1
|
Hodoameda P, Ebel GD, Mukhopadhyay S, Clem RJ. Extreme infectious titer variability in individual Aedes aegypti mosquitoes infected with Sindbis virus is associated with both differences in virus population structure and dramatic disparities in specific infectivity. PLoS Pathog 2024; 20:e1012047. [PMID: 38412195 PMCID: PMC10923411 DOI: 10.1371/journal.ppat.1012047] [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: 07/15/2023] [Revised: 03/08/2024] [Accepted: 02/13/2024] [Indexed: 02/29/2024] Open
Abstract
Variability in how individuals respond to pathogens is a hallmark of infectious disease, yet the basis for individual variation in host response is often poorly understood. The titer of infectious virus among individual mosquitoes infected with arboviruses is frequently observed to vary by several orders of magnitude in a single experiment, even when the mosquitoes are highly inbred. To better understand the basis for this titer variation, we sequenced populations of Sindbis virus (SINV) obtained from individual infected Aedes aegypti mosquitoes that, despite being from a highly inbred laboratory colony, differed in their titers of infectious virus by approximately 10,000-fold. We observed genetic differences between these virus populations that indicated the virus present in the midguts of low titer mosquitoes was less fit than that of high titer mosquitoes, possibly due to founder effects that occurred during midgut infection. Furthermore, we found dramatic differences in the specific infectivity or SI (the ratio of infectious units/viral genome equivalents) between these virus populations, with the SI of low titer mosquitoes being up to 10,000-fold lower than that of high titer mosquitoes. Despite having similar amounts of viral genomes, low titer mosquitoes appeared to contain less viral particles, suggesting that viral genomes were packaged into virions less efficiently than in high titer mosquitoes. Finally, antibiotic treatment, which has been shown to suppress mosquito antiviral immunity, caused an increase in SI. Our results indicate that the extreme variation that is observed in SINV infectious titer between individual Ae. aegypti mosquitoes is due to both genetic differences between virus populations and to differences in the proportion of genomes that are packaged into infectious particles.
Collapse
Affiliation(s)
- Peter Hodoameda
- Division of Biology, Kansas State University, Manhattan, Kansas United States of America
| | - Gregory D. Ebel
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado United States of America
| | - Suchetana Mukhopadhyay
- Department of Biology, Indiana University, Bloomington, Indiana United States of America
| | - Rollie J. Clem
- Division of Biology, Kansas State University, Manhattan, Kansas United States of America
| |
Collapse
|
2
|
Taracena-Agarwal ML, Hixson B, Nandakumar S, Girard-Mejia AP, Chen RY, Huot L, Padilla N, Buchon N. The midgut epithelium of mosquitoes adjusts cell proliferation and endoreplication to respond to physiological challenges. BMC Biol 2024; 22:22. [PMID: 38281940 PMCID: PMC10823748 DOI: 10.1186/s12915-023-01769-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 11/17/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Hematophagous mosquitoes transmit many pathogens that cause human diseases. Pathogen acquisition and transmission occur when female mosquitoes blood feed to acquire nutrients for reproduction. The midgut epithelium of mosquitoes serves as the point of entry for transmissible viruses and parasites. RESULTS We studied midgut epithelial dynamics in five major mosquito vector species by quantifying PH3-positive cells (indicative of mitotic proliferation), the incorporation of nucleotide analogs (indicative of DNA synthesis accompanying proliferation and/or endoreplication), and the ploidy (by flow cytometry) of cell populations in the posterior midgut epithelium of adult females. Our results show that the epithelial dynamics of post-emergence maturation and of mature sugar-fed guts were similar in members of the Aedes, Culex, and Anopheles genera. In the first three days post-emergence, ~ 20% of cells in the posterior midgut region of interest incorporated nucleotide analogs, concurrent with both proliferative activity and a broad shift toward higher ploidy. In mature mosquitoes maintained on sugar, an average of 3.5% of cells in the posterior midgut region of interest incorporated nucleotide analogs from five to eight days post-emergence, with a consistent presence of mitotic cells indicating constant cell turnover. Oral bacterial infection triggered a sharp increase in mitosis and nucleotide analog incorporation, suggesting that the mosquito midgut undergoes accelerated cellular turnover in response to damage. Finally, blood feeding resulted in an increase in cell proliferation, but the nature and intensity of the response varied by mosquito species and by blood source (human, bovine, avian or artificial). In An. gambiae, enterocytes appeared to reenter the cell cycle to increase ploidy after consuming blood from all sources except avian. CONCLUSIONS We saw that epithelial proliferation, differentiation, and endoreplication reshape the blood-fed gut to increase ploidy, possibly to facilitate increased metabolic activity. Our results highlight the plasticity of the midgut epithelium in mosquitoes' physiological responses to distinct challenges.
Collapse
Affiliation(s)
- M L Taracena-Agarwal
- Department of Entomology, College of Agriculture and Life Sciences, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, 14852, USA
| | - B Hixson
- Department of Entomology, College of Agriculture and Life Sciences, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, 14852, USA
| | - S Nandakumar
- Department of Entomology, College of Agriculture and Life Sciences, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, 14852, USA
| | - A P Girard-Mejia
- Grupo de Biología y Control de Vectores, Centro de Estudios en Salud, Universidad del Valle de Guatemala, Guatemala City, 01015, Guatemala
| | - R Y Chen
- Department of Entomology, College of Agriculture and Life Sciences, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, 14852, USA
| | - L Huot
- Department of Entomology, College of Agriculture and Life Sciences, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, 14852, USA
| | - N Padilla
- Grupo de Biología y Control de Vectores, Centro de Estudios en Salud, Universidad del Valle de Guatemala, Guatemala City, 01015, Guatemala
| | - N Buchon
- Department of Entomology, College of Agriculture and Life Sciences, Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY, 14852, USA.
| |
Collapse
|
3
|
Boehm EC, Jaeger AS, Ries HJ, Castañeda D, Weiler AM, Valencia CC, Weger-Lucarelli J, Ebel GD, O’Connor SL, Friedrich TC, Zamanian M, Aliota MT. Wolbachia-mediated resistance to Zika virus infection in Aedes aegypti is dominated by diverse transcriptional regulation and weak evolutionary pressures. PLoS Negl Trop Dis 2023; 17:e0011674. [PMID: 37782672 PMCID: PMC10569609 DOI: 10.1371/journal.pntd.0011674] [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: 06/26/2023] [Revised: 10/12/2023] [Accepted: 09/13/2023] [Indexed: 10/04/2023] Open
Abstract
A promising candidate for arbovirus control and prevention relies on replacing arbovirus-susceptible Aedes aegypti populations with mosquitoes that have been colonized by the intracellular bacterium Wolbachia and thus have a reduced capacity to transmit arboviruses. This reduced capacity to transmit arboviruses is mediated through a phenomenon referred to as pathogen blocking. Pathogen blocking has primarily been proposed as a tool to control dengue virus (DENV) transmission, however it works against a range of viruses, including Zika virus (ZIKV). Despite years of research, the molecular mechanisms underlying pathogen blocking still need to be better understood. Here, we used RNA-seq to characterize mosquito gene transcription dynamics in Ae. aegypti infected with the wMel strain of Wolbachia that are being released by the World Mosquito Program in Medellín, Colombia. Comparative analyses using ZIKV-infected, uninfected tissues, and mosquitoes without Wolbachia revealed that the influence of wMel on mosquito gene transcription is multifactorial. Importantly, because Wolbachia limits, but does not completely prevent, replication of ZIKV and other viruses in coinfected mosquitoes, there is a possibility that these viruses could evolve resistance to pathogen blocking. Therefore, to understand the influence of Wolbachia on within-host ZIKV evolution, we characterized the genetic diversity of molecularly barcoded ZIKV virus populations replicating in Wolbachia-infected mosquitoes and found that within-host ZIKV evolution was subject to weak purifying selection and, unexpectedly, loose anatomical bottlenecks in the presence and absence of Wolbachia. Together, these findings suggest that there is no clear transcriptional profile associated with Wolbachia-mediated ZIKV restriction, and that there is no evidence for ZIKV escape from this restriction in our system.
Collapse
Affiliation(s)
- Emma C. Boehm
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, United States of America
| | - Anna S. Jaeger
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, United States of America
| | - Hunter J. Ries
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - David Castañeda
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, United States of America
| | - Andrea M. Weiler
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Corina C. Valencia
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - James Weger-Lucarelli
- Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Gregory D. Ebel
- Colorado State University, Fort Collins, Colorado, United States of America
| | - Shelby L. O’Connor
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Thomas C. Friedrich
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Mostafa Zamanian
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Matthew T. Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, United States of America
| |
Collapse
|
4
|
Boehm EC, Jaeger AS, Ries HJ, Castañeda D, Weiler AM, Valencia CC, Weger-Lucarelli J, Ebel GD, O’Connor SL, Friedrich TC, Zamanian M, Aliota MT. Wolbachia -mediated resistance to Zika virus infection in Aedes aegypti is dominated by diverse transcriptional regulation and weak evolutionary pressures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.26.546271. [PMID: 37425681 PMCID: PMC10327090 DOI: 10.1101/2023.06.26.546271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
A promising candidate for arbovirus control and prevention relies on replacing arbovirus-susceptible Aedes aegypti populations with mosquitoes that have been colonized by the intracellular bacterium Wolbachia and thus have a reduced capacity to transmit arboviruses. This reduced capacity to transmit arboviruses is mediated through a phenomenon referred to as pathogen blocking. Pathogen blocking has primarily been proposed as a tool to control dengue virus (DENV) transmission, however it works against a range of viruses, including Zika virus (ZIKV). Despite years of research, the molecular mechanisms underlying pathogen blocking still need to be better understood. Here, we used RNA-seq to characterize mosquito gene transcription dynamics in Ae. aegypti infected with the w Mel strain of Wolbachia that are being released by the World Mosquito Program in Medellín, Colombia. Comparative analyses using ZIKV-infected, uninfected tissues, and mosquitoes without Wolbachia revealed that the influence of w Mel on mosquito gene transcription is multifactorial. Importantly, because Wolbachia limits, but does not completely prevent, replication of ZIKV and other viruses in coinfected mosquitoes, there is a possibility that these viruses could evolve resistance to pathogen blocking. Therefore, to understand the influence of Wolbachia on within-host ZIKV evolution, we characterized the genetic diversity of molecularly barcoded ZIKV virus populations replicating in Wolbachia -infected mosquitoes and found that within-host ZIKV evolution was subject to weak purifying selection and, unexpectedly, loose anatomical bottlenecks in the presence and absence of Wolbachia . Together, these findings suggest that there is no clear transcriptional profile associated with Wolbachia -mediated ZIKV restriction, and that there is no evidence for ZIKV escape from this restriction in our system. Author Summary When Wolbachia bacteria infect Aedes aegypti mosquitoes, they dramatically reduce the mosquitoes' susceptibility to infection with a range of arthropod-borne viruses, including Zika virus (ZIKV). Although this pathogen-blocking effect has been widely recognized, its mechanisms remain unclear. Furthermore, because Wolbachia limits, but does not completely prevent, replication of ZIKV and other viruses in coinfected mosquitoes, there is a possibility that these viruses could evolve resistance to Wolbachia -mediated blocking. Here, we use host transcriptomics and viral genome sequencing to examine the mechanisms of ZIKV pathogen blocking by Wolbachia and viral evolutionary dynamics in Ae. aegypti mosquitoes. We find complex transcriptome patterns that do not suggest a single clear mechanism for pathogen blocking. We also find no evidence that Wolbachia exerts detectable selective pressures on ZIKV in coinfected mosquitoes. Together our data suggest that it may be difficult for ZIKV to evolve Wolbachia resistance, perhaps due to the complexity of the pathogen blockade mechanism.
Collapse
Affiliation(s)
- Emma C. Boehm
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| | - Anna S. Jaeger
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| | - Hunter J. Ries
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, WI, United States
| | - David Castañeda
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| | - Andrea M. Weiler
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, United States
| | - Corina C. Valencia
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, United States
| | | | | | - Shelby L. O’Connor
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, United States
- Department of Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, WI, United States
| | - Thomas C. Friedrich
- Wisconsin National Primate Research Center, University of Wisconsin–Madison, Madison, WI, United States
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, WI, United States
| | - Mostafa Zamanian
- Department of Pathobiological Sciences, University of Wisconsin–Madison, Madison, WI, United States
| | - Matthew T. Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities
| |
Collapse
|
5
|
Tang X, Zhou J, Koski TM, Liu S, Zhao L, Sun J. Hypoxia-induced tracheal elasticity in vector beetle facilitates the loading of pinewood nematode. eLife 2023; 12:84621. [PMID: 36995744 PMCID: PMC10063229 DOI: 10.7554/elife.84621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/21/2023] [Indexed: 03/31/2023] Open
Abstract
Many pathogens rely on their insect vectors for transmission. Such pathogens are under selection to improve vector competence for their transmission by employing various tissue or cellular responses of vectors. However, whether pathogens can actively cause hypoxia in vectors and exploit hypoxia responses to promote their vector competence is still unknown. Fast dispersal of pinewood nematode (PWN), the causal agent for the destructive pine wilt disease and subsequent infection of pine trees, is characterized by the high vector competence of pine sawyer beetles (Monochamus spp.), and a single beetle can harbor over 200,000 PWNs in its tracheal system. Here, we demonstrate that PWN loading activates hypoxia in tracheal system of the vector beetles. Both PWN loading and hypoxia enhanced tracheal elasticity and thickened the apical extracellular matrix (aECM) of the tracheal tubes while a notable upregulated expression of a resilin-like mucin protein Muc91C was observed at the aECM layer of PWN-loaded and hypoxic tracheal tubes. RNAi knockdown of Muc91C reduced tracheal elasticity and aECM thickness under hypoxia conditions and thus decreasing PWN loading. Our study suggests a crucial role of hypoxia-induced developmental responses in shaping vector tolerance to the pathogen and provides clues for potential molecular targets to control pathogen dissemination.
Collapse
Affiliation(s)
- Xuan Tang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiao Zhou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Tuuli-Marjaana Koski
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Science/Hebei Basic Science Center for Biotic Interactions, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Shiyao Liu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lilin Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianghua Sun
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Science/Hebei Basic Science Center for Biotic Interactions, Institute of Life Science and Green Development, Hebei University, Baoding, China
| |
Collapse
|
6
|
Zhang Y, Li BX, Mao QZ, Zhuo JC, Huang HJ, Lu JB, Zhang CX, Li JM, Chen JP, Lu G. The JAK-STAT pathway promotes persistent viral infection by activating apoptosis in insect vectors. PLoS Pathog 2023; 19:e1011266. [PMID: 36928081 PMCID: PMC10069781 DOI: 10.1371/journal.ppat.1011266] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/03/2023] [Accepted: 03/04/2023] [Indexed: 03/18/2023] Open
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway is an evolutionarily conserved signaling pathway that can regulate various biological processes. However, the role of JAK-STAT pathway in the persistent viral infection in insect vectors has rarely been investigated. Here, using a system that comprised two different plant viruses, Rice stripe virus (RSV) and Rice black-streaked dwarf virus (RBSDV), as well as their insect vector small brown planthopper, we elucidated the regulatory mechanism of JAK-STAT pathway in persistent viral infection. Both RSV and RBSDV infection activated the JAK-STAT pathway and promoted the accumulation of suppressor of cytokine signaling 5 (SOCS5), an E3 ubiquitin ligase regulated by the transcription factor STAT5B. Interestingly, the virus-induced SOCS5 directly interacted with the anti-apoptotic B-cell lymphoma-2 (BCL2) to accelerate the BCL2 degradation through the 26S proteasome pathway. As a result, the activation of apoptosis facilitated persistent viral infection in their vector. Furthermore, STAT5B activation promoted virus amplification, whereas STAT5B suppression inhibited apoptosis and reduced virus accumulation. In summary, our results reveal that virus-induced JAK-STAT pathway regulates apoptosis to promote viral infection, and uncover a new regulatory mechanism of the JAK-STAT pathway in the persistent plant virus transmission by arthropod vectors.
Collapse
Affiliation(s)
- Yan Zhang
- College of Plant Protection, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Bo-Xue Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Qian-Zhuo Mao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Ji-Chong Zhuo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Hai-Jian Huang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Jia-Bao Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Chuan-Xi Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Jun-Min Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
| | - Jian-Ping Chen
- College of Plant Protection, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
- * E-mail: (J-PC); (GL)
| | - Gang Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key Laboratory of Biotechnology in Plant Protection of MARA and Zhejiang Province, Institute of Plant Virology, Ningbo University, Ningbo, China
- * E-mail: (J-PC); (GL)
| |
Collapse
|
7
|
Liu W, Cheng P, An S, Zhang K, Gong M, Zhang Z, Zhang R. Chromosome-level assembly of Culex pipiens molestus and improved reference genome of Culex pipiens pallens (Culicidae, Diptera). Mol Ecol Resour 2023; 23:486-498. [PMID: 36075571 DOI: 10.1111/1755-0998.13712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/06/2022] [Accepted: 09/05/2022] [Indexed: 01/04/2023]
Abstract
Culex pipiens molestus and Culex pipiens pallens are two distinct bioforms in the Culex pipiens complex that are important vectors of several pathogens and are widely distributed around the world. In the current study, we present a high-quality chromosome-level genome of Cx. pipiens f. molestus and describe the genetic characteristics of this genome. The assembly genome was 559.749 Mb with contig and scaffold N50 values of 200.952 Mb and 0.370 Mb, and more than 94.78% of the assembled bases were located on 3 chromosomes. A total of 19,399 protein-coding genes were predicted. Many gene families were expanded in the genome of Cx. pipiens f. molestus, particularly those of the chemosensory protein (CSP) and gustatory receptor (GR) gene families. In addition, utilizing Hi-C data, we improved the previously assembled draft genome of Cx. pipiens f. pallens, with scaffold N50 of 186.195 Mb and contig N50 of 0.749 Mb, and more than 97.02% of the assembled bases were located on three chromosomes. This reference genome provides a foundation for genome-based investigations of the unique ecological and evolutionary characteristics of Cx. pipiens f. molestus, and the findings in this study will help to elucidate the mechanisms involved in species divergence in the Culex pipiens complex.
Collapse
Affiliation(s)
- Wenjuan Liu
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Peng Cheng
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,Shandong Institute of Parasitic Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Jining, China
| | - Sha An
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Kexin Zhang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Maoqing Gong
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,Shandong Institute of Parasitic Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Jining, China
| | - Zhong Zhang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| | - Ruiling Zhang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China.,School of Basic Medical Science, Shandong First Medical University (Shandong Academy of Medical Sciences), Tai'an, China
| |
Collapse
|
8
|
Cardoso-Jaime V, Tikhe CV, Dong S, Dimopoulos G. The Role of Mosquito Hemocytes in Viral Infections. Viruses 2022; 14:v14102088. [PMID: 36298644 PMCID: PMC9608948 DOI: 10.3390/v14102088] [Citation(s) in RCA: 8] [Impact Index Per Article: 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
|
9
|
Wang J, Fan P, Wei Y, Wang J, Zou W, Zhou G, Zhong D, Zheng X. Isobaric tags for relative and absolute quantification-based proteomic analysis of host-pathogen protein interactions in the midgut of Aedes albopictus during dengue virus infection. Front Microbiol 2022; 13:990978. [PMID: 36187964 PMCID: PMC9515977 DOI: 10.3389/fmicb.2022.990978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
Aedes albopictus (Ae. albopictus), an important vector of dengue virus (DENV), is distributed worldwide. Identifying host proteins involved in flavivirus replication in Ae. albopictus and determining their natural antiviral mechanisms are critical to control virus transmission. Revealing the key proteins related to virus replication and exploring the host-pathogen interaction are of great significance in finding new pathways of the natural immune response in Ae. albopictus. Isobaric tags for relative and absolute quantification (iTRAQ) was used to perform a comparative proteomic analysis between the midgut of Ae. albopictus infected with DENV and the control. 3,419 proteins were detected, of which 162 were ≥ 1.2-fold differentially upregulated or ≤ 0.8-fold differentially downregulated (p < 0.05) during DENV infections. Differentially expressed proteins (DEPs) were mainly enriched in ubiquitin ligase complex, structural constituent of cuticle, carbohydrate metabolism, and lipid metabolism pathways. We found that one of the DEPs, a putative pupal cuticle (PC) protein could inhibit the replication of DENV and interact with the DENV-E protein. In addition, the result of immunofluorescence (IF) test showed that there was co-localization between ubiquitin carboxyl-terminal hydrolase (UCH) protein and the DENV-E protein, and virus infection reduced the level of this protein. iTRAQ-based proteomic analysis of the Ae. albopictus midgut identified dengue infection-induced upregulated and downregulated proteins. The interaction between the PC and UCH proteins in the midgut of Ae. albopictus might exert a natural antiviral mechanism in mosquito.
Collapse
Affiliation(s)
- Jiatian Wang
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Peiyang Fan
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yong Wei
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jiaqi Wang
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Weihao Zou
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guofa Zhou
- Program in Public Health, College of Health Sciences, University of California, Irvine, Irvine, CA, United States
| | - Daibin Zhong
- Program in Public Health, College of Health Sciences, University of California, Irvine, Irvine, CA, United States
| | - Xueli Zheng
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
- *Correspondence: Xueli Zheng,
| |
Collapse
|
10
|
Expressing the Pro-Apoptotic Reaper Protein via Insertion into the Structural Open Reading Frame of Sindbis Virus Reduces the Ability to Infect Aedes aegypti Mosquitoes. Viruses 2022; 14:v14092035. [PMID: 36146841 PMCID: PMC9501589 DOI: 10.3390/v14092035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Arboviruses continue to threaten a significant portion of the human population, and a better understanding is needed of the determinants of successful arbovirus infection of arthropod vectors. Avoiding apoptosis has been shown to be one such determinant. Previous work showed that a Sindbis virus (SINV) construct called MRE/rpr that expresses the Drosophila pro-apoptotic protein Reaper via a duplicated subgenomic promoter had a reduced ability to orally infect Aedes aegypti mosquitoes at 3 days post-blood meal (PBM), but this difference diminished over time as virus variants containing deletions in the inserted reaper gene rapidly predominated. In order to further clarify the effect of midgut apoptosis on disseminated infection in Ae. aegypti, we constructed MRE/rprORF, a version of SINV containing reaper inserted into the structural open reading frame (ORF) as an in-frame fusion. MRE/rprORF successfully expressed Reaper, replicated similarly to MRE/rpr in cell lines, induced apoptosis in cultured cells, and caused increased effector caspase activity in mosquito midgut tissue. Mosquitoes that fed on blood containing MRE/rprORF developed significantly less midgut and disseminated infection when compared to MRE/rpr or a control virus up to at least 7 days PBM, when less than 50% of mosquitoes that ingested MRE/rprORF had detectable disseminated infection, compared with around 80% or more of mosquitoes fed with MRE/rpr or control virus. However, virus titer in the minority of mosquitoes that became infected with MRE/rprORF was not significantly different from control virus. Deep sequencing of virus populations from ten mosquitoes infected with MRE/rprORF indicated that the reaper insert was stable, with only a small number of point mutations and no deletions being observed at frequencies greater than 1%. Our results indicate that expression of Reaper by this method significantly reduces infection prevalence, but if infection is established then Reaper expression has limited ability to continue to suppress replication.
Collapse
|
11
|
Investigation of Biological Factors Contributing to Individual Variation in Viral Titer after Oral Infection of Aedes aegypti Mosquitoes by Sindbis Virus. Viruses 2022; 14:v14010131. [PMID: 35062335 PMCID: PMC8780610 DOI: 10.3390/v14010131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/21/2022] Open
Abstract
The mechanisms involved in determining arbovirus vector competence, or the ability of an arbovirus to infect and be transmitted by an arthropod vector, are still incompletely understood. It is well known that vector competence for a particular arbovirus can vary widely among different populations of a mosquito species, which is generally attributed to genetic differences between populations. What is less understood is the considerable variability (up to several logs) that is routinely observed in the virus titer between individual mosquitoes in a single experiment, even in mosquitoes from highly inbred lines. This extreme degree of variation in the virus titer between individual mosquitoes has been largely ignored in past studies. We investigated which biological factors can affect titer variation between individual mosquitoes of a laboratory strain of Aedes aegypti, the Orlando strain, after Sindbis virus infection. Greater titer variation was observed after oral versus intrathoracic infection, suggesting that the midgut barrier contributes to titer variability. Among the other factors tested, only the length of the incubation period affected the degree of titer variability, while virus strain, mosquito strain, mosquito age, mosquito weight, amount of blood ingested, and virus concentration in the blood meal had no discernible effect. We also observed differences in culture adaptability and in the ability to orally infect mosquitoes between virus populations obtained from low and high titer mosquitoes, suggesting that founder effects may affect the virus titer in individual mosquitoes, although other explanations also remain possible.
Collapse
|
12
|
Infection of Aedes aegypti Mosquitoes with Midgut-Attenuated Sindbis Virus Reduces, but Does Not Eliminate, Disseminated Infection. J Virol 2021; 95:e0013621. [PMID: 33853958 DOI: 10.1128/jvi.00136-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Arboviruses are transmitted by specific vectors, and the reasons for this specificity are not fully understood. One contributing factor is the existence of tissue barriers within the vector such as the midgut escape barrier. We used microRNA (miRNA) targeting of Sindbis virus (SINV) to study how replication in midgut cells contributes to overcoming this barrier in the mosquito Aedes aegypti. SINV constructs were designed to be attenuated specifically in midgut cells by inserting binding sites for midgut-specific miRNAs into either the 3' untranslated region (MRE3'miRT) or the structural open reading frame (MRE-ORFmiRT) of the SINV genome. Both miRNA-targeted viruses replicated less efficiently than control viruses in the presence of these miRNAs. When mosquitoes were given infectious blood meals containing miRNA-targeted viruses, only around 20% (MRE3'miRT) or 40% (MRE-ORFmiRT) of mosquitoes developed disseminated infection. In contrast, dissemination occurred in almost all mosquitoes fed control viruses. Deep sequencing of virus populations from individual mosquitoes ruled out selection for mutations in the inserted target sequences as the cause for dissemination in these mosquitoes. In mosquitoes that became infected with miRNA-targeted viruses, titers were equivalent to those of mosquitoes infected with control virus in both the midgut and the carcass, and there was no evidence of a threshold titer necessary for dissemination. Instead, it appeared that if infection was successfully established in the midgut, replication and dissemination were largely normal. Our results support the hypothesis that replication is an important factor in allowing SINV to overcome the midgut escape barrier but hint that other factors are also likely involved. IMPORTANCE When a mosquito ingests an arbovirus during a blood meal, the arbovirus must escape from the midgut of the vector and infect the salivary glands in order to be transmitted to a new host. We used tissue-specific miRNA targeting to examine the requirement for Sindbis virus (SINV) to replicate in midgut epithelium in order to cause disseminated infection in the mosquito Aedes aegypti. Our results indicate that specifically reducing the ability of SINV to replicate in the mosquito midgut reduces its overall ability to establish infection in the mosquito, but if infection is established, replication and dissemination occur normally. These results are consistent with an importance for replication in the midgut epithelium in aiding arboviruses in crossing the midgut barrier.
Collapse
|
13
|
Liu Y, Garron TM, Chang Q, Su Z, Zhou C, Qiu Y, Gong EC, Zheng J, Yin YW, Ksiazek T, Brasel T, Jin Y, Boor P, Comer JE, Gong B. Cell-Type Apoptosis in Lung during SARS-CoV-2 Infection. Pathogens 2021; 10:pathogens10050509. [PMID: 33922476 PMCID: PMC8145065 DOI: 10.3390/pathogens10050509] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/12/2021] [Accepted: 04/21/2021] [Indexed: 12/23/2022] Open
Abstract
The SARS-CoV-2 pandemic has inspired renewed interest in understanding the fundamental pathology of acute respiratory distress syndrome (ARDS) following infection. However, the pathogenesis of ARDS following SRAS-CoV-2 infection remains largely unknown. In the present study, we examined apoptosis in postmortem lung sections from COVID-19 patients and in lung tissues from a non-human primate model of SARS-CoV-2 infection, in a cell-type manner, including type 1 and 2 alveolar cells and vascular endothelial cells (ECs), macrophages, and T cells. Multiple-target immunofluorescence assays and Western blotting suggest both intrinsic and extrinsic apoptotic pathways are activated during SARS-CoV-2 infection. Furthermore, we observed that SARS-CoV-2 fails to induce apoptosis in human bronchial epithelial cells (i.e., BEAS2B cells) and primary human umbilical vein endothelial cells (HUVECs), which are refractory to SARS-CoV-2 infection. However, infection of co-cultured Vero cells and HUVECs or Vero cells and BEAS2B cells with SARS-CoV-2 induced apoptosis in both Vero cells and HUVECs/BEAS2B cells but did not alter the permissiveness of HUVECs or BEAS2B cells to the virus. Post-exposure treatment of the co-culture of Vero cells and HUVECs with a novel non-cyclic nucleotide small molecule EPAC1-specific activator reduced apoptosis in HUVECs. These findings may help to delineate a novel insight into the pathogenesis of ARDS following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yakun Liu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.L.); (Q.C.); (Z.S.); (C.Z.); (Y.Q.); (E.C.G.); (T.K.)
| | - Tania M. Garron
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (T.M.G.); (T.B.)
| | - Qing Chang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.L.); (Q.C.); (Z.S.); (C.Z.); (Y.Q.); (E.C.G.); (T.K.)
| | - Zhengchen Su
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.L.); (Q.C.); (Z.S.); (C.Z.); (Y.Q.); (E.C.G.); (T.K.)
| | - Changcheng Zhou
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.L.); (Q.C.); (Z.S.); (C.Z.); (Y.Q.); (E.C.G.); (T.K.)
| | - Yuan Qiu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.L.); (Q.C.); (Z.S.); (C.Z.); (Y.Q.); (E.C.G.); (T.K.)
| | - Eric C. Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.L.); (Q.C.); (Z.S.); (C.Z.); (Y.Q.); (E.C.G.); (T.K.)
| | - Junying Zheng
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Y. Whitney Yin
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Thomas Ksiazek
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.L.); (Q.C.); (Z.S.); (C.Z.); (Y.Q.); (E.C.G.); (T.K.)
| | - Trevor Brasel
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (T.M.G.); (T.B.)
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, MA 02118, USA;
| | - Paul Boor
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.L.); (Q.C.); (Z.S.); (C.Z.); (Y.Q.); (E.C.G.); (T.K.)
- Correspondence: (P.B.); (J.E.C.); (B.G.); Tel.: +1-409-772-2813 (P.B.); +1-409-266-6915 (J.E.C.); +1-409-266-6682 (B.G.)
| | - Jason E. Comer
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (T.M.G.); (T.B.)
- Correspondence: (P.B.); (J.E.C.); (B.G.); Tel.: +1-409-772-2813 (P.B.); +1-409-266-6915 (J.E.C.); +1-409-266-6682 (B.G.)
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (Y.L.); (Q.C.); (Z.S.); (C.Z.); (Y.Q.); (E.C.G.); (T.K.)
- Correspondence: (P.B.); (J.E.C.); (B.G.); Tel.: +1-409-772-2813 (P.B.); +1-409-266-6915 (J.E.C.); +1-409-266-6682 (B.G.)
| |
Collapse
|
14
|
Zhao W, Zhu J, Lu H, Zhu J, Jiang F, Wang W, Luo L, Kang L, Cui F. The nucleocapsid protein of rice stripe virus in cell nuclei of vector insect regulates viral replication. Protein Cell 2021; 13:360-378. [PMID: 33675514 PMCID: PMC7936609 DOI: 10.1007/s13238-021-00822-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/08/2021] [Indexed: 01/05/2023] Open
Abstract
Rice stripe virus (RSV) transmitted by the small brown planthopper causes severe rice yield losses in Asian countries. Although viral nuclear entry promotes viral replication in host cells, whether this phenomenon occurs in vector cells remains unknown. Therefore, in this study, we systematically evaluated the presence and roles of RSV in the nuclei of vector insect cells. We observed that the nucleocapsid protein (NP) and viral genomic RNAs were partially transported into vector cell nuclei by utilizing the importin α nuclear transport system. When blocking NP nuclear localization, cytoplasmic RSV accumulation significantly increased. In the vector cell nuclei, NP bound the transcription factor YY1 and affected its positive regulation to FAIM. Subsequently, decreased FAIM expression triggered an antiviral caspase-dependent apoptotic reaction. Our results reveal that viral nuclear entry induces completely different immune effects in vector and host cells, providing new insights into the balance between viral load and the immunity pressure in vector insects.
Collapse
Affiliation(s)
- Wan Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junjie Zhu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hong Lu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiaming Zhu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fei Jiang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lan Luo
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Le Kang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Feng Cui
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
15
|
Clustered rapid induction of apoptosis limits ZIKV and DENV-2 proliferation in the midguts of Aedes aegypti. Commun Biol 2021; 4:69. [PMID: 33452408 PMCID: PMC7810730 DOI: 10.1038/s42003-020-01614-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 12/15/2020] [Indexed: 11/29/2022] Open
Abstract
Inter-host transmission of pathogenic arboviruses such as dengue virus (DENV) and Zika virus (ZIKV) requires systemic infection of the mosquito vector. Successful systemic infection requires initial viral entry and proliferation in the midgut cells of the mosquito followed by dissemination to secondary tissues and eventual entry into salivary glands1. Lack of arbovirus proliferation in midgut cells has been observed in several Aedes aegypti strains2, but the midgut antiviral responses underlying this phenomenon are not yet fully understood. We report here that there is a rapid induction of apoptosis (RIA) in the Aedes aegypti midgut epithelium within 2 hours of infection with DENV-2 or ZIKV in both in vivo blood-feeding and ex vivo midgut infection models. Inhibition of RIA led to increased virus proliferation in the midgut, implicating RIA as an innate immune mechanism mediating midgut infection in this mosquito vector. Ayers et al. report rapid induction of apoptosis in the midgut of Aedes aegypti mosquitoes within 2 hours of infection by dengue and Zika viruses, and find that inhibiting apoptosis led to increased virus proliferation in the midgut. These results suggest rapid induction of apoptosis as an innate immune mechanism mediating midgut infection in this mosquito vector.
Collapse
|
16
|
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
|
17
|
Infection of Mammals and Mosquitoes by Alphaviruses: Involvement of Cell Death. Cells 2020; 9:cells9122612. [PMID: 33291372 PMCID: PMC7762023 DOI: 10.3390/cells9122612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
Alphaviruses, such as the chikungunya virus, are emerging and re-emerging viruses that pose a global public health threat. They are transmitted by blood-feeding arthropods, mainly mosquitoes, to humans and animals. Although alphaviruses cause debilitating diseases in mammalian hosts, it appears that they have no pathological effect on the mosquito vector. Alphavirus/host interactions are increasingly studied at cellular and molecular levels. While it seems clear that apoptosis plays a key role in some human pathologies, the role of cell death in determining the outcome of infections in mosquitoes remains to be fully understood. Here, we review the current knowledge on alphavirus-induced regulated cell death in hosts and vectors and the possible role they play in determining tolerance or resistance of mosquitoes.
Collapse
|
18
|
A Hyperactive Kunjin Virus NS3 Helicase Mutant Demonstrates Increased Dissemination and Mortality in Mosquitoes. J Virol 2020; 94:JVI.01021-20. [PMID: 32699093 DOI: 10.1128/jvi.01021-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/19/2020] [Indexed: 01/07/2023] Open
Abstract
The unwinding of double-stranded RNA intermediates is critical for the replication and packaging of flavivirus RNA genomes. This unwinding activity is achieved by the ATP-dependent nonstructural protein 3 (NS3) helicase. In previous studies, we investigated the mechanism of energy transduction between the ATP and RNA binding pockets using molecular dynamics simulations and enzymatic characterization. Our data corroborated the hypothesis that motif V is a communication hub for this energy transduction. More specifically, mutations T407A and S411A in motif V exhibit a hyperactive helicase phenotype, leading to the regulation of translocation and unwinding during replication. However, the effect of these mutations on viral infection in cell culture and in vivo is not well understood. Here, we investigated the role of motif V in viral replication using West Nile virus (Kunjin subtype) T407A and S411A mutants (T407A and S411A Kunjin, respectively) in cell culture and in vivo We were able to recover S411A Kunjin but unable to recover T407A Kunjin. Our results indicated that S411A Kunjin decreased viral infection and increased cytopathogenicity in cell culture compared to wild-type (WT) Kunjin. Similarly, decreased infection rates in surviving S411A Kunjin-infected Culex quinquefasciatus mosquitoes were observed, but S411A Kunjin infection resulted in increased mortality compared to WT Kunjin infection. Additionally, S411A Kunjin infection increased viral dissemination and saliva positivity rates in surviving mosquitoes compared to WT Kunjin infection. These data suggest that S411A Kunjin increases viral pathogenesis in mosquitoes. Overall, these data indicate that NS3 motif V may play a role in the pathogenesis, dissemination, and transmission efficiency of Kunjin virus.IMPORTANCE Kunjin and West Nile viruses belong to the arthropod-borne flaviviruses, which can result in severe symptoms, including encephalitis, meningitis, and death. Flaviviruses have expanded into new populations and emerged as novel pathogens repeatedly in recent years, demonstrating that they remain a global threat. Currently, there are no approved antiviral therapeutics against either Kunjin or West Nile viruses. Thus, there is a pressing need for understanding the pathogenesis of these viruses in humans. In this study, we investigated the role of the Kunjin virus helicase on infection in cell culture and in vivo This work provides new insight into how flaviviruses control pathogenesis and mosquito transmission through the nonstructural protein 3 helicase.
Collapse
|
19
|
Chowdhury A, Modahl CM, Tan ST, Wong Wei Xiang B, Missé D, Vial T, Kini RM, Pompon JF. JNK pathway restricts DENV2, ZIKV and CHIKV infection by activating complement and apoptosis in mosquito salivary glands. PLoS Pathog 2020; 16:e1008754. [PMID: 32776975 PMCID: PMC7444518 DOI: 10.1371/journal.ppat.1008754] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 08/20/2020] [Accepted: 06/26/2020] [Indexed: 11/18/2022] Open
Abstract
Arbovirus infection of Aedes aegypti salivary glands (SGs) determines transmission. However, there is a dearth of knowledge on SG immunity. Here, we characterized SG immune response to dengue, Zika and chikungunya viruses using high-throughput transcriptomics. We also describe a transcriptomic response associated to apoptosis, blood-feeding and lipid metabolism. The three viruses differentially regulate components of Toll, Immune deficiency (IMD) and c-Jun N- terminal Kinase (JNK) pathways. However, silencing of the Toll and IMD pathway components showed variable effects on SG infection by each virus. In contrast, regulation of the JNK pathway produced consistent responses in both SGs and midgut. Infection by the three viruses increased with depletion of the activator Kayak and decreased with depletion of the negative regulator Puckered. Virus-induced JNK pathway regulates the complement factor, Thioester containing protein-20 (TEP20), and the apoptosis activator, Dronc, in SGs. Individual and co-silencing of these genes demonstrate their antiviral effects and that both may function together. Co-silencing either TEP20 or Dronc with Puckered annihilates JNK pathway antiviral effect. Upon infection in SGs, TEP20 induces antimicrobial peptides (AMPs), while Dronc is required for apoptosis independently of TEP20. In conclusion, we revealed the broad antiviral function of JNK pathway in SGs and showed that it is mediated by a TEP20 complement and Dronc-induced apoptosis response. These results expand our understanding of the immune arsenal that blocks arbovirus transmission. Arboviral diseases caused by dengue (DENV), Zika (ZIKV) and chikungunya (CHIKV) viruses are responsible for large number of death and debilitation around the world. These viruses are transmitted to humans by the mosquito vector, Aedes aegypti. During the bites, infected salivary glands (SGs) release saliva containing viruses, which initiate human infection. As the tissue where transmitted viruses are produced, SG infection is a key determinant of transmission. To bridge the knowledge gap in vector-virus molecular interactions in SGs, we describe the transcriptome after DENV, ZIKV and CHIKV infection using RNA-sequencing and characterized the immune response in this tissue. Our study reveals the broad antiviral function of c-Jun N-terminal kinase (JNK) pathway against DENV, ZIKV and CHIKV in SGs. We further show that it is mediated by the complement system and apoptosis, identifying the mechanism. Our study adds the JNK pathway to the immune arsenal that can be harnessed to engineer refractory vectors.
Collapse
Affiliation(s)
- Avisha Chowdhury
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Cassandra M. Modahl
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Siok Thing Tan
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | - Dorothée Missé
- MIVEGEC, IRD, CNRS, Univ. Montpellier, Montpellier, France
| | - Thomas Vial
- Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - R. Manjunatha Kini
- Department of Biological Sciences, National University of Singapore, Singapore
- * E-mail: (RMK); (JFP)
| | - Julien Francis Pompon
- Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- MIVEGEC, IRD, CNRS, Univ. Montpellier, Montpellier, France
- * E-mail: (RMK); (JFP)
| |
Collapse
|
20
|
Yap TF, Liu Z, Shveda RA, Preston DJ. A predictive model of the temperature-dependent inactivation of coronaviruses. APPLIED PHYSICS LETTERS 2020; 117:060601. [PMID: 32817726 PMCID: PMC7428726 DOI: 10.1063/5.0020782] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 07/30/2020] [Indexed: 05/06/2023]
Abstract
The COVID-19 pandemic has stressed healthcare systems and supply lines, forcing medical doctors to risk infection by decontaminating and reusing single-use personal protective equipment. The uncertain future of the pandemic is compounded by limited data on the ability of the responsible virus, SARS-CoV-2, to survive across various climates, preventing epidemiologists from accurately modeling its spread. However, a detailed thermodynamic analysis of experimental data on the inactivation of SARS-CoV-2 and related coronaviruses can enable a fundamental understanding of their thermal degradation that will help model the COVID-19 pandemic and mitigate future outbreaks. This work introduces a thermodynamic model that synthesizes existing data into an analytical framework built on first principles, including the rate law for a first-order reaction and the Arrhenius equation, to accurately predict the temperature-dependent inactivation of coronaviruses. The model provides much-needed thermal decontamination guidelines for personal protective equipment, including masks. For example, at 70 °C, a 3-log (99.9%) reduction in virus concentration can be achieved, on average, in 3 min (under the same conditions, a more conservative decontamination time of 39 min represents the upper limit of a 95% interval) and can be performed in most home ovens without reducing the efficacy of typical N95 masks as shown in recent experimental reports. This model will also allow for epidemiologists to incorporate the lifetime of SARS-CoV-2 as a continuous function of environmental temperature into models forecasting the spread of the pandemic across different climates and seasons.
Collapse
Affiliation(s)
- Te Faye Yap
- Department of Mechanical Engineering, Rice University, 6100 Main Street, Houston, Texas 77005, USA
| | - Zhen Liu
- Department of Mechanical Engineering, Rice University, 6100 Main Street, Houston, Texas 77005, USA
| | - Rachel A. Shveda
- Department of Mechanical Engineering, Rice University, 6100 Main Street, Houston, Texas 77005, USA
| | - Daniel J. Preston
- Department of Mechanical Engineering, Rice University, 6100 Main Street, Houston, Texas 77005, USA
| |
Collapse
|
21
|
González-González A, Wayne ML. Immunopathology and immune homeostasis during viral infection in insects. Adv Virus Res 2020; 107:285-314. [PMID: 32711732 DOI: 10.1016/bs.aivir.2020.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Organisms clear infections by mounting an immune response that is normally turned off once the pathogens have been cleared. However, sometimes this immune response is not properly or timely arrested, resulting in the host damaging itself. This immune dysregulation may be referred to as immunopathology. While our knowledge of immune and metabolic pathways in insects, particularly in response to viral infections, is growing, little is known about the mechanisms that regulate this immune response and hence little is known about immunopathology in this important and diverse group of organisms. In this chapter we focus both on documenting the molecular mechanisms described involved in restoring immune homeostasis in insects after viral infections and on identifying potential mechanisms for future investigation. We argue that learning about the immunopathological consequences of an improperly regulated immune response in insects will benefit both insect and human health.
Collapse
Affiliation(s)
| | - Marta L Wayne
- Department of Biology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
22
|
Ferreira PG, Tesla B, Horácio ECA, Nahum LA, Brindley MA, de Oliveira Mendes TA, Murdock CC. Temperature Dramatically Shapes Mosquito Gene Expression With Consequences for Mosquito-Zika Virus Interactions. Front Microbiol 2020; 11:901. [PMID: 32595607 PMCID: PMC7303344 DOI: 10.3389/fmicb.2020.00901] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
Vector-borne flaviviruses are emerging threats to human health. For successful transmission, the virus needs to efficiently enter mosquito cells and replicate within and escape several tissue barriers while mosquitoes elicit major transcriptional responses to flavivirus infection. This process will be affected not only by the specific mosquito-pathogen pairing but also by variation in key environmental variables such as temperature. Thus far, few studies have examined the molecular responses triggered by temperature and how these responses modify infection outcomes, despite substantial evidence showing strong relationships between temperature and transmission in a diversity of systems. To define the host transcriptional changes associated with temperature variation during the early infection process, we compared the transcriptome of mosquito midgut samples from mosquitoes exposed to Zika virus (ZIKV) and non-exposed mosquitoes housed at three different temperatures (20, 28, and 36°C). While the high-temperature samples did not show significant changes from those with standard rearing conditions (28°C) 48 h post-exposure, the transcriptome profile of mosquitoes housed at 20°C was dramatically different. The expression of genes most altered by the cooler temperature involved aspects of blood-meal digestion, ROS metabolism, and mosquito innate immunity. Further, we did not find significant differences in the viral RNA copy number between 24 and 48 h post-exposure at 20°C, suggesting that ZIKV replication is limited by cold-induced changes to the mosquito midgut environment. In ZIKV-exposed mosquitoes, vitellogenin, a lipid carrier protein, was most up-regulated at 20°C. Our results provide a deeper understanding of the temperature-triggered transcriptional changes in Aedes aegypti and can be used to further define the molecular mechanisms driven by environmental temperature variation.
Collapse
Affiliation(s)
| | - Blanka Tesla
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Elvira Cynthia Alves Horácio
- René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Brazil.,Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Laila Alves Nahum
- René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Brazil.,Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Promove College of Technology, Belo Horizonte, Brazil
| | - Melinda Ann Brindley
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| | | | - Courtney Cuinn Murdock
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States.,Odum School of Ecology, University of Georgia, Athens, GA, United States.,Center for the Ecology of Infectious Diseases, University of Georgia, Athens, GA, United States.,Center for Emerging and Global Tropical Diseases, University of Georgia, Athens, GA, United States.,River Basin Center, University of Georgia, Athens, GA, United States.,Department of Entomology, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
23
|
Slonchak A, Hugo LE, Freney ME, Hall-Mendelin S, Amarilla AA, Torres FJ, Setoh YX, Peng NYG, Sng JDJ, Hall RA, van den Hurk AF, Devine GJ, Khromykh AA. Zika virus noncoding RNA suppresses apoptosis and is required for virus transmission by mosquitoes. Nat Commun 2020; 11:2205. [PMID: 32371874 PMCID: PMC7200751 DOI: 10.1038/s41467-020-16086-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 04/12/2020] [Indexed: 12/27/2022] Open
Abstract
Flaviviruses, including Zika virus (ZIKV), utilise host mRNA degradation machinery to produce subgenomic flaviviral RNA (sfRNA). In mammalian hosts, this noncoding RNA facilitates replication and pathogenesis of flaviviruses by inhibiting IFN-signalling, whereas the function of sfRNA in mosquitoes remains largely elusive. Herein, we conduct a series of in vitro and in vivo experiments to define the role of ZIKV sfRNA in infected Aedes aegypti employing viruses deficient in production of sfRNA. We show that sfRNA-deficient viruses have reduced ability to disseminate and reach saliva, thus implicating the role for sfRNA in productive infection and transmission. We also demonstrate that production of sfRNA alters the expression of mosquito genes related to cell death pathways, and prevents apoptosis in mosquito tissues. Inhibition of apoptosis restored replication and transmission of sfRNA-deficient mutants. Hence, we propose anti-apoptotic activity of sfRNA as the mechanism defining its role in ZIKV transmission. The function on subgenomic flaviviral RNA (sfRNA) in the mosquito vector is not well understood. Here, Slonchak et al. show that sfRNA affects virus-induced apoptosis and dissemination of ZIKV in Aedes aegypti mosquitoes, suggesting a role of sfRNA in Zika virus replication and transmission.
Collapse
Affiliation(s)
- Andrii Slonchak
- The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Leon E Hugo
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Morgan E Freney
- The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Sonja Hall-Mendelin
- Public Health Virology, Forensic and Scientific Services, Department of Health, Queensland Government, Brisbane, QLD, 4108, Australia
| | | | | | - Yin Xiang Setoh
- The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Nias Y G Peng
- The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Julian D J Sng
- The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Roy A Hall
- The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Andrew F van den Hurk
- Public Health Virology, Forensic and Scientific Services, Department of Health, Queensland Government, Brisbane, QLD, 4108, Australia
| | - Gregor J Devine
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | | |
Collapse
|
24
|
Oliveira JH, Bahia AC, Vale PF. How are arbovirus vectors able to tolerate infection? DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 103:103514. [PMID: 31585195 DOI: 10.1016/j.dci.2019.103514] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/20/2019] [Accepted: 10/01/2019] [Indexed: 06/10/2023]
Abstract
One of the defining features of mosquito vectors of arboviruses such as Dengue and Zika is their ability to tolerate high levels of virus proliferation without suffering significant pathology. This adaptation is central to vector competence and disease spread. The molecular mechanisms, pathways, cellular and metabolic adaptations responsible for mosquito disease tolerance are still largely unknown and may represent effective ways to control mosquito populations and prevent arboviral diseases. In this review article, we describe the key link between disease tolerance and pathogen transmission, and how vector control methods may benefit by focusing efforts on dissecting the mechanisms underlying mosquito tolerance of arboviral infections. We briefly review recent work investigating tolerance mechanisms in other insects, describe the state of the art regarding the mechanisms of disease tolerance in mosquitos, and highlight the emerging role of gut microbiota in mosquito immunity and disease tolerance.
Collapse
Affiliation(s)
- José Henrique Oliveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianopolis, SC, Brazil.
| | - Ana Cristina Bahia
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Pedro F Vale
- Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
25
|
Chen Q, Wei T. Cell Biology During Infection of Plant Viruses in Insect Vectors and Plant Hosts. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2020; 33:18-25. [PMID: 31729283 DOI: 10.1094/mpmi-07-19-0184-cr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Plant viruses typically cause severe pathogenicity in plants, even resulting in the death of plants. Many pathogenic plant viruses are transmitted in a persistent manner via insect vectors. Interestingly, unlike in the plant hosts, persistent viruses are either nonpathogenic or show limited pathogenicity in their insect vectors, while taking advantage of the cellular machinery of insect vectors for completing their life cycles. This review discusses why persistent plant viruses are nonpathogenic or have limited pathogenicity to their insect vectors while being pathogenic to plants hosts. Current advances in cell biology of virus-insect vector interactions are summarized, including virus-induced inclusion bodies, changes of insect cellular ultrastructure, and immune response of insects to the viruses, especially autophagy and apoptosis. The corresponding findings of virus-plant interactions are compared. An integrated view of the balance strategy achieved by the interaction between viral attack and the immune response of insect is presented. Finally, we outline progress gaps between virus-insect and virus-plant interactions, thus highlighting the contributions of cultured cells to the cell biology of virus-insect interactions. Furthermore, future prospects of studying the cell biology of virus-vector interactions are presented.
Collapse
Affiliation(s)
- Qian Chen
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, PR China
| | - Taiyun Wei
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, PR China
| |
Collapse
|
26
|
Mehrbod P, Ande SR, Alizadeh J, Rahimizadeh S, Shariati A, Malek H, Hashemi M, Glover KKM, Sher AA, Coombs KM, Ghavami S. The roles of apoptosis, autophagy and unfolded protein response in arbovirus, influenza virus, and HIV infections. Virulence 2019; 10:376-413. [PMID: 30966844 PMCID: PMC6527025 DOI: 10.1080/21505594.2019.1605803] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/16/2019] [Accepted: 04/08/2019] [Indexed: 12/11/2022] Open
Abstract
Virus infection induces different cellular responses in infected cells. These include cellular stress responses like autophagy and unfolded protein response (UPR). Both autophagy and UPR are connected to programed cell death I (apoptosis) in chronic stress conditions to regulate cellular homeostasis via Bcl2 family proteins, CHOP and Beclin-1. In this review article we first briefly discuss arboviruses, influenza virus, and HIV and then describe the concepts of apoptosis, autophagy, and UPR. Finally, we focus upon how apoptosis, autophagy, and UPR are involved in the regulation of cellular responses to arboviruses, influenza virus and HIV infections. Abbreviation: AIDS: Acquired Immunodeficiency Syndrome; ATF6: Activating Transcription Factor 6; ATG6: Autophagy-specific Gene 6; BAG3: BCL Associated Athanogene 3; Bak: BCL-2-Anatagonist/Killer1; Bax; BCL-2: Associated X protein; Bcl-2: B cell Lymphoma 2x; BiP: Chaperon immunoglobulin heavy chain binding Protein; CARD: Caspase Recruitment Domain; cART: combination Antiretroviral Therapy; CCR5: C-C Chemokine Receptor type 5; CD4: Cluster of Differentiation 4; CHOP: C/EBP homologous protein; CXCR4: C-X-C Chemokine Receptor Type 4; Cyto c: Cytochrome C; DCs: Dendritic Cells; EDEM1: ER-degradation enhancing-a-mannosidase-like protein 1; ENV: Envelope; ER: Endoplasmic Reticulum; FasR: Fas Receptor;G2: Gap 2; G2/M: Gap2/Mitosis; GFAP: Glial Fibrillary Acidic Protein; GP120: Glycoprotein120; GP41: Glycoprotein41; HAND: HIV Associated Neurodegenerative Disease; HEK: Human Embryonic Kidney; HeLa: Human Cervical Epithelial Carcinoma; HIV: Human Immunodeficiency Virus; IPS-1: IFN-β promoter stimulator 1; IRE-1: Inositol Requiring Enzyme 1; IRGM: Immunity Related GTPase Family M protein; LAMP2A: Lysosome Associated Membrane Protein 2A; LC3: Microtubule Associated Light Chain 3; MDA5: Melanoma Differentiation Associated gene 5; MEF: Mouse Embryonic Fibroblast; MMP: Mitochondrial Membrane Permeabilization; Nef: Negative Regulatory Factor; OASIS: Old Astrocyte Specifically Induced Substrate; PAMP: Pathogen-Associated Molecular Pattern; PERK: Pancreatic Endoplasmic Reticulum Kinase; PRR: Pattern Recognition Receptor; Puma: P53 Upregulated Modulator of Apoptosis; RIG-I: Retinoic acid-Inducible Gene-I; Tat: Transactivator Protein of HIV; TLR: Toll-like receptor; ULK1: Unc51 Like Autophagy Activating Kinase 1; UPR: Unfolded Protein Response; Vpr: Viral Protein Regulatory; XBP1: X-Box Binding Protein 1.
Collapse
Affiliation(s)
- Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Past eur Institute of IRAN, Tehran, Iran
| | - Sudharsana R. Ande
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Javad Alizadeh
- Department of Human Anatomy & Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Children‘s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Research Institute of Oncology and Hematology, CancerCare Manitoba, University of Manitoba, Winnipeg, Canada
| | - Shahrzad Rahimizadeh
- Department of Medical Microbiology, Assiniboine Community College, School of Health and Human Services and Continuing Education, Winnipeg, MB, Canada
| | - Aryana Shariati
- Department of Human Anatomy & Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Hadis Malek
- Department of Biology, Islamic Azad University, Mashhad, Iran
| | - Mohammad Hashemi
- Department of Clinical Biochemistry, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Kathleen K. M. Glover
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Affan A. Sher
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Kevin M. Coombs
- Children‘s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, MB, Canada
| | - Saeid Ghavami
- Department of Human Anatomy & Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Children‘s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Research Institute of Oncology and Hematology, CancerCare Manitoba, University of Manitoba, Winnipeg, Canada
- Health Policy Research Centre, Shiraz Medical University of Medical Science, Shiraz, Iran
| |
Collapse
|
27
|
Engineered action at a distance: Blood-meal-inducible paralysis in Aedes aegypti. PLoS Negl Trop Dis 2019; 13:e0007579. [PMID: 31479450 PMCID: PMC6719823 DOI: 10.1371/journal.pntd.0007579] [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: 10/25/2018] [Accepted: 06/26/2019] [Indexed: 11/23/2022] Open
Abstract
Background Population suppression through mass-release of Aedes aegypti males carrying dominant-lethal transgenes has been demonstrated in the field. Where population dynamics show negative density-dependence, suppression can be enhanced if lethality occurs after the density-dependent (i.e. larval) stage. Existing molecular tools have limited current examples of such Genetic Pest Management (GPM) systems to achieving this through engineering ‘cell-autonomous effectors’ i.e. where the expressed deleterious protein is restricted to the cells in which it is expressed–usually under the control of the regulatory elements (e.g. promoter regions) used to build the system. This limits the flexibility of these technologies as regulatory regions with useful spatial, temporal or sex-specific expression patterns may only be employed if the cells they direct expression in are simultaneously sensitive to existing effectors, and also precludes the targeting of extracellular regions such as cell-surface receptors. Expanding the toolset to ‘non-cell autonomous’ effectors would significantly reduce these limitations. Methodology/Principal findings We sought to engineer female-specific, late-acting lethality through employing the Ae. aegypti VitellogeninA1 promoter to drive blood-meal-inducible, fat-body specific expression of tTAV. Initial attempts using pro-apoptotic effectors gave no evident phenotype, potentially due to the lower sensitivity of terminally-differentiated fat-body cells to programmed-death signals. Subsequently, we dissociated the temporal and spatial expression of this system by engineering a novel synthetic effector (Scorpion neurotoxin–TetO-gp67.AaHIT) designed to be secreted out of the tissue in which it was expressed (fat-body) and then affect cells elsewhere (neuro-muscular junctions). This resulted in a striking, temporary-paralysis phenotype after blood-feeding. Conclusions/Significance These results are significant in demonstrating for the first time an engineered ‘action at a distance’ phenotype in a non-model pest insect. The potential to dissociate temporal and spatial expression patterns of useful endogenous regulatory elements will extend to a variety of other pest insects and effectors. A recent addition to the toolbox for controlling populations of the disease vector Aedes aegypti is the mass-release of males engineered with dominant, lethal transgenes. The lethal effect of these transgenes is activated in the progeny of these released engineered males and wild females they mate with in the field and with continuous release of males can cause population collapse. To date, these systems have relied on the use of ‘cell-autonomous’ effectors, meaning that their action is restricted to the cells in which they are expressed, limiting the flexibility of designing new, more complex systems. Here we demonstrate that it is possible to engineer ‘non-cell autonomous’ effectors–that is where the effect (e.g. the action of a toxic protein) can act on cells distant from the tissues in which they are originally expressed. To achieve this we utilised the endogenous cell secretory pathway to engineer a novel control phenotype–blood-meal inducible (i.e. late-acting, female-specific) reversible paralysis. The logic behind engineering such ‘action at a distance’ phenotypes will extend to a variety of other pest insects and control phenotypes.
Collapse
|
28
|
Du J, Gao S, Tian Z, Guo Y, Kang D, Xing S, Zhang G, Liu G, Luo J, Chang H, Yin H. Transcriptome analysis of responses to bluetongue virus infection in Aedes albopictus cells. BMC Microbiol 2019; 19:121. [PMID: 31182015 PMCID: PMC6558886 DOI: 10.1186/s12866-019-1498-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 05/31/2019] [Indexed: 01/15/2023] Open
Abstract
Background Bluetongue virus (BTV) causes a disease among wild and domesticated ruminants which is not contagious, but which is transmitted by biting midges of the Culicoides species. BTV can induce an intense cytopathic effect (CPE) in mammalian cells after infection, although Culicoides- or mosquito-derived cell cultures cause non-lytic infection with BTV without CPE. However, little is known about the transcriptome changes in Aedes albopictus cells infected with BTV. Methods Transcriptome sequencing was used to identify the expression pattern of mRNA transcripts in A. albopictus cells infected with BTV, given the absence of the Culicoides genome sequence. Bioinformatics analyses were performed to examine the biological functions of the differentially expressed genes. Subsequently, quantitative reverse transcription–polymerase chain reaction was utilized to validate the sequencing data. Results In total, 51,850,205 raw reads were generated from the BTV infection group and 51,852,293 from the control group. A total of 5769 unigenes were common to both groups; only 779 unigenes existed exclusively in the infection group and 607 in the control group. In total, 380 differentially expressed genes were identified, 362 of which were up-regulated and 18 of which were down-regulated. Bioinformatics analyses revealed that the differentially expressed genes mainly participated in endocytosis, FoxO, MAPK, dorso-ventral axis formation, insulin resistance, Hippo, and JAK-STAT signaling pathways. Conclusion This study represents the first attempt to investigate transcriptome-wide dysregulation in A. albopictus cells infected with BTV. The understanding of BTV pathogenesis and virus–vector interaction will be improved by global transcriptome profiling. Electronic supplementary material The online version of this article (10.1186/s12866-019-1498-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Junzheng Du
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu, 730046, People's Republic of China.
| | - Shandian Gao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu, 730046, People's Republic of China
| | - Zhancheng Tian
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu, 730046, People's Republic of China
| | - Yanni Guo
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu, 730046, People's Republic of China
| | - Di Kang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu, 730046, People's Republic of China
| | - Shanshan Xing
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu, 730046, People's Republic of China
| | - Guorui Zhang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu, 730046, People's Republic of China
| | - Guangyuan Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu, 730046, People's Republic of China
| | - Jianxun Luo
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu, 730046, People's Republic of China
| | - Huiyun Chang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu, 730046, People's Republic of China
| | - Hong Yin
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu, 730046, People's Republic of China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, People's Republic of China
| |
Collapse
|
29
|
Thaker SK, Chapa T, Garcia G, Gong D, Schmid EW, Arumugaswami V, Sun R, Christofk HR. Differential Metabolic Reprogramming by Zika Virus Promotes Cell Death in Human versus Mosquito Cells. Cell Metab 2019; 29:1206-1216.e4. [PMID: 30827860 PMCID: PMC6818653 DOI: 10.1016/j.cmet.2019.01.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/04/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022]
Abstract
Zika virus is a pathogen that poses serious consequences, including congenital microcephaly. Although many viruses reprogram host cell metabolism, whether Zika virus alters cellular metabolism and the functional consequences of Zika-induced metabolic changes remain unknown. Here, we show that Zika virus infection differentially reprograms glucose metabolism in human versus C6/36 mosquito cells by increasing glucose use in the tricarboxylic acid cycle in human cells versus increasing glucose use in the pentose phosphate pathway in mosquito cells. Infection of human cells selectively depletes nucleotide triphosphate levels, leading to elevated AMP/ATP ratios, AMP-activated protein kinase (AMPK) phosphorylation, and caspase-mediated cell death. AMPK is also phosphorylated in Zika virus-infected mouse brain. Inhibiting AMPK in human cells decreases Zika virus-mediated cell death, whereas activating AMPK in mosquito cells promotes Zika virus-mediated cell death. These findings suggest that the differential metabolic reprogramming during Zika virus infection of human versus mosquito cells determines whether cell death occurs.
Collapse
Affiliation(s)
- Shivani K Thaker
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Travis Chapa
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Danyang Gong
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ernst W Schmid
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90098, USA
| | - Ren Sun
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Heather R Christofk
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
30
|
Huang YJS, Higgs S, Vanlandingham DL. Arbovirus-Mosquito Vector-Host Interactions and the Impact on Transmission and Disease Pathogenesis of Arboviruses. Front Microbiol 2019; 10:22. [PMID: 30728812 PMCID: PMC6351451 DOI: 10.3389/fmicb.2019.00022] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/09/2019] [Indexed: 12/11/2022] Open
Abstract
Hundreds of viruses, designated as arboviruses, are transmitted by arthropod vectors in complex transmission cycles between the virus, vertebrate host, and the vector. With millions of human and animal infections per year, it is critical to improve our understanding of the interactions between the biological and environmental factors that play a critical role in pathogenesis, disease outcomes, and transmission of arboviruses. This review focuses on mosquito-borne arboviruses and discusses current knowledge of the factors and underlying mechanisms that influence infection and transmission of arboviruses and discusses critical factors and pathways that can potentially become targets for intervention and therapeutics.
Collapse
Affiliation(s)
- Yan-Jang S Huang
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States.,Biosecurity Research Institute, Kansas State University, Manhattan, KS, United States
| | - Stephen Higgs
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States.,Biosecurity Research Institute, Kansas State University, Manhattan, KS, United States
| | - Dana L Vanlandingham
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, United States.,Biosecurity Research Institute, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
31
|
Chen Q, Zheng L, Mao Q, Liu J, Wang H, Jia D, Chen H, Wu W, Wei T. Fibrillar structures induced by a plant reovirus target mitochondria to activate typical apoptotic response and promote viral infection in insect vectors. PLoS Pathog 2019; 15:e1007510. [PMID: 30653614 PMCID: PMC6353215 DOI: 10.1371/journal.ppat.1007510] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 01/30/2019] [Accepted: 12/07/2018] [Indexed: 11/19/2022] Open
Abstract
Numerous plant viruses that cause significant agricultural problems are persistently transmitted by insect vectors. We wanted to see if apoptosis was involved in viral infection process in the vector. We found that a plant reovirus (rice gall dwarf virus, RGDV) induced typical apoptotic response during viral replication in the leafhopper vector and cultured vector cells, as demonstrated by mitochondrial degeneration and membrane potential decrease. Fibrillar structures formed by nonstructural protein Pns11 of RGDV targeted the outer membrane of mitochondria, likely by interaction with an apoptosis-related mitochondrial protein in virus-infected leafhopper cells or nonvector insect cells. Such association of virus-induced fibrillar structures with mitochondria clearly led to mitochondrial degeneration and membrane potential decrease, suggesting that RGDV Pns11 was the inducer of apoptotic response in insect vectors. A caspase inhibitor treatment and knockdown of caspase gene expression using RNA interference each reduced apoptosis and viral accumulation, while the knockdown of gene expression for the inhibitor of apoptosis protein improved apoptosis and viral accumulation. Thus, RGDV exploited caspase-dependent apoptotic response to promote viral infection in insect vectors. For the first time, we directly confirmed that a nonstructural protein encoded by a persistent plant virus can induce the typical apoptotic response to benefit viral transmission by insect vectors.
Collapse
Affiliation(s)
- Qian Chen
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Limin Zheng
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
- Key Laboratory of Pest Management of Horticultural Crop of Hunan Province, Hunan Plant Protection Institute, Hunan Academy of Agricultural Science, Changsha, PR China
| | - Qianzhuo Mao
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Jiejie Liu
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Haitao Wang
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Dongsheng Jia
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Hongyan Chen
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Wei Wu
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| | - Taiyun Wei
- Fujian Province Key Laboratory of Plant Virology, Institute of Plant Virology, State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Vector-borne Virus Research Center, Fujian Agriculture and Forestry University, Fuzhou, Fujian, PR China
| |
Collapse
|
32
|
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
|
33
|
Wei J, Jia D, Mao Q, Zhang X, Chen Q, Wu W, Chen H, Wei T. Complex interactions between insect-borne rice viruses and their vectors. Curr Opin Virol 2018; 33:18-23. [PMID: 30031984 DOI: 10.1016/j.coviro.2018.07.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 06/28/2018] [Accepted: 07/05/2018] [Indexed: 01/12/2023]
Abstract
Insect-borne rice viral diseases are widespread and economically important in many rice-growing countries. Long-term associations between rice viruses and their insect vectors result in evolutionary trade-offs that maintain a balance between the fitness cost of the viral infection of insects and the persistent transmission of the virus by the insect. To promote optimal replication, rice viruses activate innate immune responses, such as autophagy, apoptosis, and stress-regulated signaling pathways in the vector; meanwhile, a conserved insect small interfering RNA antiviral pathway is activated to control excessive viral replication, guaranteeing persistent virus transmission. Furthermore, growing evidence has shown that rice viruses can manipulate their vectors either directly or by inducing changes in host plants to promote the spread of viral pathogens. Thus, understanding the plant-virus-insect relationships offers important insights into how disease epidemics occur and facilitates the design of powerful new strategies for disease control.
Collapse
Affiliation(s)
- Jing Wei
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Dongsheng Jia
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China; State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops and College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Qianzhuo Mao
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Xiaofeng Zhang
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Qian Chen
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China; State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops and College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wei Wu
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Hongyan Chen
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Taiyun Wei
- Vector-borne Virus Research Center, Fujian Province Key Laboratory of Plant Virology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China; State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops and College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
34
|
Taracena ML, Bottino-Rojas V, Talyuli OAC, Walter-Nuno AB, Oliveira JHM, Angleró-Rodriguez YI, Wells MB, Dimopoulos G, Oliveira PL, Paiva-Silva GO. Regulation of midgut cell proliferation impacts Aedes aegypti susceptibility to dengue virus. PLoS Negl Trop Dis 2018; 12:e0006498. [PMID: 29782512 PMCID: PMC5983868 DOI: 10.1371/journal.pntd.0006498] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/01/2018] [Accepted: 05/03/2018] [Indexed: 01/12/2023] Open
Abstract
Aedes aegypti is the vector of some of the most important vector-borne diseases like dengue, chikungunya, zika and yellow fever, affecting millions of people worldwide. The cellular processes that follow a blood meal in the mosquito midgut are directly associated with pathogen transmission. We studied the homeostatic response of the midgut against oxidative stress, as well as bacterial and dengue virus (DENV) infections, focusing on the proliferative ability of the intestinal stem cells (ISC). Inhibition of the peritrophic matrix (PM) formation led to an increase in reactive oxygen species (ROS) production by the epithelial cells in response to contact with the resident microbiota, suggesting that maintenance of low levels of ROS in the intestinal lumen is key to keep ISCs division in balance. We show that dengue virus infection induces midgut cell division in both DENV susceptible (Rockefeller) and refractory (Orlando) mosquito strains. However, the susceptible strain delays the activation of the regeneration process compared with the refractory strain. Impairment of the Delta/Notch signaling, by silencing the Notch ligand Delta using RNAi, significantly increased the susceptibility of the refractory strains to DENV infection of the midgut. We propose that this cell replenishment is essential to control viral infection in the mosquito. Our study demonstrates that the intestinal epithelium of the blood fed mosquito is able to respond and defend against different challenges, including virus infection. In addition, we provide unprecedented evidence that the activation of a cellular regenerative program in the midgut is important for the determination of the mosquito vectorial competence. Aedes mosquitoes are important vectors of arboviruses, representing a major threat to public health. While feeding on blood, mosquitoes address the challenges of digestion and preservation of midgut homeostasis. Damaged or senescent cells must be constantly replaced by new cells to maintain midgut epithelial integrity. In this study, we show that the intestinal stem cells (ISCs) of blood-fed mosquitoes are able to respond to abiotic and biotic challenges. Exposing midgut cells to different types of stress, such as the inhibition of the peritrophic matrix formation, changes in the midgut redox state, or infection with entomopathogenic bacteria or viruses, resulted in an increased number of mitotic cells in blood-fed mosquitoes. Mosquito strains with different susceptibilities to DENV infection presented different time course of cell regeneration in response to viral infection. Knockdown of the Notch pathway in a refractory mosquito strain limited cell division after infection with DENV and resulted in increased mosquito susceptibility to the virus. Conversely, inducing midgut cell proliferation made a susceptible strain more resistant to viral infection. Therefore, the effectiveness of midgut cellular renewal during viral infection proved to be an important factor in vector competence. These findings can contribute to the understanding of virus-host interactions and help to develop more successful strategies of vector control.
Collapse
Affiliation(s)
- Mabel L. Taracena
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | - Vanessa Bottino-Rojas
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | - Octavio A. C. Talyuli
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | - Ana Beatriz Walter-Nuno
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | - José Henrique M. Oliveira
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Yesseinia I. Angleró-Rodriguez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, United States of America
| | - Michael B. Wells
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, United States of America
- The Johns Hopkins Malaria Research Institute, The Johns Hopkins Bloomberg School of Public Health, Baltimore, United States of America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, United States of America
| | - Pedro L. Oliveira
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
| | - Gabriela O. Paiva-Silva
- Programa de Biologia Molecular e Biotecnologia, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brasil
- * E-mail:
| |
Collapse
|
35
|
Swevers L, Liu J, Smagghe G. Defense Mechanisms against Viral Infection in Drosophila: RNAi and Non-RNAi. Viruses 2018; 10:E230. [PMID: 29723993 PMCID: PMC5977223 DOI: 10.3390/v10050230] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/20/2018] [Accepted: 04/27/2018] [Indexed: 12/20/2022] Open
Abstract
RNAi is considered a major antiviral defense mechanism in insects, but its relative importance as compared to other antiviral pathways has not been evaluated comprehensively. Here, it is attempted to give an overview of the antiviral defense mechanisms in Drosophila that involve both RNAi and non-RNAi. While RNAi is considered important in most viral infections, many other pathways can exist that confer antiviral resistance. It is noted that very few direct recognition mechanisms of virus infections have been identified in Drosophila and that the activation of immune pathways may be accomplished indirectly through cell damage incurred by viral replication. In several cases, protection against viral infection can be obtained in RNAi mutants by non-RNAi mechanisms, confirming the variability of the RNAi defense mechanism according to the type of infection and the physiological status of the host. This analysis is aimed at more systematically investigating the relative contribution of RNAi in the antiviral response and more specifically, to ask whether RNAi efficiency is affected when other defense mechanisms predominate. While Drosophila can function as a useful model, this issue may be more critical for economically important insects that are either controlled (agricultural pests and vectors of diseases) or protected from parasite infection (beneficial insects as bees) by RNAi products.
Collapse
Affiliation(s)
- Luc Swevers
- Institute of Biosciences & Applications, NCSR "Demokritos", 15341 Athens, Greece.
| | - Jisheng Liu
- School of Life Sciences, Guangzhou University, 510006 Guangzhou, China.
| | - Guy Smagghe
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
36
|
Ciota AT. West Nile virus and its vectors. CURRENT OPINION IN INSECT SCIENCE 2017; 22:28-36. [PMID: 28805636 DOI: 10.1016/j.cois.2017.05.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/01/2017] [Indexed: 06/07/2023]
Abstract
West Nile virus (WNV Flaviviridae; Flavivrus) is the most geographically widespread arbovirus in the world and the leading cause of arboviral encephalitis globally. Worldwide, WNV is maintained in an enzootic cycle between primarily Culex spp. mosquitoes and birds, with human infection and disease resulting from enzootic spillover. Dynamic and complex intrinsic and extrinsic factors contribute to the temporal and spatial variability in WNV transmission. The most current information on the relative contribution of each of these factors is reviewed and a case to incorporate detailed and localized environmental and genetic data into predictive models is presented.
Collapse
Affiliation(s)
- Alexander T Ciota
- The Arbovirus Laboratory, Wadsworth Center, New York State Department of Health, Slingerlands, NY, USA; Department of Biomedical Sciences, State University of New York at Albany School of Public Health, Albany, NY, USA.
| |
Collapse
|
37
|
The p53 gene with emphasis on its paralogues in mosquitoes. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2017; 50:747-754. [PMID: 28690024 DOI: 10.1016/j.jmii.2017.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/31/2017] [Accepted: 06/21/2017] [Indexed: 01/21/2023]
Abstract
The p53 gene is highly important in human cancers, as it serves as a tumor-suppressor gene. Subsequently, two p53 homologues, i.e., p73 and p63, with high identity of amino acids were identified, leading to construction of the p53 family. The p53 gene is highly important in human cancer because it usually transcribes genes that function by causing apoptosis in mammalian cells. In contrast, p63 and p73 tend to be more important in modulating development than inducing cell death, even though they share similar protein structures. Relatively recently, p53 was also identified in mosquitoes and many other insect species. Uniquely, its structure lacks the sterile alpha motif domain which is a putative protein-protein interaction domain and exclusively exists at the C-terminal region in p73 and p63 in mammals. A phylogenetic analysis revealed that the p53 gene derived from mosquitoes is composed of two paralogues, p53-1 and p53-2. Of these, only p53-2 is responsively upregulated by dengue 2 virus (DENV2) in C6/36 cells which usually survive the infection. This indicates that the p53 gene is closely related to DENV infection in mosquito cells. The specific significance of p53-2's involvement in cell survival from virus-induced stress is described and briefly discussed in this report.
Collapse
|
38
|
Vermaak E, Maree FF, Theron J. The Culicoides sonorensis inhibitor of apoptosis 1 protein protects mammalian cells from apoptosis induced by infection with African horse sickness virus and bluetongue virus. Virus Res 2017; 232:152-161. [PMID: 28267609 DOI: 10.1016/j.virusres.2017.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/13/2017] [Accepted: 03/01/2017] [Indexed: 12/14/2022]
Abstract
African horse sickness virus (AHSV) and bluetongue virus (BTV) are arboviruses of the genus Orbivirus that are transmitted to their vertebrate hosts by Culicoides biting midges. These orbiviruses exhibit lytic infection (apoptosis) in mammalian cells, but cause persistent infection with no cytopathic effects in Culicoides sonorensis cells. Although regulation of apoptosis could thus be integral for establishing persistent virus infection in midge cells, nothing is known about the presence and function of apoptosis pathways in Culicoides midges and their derived cell lines. Here, we report the cloning and functional characterization of an inhibitor of apoptosis protein (IAP), designated CsIAP1, from C. sonorensis cells. The CsIAP1 protein contains two baculoviral IAP repeat (BIR) domains and a RING domain. Silencing of the Cs iap1 gene in C. sonorensis cells caused apoptosis, indicating that CsIAP1 plays a role in cell survival. Stable expression of the CsIAP1 protein in BSR mammalian cells suppressed apoptosis induced by AHSV-4 and BTV-10 infection, and biochemical data indicated that CsIAP1 is an inhibitor of mammalian caspase-9, an initiator caspase in the intrinsic apoptotic pathway. Mutagenesis studies indicated that the BIR2 and RING domains are required for the anti-apoptotic activity of CsIAP1. The results suggest that the mechanism by which CsIAP1 suppresses apoptosis in insect cells may involve inhibition of a Culicoides caspase-9 homologue through a mechanism that requires both the BIR2 and RING domains. This study provides the first evidence that the CsIAP1 protein is a key negative regulator of apoptosis in C. sonorensis cells.
Collapse
Affiliation(s)
- Elaine Vermaak
- Department of Microbiology and Plant Pathology, University of Pretoria, Pretoria 0002, South Africa
| | - Francois F Maree
- Department of Microbiology and Plant Pathology, University of Pretoria, Pretoria 0002, South Africa; Transboundary Animal Diseases, Onderstepoort Veterinary Institute, Agricultural Research Council, Pretoria 0110, South Africa
| | - Jacques Theron
- Department of Microbiology and Plant Pathology, University of Pretoria, Pretoria 0002, South Africa.
| |
Collapse
|
39
|
Pando-Robles V, Batista CV. Aedes-Borne Virus-Mosquito Interactions: Mass Spectrometry Strategies and Findings. Vector Borne Zoonotic Dis 2017; 17:361-375. [PMID: 28192064 DOI: 10.1089/vbz.2016.2040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aedes-borne viruses are responsible for high-impact neglected tropical diseases and unpredictable outbreaks such as the ongoing Zika epidemics. Aedes mosquitoes spread different arboviruses such as Dengue virus (DENV), Chikungunya virus (CHIKV), and Zika virus, among others, and are responsible for the continuous emergence and reemergence of these pathogens. These viruses have complex transmission cycles that include two hosts, namely the Aedes mosquito as a vector and susceptible vertebrate hosts. Human infection with arboviruses causes diseases that range from subclinical or mild to febrile diseases, encephalitis, and hemorrhagic fever. Infected mosquitoes do not show detectable signs of disease, even though the virus maintains a lifelong persistent infection. The infection of the Aedes mosquito by viruses involves a molecular crosstalk between cell and viral proteins. An understanding of how mosquito vectors and viruses interact is of fundamental interest, and it also offers novel perspectives for disease control. In recent years, mass spectrometry (MS)-based strategies in combination with bioinformatics have been successfully applied to identify and quantify global changes in cellular proteins, lipids, peptides, and metabolites in response to viral infection. Although the information about proteomics in the Aedes mosquito is limited, the information that has been reported can set up the basis for future studies. This review reflects how MS-based approaches have extended our understanding of Aedes mosquito biology and the development of DENV and CHIKV infection in the vector. Finally, this review discusses future challenges in the field.
Collapse
Affiliation(s)
- Victoria Pando-Robles
- 1 Laboratorio de Proteómica, Departamento de Infección e Inmunidad, Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, México
| | - Cesar V Batista
- 2 Laboratorio Universitario de Proteómica, Instituto de Biotecnología. Universidad Nacional Autónoma de México , Cuernavaca, México
| |
Collapse
|
40
|
Ghanim M, Fattah-Hosseini S, Levy A, Cilia M. Morphological abnormalities and cell death in the Asian citrus psyllid (Diaphorina citri) midgut associated with Candidatus Liberibacter asiaticus. Sci Rep 2016; 6:33418. [PMID: 27630042 PMCID: PMC5024303 DOI: 10.1038/srep33418] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/26/2016] [Indexed: 12/03/2022] Open
Abstract
Candidatus Liberibacter asiaticus (CLas) is a phloem-limited, gram-negative, fastidious bacterium that is associated with the development of citrus greening disease, also known as Huanglongbing (HLB). CLas is transmitted by the Asian citrus psyllid (ACP) Diaphorina citri, in a circulative manner. Two major barriers to transmission within the insect are the midgut and the salivary glands. We performed a thorough microscopic analysis within the insect midgut following exposure to CLas-infected citrus trees. We observed changes in nuclear architecture, including pyknosis and karyorrhexis as well as changes to the actin cytoskeleton in CLas-exposed midgut cells. Further analyses showed that the changes are likely due to the activation of programmed cell death as assessed by Annexin V staining and DNA fragmentation assays. These results suggest that exposure to CLas-infected trees induces apoptotic responses in the psyllid midgut that should be further investigated. Understanding the adaptive significance of the apoptotic response has the potential to create new approaches for controlling HLB.
Collapse
Affiliation(s)
- Murad Ghanim
- Department of Entomology, Volcani Center, P.O. Box 6, Bet Dagnan 50250, Israel
| | | | - Amit Levy
- Boyce Thompson Institute for Plant Research, Ithaca, NY 14853, United States
| | - Michelle Cilia
- Boyce Thompson Institute for Plant Research, Ithaca, NY 14853, United States
- Plant Pathology and Plant-Microbe Biology Section, School of Integrative Plant Science, Cornell University, Ithaca, NY 14853, United States
- USDA Agricultural Research Service, Emerging Pests and Pathogens Research Unit, Ithaca, NY 14853, United States
| |
Collapse
|
41
|
Eng MW, van Zuylen MN, Severson DW. Apoptosis-related genes control autophagy and influence DENV-2 infection in the mosquito vector, Aedes aegypti. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 76:70-83. [PMID: 27418459 PMCID: PMC5010484 DOI: 10.1016/j.ibmb.2016.07.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/06/2016] [Accepted: 07/10/2016] [Indexed: 05/12/2023]
Abstract
The mosquito Aedes aegypti is the primary urban vector for dengue virus (DENV) worldwide. Insight into interactions occurring between host and pathogen is important in understanding what factors contribute to vector competence. However, many of the molecular mechanisms for vector competence remain unknown. Our previous global transcriptional analysis suggested that differential expression of apoptotic proteins is involved in determining refractoriness vs susceptibility to DENV-2 infection in Ae. aegypti females following a DENV-infected blood meal. To determine whether DENV-refractory Ae. aegypti showed more robust apoptosis upon infection, we compared numbers of apoptotic cells from midguts of refractory and susceptible strains and observed increased numbers of apoptotic cells in only the refractory strain upon DENV-2 infection. Thereafter, we manipulated apoptosis through dsRNA interference of the initiator caspase, Aedronc. Unexpectedly, dsAedronc-treated females showed both decreased frequency of disseminated infection and decreased virus titer in infected individuals. Insect caspases have also previously been identified as regulators of the cellular recycling process known as autophagy. We observed activation of autophagy in midgut and fat body tissues following a blood meal, as well as programmed activation of several apoptosis-related genes, including the effector caspase, Casps7. To determine whether autophagy was affected by caspase knockdown, we silenced Aedronc and Casps7, and observed reduced activation of autophagy upon silencing. Our results provide evidence that apoptosis-related genes are also involved in regulating autophagy, and that Aedronc may play an important role in DENV-2 infection success in Ae. aegypti, possibly through its regulation of autophagy.
Collapse
Affiliation(s)
- Matthew W Eng
- Eck Institute for Global Health and Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Madeleine N van Zuylen
- Eck Institute for Global Health and Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - David W Severson
- Eck Institute for Global Health and Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
42
|
Feng L, Liu H, Li X, Qiao J, Wang S, Guo D, Liu Q. Identification of AaCASPS7, an effector caspase in Aedes albopictus. Gene 2016; 593:117-125. [PMID: 27502418 DOI: 10.1016/j.gene.2016.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 08/01/2016] [Accepted: 08/04/2016] [Indexed: 11/28/2022]
Abstract
Aedes albopictus mosquito is a vector of various arboviruses and is becoming a significant threat to public health due to its rapid global expansion. Several reports suggest that apoptosis could be a factor limiting arbovirus infection in mosquitoes. Thus, it is significant to identify apoptosis pathway and study the correlation between apoptosis and virus infection in mosquitoes. Apoptosis is a type of programmed cell death that plays a vital role in immunity, development, and tissue homeostasis. Caspases are a family of conserved proteases playing important roles in apoptosis. In this study, we identified Aedes albopictus AaCASPS7, a caspase shared high identity with dipteran insect drICE orthologs. Phylogenetic analysis showed the closest relative of AaCASPS7 was Aedes aegypti AeCASPS7. AaCASPS7 displayed several features that were typical of an effector caspase and showed significant activity to effector caspase substrates. Aacasps7 transcripts were expressed ubiquitously in developmental and adult stages in Aedes albopictus mosquitoes. Transient expression of AaCASPS7 induced caspase-dependent apoptosis in C6/36 cells. Taken together the above data, this study identified a novel caspase, AaCASPS7, which might function as an apoptotic caspase. Further study the function of AaCASPS7 would facilitate better understanding the apoptotic mechanism in Aedes albopictus mosquito.
Collapse
Affiliation(s)
- Lingyan Feng
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Hao Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Xiaomei Li
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Jialu Qiao
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Shengya Wang
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Deyin Guo
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Qingzhen Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China.
| |
Collapse
|
43
|
Meng K, Li X, Wang S, Zhong C, Yang Z, Feng L, Liu Q. The Strica Homolog AaCASPS16 Is Involved in Apoptosis in the Yellow Fever Vector, Aedes albopictus. PLoS One 2016; 11:e0157846. [PMID: 27351972 PMCID: PMC4924790 DOI: 10.1371/journal.pone.0157846] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/06/2016] [Indexed: 12/20/2022] Open
Abstract
Caspases are a family of cysteine proteases playing essential roles during apoptosis. Seven caspases identified in Drosophila were Dronc, Dredd, Strica, Dcp-1, Decay, Drice and Damm. Among them, Strica is an insect-specific caspase containing a long serine- and threonine- rich prodomain, of which function is not yet well studied. Here we identified a homolog of strica from Aedes albopictus, named as Aacasps16. Aacasps16 encoded a protein containing a putative serine- and threonine-rich prodomain and a well conserved caspase catalytic domain. AaCASPS16 shared high identity with dipteran insects Strica homologs. Alignment showed that the closest relative of AaCASPS16 was Aedes aegypti AeCASPS16. The expression profiles of Aacasps16 during developmental and adult stages were analyzed. Purified recombinant AaCASPS16 exhibited the highest caspase activity to WEHD, which is the substrate preferred by human caspase-9. AaCASPS16 induced apoptosis when over-expressed in C6/36 cells. AaCASPS16 was processed during apoptosis induced by actinomycin D and ultraviolet irradiation treatment, whereas partial silencing of Aacasps16 reduced actinomycin D- and ultraviolet irradiation-triggered apoptosis in C6/36 cells. Taken together, our study identified AaCASPS16 as a novel apoptotic caspase in Aedes albopictus.
Collapse
Affiliation(s)
- Kun Meng
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Xiaomei Li
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Shengya Wang
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Chunyan Zhong
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Zhouning Yang
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Lingyan Feng
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Qingzhen Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
- * E-mail:
| |
Collapse
|
44
|
Dong S, Kantor AM, Lin J, Passarelli AL, Clem RJ, Franz AWE. Infection pattern and transmission potential of chikungunya virus in two New World laboratory-adapted Aedes aegypti strains. Sci Rep 2016; 6:24729. [PMID: 27102548 PMCID: PMC4840389 DOI: 10.1038/srep24729] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 04/05/2016] [Indexed: 12/02/2022] Open
Abstract
Chikungunya virus (CHIKV) is an emerging mosquito-borne virus belonging to the Togaviridae, which is transmitted to humans by Aedes aegypti and Ae. albopictus. We describe the infection pattern of CHIKV in two New World Ae. aegypti strains, HWE and ORL. Both mosquito strains were susceptible to the virus but showed different infection patterns in midguts and salivary glands. Even though acquisition of a bloodmeal showed moderate levels of apoptosis in midgut tissue, there was no obvious additional CHIKV-induced apoptosis detectable during midgut infection. Analysis of expression of apoptosis-related genes suggested that CHIKV infection dampens rather than promotes apoptosis in the mosquito midgut. In both mosquito strains, the virus was present in saliva within two days post-oral infection. HWE and ORL mosquitoes exhibited no salivary gland infection barrier; however, only 60% (HWE) to 65% (ORL) of the females had released the virus in their saliva at one week post-oral acquisition, suggesting a salivary gland escape barrier. CHIKV induced an apoptotic response in salivary glands of HWE and ORL mosquitoes, demonstrating that the virus caused pathology in its natural vector.
Collapse
Affiliation(s)
- Shengzhang Dong
- 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
| | - 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
|
45
|
Clem RJ. Arboviruses and apoptosis: the role of cell death in determining vector competence. J Gen Virol 2016; 97:1033-1036. [PMID: 26872460 DOI: 10.1099/jgv.0.000429] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A relatively small number of mosquito species transmit arboviruses such as dengue, yellow fever, chikungunya and West Nile viruses to hundreds of millions of people each year, yet we still lack a thorough understanding of the molecular factors that determine vector competence. Apoptosis has been shown to be an important factor in determining the outcome of virus infection for many viruses. However, until recently, it was not clear whether apoptosis plays a role in determining the outcome of arbovirus infections in mosquitoes. Recent work has begun to shed light on the roles of apoptosis in this important process.
Collapse
Affiliation(s)
- Rollie J Clem
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
46
|
Xing S, Du J, Gao S, Tian Z, Zheng Y, Liu G, Luo J, Yin H. Analysis of the miRNA expression profile in an Aedes albopictus cell line in response to bluetongue virus infection. INFECTION GENETICS AND EVOLUTION 2016; 39:74-84. [PMID: 26774367 DOI: 10.1016/j.meegid.2016.01.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 12/14/2015] [Accepted: 01/11/2016] [Indexed: 12/25/2022]
Abstract
Cellular microRNAs (miRNAs) have been reported to be key regulators of virus-host interactions. Bluetongue virus (BTV) is an insect-borne virus that causes huge economic losses in the livestock industry worldwide. Aedes albopictus cell lines have become powerful and convenient tools for studying BTV-vector interactions. However, the role of miRNAs in A. albopictus cells during BTV infection is not well understood. In this study, we performed a deep sequencing analysis of small RNA libraries of BTV-infected and mock-infected A. albopictus cells, and a total of 11,206,854 and 12,125,274 clean reads were identified, respectively. A differential expression analysis showed that 140 miRNAs, including 15 known and 125 novel miRNAs, were significantly dysregulated after infection, and a total of 414 and 2307 target genes were annotated, respectively. Real-time quantitative reverse transcription-polymerase chain reaction validated the expression patterns of 11 selected miRNAs and their mRNA targets. Functional annotation of the target genes suggested that these target genes were mainly involved in metabolic pathways, oxidative phosphorylation, endocytosis, RNA transport, as well as the FoxO, Hippo, Jak-STAT, and MAPK signaling pathways. This is the first systematic study on the effect of BTV infection on miRNA expression in A. albopictus cells. This investigation provides information concerning the cellular miRNA expression profile in response to BTV infection, and it offers clues for identifying potential candidates for vector-based antiviral strategies.
Collapse
Affiliation(s)
- Shanshan Xing
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Junzheng Du
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China.
| | - Shandian Gao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Zhancheng Tian
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Yadong Zheng
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Guangyuan Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Jianxun Luo
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China
| | - Hong Yin
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu 730046, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, PR China.
| |
Collapse
|
47
|
Carter JR, Taylor S, Fraser TS, Kucharski CA, Dawson JL, Fraser MJ. Suppression of the Arboviruses Dengue and Chikungunya Using a Dual-Acting Group-I Intron Coupled with Conditional Expression of the Bax C-Terminal Domain. PLoS One 2015; 10:e0139899. [PMID: 26580561 PMCID: PMC4651551 DOI: 10.1371/journal.pone.0139899] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 09/18/2015] [Indexed: 11/19/2022] Open
Abstract
In portions of South Asia, vectors and patients co-infected with dengue (DENV) and chikungunya (CHIKV) are on the rise, with the potential for this occurrence in other regions of the world, for example the United States. Therefore, we engineered an antiviral approach that suppresses the replication of both arboviruses in mosquito cells using a single antiviral group I intron. We devised unique configurations of internal, external, and guide sequences that permit homologous recognition and splicing with conserved target sequences in the genomes of both viruses using a single trans-splicing Group I intron, and examined their effectiveness to suppress infections of DENV and CHIKV in mosquito cells when coupled with a proapoptotic 3' exon, ΔN Bax. RT-PCR demonstrated the utility of these introns in trans-splicing the ΔN Bax sequence downstream of either the DENV or CHIKV target site in transformed Aedes albopictus C6/36 cells, independent of the order in which the virus specific targeting sequences were inserted into the construct. This trans-splicing reaction forms DENV or CHIKV ΔN Bax RNA fusions that led to apoptotic cell death as evidenced by annexin V staining, caspase, and DNA fragmentation assays. TCID50-IFA analyses demonstrate effective suppression of DENV and CHIKV infections by our anti-arbovirus group I intron approach. This represents the first report of a dual-acting Group I intron, and demonstrates that we can target DENV and CHIKV RNAs in a sequence specific manner with a single, uniquely configured CHIKV/DENV dual targeting group I intron, leading to replication suppression of both arboviruses, and thus providing a promising single antiviral for the transgenic suppression of multiple arboviruses.
Collapse
Affiliation(s)
- James R. Carter
- Department of Biological Sciences, Eck Institute of Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Samantha Taylor
- Department of Biological Sciences, Eck Institute of Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Tresa S. Fraser
- Department of Biological Sciences, Eck Institute of Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Cheryl A. Kucharski
- Department of Biological Sciences, Eck Institute of Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - James L. Dawson
- Department of Biological Sciences, Eck Institute of Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Malcolm J. Fraser
- Department of Biological Sciences, Eck Institute of Global Health, University of Notre Dame, Notre Dame, Indiana, United States of America
- * E-mail:
| |
Collapse
|
48
|
Abstract
Inhibitors of apoptosis (IAPs) family of genes encode baculovirus IAP-repeat domain-containing proteins with antiapoptotic function. These proteins also contain RING or UBC domains and act by binding to major proapoptotic factors and ubiquitylating them. High levels of IAPs inhibit caspase-mediated apoptosis. For these cells to undergo apoptosis, IAP function must be neutralized by IAP-antagonists. Mammalian IAP knockouts do not exhibit obvious developmental phenotypes, but the cells are more sensitized to apoptosis in response to injury. Loss of the mammalian IAP-antagonist ARTS results in reduced stem cell apoptosis. In addition to the antiapoptotic properties, IAPs regulate the innate immune response, and the loss of IAP function in humans is associated with immunodeficiency. The roles of IAPs in Drosophila apoptosis regulation are more apparent, where the loss of IAP1, or the expression of IAP-antagonists in Drosophila cells, is sufficient to trigger apoptosis. In this organism, apoptosis as a fate is conferred by the transcriptional induction of the IAP-antagonists. Many signaling pathways often converge on shared enhancer regions of IAP-antagonists. Cell death sensitivity is further regulated by posttranscriptional mechanisms, including those regulated by kinases, miRs, and ubiquitin ligases. These mechanisms are employed to eliminate damaged or virus-infected cells, limit neuroblast (neural stem cell) numbers, generate neuronal diversity, and sculpt tissue morphogenesis.
Collapse
Affiliation(s)
- Deepika Vasudevan
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Hyung Don Ryoo
- Department of Cell Biology, New York University School of Medicine, New York, New York, USA.
| |
Collapse
|
49
|
Franz AWE, Kantor AM, Passarelli AL, Clem RJ. Tissue Barriers to Arbovirus Infection in Mosquitoes. Viruses 2015; 7:3741-67. [PMID: 26184281 PMCID: PMC4517124 DOI: 10.3390/v7072795] [Citation(s) in RCA: 293] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/01/2015] [Accepted: 07/03/2015] [Indexed: 12/24/2022] Open
Abstract
Arthropod-borne viruses (arboviruses) circulate in nature between arthropod vectors and vertebrate hosts. Arboviruses often cause devastating diseases in vertebrate hosts, but they typically do not cause significant pathology in their arthropod vectors. Following oral acquisition of a viremic bloodmeal from a vertebrate host, the arbovirus disease cycle requires replication in the cellular environment of the arthropod vector. Once the vector has become systemically and persistently infected, the vector is able to transmit the virus to an uninfected vertebrate host. In order to systemically infect the vector, the virus must cope with innate immune responses and overcome several tissue barriers associated with the midgut and the salivary glands. In this review we describe, in detail, the typical arbovirus infection route in competent mosquito vectors. Based on what is known from the literature, we explain the nature of the tissue barriers that arboviruses are confronted with in a mosquito vector and how arboviruses might surmount these barriers. We also point out controversial findings to highlight particular areas that are not well understood and require further research efforts.
Collapse
Affiliation(s)
- Alexander W E Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA.
| | - Asher M Kantor
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA.
| | | | - Rollie J Clem
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|