, the presence of two additional classes of TEPs has been revealed in arthropods. In this review, we discuss the common structural features TEPs and how this knowledge can be applied to the many arthropod TEPs of unknown function. TEPs perform a wide variety of functions that are driven by different quaternary structures and protein-protein interactions between a common set of folded domains. A common theme is regulated conformational change triggered by proteolysis. Structure-function analysis of the diverse arthropod TEPs may identify not just new mechanisms in innate immunity but also interfaces between immunity, development and cell death.
Collapse
Affiliation(s)
- Marni Williams
- Department. of Chemistry, Yale University, New Haven, CT, USA
| | - Richard Baxter
- Department. of Chemistry, Yale University, New Haven, CT, USA.
| |
Collapse
|
32
|
Suzuki Y, Niu G, Hughes GL, Rasgon JL. A viral over-expression system for the major malaria mosquito Anopheles gambiae. Sci Rep 2014; 4:5127. [PMID: 24875042 PMCID: PMC4038844 DOI: 10.1038/srep05127] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/14/2014] [Indexed: 11/08/2022] Open
Abstract
Understanding pathogen/mosquito interactions is essential for developing novel strategies to control mosquito-borne diseases. Technical advances in reverse-genetics, such as RNA interference (RNAi), have facilitated elucidation of components of the mosquito immune system that are antagonistic to pathogen development, and host proteins essential for parasite development. Forward genetic approaches, however, are limited to generation of transgenic insects, and while powerful, mosquito transgenesis is a resource- and time-intensive technique that is not broadly available to most laboratories. The ability to easily "over-express" genes would enhance molecular studies in vector biology and expedite elucidation of pathogen-refractory genes without the need to make transgenic insects. We developed and characterized an efficient Anopheles gambiae densovirus (AgDNV) over-expression system for the major malaria vector Anopheles gambiae. High-levels of gene expression were detected at 3 days post-infection and increased over time, suggesting this is an effective system for gene induction. Strong expression was observed in the fat body and ovaries. We validated multiple short promoters for gene induction studies. Finally, we developed a polycistronic system to simultaneously express multiple genes of interest. This AgDNV-based toolset allows for consistent transduction of genes of interest and will be a powerful molecular tool for research in Anopheles gambiae mosquitoes.
Collapse
Affiliation(s)
- Yasutsugu Suzuki
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| | - Guodong Niu
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
- Current address: Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, 73019, United States of America
| | - Grant L. Hughes
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| | - Jason L. Rasgon
- Department of Entomology, Center for Infectious Disease Dynamics and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| |
Collapse
|
33
|
Xiao X, Liu Y, Zhang X, Wang J, Li Z, Pang X, Wang P, Cheng G. Complement-related proteins control the flavivirus infection of Aedes aegypti by inducing antimicrobial peptides. PLoS Pathog 2014; 10:e1004027. [PMID: 24722701 PMCID: PMC3983052 DOI: 10.1371/journal.ppat.1004027] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 02/09/2014] [Indexed: 01/08/2023] Open
Abstract
The complement system functions during the early phase of infection and directly mediates pathogen elimination. The recent identification of complement-like factors in arthropods indicates that this system shares common ancestry in vertebrates and invertebrates as an immune defense mechanism. Thioester (TE)-containing proteins (TEPs), which show high similarity to mammalian complement C3, are thought to play a key role in innate immunity in arthropods. Herein, we report that a viral recognition cascade composed of two complement-related proteins limits the flaviviral infection of Aedes aegypti. An A. aegypti macroglobulin complement-related factor (AaMCR), belonging to the insect TEP family, is a crucial effector in opposing the flaviviral infection of A. aegypti. However, AaMCR does not directly interact with DENV, and its antiviral effect requires an A. aegypti homologue of scavenger receptor-C (AaSR-C), which interacts with DENV and AaMCR simultaneously in vitro and in vivo. Furthermore, recognition of DENV by the AaSR-C/AaMCR axis regulates the expression of antimicrobial peptides (AMPs), which exerts potent anti-DENV activity. Our results both demonstrate the existence of a viral recognition pathway that controls the flaviviral infection by inducing AMPs and offer insights into a previously unappreciated antiviral function of the complement-like system in arthropods. Hosts are equipped with sophisticated machineries for detecting and eliminating invading viruses before they cause significant physiological damage. Unlike mammals which have both innate and adaptive immune systems, insects rely solely on the innate immune system to limit viral infection. Mosquitoes are natural vectors for many human pathogenic viruses, such as dengue virus (DENV) and yellow fever virus. Despite lacking an immunoglobulin-based humoral response, mosquitoes arm themselves with a functional complement-like system to ward off invading pathogens. Herein, we show that a system composed of complement-related factors recognizes and limits flaviviruses by inducing antimicrobial peptides expression. Understanding antiviral mechanisms in arthropods may provide novel strategies for limiting arboviral transmission in nature.
Collapse
Affiliation(s)
- Xiaoping Xiao
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
| | - Yang Liu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
| | - Xiaoyan Zhang
- School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| | - Jing Wang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
| | - Zuofeng Li
- School of Life Sciences and Technology, Tongji University, Shanghai, People's Republic of China
| | - Xiaojing Pang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
| | - Penghua Wang
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Gong Cheng
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, People's Republic of China
- * E-mail:
| |
Collapse
|
34
|
Liu Y, Zhang F, Liu J, Xiao X, Zhang S, Qin C, Xiang Y, Wang P, Cheng G. Transmission-blocking antibodies against mosquito C-type lectins for dengue prevention. PLoS Pathog 2014; 10:e1003931. [PMID: 24550728 PMCID: PMC3923773 DOI: 10.1371/journal.ppat.1003931] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 12/31/2013] [Indexed: 12/29/2022] Open
Abstract
C-type lectins are a family of proteins with carbohydrate-binding activity. Several C-type lectins in mammals or arthropods are employed as receptors or attachment factors to facilitate flavivirus invasion. We previously identified a C-type lectin in Aedes aegypti, designated as mosquito galactose specific C-type lectin-1 (mosGCTL-1), facilitating the attachment of West Nile virus (WNV) on the cell membrane. Here, we first identified that 9 A. aegypti mosGCTL genes were key susceptibility factors facilitating DENV-2 infection, of which mosGCTL-3 exhibited the most significant effect. We found that mosGCTL-3 was induced in mosquito tissues with DENV-2 infection, and that the protein interacted with DENV-2 surface envelop (E) protein and virions in vitro and in vivo. In addition, the other identified mosGCTLs interacted with the DENV-2 E protein, indicating that DENV may employ multiple mosGCTLs as ligands to promote the infection of vectors. The vectorial susceptibility factors that facilitate pathogen invasion may potentially be explored as a target to disrupt the acquisition of microbes from the vertebrate host. Indeed, membrane blood feeding of antisera against mosGCTLs dramatically reduced mosquito infective ratio. Hence, the immunization against mosGCTLs is a feasible approach for preventing dengue infection. Our study provides a future avenue for developing a transmission-blocking vaccine that interrupts the life cycle of dengue virus and reduces disease burden. Dengue virus (DENV), a mosquito-borne flavivirus, is currently the most significant arbovirus afflicting tropical and sub-tropical countries worldwide. No vaccine or therapeutics are available, and dengue has rapidly spread over the last decade. Therefore, additional strategies to combat dengue are urgently needed. In this study, we characterized multiple C-type lectins as susceptibility factors for dengue infection in A. aegypti. These mosGCTLs directly interacted with dengue virus in vitro and in vivo. The combination of antisera against multiple mosGCTLs efficiently reduced DENV-2 infection after a blood meal, suggesting that it is feasible to develop a mosGCTL-based transmission-blocking vaccine to interrupt the life cycle of dengue virus and control disease burden in nature. This study substantially extends our understanding of dengue replication in vectors and provides a research avenue by which the development of therapeutics for preventing the dissemination of mosquito-borne viral diseases can be pursued in the future.
Collapse
Affiliation(s)
- Yang Liu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
| | - Fuchun Zhang
- Guangzhou 8th People's Hospital, Guangzhou, People's Republic of China
| | - Jianying Liu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
| | - Xiaoping Xiao
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
| | - Siyin Zhang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
| | - Chengfeng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, People's Republic of China
| | - Ye Xiang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
| | - Penghua Wang
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Gong Cheng
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, People's Republic of China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, People's Republic of China
- * E-mail:
| |
Collapse
|
35
|
Pinto SB, Stainton K, Harris S, Kambris Z, Sutton ER, Bonsall MB, Parkhill J, Sinkins SP. Transcriptional regulation of Culex pipiens mosquitoes by Wolbachia influences cytoplasmic incompatibility. PLoS Pathog 2013; 9:e1003647. [PMID: 24204251 PMCID: PMC3814344 DOI: 10.1371/journal.ppat.1003647] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 08/06/2013] [Indexed: 11/23/2022] Open
Abstract
Cytoplasmic incompatibility (CI) induced by the endosymbiont Wolbachia pipientis causes complex patterns of crossing sterility between populations of the Culex pipiens group of mosquitoes. The molecular basis of the phenotype is yet to be defined. In order to investigate what host changes may underlie CI at the molecular level, we examined the transcription of a homolog of the Drosophila melanogaster gene grauzone that encodes a zinc finger protein and acts as a regulator of female meiosis, in which mutations can cause sterility. Upregulation was observed in Wolbachia-infected C. pipiens group individuals relative to Wolbachia-cured lines and the level of upregulation differed between lines that were reproductively incompatible. Knockdown analysis of this gene using RNAi showed an effect on hatch rates in a Wolbachia infected Culex molestus line. Furthermore, in later stages of development an effect on developmental progression in CI embryos occurs in bidirectionally incompatible crosses. The genome of a wPip Wolbachia strain variant from Culex molestus was sequenced and compared with the genome of a wPip variant with which it was incompatible. Three genes in inserted or deleted regions were newly identified in the C. molestus wPip genome, one of which is a transcriptional regulator labelled wtrM. When this gene was transfected into adult Culex mosquitoes, upregulation of the grauzone homolog was observed. These data suggest that Wolbachia-mediated regulation of host gene expression is a component of the mechanism of cytoplasmic incompatibility. Wolbachia are maternally inherited bacteria that manipulate invertebrate reproduction. Cytoplasmic incompatibility is embryo death that occurs when males carrying Wolbachia mate with females that do not, or that carry a different Wolbachia variant; its mechanism is poorly understood. In Culex mosquitoes, in the presence of Wolbachia a gene related to a Drosophila melanogaster gene, grauzone, which has been shown to act as a regulator of the meiotic cell cycle, showed an elevated level of expression. When lower levels of expression were achieved through RNA interference, embryo hatch rates were affected and the stage of development at which embryo death occurs was altered. To find Wolbachia genes that influence cytoplasmic incompatibility, we compared the genomes of two variants of Wolbachia from Culex that produce cytoplasmic incompatibility with one another. Although most segments of these genomes were very similar, one newly identified gene is predicted to be a regulator of gene transcription. We cloned this gene into a plasmid, expressed it in adult mosquitoes and found higher levels of expression of the Culex grauzone homolog. This suggests that the Wolbachia transcriptional regulator may play an important role in manipulating the host in order to induce cytoplasmic incompatibility.
Collapse
Affiliation(s)
- Sofia B. Pinto
- Peter Medawar Building for Pathogen Research and Nuffield Department of Medicine (NDM), University of Oxford, Oxford, United Kingdom
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Kirsty Stainton
- Peter Medawar Building for Pathogen Research and Nuffield Department of Medicine (NDM), University of Oxford, Oxford, United Kingdom
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Simon Harris
- Pathogen Genomics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Zakaria Kambris
- Peter Medawar Building for Pathogen Research and Nuffield Department of Medicine (NDM), University of Oxford, Oxford, United Kingdom
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Elizabeth R. Sutton
- Peter Medawar Building for Pathogen Research and Nuffield Department of Medicine (NDM), University of Oxford, Oxford, United Kingdom
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | | | - Julian Parkhill
- Pathogen Genomics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Steven P. Sinkins
- Peter Medawar Building for Pathogen Research and Nuffield Department of Medicine (NDM), University of Oxford, Oxford, United Kingdom
- Department of Zoology, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
36
|
Wilke ABB, Scaife S, Alphey L, Marrelli MT. DsRed2 transient expression in Culex quinquefasciatus mosquitoes. Mem Inst Oswaldo Cruz 2013; 108:529-31. [PMID: 23828005 PMCID: PMC3970632 DOI: 10.1590/s0074-02762013000400023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 12/07/2012] [Indexed: 11/21/2022] Open
Abstract
Culex quinquefasciatus mosquitoes have been successfully genetically modified only once, despite the efforts of several laboratories to transform and establish a stable strain. We have developed a transient gene expression method, in Culex, that delivers plasmid DNA directly to the mosquito haemolymph and additional tissues. We were able to express DsRed2 fluorescent protein in adult Cx. quinquefasciatus mosquitoes by injecting plasmids directly into their thorax. The expression of DsRed2 in adult Cx. quinquefasciatus mosquitoes is an important stepping stone to genetic transformation and the potential use of new control strategies and genetic interactions.
Collapse
Affiliation(s)
- André Barretto Bruno Wilke
- Departamento de Epidemiologia, Faculdade de Saúde Pública, Universidade de São Paulo, São Paulo, SP, Brasil.
| | | | | | | |
Collapse
|
37
|
Wilke ABB, Scaife S, Alphey L, Marrelli MT. DsRed2 transient expression in Culex quinquefasciatus mosquitoes. Mem Inst Oswaldo Cruz 2013; 108. [PMID: 23828005 PMCID: PMC3970632 DOI: 10.1590/0074-0276108042013023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Culex quinquefasciatus mosquitoes have been successfully genetically modified only once, despite the efforts of several laboratories to transform and establish a stable strain. We have developed a transient gene expression method, in Culex, that delivers plasmid DNA directly to the mosquito haemolymph and additional tissues. We were able to express DsRed2 fluorescent protein in adult Cx. quinquefasciatus mosquitoes by injecting plasmids directly into their thorax. The expression of DsRed2 in adult Cx. quinquefasciatus mosquitoes is an important stepping stone to genetic transformation and the potential use of new control strategies and genetic interactions.
Collapse
Affiliation(s)
- André Barretto Bruno Wilke
- Departamento de Epidemiologia, Faculdade de Saúde Pública,
Universidade de São Paulo, São Paulo, SP, Brasil,Corresponding author:
| | | | - Luke Alphey
- Oxitec Ltd, Oxford, United Kingdom,Department of Zoology, University of Oxford, Oxford, United
Kingdom
| | - Mauro Toledo Marrelli
- Departamento de Epidemiologia, Faculdade de Saúde Pública,
Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
38
|
XIA LI, YIN SHANKAI. Local gene transfection in the cochlea (Review). Mol Med Rep 2013; 8:3-10. [DOI: 10.3892/mmr.2013.1496] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 12/13/2012] [Indexed: 11/06/2022] Open
|