1
|
Terhzaz S, Kerrigan D, Almire F, Szemiel AM, Hughes J, Parvy JP, Palmarini M, Kohl A, Shi X, Pondeville E. NSm is a critical determinant for bunyavirus transmission between vertebrate and mosquito hosts. Nat Commun 2025; 16:1214. [PMID: 39890788 DOI: 10.1038/s41467-024-54809-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 11/21/2024] [Indexed: 02/03/2025] Open
Abstract
Bunyavirales is a very large order including viruses infecting a variety of taxonomic groups such as arthropods, vertebrates, plants, and protozoa. Some bunyaviruses are transmitted between vertebrate hosts by blood-sucking arthropods and cause major diseases in humans and animals. It is not understood why only some bunyaviruses have evolved the capacity to be transmitted by arthropod vectors. Here we show that only vector-borne bunyaviruses express a non-structural protein, NSm, whose function has so far remained largely elusive. Using as experimental system Bunyamwera virus (BUNV) and its invertebrate host, Aedes aegypti, we show that NSm is dispensable for viral replication in mosquito cells in vitro but is absolutely required for successful infection in the female mosquito following a blood meal. More specifically, NSm is required for cell-to-cell spread and egress from the mosquito midgut, a known barrier to viral infection. Notably, the requirement for NSm is specific to the midgut; bypassing this barrier by experimental intrathoracic infection of the mosquito eliminates the necessity of NSm for virus spread in other tissues, including the salivary glands. Overall, we unveiled a key evolutionary process that allows the transmission of vector-borne bunyaviruses between arthropod and vertebrate hosts.
Collapse
Affiliation(s)
- Selim Terhzaz
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - David Kerrigan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Floriane Almire
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Agnieszka M Szemiel
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Jean-Philippe Parvy
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK
- Departments of Tropical Disease Biology and Vector Biology, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| | - Xiaohong Shi
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Emilie Pondeville
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| |
Collapse
|
2
|
Riana E, Sri-In C, Songkasupa T, Bartholomay LC, Thontiravong A, Tiawsirisup S. Infection, dissemination, and transmission of lumpy skin disease virus in Aedes aegypti (Linnaeus), Culex tritaeniorhynchus (Giles), and Culex quinquefasciatus (Say) mosquitoes. Acta Trop 2024; 254:107205. [PMID: 38579960 DOI: 10.1016/j.actatropica.2024.107205] [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: 02/16/2024] [Revised: 03/24/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
Lumpy skin disease virus (LSDV) is a transboundary viral disease in cattle and water buffaloes. Although this Poxvirus is supposedly transmitted by mechanical vectors, only a few studies have investigated the role of local vectors in the transmission of LSDV. This study examined the infection, dissemination, and transmission rates of LSDV in Aedes aegypti, Culex tritaeniorhynchus, and Culex quinquefasciatus following artificial membrane feeding of 102.7, 103.7, 104.7 TCID50/mL LSDV in sheep blood. The results demonstrated that these mosquito species were susceptible to LSDV, with Cx tritaeniorhynchus exhibiting significantly different characteristics from Ae. aegypti and Cx. quinquefasciatus. These three mosquito species were susceptible to LSDV. Ae. aegypti showed it as early as 2 days post-infection (dpi), indicating swift dissemination in this particular species. The extrinsic incubation period (EIP) of LSDV in Cx. tritaeniorhynchus and Cx. quinquefasciatus was 8 and 14 dpi, respectively. Ingestion of different viral titers in blood did not affect the infection, dissemination, or transmission rates of Cx. tritaeniorhynchus and Cx. quinquefasciatus. All rates remained consistently high at 8-14 dpi for Cx. tritaeniorhynchus. In all three species, LSDV remained detectable until 14 dpi. The present findings indicate that, Ae. aegypti, Cx. tritaeniorhynchus, and Cx. quinquefasciatus may act as vectors during the LSDV outbreak; their involvement may extend beyond being solely mechanical vectors.
Collapse
Affiliation(s)
- Elizabeth Riana
- The International Graduate Program of Veterinary Science and Technology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand; Center of Excellence in Animal Vector-Borne Diseases, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Chalida Sri-In
- Center of Excellence in Animal Vector-Borne Diseases, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Tapanut Songkasupa
- Virology section, National Institute of Animal Health, Department of Livestock Development, Bangkok, Thailand
| | - Lyric C Bartholomay
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Aunyaratana Thontiravong
- Center of Excellence in Animal Vector-Borne Diseases, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand; Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Sonthaya Tiawsirisup
- Center of Excellence in Animal Vector-Borne Diseases, Veterinary Parasitology Unit, Department of Veterinary Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
3
|
Rosales Rosas AL, Wang L, Goossens S, Cuvry A, Li LH, Santos-Ferreira N, Soto A, Dallmeier K, Rocha-Pereira J, Delang L. Ex vivo gut cultures of Aedes aegypti are efficiently infected by mosquito-borne alpha- and flaviviruses. Microbiol Spectr 2023; 11:e0519522. [PMID: 37540021 PMCID: PMC10580962 DOI: 10.1128/spectrum.05195-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 06/27/2023] [Indexed: 08/05/2023] Open
Abstract
Aedes aegypti mosquitoes can transmit several arboviruses, including chikungunya virus (CHIKV), dengue virus (DENV), and Zika virus (ZIKV). When blood-feeding on a virus-infected human, the mosquito ingests the virus into the midgut (stomach), where it replicates and must overcome the midgut barrier to disseminate to other organs and ultimately be transmitted via the saliva. Current tools to study mosquito-borne viruses (MBVs) include 2D-cell culture systems and in vivo mosquito infection models, which offer great advantages, yet have some limitations. Here, we describe a long-term ex vivo culture of Ae. aegypti guts. Cultured guts were metabolically active for 7 d in a 96-well plate at 28°C and were permissive to ZIKV, DENV, Ross River virus, and CHIKV. Ex vivo guts from Culex pipiens mosquitoes were found to be permissive to Usutu virus. Immunofluorescence staining confirmed viral protein synthesis in CHIKV-infected guts of Ae. aegypti. Furthermore, fluorescence microscopy revealed replication and spread of a reporter DENV in specific regions of the midgut. In addition, two known antiviral molecules, β-d-N4-hydroxycytidine and 7-deaza-2'-C-methyladenosine, were able to inhibit CHIKV and ZIKV replication, respectively, in the ex vivo model. Together, our results show that ex vivo guts can be efficiently infected with mosquito-borne alpha- and flaviviruses and employed to evaluate antiviral drugs. Furthermore, the setup can be extended to other mosquito species. Ex vivo gut cultures could thus be a new model to study MBVs, offering the advantage of reduced biosafety measures compared to infecting living mosquitoes. IMPORTANCE Mosquito-borne viruses (MBVs) are a significant global health threat since they can cause severe diseases in humans, such as hemorrhagic fever, encephalitis, and chronic arthritis. MBVs rely on the mosquito vector to infect new hosts and perpetuate virus transmission. No therapeutics are currently available. The study of arbovirus infection in the mosquito vector can greatly contribute to elucidating strategies for controlling arbovirus transmission. This work investigated the infection of guts from Aedes aegypti mosquitoes in an ex vivo platform. We found several MBVs capable of replicating in the gut tissue, including viruses of major health importance, such as dengue, chikungunya, and Zika viruses. In addition, antiviral compounds reduced arbovirus infection in the cultured gut tissue. Overall, the gut model emerges as a useful tool for diverse applications such as studying tissue-specific responses to virus infection and screening potential anti-arboviral molecules.
Collapse
Affiliation(s)
- Ana Lucia Rosales Rosas
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Lanjiao Wang
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Sara Goossens
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Arno Cuvry
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Li-Hsin Li
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Nanci Santos-Ferreira
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Alina Soto
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Joana Rocha-Pereira
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Leen Delang
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| |
Collapse
|
4
|
Darby CS, Featherston KM, Lin J, Franz AWE. Detection of La Crosse Virus In Situ and in Individual Progeny to Assess the Vertical Transmission Potential in Aedes albopictus and Aedes aegypti. INSECTS 2023; 14:601. [PMID: 37504607 PMCID: PMC10380845 DOI: 10.3390/insects14070601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023]
Abstract
La Crosse virus (LACV) is circulating in the midwestern and southeastern states of the United States and can cause human encephalitis. The main vector of the virus is the eastern tree-hole mosquito, Aedes triseriatus. Ae. albopictus has been also described as a natural LACV vector, while Ae. aegypti has been infected with the virus under laboratory conditions. Here, we compare the vertical transmission potential of LACV in Ae. albopictus and Ae. aegypti, with emphasis given to the ovarian infection patterns that the virus generates in both species. Both mosquito species received artificial bloodmeals containing LACV. At defined time points post-infection/bloodmeal, midguts, head tissue, and ovaries were analyzed for the presence of virus. Viral infection patterns in the ovaries were visualized via immunofluorescence confocal microscopy and immunohistopathology assays using an LACV-specific monoclonal antibody. In Ae. aegypti, LACV was confronted with midgut infection and escape barriers, which were much less pronounced in Ae. albopictus, resulting in a significantly higher prevalence of infection in the latter. Following the ingestion of a single virus-containing bloodmeal, no progeny larvae were found to be virus-infected. Regardless, females of both species showed the presence of LACV antigen in their ovariole sheaths. Furthermore, in a single Ae. albopictus female, viral antigen was associated with the nurse cells inside the primary follicles. Following the ingestion of a second non-infectious bloodmeal at 7- or 10-days post-ingestion of an LACV-containing bloodmeal, more progeny larvae of Ae. albopictus than of Ae. aegypti were virus-infected. LACV antigen was detected in the egg chambers and ovariole sheaths of both mosquito species. Traces of viral antigen were also detected in a few oocytes from Ae. albopictus. The low level of vertical transmission and the majority of the ovarian infection patterns suggested the transovum rather than transovarial transmission (TOT) of the virus in both vector species. However, based on the detection of LACV antigen in follicular tissue and oocytes, there was the potential for TOT among several Ae. albopictus females. Thus, TOT is not a general feature of LACV infection in mosquitoes. Instead, the TOT of LACV seems to be dependent on its particular interaction with the reproductive tissues of a female.
Collapse
Affiliation(s)
| | | | | | - Alexander W. E. Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (C.S.D.); (K.M.F.); (J.L.)
| |
Collapse
|
5
|
Fu JYL, Chua CL, Abu Bakar AS, Vythilingam I, Wan Sulaiman WY, Alphey L, Chan YF, Sam IC. Susceptibility of Aedes albopictus, Ae. aegypti and human populations to Ross River virus in Kuala Lumpur, Malaysia. PLoS Negl Trop Dis 2023; 17:e0011423. [PMID: 37307291 DOI: 10.1371/journal.pntd.0011423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 05/28/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Emerging arboviruses such as chikungunya and Zika viruses have unexpectedly caused widespread outbreaks in tropical and subtropical regions recently. Ross River virus (RRV) is endemic in Australia and has epidemic potential. In Malaysia, Aedes mosquitoes are abundant and drive dengue and chikungunya outbreaks. We assessed risk of an RRV outbreak in Kuala Lumpur, Malaysia by determining vector competence of local Aedes mosquitoes and local seroprevalence as a proxy of human population susceptibility. METHODOLOGY/PRINCIPAL FINDINGS We assessed oral susceptibility of Malaysian Ae. aegypti and Ae. albopictus by real-time PCR to an Australian RRV strain SW2089. Replication kinetics in midgut, head and saliva were determined at 3 and 10 days post-infection (dpi). With a 3 log10 PFU/ml blood meal, infection rate was higher in Ae. albopictus (60%) than Ae. aegypti (15%; p<0.05). Despite similar infection rates at 5 and 7 log10 PFU/ml blood meals, Ae. albopictus had significantly higher viral loads and required a significantly lower median oral infectious dose (2.7 log10 PFU/ml) than Ae. aegypti (4.2 log10 PFU/ml). Ae. albopictus showed higher vector competence, with higher viral loads in heads and saliva, and higher transmission rate (RRV present in saliva) of 100% at 10 dpi, than Ae. aegypti (41%). Ae. aegypti demonstrated greater barriers at either midgut escape or salivary gland infection, and salivary gland escape. We then assessed seropositivity against RRV among 240 Kuala Lumpur inpatients using plaque reduction neutralization, and found a low rate of 0.8%. CONCLUSIONS/SIGNIFICANCE Both Ae. aegypti and Ae. albopictus are susceptible to RRV, but Ae. albopictus displays greater vector competence. Extensive travel links with Australia, abundant Aedes vectors, and low population immunity places Kuala Lumpur, Malaysia at risk of an imported RRV outbreak. Surveillance and increased diagnostic awareness and capacity are imperative to prevent establishment of new arboviruses in Malaysia.
Collapse
Affiliation(s)
- Jolene Yin Ling Fu
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Chong Long Chua
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | | | - Indra Vythilingam
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | | | - Luke Alphey
- Arthropod Genetics Group, The Pirbright Institute, Woking, United Kingdom
| | - Yoke Fun Chan
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - I-Ching Sam
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Mulatier M, Boullis A, Dollin C, Cebrián-Torrejón G, Vega-Rúa A. Chikungunya Virus Infection and Gonotrophic Cycle Shape Aedes aegypti Oviposition Behavior and Preferences. Viruses 2023; 15:v15051043. [PMID: 37243130 DOI: 10.3390/v15051043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Targeting gravid females through chemical lures is a promising strategy in vector control; however, it requires the understanding of the factors susceptible to alter female oviposition behavior. Here, we evaluated the effect of infection with chikungunya virus (CHIKV) and the number of gonotrophic cycles (GCs) on oviposition activity in A. aegypti. Dual choice oviposition assays were performed, where dodecanoic acid, pentadecanoic acid, n-heneicosane and a Sargasssum fluitans (Børgesen) Børgesen extract were tested in uninfected females and females infected with CHIKV, at the 1st and 2nd GC. Infected females displayed a lower percentage of oviposition and a higher number of eggs laid at the 1st GC. Then, the combined effects of GC and CHIKV were observed on oviposition preferences, with a chemical-dependent effect. For instance, the deterrent effect of n-heneicosane and pentadecanoic acid increased at the 2nd GC in infected females. These results allow for a deeper understanding of the mechanisms involved in oviposition site selection and highlight the need for taking into account physiological stage changes to increase the control programs' efficacy.
Collapse
Affiliation(s)
- Margaux Mulatier
- Laboratory of Vector Control Research, Pasteur Institute of Guadeloupe-Lieu-dit Morne Jolivière, 97139 Les Abymes, France
| | - Antoine Boullis
- Laboratory of Vector Control Research, Pasteur Institute of Guadeloupe-Lieu-dit Morne Jolivière, 97139 Les Abymes, France
| | - Christelle Dollin
- Laboratory of Vector Control Research, Pasteur Institute of Guadeloupe-Lieu-dit Morne Jolivière, 97139 Les Abymes, France
| | | | - Anubis Vega-Rúa
- Laboratory of Vector Control Research, Pasteur Institute of Guadeloupe-Lieu-dit Morne Jolivière, 97139 Les Abymes, France
| |
Collapse
|
7
|
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: 1.5] [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
|
8
|
Peinado RDS, Eberle RJ, Arni RK, Coronado MA. A Review of Omics Studies on Arboviruses: Alphavirus, Orthobunyavirus and Phlebovirus. Viruses 2022; 14:2194. [PMID: 36298749 PMCID: PMC9607206 DOI: 10.3390/v14102194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
Since the intricate and complex steps in pathogenesis and host-viral interactions of arthropod-borne viruses or arboviruses are not completely understood, the multi-omics approaches, which encompass proteomics, transcriptomics, genomics and metabolomics network analysis, are of great importance. We have reviewed the omics studies on mosquito-borne viruses of the Togaviridae, Peribuyaviridae and Phenuiviridae families, specifically for Chikungunya, Mayaro, Oropouche and Rift Valley Fever viruses. Omics studies can potentially provide a new perspective on the pathophysiology of arboviruses, contributing to a better comprehension of these diseases and their effects and, hence, provide novel insights for the development of new antiviral drugs or therapies.
Collapse
Affiliation(s)
- Rafaela dos S. Peinado
- Multiuser Center for Biomolecular Innovation, Department of Physics, Sao Paulo State University, Sao Jose do Rio Preto 15054-000, SP, Brazil
| | - Raphael J. Eberle
- Institute of Biological Information Processing, IBI-7: Structural Biochemistry, Forschungszentrum Jülich, 52425 Jülich, Germany
- Institut für Physikalische Biologie, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | - Raghuvir K. Arni
- Multiuser Center for Biomolecular Innovation, Department of Physics, Sao Paulo State University, Sao Jose do Rio Preto 15054-000, SP, Brazil
| | - Mônika A. Coronado
- Institute of Biological Information Processing, IBI-7: Structural Biochemistry, Forschungszentrum Jülich, 52425 Jülich, Germany
| |
Collapse
|
9
|
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: 2.7] [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
|
10
|
Choy MM, Ng DHL, Siriphanitchakorn T, Ng WC, Sundstrom KB, Tan HC, Zhang SL, Chan KWK, Manuel M, Kini RM, Chan KR, Vasudevan SG, Ooi EE. A Non-structural 1 Protein G53D Substitution Attenuates a Clinically Tested Live Dengue Vaccine. Cell Rep 2021; 31:107617. [PMID: 32402284 DOI: 10.1016/j.celrep.2020.107617] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 03/03/2020] [Accepted: 04/15/2020] [Indexed: 12/26/2022] Open
Abstract
The molecular basis of dengue virus (DENV) attenuation remains ambiguous and hampers a targeted approach to derive safe but nonetheless immunogenic live vaccine candidates. Here, we take advantage of DENV serotype 2 PDK53 vaccine strain, which recently and successfully completed a phase-3 clinical trial, to identify how this virus is attenuated compared to its wild-type parent, DENV2 16681. Site-directed mutagenesis on a 16681 infectious clone identifies a single G53D substitution in the non-structural 1 (NS1) protein that reduces 16681 infection and dissemination in both Aedes aegypti, as well as in mammalian cells to produce the characteristic phenotypes of PDK53. Mechanistically, NS1 G53D impairs the function of a known host factor, the endoplasmic reticulum (ER)-resident ribophorin 1 protein, to properly glycosylate NS1 and thus induce a host antiviral gene through ER stress responses. Our findings provide molecular insights on DENV attenuation on a clinically tested strain.
Collapse
Affiliation(s)
- Milly M Choy
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Dorothy H L Ng
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore; Department of Infectious Diseases, Singapore General Hospital, Singapore 169108, Singapore
| | - Tanamas Siriphanitchakorn
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
| | - Wy Ching Ng
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Karin B Sundstrom
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Hwee Cheng Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Summer L Zhang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Kitti W K Chan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Menchie Manuel
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - R Manjunatha Kini
- Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
| | - Kuan Rong Chan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Subhash G Vasudevan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Eng Eong Ooi
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore.
| |
Collapse
|
11
|
Dong S, Dimopoulos G. Antiviral Compounds for Blocking Arboviral Transmission in Mosquitoes. Viruses 2021; 13:v13010108. [PMID: 33466915 PMCID: PMC7830659 DOI: 10.3390/v13010108] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/06/2021] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Mosquito-borne arthropod-borne viruses (arboviruses) such as the dengue virus (DENV), Zika virus (ZIKV), and chikungunya virus (CHIKV) are important human pathogens that are responsible for significant global morbidity and mortality. The recent emergence and re-emergence of mosquito-borne viral diseases (MBVDs) highlight the urgent need for safe and effective vaccines, therapeutics, and vector-control approaches to prevent MBVD outbreaks. In nature, arboviruses circulate between vertebrate hosts and arthropod vectors; therefore, disrupting the virus lifecycle in mosquitoes is a major approach for combating MBVDs. Several strategies were proposed to render mosquitoes that are refractory to arboviral infection, for example, those involving the generation of genetically modified mosquitoes or infection with the symbiotic bacterium Wolbachia. Due to the recent development of high-throughput screening methods, an increasing number of drugs with inhibitory effects on mosquito-borne arboviruses in mammalian cells were identified. These antivirals are useful resources that can impede the circulation of arboviruses between arthropods and humans by either rendering viruses more vulnerable in humans or suppressing viral infection by reducing the expression of host factors in mosquitoes. In this review, we summarize recent advances in small-molecule antiarboviral drugs in mammalian and mosquito cells, and discuss how to use these antivirals to block the transmission of MBVDs.
Collapse
|
12
|
Merwaiss F, Filomatori CV, Susuki Y, Bardossy ES, Alvarez DE, Saleh MC. Chikungunya Virus Replication Rate Determines the Capacity of Crossing Tissue Barriers in Mosquitoes. J Virol 2021; 95:e01956-20. [PMID: 33148794 PMCID: PMC7925089 DOI: 10.1128/jvi.01956-20] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
Chikungunya virus (CHIKV) is a reemerging and rapidly spreading pathogen transmitted by mosquitoes. The emergence of new epidemic variants of the virus is associated with genetic evolutionary traits, including duplication of repeated RNA elements in the 3' untranslated region (UTR) that seemingly favor transmission by mosquitoes. The transmission potential of a given variant results from a complex interplay between virus populations and anatomical tissue barriers in the mosquito. Here, we used the wild-type CHIKV Caribbean strain and an engineered mutant harboring a deletion in the 3' UTR to dissect the interactions of virus variants with the anatomical barriers that impede transmission during the replication cycle of the virus in Aedes mosquitoes. Compared to the 3'-UTR mutant, we observed that the wild-type virus had a short extrinsic incubation period (EIP) after an infectious blood meal and was expectorated into mosquito saliva much more efficiently. We found that high viral titers in the midgut are not sufficient to escape the midgut escape barrier. Rather, viral replication kinetics play a crucial role in determining midgut escape and the transmission ability of CHIKV. Finally, competition tests in mosquitoes coinfected with wild-type and mutant viruses revealed that both viruses successfully colonized the midgut, but wild-type viruses effectively displaced mutant viruses during systemic infection due to their greater efficiency of escaping from the midgut into secondary tissues. Overall, our results uncover a link between CHIKV replication kinetics and the effect of bottlenecks on population diversity, as slowly replicating variants are less able to overcome the midgut escape barrier.IMPORTANCE It is well established that selective pressures in mosquito vectors impose population bottlenecks for arboviruses. Here, we used a CHIKV Caribbean lineage mutant carrying a deletion in the 3' UTR to study host-virus interactions in vivo in the epidemic mosquito vector Aedes aegypti We found that the mutant virus had a delayed replication rate in mosquitoes, which lengthened the extrinsic incubation period (EIP) and reduced fitness relative to the wild-type virus. As a result, the mutant virus displayed a reduced capacity to cross anatomical barriers during the infection cycle in mosquitoes, thus reducing the virus transmission rate. Our findings show how selective pressures act on CHIKV noncoding regions to select variants with shorter EIPs that are preferentially transmitted by the mosquito vector.
Collapse
Affiliation(s)
- Fernando Merwaiss
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
- Institut Pasteur, Viruses and RNA Interference Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Claudia V Filomatori
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Yasutsugu Susuki
- Institut Pasteur, Viruses and RNA Interference Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Eugenia S Bardossy
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Diego E Alvarez
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - María-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| |
Collapse
|
13
|
Effects of Manipulating Fibroblast Growth Factor Expression on Sindbis Virus Replication In Vitro and in Aedes aegypti Mosquitoes. Viruses 2020; 12:v12090943. [PMID: 32858937 PMCID: PMC7552049 DOI: 10.3390/v12090943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/14/2020] [Accepted: 08/23/2020] [Indexed: 12/05/2022] Open
Abstract
Fibroblast growth factors (FGFs) are conserved among vertebrate and invertebrate animals and function in cell proliferation, cell differentiation, tissue repair, and embryonic development. A viral fibroblast growth factor (vFGF) homolog encoded by baculoviruses, a group of insect viruses, is involved in escape of baculoviruses from the insect midgut by stimulating basal lamina remodeling. This led us to investigate whether cellular FGF is involved in the escape of an arbovirus from mosquito midgut. In this study, the effects of manipulating FGF expression on Sindbis virus (SINV) replication and escape from the midgut of the mosquito vector Aedes aegypti were examined. RNAi-mediated silencing of either Ae. aegypti FGF (AeFGF) or FGF receptor (AeFGFR) expression reduced SINV replication following oral infection of Ae. aegypti mosquitoes. However, overexpression of baculovirus vFGF using recombinant SINV constructs had no effect on replication of these viruses in cultured mosquito or vertebrate cells, or in orally infected Ae. aegypti mosquitoes. We conclude that reducing FGF signaling decreases the ability of SINV to replicate in mosquitoes, but that overexpression of vFGF has no effect, possibly because endogenous FGF levels are already sufficient for optimal virus replication. These results support the hypothesis that FGF signaling, possibly by inducing remodeling of midgut basal lamina, is involved in arbovirus midgut escape following virus acquisition from a blood meal.
Collapse
|
14
|
Filomatori CV, Merwaiss F, Bardossy ES, Alvarez DE. Impact of alphavirus 3'UTR plasticity on mosquito transmission. Semin Cell Dev Biol 2020; 111:148-155. [PMID: 32665176 DOI: 10.1016/j.semcdb.2020.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022]
Abstract
Alphaviruses such as chikungunya and western equine encephalitis viruses are important human pathogens transmitted by mosquitoes that have recently caused large epidemic and epizootic outbreaks. The epidemic potential of alphaviruses is often related to enhanced mosquito transmission. Tissue barriers and antiviral responses impose bottlenecks to viral populations in mosquitoes. Substitutions in the envelope proteins and the presence of repeated sequence elements (RSEs) in the 3'UTR of epidemic viruses were proposed to be specifically associated to efficient replication in mosquito vectors. Here, we discuss the molecular mechanisms that originated RSEs, the evolutionary forces that shape the 3'UTR of alphaviruses, and the significance of RSEs for mosquito transmission. Finally, the presence of RSEs in the 3'UTR of viral genomes appears as evolutionary trait associated to mosquito adaptation and emerges as a common feature among viruses from the alphavirus and flavivirus genera.
Collapse
Affiliation(s)
- Claudia V Filomatori
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín-CONICET, Argentina
| | - Fernando Merwaiss
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín-CONICET, Argentina
| | - Eugenia S Bardossy
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín-CONICET, Argentina
| | - Diego E Alvarez
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín-CONICET, Argentina.
| |
Collapse
|
15
|
Cui Y, Liu P, Mooney BP, Franz AWE. Quantitative Proteomic Analysis of Chikungunya Virus-Infected Aedes aegypti Reveals Proteome Modulations Indicative of Persistent Infection. J Proteome Res 2020; 19:2443-2456. [PMID: 32375005 DOI: 10.1021/acs.jproteome.0c00173] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The mosquito-borne chikungunya virus (CHIKV) poses a threat to human health in tropical countries throughout the world. The molecular interactions of CHIKV with its mosquito vector Aedes aegypti are not fully understood. Following oral acquisition of CHIKV via salinemeals, we analyzed changes in the proteome of Ae. aegypti in 12 h intervals by label-free quantification using a timsTOF Pro mass spectrometer. For each of the seven time points, between 2647 and 3167 proteins were identified among CHIKV-infected and noninfected mosquito samples, and fewer than 6% of those identified proteins were affected by the virus. Functional enrichment analysis revealed that the three pathways, Endocytosis, Oxidative phosphorylation, and Ribosome biogenesis, were enriched during CHIKV infection. On the other hand, three pathways of the cellular RNA machinery and five metabolism related pathways were significantly attenuated in the CHIKV-infected samples. Furthermore, proteins associated with cytoskeleton and vesicular transport, as well as various serine-type endopeptidases and metallo-proteinases, were modulated in the presence of CHIKV. Our study reveals biological pathways and novel proteins interacting with CHIKV in the mosquito. Overall, CHIKV infection caused minor changes to the mosquito proteome demonstrating a high level of adaption between the vector and the virus, essentially coexisting in a nonpathogenic relationship. The mass spectrometry data have been deposited to the MassIVE repository (https://massive.ucsd.edu/ProteoSAFe/dataset.jsp?task=abfd14f7015243c69854731998d55df1) with the data set identifier MSV000085115.
Collapse
Affiliation(s)
- Yingjun Cui
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri 65211, United States
| | - Pei Liu
- Charles W. Gehrke Proteomics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Brian P Mooney
- Department of Biochemistry and Charles W. Gehrke Proteomics Center, University of Missouri, Columbia, Missouri 65211, United States
| | - Alexander W E Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri 65211, United States
| |
Collapse
|
16
|
McFarlane M, Almire F, Kean J, Donald CL, McDonald A, Wee B, Lauréti M, Varjak M, Terry S, Vazeille M, Gestuveo RJ, Dietrich I, Loney C, Failloux AB, Schnettler E, Pondeville E, Kohl A. The Aedes aegypti Domino Ortholog p400 Regulates Antiviral Exogenous Small Interfering RNA Pathway Activity and ago-2 Expression. mSphere 2020; 5:e00081-20. [PMID: 32269152 PMCID: PMC7142294 DOI: 10.1128/msphere.00081-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/13/2020] [Indexed: 02/03/2023] Open
Abstract
Arboviruses are pathogens of humans and animals. A better understanding of the interactions between these pathogens and the arthropod vectors, such as mosquitoes, that transmit them is necessary to develop novel control measures. A major antiviral pathway in the mosquito vector is the exogenous small interfering RNA (exo-siRNA) pathway, which is induced by arbovirus-derived double-stranded RNA in infected cells. Although recent work has shown the key role played by Argonaute-2 (Ago-2) and Dicer-2 (Dcr-2) in this pathway, the regulatory mechanisms that govern these pathways have not been studied in mosquitoes. Here, we show that the Domino ortholog p400 has antiviral activity against the alphavirus Semliki Forest virus (Togaviridae) both in Aedes aegypti-derived cells and in vivo Antiviral activity of p400 was also demonstrated against chikungunya virus (Togaviridae) and Bunyamwera virus (Peribunyaviridae) but not Zika virus (Flaviviridae). p400 was found to be expressed across mosquito tissues and regulated ago-2 but not dcr-2 transcript levels in A. aegypti mosquitoes. These findings provide novel insights into the regulation of an important aedine exo-siRNA pathway effector protein, Ago-2, by the Domino ortholog p400. They add functional insights to previous observations of this protein's antiviral and RNA interference regulatory activities in Drosophila melanogasterIMPORTANCE Female Aedes aegypti mosquitoes are vectors of human-infecting arthropod-borne viruses (arboviruses). In recent decades, the incidence of arthropod-borne viral infections has grown dramatically. Vector competence is influenced by many factors, including the mosquito's antiviral defenses. The exogenous small interfering RNA (siRNA) pathway is a major antiviral response restricting arboviruses in mosquitoes. While the roles of the effectors of this pathway, Argonaute-2 and Dicer-2 are well characterized, nothing is known about its regulation in mosquitoes. In this study, we demonstrate that A. aegypti p400, whose ortholog Domino in Drosophila melanogaster is a chromatin-remodeling ATPase member of the Tip60 complex, regulates siRNA pathway activity and controls ago-2 expression levels. In addition, we found p400 to have antiviral activity against different arboviruses. Therefore, our study provides new insights into the regulation of the antiviral response in A. aegypti mosquitoes.
Collapse
Affiliation(s)
- Melanie McFarlane
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Floriane Almire
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Joy Kean
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Claire L Donald
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Alma McDonald
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Bryan Wee
- Usher Institute for Population Health Sciences & Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Mathilde Lauréti
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Margus Varjak
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Sandra Terry
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Marie Vazeille
- Arboviruses and Insect Vectors Unit, Department of Virology, Institut Pasteur, Paris, France
| | - Rommel J Gestuveo
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
- Division of Biological Sciences, University of the Philippines Visayas, Miagao, Philippines
| | - Isabelle Dietrich
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Colin Loney
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Anna-Bella Failloux
- Arboviruses and Insect Vectors Unit, Department of Virology, Institut Pasteur, Paris, France
| | - Esther Schnettler
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Emilie Pondeville
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland
| |
Collapse
|
17
|
King JG. Developmental and comparative perspectives on mosquito immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 103:103458. [PMID: 31377103 DOI: 10.1016/j.dci.2019.103458] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 06/10/2023]
Abstract
Diseases spread by mosquitoes have killed more people than those spread by any other group of arthropod vectors and remain an important factor in determining global health and economic stability. The mosquito innate immune system can act to either modulate infection with human pathogens or fight off entomopathogens and increase the fitness and longevity of infected mosquitoes. While work remains towards understanding the larval immune system and the development of the mosquito immune system, it has recently become clearer that environmental factors heavily shape the developing mosquito immune system and continue to influence the adult immune system as well. The adult immune system has been well-studied and is known to involve multiple tissues and diverse molecular mechanisms. This review summarizes and synthesizes what is currently understood about the development of the mosquito immune system and includes comparisons of immune components unique to mosquitoes among the blood-feeding arthropods as well as important distinguishing factors between the anopheline and culicine mosquitoes. An explanation is included for how mosquito immunity factors into vector competence and vectorial capacity is presented along with a model for the interrelationships between nutrition, microbiome, pathogen interactions and behavior as they relate to mosquito development, immune status, adult female fitness and ultimately, vectorial capacity. Novel discoveries in the fields of mosquito ecoimmunology, neuroimmunology, and intracellular antiviral responses are highlighted.
Collapse
Affiliation(s)
- Jonas G King
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, 32 Creelman Street, Dorman 402, Mississippi State, MS 39762, USA.
| |
Collapse
|
18
|
Abdeltawab MSA, Rifaie SA, Shoeib EY, El-Latif HAA, Badawi M, Salama WH, El-Aal AAA. Insights into the impact of Ivermectin on some protein aspects linked to Culex pipiens digestion and immunity. Parasitol Res 2019; 119:55-62. [DOI: 10.1007/s00436-019-06539-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 11/03/2019] [Indexed: 02/08/2023]
|
19
|
Chikungunya virus populations experience diversity- dependent attenuation and purifying intra-vector selection in Californian Aedes aegypti mosquitoes. PLoS Negl Trop Dis 2019; 13:e0007853. [PMID: 31751338 PMCID: PMC6894883 DOI: 10.1371/journal.pntd.0007853] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 12/05/2019] [Accepted: 10/16/2019] [Indexed: 12/15/2022] Open
Abstract
Chikungunya virus (Togaviridae, Alphavirus; CHIKV) is a mosquito-borne global health threat that has been transmitted transiently in the southeastern United States. A primary CHIKV mosquito vector, Aedes aegypti, was recently established in the populous state of California, but the vector competence of Californian mosquitoes is unknown. Explosive CHIKV epidemics since 2004 have been associated with the acquisition of mosquito-adaptive mutations that enhance transmission by Ae. aegypti or Ae. albopictus. As a highly mutable RNA virus, CHIKV has the potential for extensive and rapid genetic diversification in vertebrate hosts and mosquito vectors. We previously demonstrated that expansion of CHIKV diversity in cell culture allows for greater adaptability to novel selection pressures, and that CHIKV fidelity variants are able to diversify more than wildtype (WT) CHIKV in mice. The evolution of intra-vector CHIKV populations and the correlation between CHIKV population diversity and infectivity and transmissibility in mosquitoes has not yet been studied. Here, we address these gaps in knowledge via experimental infection of Ae. aegypti from California with WT and fidelity variant CHIKV. We show that Ae. aegypti from California are highly competent vectors for CHIKV. We also report that CHIKV fidelity variants diversify more than WT in mosquitoes and exhibit attenuated infectivity at the level of the midgut. Furthermore, we demonstrate that intra-vector populations of CHIKV are subjected to purifying selection in mosquito bodies, and sequences of non-coding CHIKV regions are highly conserved. These findings will inform public health risk assessment for CHIKV in California and improve our understanding of constraints to CHIKV evolution in mosquitoes. Chikungunya virus (CHIKV) is transmitted by Aedes aegypti mosquitoes and has caused explosive epidemics in Asia and the Americas since 2004. During mosquito infection, the CHIKV genome replicates with a high mutation rate to produce virus populations with high genetic diversity that facilitate virus evolution. With this study, we address three gaps in knowledge: 1) are Ae. aegypti mosquitoes from Los Angeles, California, capable of transmitting CHIKV, 2) what effect does increased CHIKV population diversity have on virus infection and transmission by mosquitoes, and 3) are there constraints to CHIKV evolution in mosquitoes? We use oral infection of Ae. aegypti mosquitoes originating from Los Angeles, California to demonstrate high laboratory transmission competence of CHIKV. We also show that oral infection of mosquitoes with CHIKV variants that produce more diverse populations are less able to infect mosquitoes than wildtype CHIKV populations. Lastly, our study provides evidence of genome-wide and regional constraints to CHIKV evolution within Ae. aegypti mosquitoes. Our results will inform public health risk assessments for potential CHIKV introduction in southern California and advance our understanding of the role of mosquitoes in CHIKV evolution.
Collapse
|
20
|
Zika Virus Dissemination from the Midgut of Aedes aegypti is Facilitated by Bloodmeal-Mediated Structural Modification of the Midgut Basal Lamina. Viruses 2019; 11:v11111056. [PMID: 31739432 PMCID: PMC6893695 DOI: 10.3390/v11111056] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/07/2019] [Accepted: 11/12/2019] [Indexed: 12/28/2022] Open
Abstract
The arboviral disease cycle requires that key tissues in the arthropod vector become persistently infected with the virus. The midgut is the first organ in the mosquito that needs to be productively infected with an orally acquired virus. Following midgut infection, the virus then disseminates to secondary tissues including the salivary glands. Once these are productively infected, the mosquito is able to transmit the virus to a vertebrate host. Recently, we described the midgut dissemination pattern for chikungunya virus in Aedes aegypti. Here we assess the dissemination pattern in the same mosquito species for Zika virus (ZIKV), a human pathogenic virus belonging to the Flaviviridae. ZIKV infection of secondary tissues, indicative of dissemination from the midgut, was not observed before 72 h post infectious bloodmeal (pibm). Virion accumulation at the midgut basal lamina (BL) was only sporadic, although at 96–120 h pibm, virions were frequently observed between strands of the BL indicative of their dissemination. Our data suggest that ZIKV dissemination from the mosquito midgut occurs after digestion of the bloodmeal. Using gold-nanoparticles of 5 nm and 50 nm size, we show that meal ingestion leads to severe midgut tissue distention, causing the mesh width of the BL to remain enlarged after complete digestion of the meal. This could explain how ZIKV can exit the midgut via the BL after bloodmeal digestion. Ingestion of a subsequent, non-infectious bloodmeal five days after acquisition of an initial, dengue 4 virus containing bloodmeal resulted in an increased number of virions present in the midgut epithelium adjacent to the BL. Thus, subsequent bloodmeal ingestion by an infected mosquito may primarily stimulate de novo synthesis of virions leading to increased viral titers in the vector.
Collapse
|
21
|
Ghosh A, Mullapudi T, Bomanna S, Tyagi BK, Ravi V, Desai A. Understanding the mechanism of Chikungunya virus vector competence in three species of mosquitoes. MEDICAL AND VETERINARY ENTOMOLOGY 2019; 33:375-387. [PMID: 30913314 DOI: 10.1111/mve.12376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/28/2018] [Accepted: 02/25/2019] [Indexed: 06/09/2023]
Abstract
Chikungunya virus (CHIKV) is primarily transmitted by Aedes spp. mosquitoes. The present study investigated vector competence for CHIKV in Aedes aegypti and Aedes albopictus mosquitoes found in Madurai, South India. The role of receptor proteins on midguts contributing to permissiveness of CHIKV to Aedes spp. mosquitoes was also undertaken. Mosquitoes were orally infected with CHIKV DRDE-06. Infection of midguts and dissemination to heads was confirmed by immunofluorescence assay at different time points. A plaque assay was performed from mosquito homogenates at different time points to study CHIKV replication. Presence of putative CHIKV receptor proteins on mosquito midgut epithelial cells was detected by virus overlay protein binding assay (VOPBA). The identity of these proteins was established using mass spectrometry. CHIKV infection of Ae. aegypti and Ae. albopictus midguts and dissemination to heads was observed to be similar. A plaque assay performed with infected mosquito homogenates revealed that CHIKV replication dynamics was similar in Aedes sp. mosquitoes until 28 days post infection. VOPBA performed with mosquito midgut membrane proteins revealed that prohibitin could serve as a putative CHIKV receptor on Aedes mosquito midguts, whereas an absence of CHIKV binding protein/s on Culex quinquefasciatus midguts can partially explain the non-permissiveness of these mosquitoes to infection.
Collapse
Affiliation(s)
- A Ghosh
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | - T Mullapudi
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | - S Bomanna
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | - B K Tyagi
- Centre for Research in Medical Entomology, Madurai, Tamil Nadu, India
| | - V Ravi
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | - A Desai
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| |
Collapse
|
22
|
RNASeq Analysis of Aedes albopictus Mosquito Midguts after Chikungunya Virus Infection. Viruses 2019; 11:v11060513. [PMID: 31167461 PMCID: PMC6631752 DOI: 10.3390/v11060513] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 06/02/2019] [Indexed: 01/01/2023] Open
Abstract
Chikungunya virus (CHIKV) is an emerging pathogen around the world and causes significant morbidity in patients. A single amino acid mutation in the envelope protein of CHIKV has led to a shift in vector preference towards Aedesalbopictus. While mosquitoes are known to mount an antiviral immune response post-infection, molecular interactions during the course of infection at the tissue level remain largely uncharacterised. We performed whole transcriptome analysis on dissected midguts of Aedes albopictus infected with CHIKV to identify differentially expressed genes. For this, RNA was extracted at two days post-infection (2-dpi) from pooled midguts. We initially identified 25 differentially expressed genes (p-value < 0.05) when mapped to a reference transcriptome. Further, multiple differentially expressed genes were identified from a custom de novo transcriptome, which was assembled using the reads that did not align with the reference genome. Thirteen of the identified transcripts, possibly involved in immunity, were validated by qRT-PCR. Homologues of seven of these genes were also found to be significantly upregulated in Aedes aegypti midguts 2 dpi, indicating a conserved mechanism at play. These results will help us to characterise the molecular interaction between Aedes albopictus and CHIKV and can be utilised to reduce the impact of this viral infection.
Collapse
|
23
|
Kantor AM, Lin J, Wang A, Thompson DC, Franz AWE. Infection Pattern of Mayaro Virus in Aedes aegypti (Diptera: Culicidae) and Transmission Potential of the Virus in Mixed Infections With Chikungunya Virus. JOURNAL OF MEDICAL ENTOMOLOGY 2019; 56:832-843. [PMID: 30668762 PMCID: PMC6467640 DOI: 10.1093/jme/tjy241] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Indexed: 06/09/2023]
Abstract
Mayaro virus (MAYV; Togaviridae; Alphavirus) has drawn increasing attention as an arthropod-borne virus with potential to cause outbreaks among the human populations of the Western Hemisphere. In the tropical regions of Central and South America, the virus exists in sylvatic cycles between mosquitoes and primate reservoirs such as marmosets. Although forest-dwelling mosquitoes are regarded as important vectors for MAYV, it has been shown previously that the virus can infect and potentially be transmitted by the mosquitoes, Aedes aegypti and Aedes albopictus (Diptera: Culicidae). Here, we compare the infection and transmission efficiencies of two MAYV strains, IQT 4235 from Iquitos, Peru ('IQT') and the type strain of MAYV from Trinidad, TRVL 4675 ('TRVL') in two laboratory-adapted Ae. aegypti strains, Higgs White Eye and Orlando. The TRVL strain was less efficiently transmitted by both mosquito strains than MAYV IQT. Based on the full-length nucleotide sequences of the two viral genomes, we show that the TRVL prototype strain of MAYV is phylogenetically ancestral and more distantly related to the IQT strain. The TRVL strain efficiently infected wild-type Ae. albopictus from Missouri and readily disseminated in those. Considering scenarios in which natural MAYV transmission cycles may overlap with those of chikungunya virus (CHIKV; Togaviridae; Alphavirus), we assessed the effects of mixed infections of the two viruses in mosquitoes based on coinfection or superinfection. Although coinfection had no measurable effect on the transmission potential of either virus, we observed superinfection exclusion for CHIKV in MAYV-infected mosquitoes but not for MAYV in CHIKV-infected mosquitoes.
Collapse
Affiliation(s)
- Asher M Kantor
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO
| | - Jingyi Lin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO
| | - Allen Wang
- Department of Biological Sciences, University of Missouri, Columbia, MO
| | - Dana C Thompson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO
| | | |
Collapse
|
24
|
Matusali G, Colavita F, Bordi L, Lalle E, Ippolito G, Capobianchi MR, Castilletti C. Tropism of the Chikungunya Virus. Viruses 2019; 11:v11020175. [PMID: 30791607 PMCID: PMC6410217 DOI: 10.3390/v11020175] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/16/2019] [Accepted: 02/17/2019] [Indexed: 12/12/2022] Open
Abstract
Chikungunya virus (CHIKV) is a re-emerging mosquito-borne virus that displays a large cell and organ tropism, and causes a broad range of clinical symptoms in humans. It is maintained in nature through both urban and sylvatic cycles, involving mosquito vectors and human or vertebrate animal hosts. Although CHIKV was first isolated in 1953, its pathogenesis was only more extensively studied after its re-emergence in 2004. The unexpected spread of CHIKV to novel tropical and non-tropical areas, in some instances driven by newly competent vectors, evidenced the vulnerability of new territories to this infectious agent and its associated diseases. The comprehension of the exact CHIKV target cells and organs, mechanisms of pathogenesis, and spectrum of both competitive vectors and animal hosts is pivotal for the design of effective therapeutic strategies, vector control measures, and eradication actions.
Collapse
Affiliation(s)
- Giulia Matusali
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Francesca Colavita
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Licia Bordi
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Eleonora Lalle
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Giuseppe Ippolito
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Maria R Capobianchi
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| | - Concetta Castilletti
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, 00149 Rome, Italy.
| |
Collapse
|
25
|
Transcriptional Profile of Aedes aegypti Leucine-Rich Repeat Proteins in Response to Zika and Chikungunya Viruses. Int J Mol Sci 2019; 20:ijms20030615. [PMID: 30708982 PMCID: PMC6386990 DOI: 10.3390/ijms20030615] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
Aedes aegypti (L.) is the primary vector of chikungunya, dengue, yellow fever, and Zika viruses. The leucine-rich repeats (LRR)-containing domain is evolutionarily conserved in many proteins associated with innate immunity in invertebrates and vertebrates, as well as plants. We focused on the AaeLRIM1 and AaeAPL1 gene expressions in response to Zika virus (ZIKV) and chikungunya virus (CHIKV) infection using a time course study, as well as the developmental expressions in the eggs, larvae, pupae, and adults. RNA-seq analysis data provided 60 leucine-rich repeat related transcriptions in Ae. aegypti in response to Zika virus (Accession number: GSE118858, accessed on: August 22, 2018, GEO DataSets). RNA-seq analysis data showed that AaeLRIM1 (AAEL012086-RA) and AaeAPL1 (AAEL009520-RA) were significantly upregulated 2.5 and 3-fold during infection by ZIKV 7-days post infection (dpi) of an Ae. aegypti Key West strain compared to an Orlando strain. The qPCR data showed that LRR-containing proteins related genes, AaeLRIM1 and AaeAPL1, and five paralogues were expressed 100-fold lower than other nuclear genes, such as defensin, during all developmental stages examined. Together, these data provide insights into the transcription profiles of LRR proteins of Ae. aegypti during its development and in response to infection with emergent arboviruses.
Collapse
|
26
|
Sirisena PDNN, Kumar A, Sunil S. Evaluation of Aedes aegypti (Diptera: Culicidae) Life Table Attributes Upon Chikungunya Virus Replication Reveals Impact on Egg-Laying Pathways. JOURNAL OF MEDICAL ENTOMOLOGY 2018; 55:1580-1587. [PMID: 29931258 DOI: 10.1093/jme/tjy097] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Indexed: 06/08/2023]
Abstract
Arthropod-borne virus disease cycles constitute interactions among three primary players-the host, the vector, and the virus-in which the virus needs to interact with the host and the vector to establish its survival. While in the host, some arboviruses replicate aggressively, resulting in host pathogenicity, and manifest as a disease condition. These viruses more often utilize the vectors as reservoirs before they are transmitted to the host and therefore do not amplify to as large titers as they do in the hosts. In spite of this, the vector undergoes stress and activates several of its defense systems, resulting in alterations in its physiology. The present study was undertaken to evaluate the physiological changes that the mosquito vector Aedes aegypti (L.) (Diptera: Culicidae) undergoes during the replication of an arbovirus, Chikungunya virus (CHIKV). After the mosquitoes were infected with CHIKV, dissemination of the virus into various tissues and physiological parameters such as fecundity, vector mortality, egg laying, survival rate, overall fitness were monitored throughout the lifespan of the mosquitoes. Our study reveals that there is a fitness cost to the mosquitoes due to the infection of CHIKV. This fitness cost is manifested as higher mortality and low survival rate of the CHIKV-infected mosquitoes. Further evaluation revealed that the egg-laying pathway was affected, resulting in lower number of eggs. Expression analysis of six transcripts in the egg-laying pathway revealed that these transcripts were downregulated during the gonotrophic cycles in CHIKV-infected mosquitoes as compared to normal blood-fed mosquitoes.
Collapse
Affiliation(s)
- P D N N Sirisena
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Ankit Kumar
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Sujatha Sunil
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| |
Collapse
|
27
|
Kantor AM, Grant DG, Balaraman V, White TA, Franz AWE. Ultrastructural Analysis of Chikungunya Virus Dissemination from the Midgut of the Yellow Fever Mosquito, Aedes aegypti. Viruses 2018; 10:E571. [PMID: 30340365 PMCID: PMC6213114 DOI: 10.3390/v10100571] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 11/16/2022] Open
Abstract
The transmission cycle of chikungunya virus (CHIKV) requires that mosquito vectors get persistently infected with the virus, following its oral acqsuisition from a vertebrate host. The mosquito midgut is the initial organ that gets infected with orally acquired CHIKV. Following its replication in the midgut epithelium, the virus exits the midgut and infects secondary tissues including the salivary glands before being transmitted to another host. Here, we investigate the pattern of CHIKV dissemination from the midgut of Aedes aegypti at the ultrastructural level. Bloodmeal ingestion caused overstretching of the midgut basal lamina (BL), which was disrupted in areas adjacent to muscles surrounding the midgut as shown by scanning electron microscopy (SEM). Using both transmission electron microscopy (TEM) and focused ion beam scanning electron microscopy (FIB-SEM) to analyze midgut preparations, mature chikungunya (CHIK) virions were found accumulating at the BL and within strands of the BL at 24⁻32 h post-infectious bloodmeal (pibm). From 48 h pibm onwards, virions no longer congregated at the BL and became dispersed throughout the basal labyrinth of the epithelial cells. Ingestion of a subsequent, non-infectious bloodmeal caused mature virions to congregate again at the midgut BL. Our study suggests that CHIKV needs a single replication cycle in the midgut epithelium before mature virions directly traverse the midgut BL during a relatively narrow time window, within 48 h pibm.
Collapse
Affiliation(s)
- Asher M. Kantor
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (A.M.K.); (V.B.)
| | - DeAna G. Grant
- Electron Microscopy Core Facility, University of Missouri, Columbia, MO 65211, USA; (D.G.G.); (T.A.W.)
| | - Velmurugan Balaraman
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (A.M.K.); (V.B.)
| | - Tommi A. White
- Electron Microscopy Core Facility, University of Missouri, Columbia, MO 65211, USA; (D.G.G.); (T.A.W.)
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Alexander W. E. Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (A.M.K.); (V.B.)
| |
Collapse
|
28
|
Singh A, Kumar A, Yadav R, Uversky VN, Giri R. Deciphering the dark proteome of Chikungunya virus. Sci Rep 2018; 8:5822. [PMID: 29643398 PMCID: PMC5895634 DOI: 10.1038/s41598-018-23969-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 03/21/2018] [Indexed: 12/24/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus. The outbreak of CHIKV infection has been seen in many tropical and subtropical regions of the biosphere. Current reports evidenced that after outbreaks in 2005-06, the fitness of this virus propagating in Aedes albopictus enhanced due to the epistatic mutational changes in its envelope protein. In our study, we evaluated the prevalence of intrinsically disordered proteins (IDPs) and IDP regions (IDPRs) in CHIKV proteome. IDPs/IDPRs are known as members of a 'Dark Proteome' that defined as a set of polypeptide segments or whole protein without unique three-dimensional structure within the cellular milieu but with significant biological functions, such as cell cycle regulation, control of signaling pathways, and maintenance of viral proteomes. However, the intrinsically disordered aspects of CHIKV proteome and roles of IDPs/IDPRs in the pathogenic mechanism of this important virus have not been evaluated as of yet. There are no existing reports on the analysis of intrinsic disorder status of CHIKV. To fulfil this goal, we have analyzed the abundance and functionality of IDPs/IDPRs in CHIKV proteins, involved in the replication and maturation. It is likely that these IDPs/IDPRs can serve as novel targets for disorder based drug design.
Collapse
Affiliation(s)
- Ankur Singh
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh, 175005, India
| | - Ankur Kumar
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh, 175005, India
| | - Rakhi Yadav
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh, 175005, India
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, Himachal Pradesh, 175005, India.
- BioX Centre, Indian Institute of Technology Mandi, VPO Kamand, 175005, India.
| |
Collapse
|
29
|
Lim EXY, Lee WS, Madzokere ET, Herrero LJ. Mosquitoes as Suitable Vectors for Alphaviruses. Viruses 2018; 10:v10020084. [PMID: 29443908 PMCID: PMC5850391 DOI: 10.3390/v10020084] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/07/2018] [Accepted: 02/08/2018] [Indexed: 12/28/2022] Open
Abstract
Alphaviruses are arthropod-borne viruses and are predominantly transmitted via mosquito vectors. This vector preference by alphaviruses raises the important question of the determinants that contribute to vector competence. There are several tissue barriers of the mosquito that the virus must overcome in order to establish a productive infection. Of importance are the midgut, basal lamina and the salivary glands. Infection of the salivary glands is crucial for virus transmission during the mosquito’s subsequent bloodfeed. Other factors that may contribute to vector competence include the microflora and parasites present in the mosquito, environmental conditions, the molecular determinants of the virus to adapt to the vector, as well as the effect of co-infection with other viruses. Though mosquito innate immunity is a contributing factor to vector competence, it will not be discussed in this review. Detailed understanding of these factors will be instrumental in minimising transmission of alphaviral diseases.
Collapse
Affiliation(s)
- Elisa X Y Lim
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD 4215, Australia.
| | - Wai Suet Lee
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD 4215, Australia.
| | - Eugene T Madzokere
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD 4215, Australia.
| | - Lara J Herrero
- Institute for Glycomics, Griffith University, Gold Coast Campus, Southport, QLD 4215, Australia.
| |
Collapse
|
30
|
Bartholomay LC, Michel K. Mosquito Immunobiology: The Intersection of Vector Health and Vector Competence. ANNUAL REVIEW OF ENTOMOLOGY 2018; 63:145-167. [PMID: 29324042 DOI: 10.1146/annurev-ento-010715-023530] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
As holometabolous insects that occupy distinct aquatic and terrestrial environments in larval and adult stages and utilize hematophagy for nutrient acquisition, mosquitoes are subjected to a wide variety of symbiotic interactions. Indeed, mosquitoes play host to endosymbiotic, entomopathogenic, and mosquito-borne organisms, including protozoa, viruses, bacteria, fungi, fungal-like organisms, and metazoans, all of which trigger and shape innate infection-response capacity. Depending on the infection or interaction, the mosquito may employ, for example, cellular and humoral immune effectors for septic infections in the hemocoel, humoral infection responses in the midgut lumen, and RNA interference and programmed cell death for intracellular pathogens. These responses often function in concert, regardless of the infection type, and provide a robust front to combat infection. Mosquito-borne pathogens and entomopathogens overcome these immune responses, employing avoidance or suppression strategies. Burgeoning methodologies are capitalizing on this concerted deployment of immune responses to control mosquito-borne disease.
Collapse
Affiliation(s)
- Lyric C Bartholomay
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Wisconsin 53706;
| | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, Kansas 66506;
| |
Collapse
|
31
|
Dong S, Balaraman V, Kantor AM, Lin J, Grant DG, Held NL, Franz AWE. Chikungunya virus dissemination from the midgut of Aedes aegypti is associated with temporal basal lamina degradation during bloodmeal digestion. PLoS Negl Trop Dis 2017; 11:e0005976. [PMID: 28961239 PMCID: PMC5636170 DOI: 10.1371/journal.pntd.0005976] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 10/11/2017] [Accepted: 09/19/2017] [Indexed: 11/21/2022] Open
Abstract
In the mosquito, the midgut epithelium is the initial tissue to become infected with an arthropod-borne virus (arbovirus) that has been acquired from a vertebrate host along with a viremic bloodmeal. Following its replication in midgut epithelial cells, the virus needs to exit the midgut and infect secondary tissues including the salivary glands before it can be transmitted to another vertebrate host. The viral exit mechanism from the midgut, the midgut escape barrier (MEB), is poorly understood although it is an important determinant of mosquito vector competence for arboviruses. Using chikungunya virus (CHIKV) as a model in Aedes aegypti, we demonstrate that the basal lamina (BL) of the extracellular matrix (ECM) surrounding the midgut constitutes a potential barrier for the virus. The BL, predominantly consisting of collagen IV and laminin, becomes permissive during bloodmeal digestion in the midgut lumen. Bloodmeal digestion, BL permissiveness, and CHIKV dissemination are coincident with increased collagenase activity, diminished collagen IV abundance, and BL shredding in the midgut between 24–32 h post-bloodmeal. This indicates that there may be a window-of-opportunity during which the MEB in Ae. aegypti becomes permissive for CHIKV. Matrix metalloproteinases (MMPs) are the principal extracellular endopeptidases responsible for the degradation/remodeling of the ECM including the BL. We focused on Ae. aegypti (Ae)MMP1, which is expressed in midgut epithelial cells, is inducible upon bloodfeeding, and shows collagenase (gelatinase) activity. However, attempts to inhibit AeMMP activity in general or specifically that of AeMMP1 did not seem to affect its function nor produce an altered midgut escape phenotype. As an alternative, we silenced and overexpressed the Ae. aegyptitissue inhibitor of metalloproteinases (AeTIMP) in the mosquito midgut. AeTIMP was highly upregulated in the midgut during bloodmeal digestion and was able to inhibit MMP activity in vitro. Bloodmeal-inducible, midgut-specific overexpression of AeTIMP or its expression via a recombinant CHIKV significantly increased midgut dissemination rates of the virus. Possibly, AeTIMP overexpression affected BL degradation and/or restoration thereby increasing the midgut dissemination efficiency of the virus. The biological nature of the midgut escape barrier in insects for arthropod-borne viruses has been a mystery for decades. Here we show that the basal lamina (BL) surrounding the mosquito midgut acts as a barrier for chikungunya virus, an alphavirus, which has emerged in the New World hemisphere around three years ago. The barrier became permissive for the virus during digestion of a viremic bloodmeal inside the midgut lumen. Concurrent with BL permissiveness, we observed that collagen IV, a major component of the BL became temporally degraded while the BL was visibly damaged. Based on previous findings, we hypothesized that matrix metalloproteinases such as Ae. aegypti (Ae)MMP1 may be involved in BL degradation. We confirmed that recombinant AeMMP1 exhibited strong gelatinase activity, which was profoundly reduced when recombinant AeMMP1 interacted in vitro with the recombinant Ae. aegypti tissue inhibitor of metalloproteinases (AeTIMP). When transgenically overexpressing AeTIMP in an attempt to temporally inhibit general MMP activity in the mosquito midgut, we observed that the dissemination efficiency of chikungunya virus became significantly increased, while its midgut infection was not affected. It is possible that AeTIMP overexpression affected BL degradation/restoration permitting increased quantities of virus to escape from the midgut.
Collapse
Affiliation(s)
- Shengzhang Dong
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Velmurugan Balaraman
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Asher M. Kantor
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Jingyi Lin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - DeAna G. Grant
- Electron Microscopy Core Facility, University of Missouri, Columbia, Missouri, United States of America
| | - Nicole L. Held
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Alexander W. E. Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
32
|
Göertz GP, Vogels CBF, Geertsema C, Koenraadt CJM, Pijlman GP. Mosquito co-infection with Zika and chikungunya virus allows simultaneous transmission without affecting vector competence of Aedes aegypti. PLoS Negl Trop Dis 2017; 11:e0005654. [PMID: 28570693 PMCID: PMC5469501 DOI: 10.1371/journal.pntd.0005654] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/13/2017] [Accepted: 05/19/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Zika virus (ZIKV) and chikungunya virus (CHIKV) are highly pathogenic arthropod-borne viruses that are currently a serious health burden in the Americas, and elsewhere in the world. ZIKV and CHIKV co-circulate in the same geographical regions and are mainly transmitted by Aedes aegypti mosquitoes. There is a growing number of case reports of ZIKV and CHIKV co-infections in humans, but it is uncertain whether co-infection occurs via single or multiple mosquito bites. Here we investigate the potential of Ae. aegypti mosquitoes to transmit both ZIKV and CHIKV in one bite, and we assess the consequences of co-infection on vector competence. METHODOLOGY/PRINCIPAL FINDINGS First, growth curves indicated that co-infection with CHIKV negatively affects ZIKV production in mammalian, but not in mosquito cells. Next, Ae. aegypti mosquitoes were infected with ZIKV, CHIKV, or co-infected via an infectious blood meal or intrathoracic injections. Infection and transmission rates, as well as viral titers of positive mosquitoes, were determined at 14 days after blood meal or 7 days after injection. Saliva and bodies of (co-)infected mosquitoes were scored concurrently for the presence of ZIKV and/or CHIKV using a dual-colour immunofluorescence assay. The results show that orally exposed Ae. aegypti mosquitoes are highly competent, with transmission rates of up to 73% for ZIKV, 21% for CHIKV, and 12% of mosquitoes transmitting both viruses in one bite. However, simultaneous oral exposure to both viruses did not change infection and transmission rates compared to exposure to a single virus. Intrathoracic injections indicate that the selected strain of Ae. aegypti has a strong salivary gland barrier for CHIKV, but a less profound barrier for ZIKV. CONCLUSIONS/SIGNIFICANCE This study shows that Ae. aegypti can transmit both ZIKV and CHIKV via a single bite. Furthermore, co-infection of ZIKV and CHIKV does not influence the vector competence of Ae. aegypti.
Collapse
Affiliation(s)
- Giel P. Göertz
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
| | - Chantal B. F. Vogels
- Laboratory of Entomology, Wageningen University & Research, Wageningen, The Netherlands
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
| | | | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
33
|
Dong S, Behura SK, Franz AWE. The midgut transcriptome of Aedes aegypti fed with saline or protein meals containing chikungunya virus reveals genes potentially involved in viral midgut escape. BMC Genomics 2017; 18:382. [PMID: 28506207 PMCID: PMC5433025 DOI: 10.1186/s12864-017-3775-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/09/2017] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The mosquito Aedes aegypti is the primary vector for medically important arthropod-borne viruses, including chikungunya virus (CHIKV). Following oral acquisition, an arbovirus has to persistently infect several organs in the mosquito before becoming transmissible to another vertebrate host. A major obstacle an arbovirus has to overcome during its infection cycle inside the mosquito is the midgut escape barrier, representing the exit mechanism arboviruses utilize when disseminating from the midgut. To understand the transcriptomic basis of midgut escape and to reveal genes involved in the process, we conducted a comparative transcriptomic analysis of midgut samples from mosquitoes which had received a saline meal (SM) or a protein meal (PM) (not) containing CHIKV. RESULTS CHIKV which was orally acquired by a mosquito along with a SM or PM productively infected the midgut epithelium and disseminated to secondary tissues. A total of 27 RNA-Seq libraries from midguts of mosquitoes that had received PM or SM (not) containing CHIKV at 1 and 2 days post-feeding were generated and sequenced. Fewer than 80 genes responded differentially to the presence of CHIKV in midguts of mosquitoes that had acquired the virus along with SM or PM. SM feeding induced differential expression (DE) of 479 genes at day 1 and 314 genes at day 2 when compared to midguts of sugarfed mosquitoes. By comparison, PM feeding induced 6029 DE genes at day 1 and 7368 genes at day 2. Twenty-three DE genes encoding trypsins, metalloproteinases, and serine-type endopeptidases were significantly upregulated in midguts of mosquitoes at day 1 following SM or PM ingestion. Two of these genes were Ae. aegypti late trypsin (AeLT) and serine collagenase 1 precursor (AeSP1). In vitro, recombinant AeLT showed strong matrix metalloproteinase activity whereas recombinant AeSP1 did not. CONCLUSIONS By substituting a bloodmeal for SM, we identified midgut-expressed genes not involved in blood or protein digestion. These included genes coding for trypsins, metalloproteinases, and serine-type endopeptidases, which could be involved in facilitating midgut escape for arboviruses in Ae. aegypti. The presence of CHIKV in any of the ingested meals had relatively minor effects on the overall gene expression profiles in midguts.
Collapse
Affiliation(s)
- Shengzhang Dong
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Susanta K Behura
- Department of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Alexander W E Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
34
|
Mills MK, Nayduch D, McVey DS, Michel K. Functional Validation of Apoptosis Genes IAP1 and DRONC in Midgut Tissue of the Biting Midge Culicoides sonorensis (Diptera: Ceratopogonidae) by RNAi. JOURNAL OF MEDICAL ENTOMOLOGY 2017; 54:559-567. [PMID: 28399198 PMCID: PMC5502902 DOI: 10.1093/jme/tjw225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Indexed: 05/02/2023]
Abstract
Culicoides biting midges transmit multiple ruminant viruses, including bluetongue virus and epizootic hemorrhagic disease virus, causing significant economic burden worldwide. To further enhance current control techniques, understanding vector-virus interactions within the midge is critical. We developed previously a double-stranded RNA (dsRNA) delivery method to induce RNA interference (RNAi) for targeted gene knockdown in adult Culicoides sonorensis Wirth & Jones. Here, we confirm the C. sonorensis inhibitor of apoptosis 1 (CsIAP1) as an anti-apoptotic functional ortholog of IAP1 in Drosophila, identify the ortholog of the Drosophila initiator caspase DRONC (CsDRONC), and demonstrate that injection of dsRNA into the hemocoel can be used for targeted knockdown in the midgut in C. sonorensis. We observed CsIAP1 transcript reduction in whole midges, with highest transcript reduction in midgut tissues. IAP1knockdown (kd) resulted in pro-apoptotic caspase activation in midgut tissues. In IAP1kd midges, midgut tissue integrity and size were severely compromised. This phenotype, as well as reduced longevity, was partially reverted by co-RNAi suppression of CsDRONC and CsIAP1. Therefore, RNAi can be directed to the midgut of C. sonorensis, the initial site of virus infection, using dsRNA injection into the hemocoel. In addition, we provide evidence that the core apoptosis pathway is conserved in C. sonorensis and can be experimentally activated in the midgut to reduce longevity in C. sonorensis. This study thus paves the way for future reverse genetic analyses of midgut-virus interactions in C. sonorensis, including the putative antiviral properties of RNAi and apoptosis pathways.
Collapse
Affiliation(s)
- M K Mills
- Division of Biology, Kansas State University, Manhattan, KS 66506 (; )
| | - D Nayduch
- United States Department of Agriculture, Agricultural Research Service Arthropod Borne Animal Disease Research Unit, Manhattan, KS 66502
| | - D S McVey
- United States Department of Agriculture, Agricultural Research Service Arthropod Borne Animal Disease Research Unit, Manhattan, KS 66502
| | - K Michel
- Division of Biology, Kansas State University, Manhattan, KS 66506 (; )
| |
Collapse
|
35
|
Costa-da-Silva AL, Ioshino RS, de Araújo HRC, Kojin BB, Zanotto PMDA, Oliveira DBL, Melo SR, Durigon EL, Capurro ML. Laboratory strains of Aedes aegypti are competent to Brazilian Zika virus. PLoS One 2017; 12:e0171951. [PMID: 28187183 PMCID: PMC5302382 DOI: 10.1371/journal.pone.0171951] [Citation(s) in RCA: 37] [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: 08/20/2016] [Accepted: 01/28/2017] [Indexed: 11/18/2022] Open
Abstract
The Zika virus outbreaks are unprecedented human threat in relation to congenital malformations and neurological/autoimmune complications. Since this virus has high potential to spread in regions presenting the vectors, improvement in mosquito control is a top priority. Thus, Aedes aegypti laboratory strains will be fundamental to support studies in different research fields implicated on Zika-mosquito interactions which are the basis for the development of innovative control methods. In this sense, our aim was to determine the main infection aspects of a Brazilian Zika strain in reference Aedes aegypti laboratory mosquitoes. We orally exposed Rockefeller, Higgs and Rexville mosquitoes to the Brazilian ZIKV (ZIKVBR) and qRT-PCR was applied to determine the infection, dissemination and detection rates of ZIKV in the collected saliva as well as viral levels in mosquito tissues. The three strains sustain the virus development but Higgs showed significantly lower viral loads in bodies at 14 days post-infection (dpi) and the lowest prevalences in bodies and heads. The Rockefeller strain was the most susceptible at 7 dpi but similar dissemination rates were observed at 14 dpi. Although variations exist, the ZIKVBR RNA shows detectable levels in saliva of the three strains at 14 dpi but is only detected in Rockefeller at 7 dpi. Moreover, saliva samples from the three strains were confirmed to be infectious when intrathoracically injected into mosquitoes. The ZIKVBR kinetics was monitored in Rockefeller mosquitoes and virus could be identified in the heads at 4 dpi but was more consistently detected late in infection. Our study presents the first evaluation on how Brazilian Zika virus behaves in reference Aedes aegypti strains and shed light on how the infection evolves over time. Vector competence and hallmarks of the ZIKVBR development were revealed in laboratory mosquitoes, providing additional information to accelerate studies focused on ZIKV-mosquito interactions.
Collapse
Affiliation(s)
- André Luis Costa-da-Silva
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, INCT-EM, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail: (ALCS); (MLC)
| | - Rafaella Sayuri Ioshino
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, INCT-EM, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Helena Rocha Corrêa de Araújo
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, INCT-EM, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bianca Burini Kojin
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, INCT-EM, Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Stella Rezende Melo
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brasil
| | - Edison Luiz Durigon
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brasil
| | - Margareth Lara Capurro
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, INCT-EM, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail: (ALCS); (MLC)
| |
Collapse
|
36
|
Kantor AM, Dong S, Held NL, Ishimwe E, Passarelli AL, Clem RJ, Franz AW. Identification and initial characterization of matrix metalloproteinases in the yellow fever mosquito, Aedes aegypti. INSECT MOLECULAR BIOLOGY 2017; 26:113-126. [PMID: 28054419 PMCID: PMC5216420 DOI: 10.1111/imb.12275] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Aedes aegypti is a major vector for arboviruses such as dengue, chikungunya and Zika viruses. During acquisition of a viremic bloodmeal, an arbovirus infects mosquito midgut cells before disseminating to secondary tissues, including the salivary glands. Once virus is released into the salivary ducts it can be transmitted to another vertebrate host. The midgut is surrounded by a basal lamina (BL) in the extracellular matrix, consisting of a proteinaceous mesh composed of collagen IV and laminin. BL pore size exclusion limit prevents virions from passing through. Thus, the BL probably requires remodelling via enzymatic activity to enable efficient virus dissemination. Matrix metalloproteinases (MMPs) are extracellular endopeptidases that are involved in remodelling of the extracellular matrix. Here, we describe and characterize the nine Ae. aegypti encoded MMPs, AeMMPs 1-9, which share common features with other invertebrate and vertebrate MMPs. Expression profiling in Ae. aegypti revealed that Aemmp4 and Aemmp6 were upregulated during metamorphosis, whereas expression of Aemmp1 and Aemmp2 increased during bloodmeal digestion. Aemmp1 expression was also upregulated in the presence of a bloodmeal containing chikungunya virus. Using polyclonal antibodies, AeMMP1 and AeMMP2 were specifically detected in tissues associated with the mosquito midgut.
Collapse
Affiliation(s)
- Asher M. Kantor
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Shengzhang Dong
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Nicole L. Held
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Egide Ishimwe
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - A. Lorena Passarelli
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Rollie J. Clem
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Alexander W.E. Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| |
Collapse
|