1
|
Paludan SR, Pradeu T, Pichlmair A, Wray KB, Mikkelsen JG, Olagnier D, Mogensen TH. Early host defense against virus infections. Cell Rep 2024; 43:115070. [PMID: 39675007 DOI: 10.1016/j.celrep.2024.115070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/25/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024] Open
Abstract
Early host defense eliminates many viruses before infections are established while clearing others so they remain subclinical or cause only mild disease. The field of immunology has been shaped by broad concepts, including the pattern recognition theory that currently dominates innate immunology. Focusing on early host responses to virus infections, we analyze the literature to build a working hypothesis for the principles that govern the early line of cellular antiviral defense. Aiming to ultimately arrive at a criteria-based theory with strong explanatory power, we propose that both controlling infection and limiting inflammation are key drivers for the early cellular antiviral response. This response, which we suggest is exerted by a set of "microbe- and inflammation-restricting mechanisms," directly restrict viral replication while also counteracting inflammation. Exploring the mechanisms and physiological importance of the early layer of cellular antiviral defense may open further lines of research in immunology.
Collapse
Affiliation(s)
- Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Thomas Pradeu
- CNRS UMR 5164 ImmunoConcept, University of Bordeaux, Bordeaux, France; Department of Biological and Medical Sciences, University of Bordeaux, Bordeaux, France; Chapman University, Orange, CA, USA
| | - Andreas Pichlmair
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Technical University of Munich, School of Medicine, Institute of Virology, Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - K Brad Wray
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Centre for Science Studies, Aarhus University, Aarhus, Denmark; Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| | - Jacob Giehm Mikkelsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - David Olagnier
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Trine H Mogensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
2
|
Chen J, Liu YJ, Wang Q, Zhang L, Yang S, Feng WJ, Shi M, Gao J, Dai PL, Wu YY. Multiple stresses induced by chronic exposure to flupyradifurone affect honey bee physiological states. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 935:173418. [PMID: 38788938 DOI: 10.1016/j.scitotenv.2024.173418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/16/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
Flupyradifurone (FPF) has been reported to have a potential risk to terrestrial and aquatic ecosystems. In the present study, the effects of chronic FPF exposure on bees were systematically investigated at the individual behavioral, tissue, cell, enzyme activity, and the gene expression levels. Chronic exposure (14 d) to FPF led to reduced survival (12 mg/L), body weight gain (4 and 12 mg/L), and food utilization efficiency (4 and 12 mg/L). Additionally, FPF exposure (12 mg/L) impaired sucrose sensitivity and memory of bees. Morphological analysis revealed significant cellular and subcellular changes in brain neurons and midgut epithelial cells, including mitochondrial damage, nuclear disintegration, and apoptosis. FPF exposure (4 and 12 mg/L) led to oxidative stress, as evidenced by increased lipid peroxidation and alterations in antioxidant enzyme activity. Notably, gene expression analysis indicated significant dysregulation of apoptosis, immune, detoxification, sucrose responsiveness and memory-related genes, suggesting the involvement of different pathways in FPF-induced toxicity. The multiple stresses and potential mechanisms described here provide a basis for determining the intrinsic toxicity of FPF.
Collapse
Affiliation(s)
- Jin Chen
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Yong-Jun Liu
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Qiang Wang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Li Zhang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Sa Yang
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Wang-Jiang Feng
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - Min Shi
- Development Center of Plant Germplasm Resources, College of Life Sciences, Shanghai Normal University, Shanghai 201418, China
| | - Jing Gao
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China.
| | - Ping-Li Dai
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China.
| | - Yan-Yan Wu
- State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China.
| |
Collapse
|
3
|
Li M, Zhou Y, Cheng J, Wang Y, Lan C, Shen Y. Response of the mosquito immune system and symbiotic bacteria to pathogen infection. Parasit Vectors 2024; 17:69. [PMID: 38368353 PMCID: PMC10874582 DOI: 10.1186/s13071-024-06161-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/24/2024] [Indexed: 02/19/2024] Open
Abstract
Mosquitoes are the deadliest animal in the word, transmitting a variety of insect-borne infectious diseases, such as malaria, dengue fever, yellow fever, and Zika, causing more deaths than any other vector-borne pathogen. Moreover, in the absence of effective drugs and vaccines to prevent and treat insect-borne diseases, mosquito control is particularly important as the primary measure. In recent decades, due to the gradual increase in mosquito resistance, increasing attention has fallen on the mechanisms and effects associated with pathogen infection. This review provides an overview of mosquito innate immune mechanisms in terms of physical and physiological barriers, pattern recognition receptors, signalling pathways, and cellular and humoral immunity, as well as the antipathogenic effects of mosquito symbiotic bacteria. This review contributes to an in-depth understanding of the interaction process between mosquitoes and pathogens and provides a theoretical basis for biological defence strategies against mosquito-borne infectious diseases.
Collapse
Affiliation(s)
- Manjin Li
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yang Zhou
- Nanjing Medical University, Nanjing, 211166, China
| | - Jin Cheng
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yiqing Wang
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Cejie Lan
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
| | - Yuan Shen
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
- Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
4
|
Hu W, Zhao C, Zheng R, Duan S, Lu Z, Zhang Z, Yi L, Zhang N. Serratia marcescens induces apoptosis in Diaphorina citri gut cells via reactive oxygen species-mediated oxidative stress. PEST MANAGEMENT SCIENCE 2024; 80:602-612. [PMID: 37740936 DOI: 10.1002/ps.7787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/17/2023] [Accepted: 09/23/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND Asian citrus psyllid, Diaphorina citri, is a notorious pest in the citrus industry because it transmits Candidatus Liberibacter asiaticus, which causes an uncurable, devastating disease in citrus worldwide. Serratia marcescens is widely distributed in various environments that exhibits toxic effects to many insects. To develop strategies for enhancing the efficiency of pathogen-induced host mortality, a better understanding of the toxicity mechanism of Serratia marcescens on Diaphorina citri is critical. RESULTS Serratia marcescens KH-001 successfully colonized Diaphorina citri gut by feeding artificial diets, resulting in the damage of cells including nucleus, mitochondria, vesicles, and microvilli. Oral ingestion of Serratia marcescens KH-001 strongly induced apoptosis in gut cells by enhancing levels of Cyt c, p53 and caspase-1 and decreasing levels of inhibitors of apoptosis (IAP) and Bax inhibitor-1 (BI-1). The expression of dual oxidase (Duox) and nitric oxide synthase (Nos) was up-regulated by Serratia marcescens KH-001, which increased hydrogen peroxide (H2 O2 ) levels in the gut. Injection of abdomen of Diaphorina citri with H2 O2 accelerated the death of the adults and induced apoptosis in the gut cells by activating Cyt c, p53 and caspase-1 and suppressing IAP and BI-1. Pretreatment of infected Diaphorina citri with vitamin c (Vc) increased the adult survival and diminished the apoptosis-inducing effect. CONCLUSIONS The colonization of Serratia marcescens KH-001 in the guts of Diaphorina citri increased H2 O2 accumulation, leading to severe changes and apoptosis in intestinal cells, which enhanced a higher mortality level of D. citr. This study identifies the underlying virulence mechanism of Serratia marcescens KH-001 on Diaphorina citri that contributes to a widespread application in the integrated management of citrus psyllid. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Wei Hu
- National Navel Orange Engineering Research Center, College of Life Sciences, Gannan Normal University, Ganzhou, China
| | - Chongfei Zhao
- National Navel Orange Engineering Research Center, College of Life Sciences, Gannan Normal University, Ganzhou, China
| | - Rongkun Zheng
- National Navel Orange Engineering Research Center, College of Life Sciences, Gannan Normal University, Ganzhou, China
| | - Shuo Duan
- National Navel Orange Engineering Research Center, College of Life Sciences, Gannan Normal University, Ganzhou, China
| | - Zhanjun Lu
- National Navel Orange Engineering Research Center, College of Life Sciences, Gannan Normal University, Ganzhou, China
| | - Zhixiang Zhang
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China
| | - Long Yi
- National Navel Orange Engineering Research Center, College of Life Sciences, Gannan Normal University, Ganzhou, China
| | - Ning Zhang
- National Navel Orange Engineering Research Center, College of Life Sciences, Gannan Normal University, Ganzhou, China
| |
Collapse
|
5
|
Zhang J, Zafar J, Kong J, Wang F, Shao X, Zhang R, Pang R, Xu H, Xu X, Jin F. MicroRNA-Mediated Host Immune Genes Manipulation Benefits AcMNPV Proliferation in Spodoptera frugiperda. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71. [PMID: 37917564 PMCID: PMC10655178 DOI: 10.1021/acs.jafc.3c05012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023]
Abstract
Spodoptera frugiperda is a highly destructive migratory pest that threatens various crops globally. Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is an effective biocontrol agent against lepidopteran pests. Here, we explored the molecular mechanisms underlying the immune response to AcMNPV infection in S. frugiperda. RNA-seq and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analyses identified the Toll, IMD, and apoptosis pathways as primary immune responses. Investigation into AcMNPV-induced apoptosis in the S. frugiperda cell line (Sf9) revealed that the Toll pathway activated the JNK via the TRAF6 (TNF receptor-associated factor 6) adapter. In addition, AcMNPV-induced the differential expression of several host-encoded microRNAs (miRNAs), with significant negative regulatory effects, on S. frugiperda antiviral immune genes. RNAi and miRNA-mimic mediated silencing of these genes resulted in increased AcMNPV proliferation. Our findings reinforce the potential of AcMNPV as a potent biocontrol agent and further our understanding of developing biotechnology-based targeted pest control agents.
Collapse
Affiliation(s)
- Jie Zhang
- National
Key Laboratory of Green Pesticide, “Belt and Road” Technology
Industry and Innovation Institute for Green and Biological Control
of Agricultural Pests, College of Plant
Protection, South China Agricultural University, Guangzhou 510642, China
| | - Junaid Zafar
- National
Key Laboratory of Green Pesticide, “Belt and Road” Technology
Industry and Innovation Institute for Green and Biological Control
of Agricultural Pests, College of Plant
Protection, South China Agricultural University, Guangzhou 510642, China
| | - Jinrong Kong
- National
Key Laboratory of Green Pesticide, “Belt and Road” Technology
Industry and Innovation Institute for Green and Biological Control
of Agricultural Pests, College of Plant
Protection, South China Agricultural University, Guangzhou 510642, China
| | - Fei Wang
- National
Key Laboratory of Green Pesticide, “Belt and Road” Technology
Industry and Innovation Institute for Green and Biological Control
of Agricultural Pests, College of Plant
Protection, South China Agricultural University, Guangzhou 510642, China
| | - Xuehua Shao
- Institute
of Fruit Tree Research, Guangdong Academy of Agricultural Sciences,
Key Laboratory of South Subtropical Fruit Biology and Genetic Resource
Utilization, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Tropical and
Subtropical Fruit Tree Research, Guangzhou 510640, China
| | - Ruonan Zhang
- National
Key Laboratory of Green Pesticide, “Belt and Road” Technology
Industry and Innovation Institute for Green and Biological Control
of Agricultural Pests, College of Plant
Protection, South China Agricultural University, Guangzhou 510642, China
| | - Rui Pang
- National
Key Laboratory of Green Pesticide, “Belt and Road” Technology
Industry and Innovation Institute for Green and Biological Control
of Agricultural Pests, College of Plant
Protection, South China Agricultural University, Guangzhou 510642, China
| | - Hanhong Xu
- National
Key Laboratory of Green Pesticide, “Belt and Road” Technology
Industry and Innovation Institute for Green and Biological Control
of Agricultural Pests, College of Plant
Protection, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoxia Xu
- National
Key Laboratory of Green Pesticide, “Belt and Road” Technology
Industry and Innovation Institute for Green and Biological Control
of Agricultural Pests, College of Plant
Protection, South China Agricultural University, Guangzhou 510642, China
| | - Fengliang Jin
- National
Key Laboratory of Green Pesticide, “Belt and Road” Technology
Industry and Innovation Institute for Green and Biological Control
of Agricultural Pests, College of Plant
Protection, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
6
|
Hrithrik TH, Lee DH, Singh N, Vik A, Hammock BD, Kim Y. Insect immune resolution with EpOME/DiHOME and its dysregulation by their analogs leading to pathogen hypersensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.07.548078. [PMID: 37461499 PMCID: PMC10350063 DOI: 10.1101/2023.07.07.548078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Epoxyoctadecamonoenoic acids (EpOMEs) are epoxide derivatives of linoleic acid (9,12-octadecadienoic acid: LA). They are metabolized into dihydroxyoctadecamonoenoic acids (DiHOMEs) in mammals. Unlike in mammals where they act as adipokines or lipokines, EpOMEs act as immunosuppressants in insects. However, the functional link between EpOMEs and pro-immune mediators such as PGE2 is not known. In addition, the physiological significance of DiHOMEs is not clear in insects. This study analyzed the physiological role of these C18 oxylipins using a lepidopteran insect pest, Spodoptera exigua. Immune challenge of S. exigua rapidly upregulated the expression of the phospholipase A2 gene to trigger C20 oxylipin biosynthesis, followed by the upregulation of genes encoding EpOME synthase (SE51385) and a soluble epoxide hydrolase (Se-sEH). The sequential gene expression resulted in the upregulations of the corresponding gene products such as PGE2, EpOMEs, and DiHOMEs. Interestingly, only PGE2 injection without the immune challenge significantly upregulated the gene expression of SE51825 and Se-sEH. The elevated levels of EpOMEs acted as immunosuppressants by inhibiting cellular and humoral immune responses induced by the bacterial challenge, in which 12,13-EpOME was more potent than 9,10-EpOME. However, DiHOMEs did not inhibit the cellular immune responses but upregulated the expression of antimicrobial peptides selectively suppressed by EpOMEs. The negative regulation of insect immunity by EpOMEs and their inactive DiHOMEs were further validated by synthetic analogs of the linoleate epoxide and corresponding diol. Furthermore, inhibitors specific to Se-sEH used to prevent EpOME degradation significantly suppressed the immune responses. The data suggest a physiological role of C18 oxylipins in resolving insect immune response. Any immune dysregulation induced by EpOME analogs or sEH inhibitors significantly enhanced insect susceptibility to the entomopathogen, Bacillus thuringiensis.
Collapse
Affiliation(s)
| | - Dong-Hee Lee
- Industry Academy Cooperation Foundation, Andong National University, Andong 36729, Korea
| | - Nalin Singh
- Department of Entomology and Nematology, University of California, Davis, CA 95616, USA
- UCD Comprehensive Cancer Center, University of California, Sacramento, CA 95817, USA
| | - Anders Vik
- Department of Pharmacy, Section for Pharmaceutical Chemistry, University of Oslo, PO Box 1068 Blindern, N-0316 Oslo, Norway
| | - Bruce D Hammock
- Department of Entomology and Nematology, University of California, Davis, CA 95616, USA
- UCD Comprehensive Cancer Center, University of California, Sacramento, CA 95817, USA
| | - Yonggyun Kim
- Department of Plant Medicals, Andong National University, Andong 36729, Korea
| |
Collapse
|
7
|
Vidal-Quist JC, Declercq J, Vanhee S, Lambrecht BN, Gómez-Rial J, Vidal C, Aydogdu E, Rombauts S, Hernández-Crespo P. RNA viruses alter house dust mite physiology and allergen production with no detected consequences for allergenicity. INSECT MOLECULAR BIOLOGY 2023; 32:173-186. [PMID: 36511188 DOI: 10.1111/imb.12822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
RNA viruses have recently been detected in association with house dust mites, including laboratory cultures, dust samples, and mite-derived pharmaceuticals used for allergy diagnosis. This study aimed to assess the incidence of viral infection on Dermatophagoides pteronyssinus physiology and on the allergenic performance of extracts derived from its culture. Transcriptional changes between genetically identical control and virus-infected mite colonies were analysed by RNAseq with the support of a new D. pteronyssinus high-quality annotated genome (56.8 Mb, 108 scaffolds, N50 = 2.73 Mb, 96.7% BUSCO-completeness). Extracts of cultures and bodies from both colonies were compared by inspecting major allergen accumulation by enzyme-linked immunosorbent assay (ELISA), allergen-related enzymatic activities by specific assays, airway inflammation in a mouse model of allergic asthma, and binding to allergic patient's sera IgE by ImmunoCAP. Viral infection induced a significant transcriptional response, including several immunity and stress-response genes, and affected the expression of seven allergens, putative isoallergens and allergen orthologs. Major allergens were unaffected except for Der p 23 that was upregulated, increasing ELISA titers up to 29% in infected-mite extracts. By contrast, serine protease allergens Der p 3, 6 and 9 were downregulated, being trypsin and chymotrypsin enzymatic activities reduced up to 21% in extracts. None of the parameters analysed in our mouse model, nor binding to human IgE were significantly different when comparing control and infected-mite extracts. Despite the described physiological impact of viral infection on the mites, no significant consequences for the allergenicity of derived extracts or their practical use in allergy diagnosis have been detected.
Collapse
Affiliation(s)
- José Cristian Vidal-Quist
- Entomología Aplicada a la Agricultura y la Salud, Departamento de Biotecnología Microbiana y de Plantas, Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid, Spain
| | - Jozefien Declercq
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Stijn Vanhee
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - José Gómez-Rial
- Laboratorio de Inmunogenética, Unidad de Inmunología, Complejo Hospitalario Universitario de Santiago (CHUS), Santiago de Compostela, Spain
| | - Carmen Vidal
- Servicio de Alergología, Complejo Hospitalario Universitario de Santiago (CHUS), Santiago de Compostela, Spain
| | - Eylem Aydogdu
- Center for Plant Systems Biology, VIB, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Stephane Rombauts
- Center for Plant Systems Biology, VIB, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Pedro Hernández-Crespo
- Entomología Aplicada a la Agricultura y la Salud, Departamento de Biotecnología Microbiana y de Plantas, Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid, Spain
| |
Collapse
|
8
|
Attarianfar M, Mikani A, Mehrabadi M. Fenoxycarb exposure affects antiviral immunity and HaNPV infection in the cotton bollworm, Helicoverpa armigera. PEST MANAGEMENT SCIENCE 2023; 79:1078-1085. [PMID: 36424349 DOI: 10.1002/ps.7301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/25/2022] [Accepted: 11/25/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Application of insect growth regulators (IGR) is a good option for insect pest management because of their fewer adverse effects on humans and domestic animals. These compounds are capable of interfering with normal growth and development by mimicking the actions of hormones such as juvenile hormone (JH) or ecdysone. The effect of JH and its analogs on some aspects of insect immunity has been determined, yet their possible effects on antiviral immunity response has not been investigated yet. Considering the importance of antiviral response in viral replication, in this study the effects of the JH analog (JHA), fenoxycarb on the antiviral immunity pathway core genes [i.e. micro (mi)RNA, small interfering (si)RNA and apoptosis] of Helicoverpa armigera (Hubner) larvae were investigated. The effect of fenoxycarb on the susceptibility of the larvae to H. armigera nuclear polyhedrosis virus (HaNPV) also was assessed. RESULTS The results showed that the transcription level of miRNA (Dicer1, Ago1), siRNA (Dicer2, Ago2) and apoptosis (Caspase1, Caspase5) core genes in H. armigera larvae were decreased significantly after 24, 48 and 96 h feeding on a diet containing lethal and sublethal doses of fenoxycarb. Moreover, the mortality rate to HaNPV in the larvae treated with fenoxycarb increased compared to the control, leading to an increased replication of HaNPV. CONCLUSION Together, our results suggest that the antiviral immune system could be modulated by JHA and facilitate HaNPV replication in the larvae, increasing the mortality rate of the insect larvae. Understanding the effect of JHA on antiviral immunity is an important step toward the process of exploiting JHAs and viral pathogens to control insect pests. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Marzieh Attarianfar
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Azam Mikani
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Mehrabadi
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
9
|
Huang Z, Wang W, Xu P, Gong S, Hu Y, Liu Y, Su F, Anjum KM, Deng WM, Yang S, Liu J, Jiao R, Chen J. Drosophila Ectoderm-expressed 4 modulates JAK/STAT pathway and protects flies against Drosophila C virus infection. Front Immunol 2023; 14:1135625. [PMID: 36817462 PMCID: PMC9937023 DOI: 10.3389/fimmu.2023.1135625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Sterile alpha and HEAT/Armadillo motif-containing protein (SARM) is conserved in evolution and negatively regulates TRIF-dependent Toll signaling in mammals. The SARM protein from Litopenaeus vannamei and its Drosophila orthologue Ectoderm-expressed (Ect4) are also involved in immune defense against pathogen infection. However, the functional mechanism of the protective effect remains unclear. In this study, we show that Ect4 is essential for the viral load in flies after a Drosophila C virus (DCV) infection. Viral load is increased in Ect4 mutants resulting in higher mortality rates than wild-type. Overexpression of Ect4 leads to a suppression of virus replication and thus improves the survival rate of the animals. Ect4 is required for the viral induction of STAT-responsive genes, TotA and TotM. Furthermore, Ect4 interacts with Stat92E, affecting the tyrosine phosphorylation and nuclear translocation of Stat92E in S2 cells. Altogether, our study identifies the adaptor protein Ect4 of the Toll pathway contributes to resistance to viral infection and regulates JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Zongliang Huang
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China,Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Wang
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China,Department of Bioengineering and Biotechnology, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China
| | - Pengpeng Xu
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shangyu Gong
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yingshan Hu
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yan Liu
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Fang Su
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Khalid Mahmood Anjum
- Department of Wildlife and Ecology, University of Veterinary and Animal Sciences, Lahore, Punjab, Pakistan
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| | - Suping Yang
- Department of Bioengineering and Biotechnology, College of Chemical Engineering, Huaqiao University, Xiamen, Fujian, China
| | - Jiyong Liu
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China,*Correspondence: Jiyong Liu, ; Renjie Jiao, ; Jianming Chen,
| | - Renjie Jiao
- Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China,*Correspondence: Jiyong Liu, ; Renjie Jiao, ; Jianming Chen,
| | - Jianming Chen
- Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, Fujian, China,Sino-French Hoffmann Institute, School of Basic Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China,*Correspondence: Jiyong Liu, ; Renjie Jiao, ; Jianming Chen,
| |
Collapse
|
10
|
Nainu F, Ophinni Y, Shiratsuchi A, Nakanishi Y. Apoptosis and Phagocytosis as Antiviral Mechanisms. Subcell Biochem 2023; 106:77-112. [PMID: 38159224 DOI: 10.1007/978-3-031-40086-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Viruses are infectious entities that make use of the replication machinery of their hosts to produce more progenies, causing disease and sometimes death. To counter viral infection, metazoan hosts are equipped with various defense mechanisms, from the rapid-evoking innate immune responses to the most advanced adaptive immune responses. Previous research demonstrated that cells in fruit flies and mice infected with Drosophila C virus and influenza, respectively, undergo apoptosis, which triggers the engulfment of apoptotic virus-infected cells by phagocytes. This process involves the recognition of eat-me signals on the surface of virus-infected cells by receptors of specialized phagocytes, such as macrophages and neutrophils in mice and hemocytes in fruit flies, to facilitate the phagocytic elimination of virus-infected cells. Inhibition of phagocytosis led to severe pathologies and death in both species, indicating that apoptosis-dependent phagocytosis of virus-infected cells is a conserved antiviral mechanism in multicellular organisms. Indeed, our understanding of the mechanisms underlying apoptosis-dependent phagocytosis of virus-infected cells has shed a new perspective on how hosts defend themselves against viral infection. This chapter explores the mechanisms of this process and its potential for developing new treatments for viral diseases.
Collapse
Affiliation(s)
- Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia.
| | - Youdiil Ophinni
- Division of Clinical Virology, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan
- Laboratory of Host Defense, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Akiko Shiratsuchi
- Center for Medical Education, Sapporo Medical University, Sapporo, Japan
- Division of Biological Function and Regulation, Graduate School of Medicine, Sapporo Medical University, Sapporo, Japan
| | | |
Collapse
|
11
|
Zhang X, Mei Y, Li H, Tang M, He K, Xiao Q. Larval-Transcriptome Dynamics of Ectropis grisescens Reveals Differences in Virulence Mechanism between Two EcobNPV Strains. INSECTS 2022; 13:1088. [PMID: 36554998 PMCID: PMC9781159 DOI: 10.3390/insects13121088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
The biological insecticide, Ectropis obliqua nucleopolyhedrovirus (EcobNPV), has been applied to control the major tea-pest Ectropis grisescens. Previously, the virus strain EcobNPV-QF4 showed higher a mortality rate (58.2% vs. 88.2%) and shorter median lethal-time (13.9 d vs. 15.4 d) on E. grisescens than the strain EcobNPV-QV. However, the mechanism of the difference in virulence between the two strains remains unclear. Using the leaf-disc method, we detected the virulence of the two strains on 3rd-instar larvae, and found that median lethal-dose (LD50) of EcobNPV-QF4 is 55-fold higher than that of EcobNPV-QV (4.35 × 108 vs. 7.89 × 106). Furthermore, fourteen larva transcriptomes of E. grisescens were subsequently sequenced at seven time-points after ingestion of the two virus strains, yielding 410.72 Gb of raw reads. Differential gene-expression analysis shows that 595, 87, 27, 108, 0, 12, and 290 genes were up-regulated in EcobNPV-QF4 at 0, 2, 6, 12, 24, 36 h and 48 h post ingestion (hpi), while 744, 68, 152, 8, 1, 0, 225 were down-regulated. KEGG enrichment showed that when the virus first invades (eats the leaf-discs), EcobNPV-QF4 mainly affects pathways such as ribosome (p-value = 2.47 × 10-29), and at 48 hpi EcobNPV-QF4, causes dramatic changes in the amino-acid-synthesis pathway and ribosome pathway (p-value = 6.94 × 10-13) in E. grisescens. Among these, thirteen key genes related to immunity were screened. The present study provides the first ever comprehensive analysis of transcriptional changes in E. grisescens after ingestion of the two strains of EcobNPV.
Collapse
Affiliation(s)
- Xinxin Zhang
- Ministry of Agriculture Key Laboratory of Tea Quality and Safety Control, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou 310008, China
| | - Yang Mei
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Hong Li
- Ministry of Agriculture Key Laboratory of Tea Quality and Safety Control, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou 310008, China
| | - Meijun Tang
- Ministry of Agriculture Key Laboratory of Tea Quality and Safety Control, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou 310008, China
| | - Kang He
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, China
| | - Qiang Xiao
- Ministry of Agriculture Key Laboratory of Tea Quality and Safety Control, Tea Research Institute, Chinese Academy of Agricultural Sciences, Hangzhou 310008, China
| |
Collapse
|
12
|
Pathogen infection routes and host innate immunity: Lessons from insects. Immunol Lett 2022; 247:46-51. [PMID: 35667452 DOI: 10.1016/j.imlet.2022.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/19/2022] [Accepted: 05/30/2022] [Indexed: 11/23/2022]
Abstract
Recent advances in insect-pathogen interactions have started to reveal the role of insect tissues and organs as natural infection routes for parasites and microbial pathogens. Here we summarize this information highlighting the micro- and macro-parasites that enter insects through distinct infection routes and link them to innate immune activity. We also examine whether the infection route determines the insect immune response and if the resulting immunological and physiological processes underpinning these different routes of infection are clearly distinct. Understanding how the infection route is associated with the robustness in insect host defense will help us identify conserved evolutionary and ecological patterns in order to design novel strategies for the management of destructive agricultural pests and disease vectors.
Collapse
|
13
|
Harsh S, Tafesh-Edwards G, Eleftherianos I. Zika virus infection triggers the melanization response in Drosophila. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166424. [DOI: 10.1016/j.bbadis.2022.166424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/20/2022] [Indexed: 11/30/2022]
|
14
|
Godoy RSM, Felix LDS, Orfanó ADS, Chaves BA, Nogueira PM, Costa BDA, Soares AS, Oliveira CCA, Nacif-Pimenta R, Silva BM, Duarte AP, de Lacerda MVG, Monteiro WM, Secundino NFC, Pimenta PFP. Dengue and Zika virus infection patterns vary among Aedes aegypti field populations from Belo Horizonte, a Brazilian endemic city. PLoS Negl Trop Dis 2021; 15:e0009839. [PMID: 34727099 PMCID: PMC8562804 DOI: 10.1371/journal.pntd.0009839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 09/24/2021] [Indexed: 01/21/2023] Open
Abstract
Dengue virus (DENV) and Zika virus (ZIKV) belong to the same viral family, the Flaviviridae. They cause recurring threats to the public health systems of tropical countries such as Brazil. The primary Brazilian vector of both viruses is the mosquito Aedes aegypti. After the mosquito ingests a blood meal from an infected person, the viruses infect and replicate in the midgut, disseminate to secondary tissues and reach the salivary gland (SG), where they are ready to be transmitted to a vertebrate host. It is thought that the intrinsic discrepancies among mosquitoes could affect their ability to deal with viral infections. This study confirms that the DENV and ZIKV infection patterns of nine Ae. aegypti field populations found in geographically separate health districts of an endemic Brazilian city vary. We analyzed the infection rate, disseminated infection, vector competence, and viral load through quantitative PCR. Mosquitoes were challenged using the membrane-feeding assay technique and were tested seven and fourteen days post-infection (early and late infection phases, respectively). The infection responses varied among the Ae. aegypti populations for both flaviviruses in the two infection phases. There was no similarity between DENV and ZIKV vector competencies or viral loads. According to the results of our study, the risk of viral transmission overtime after infection either increases or remains unaltered in ZIKV infected vectors. However, the risk may increase, decrease, or remain unaltered in DENV-infected vectors depending on the mosquito population. For both flaviviruses, the viral load persisted in the body even until the late infection phase. In contrast to DENV, the ZIKV accumulated in the SG over time in all the mosquito populations. These findings are novel and may help direct the development of control strategies to fight dengue and Zika outbreaks in endemic regions, and provide a warning about the importance of understanding mosquito responses to arboviral infections. Dengue and Zika are neglected diseases caused by viruses transmitted to humans by mosquitoes (vector-borne diseases). The primary vector of both diseases is Aedes aegypti, a highly abundant mosquito in tropical countries and adapted to the urban habitat. The viral cycle in the vector starts when the mosquito bites an infected person and acquires the viruses through the blood meal. When the infected blood reaches the mosquito’s midgut, the viruses invade the epithelial cells and disseminate in several organs until they reach the salivary glands, enabling viral transmission to the next person. However, the mosquitoes have developed strategies to combat the viral invasion and dissemination in their body, making this journey a challenge to the viruses. Herein, we show that the mosquito responses against dengue and Zika viruses are distinct. In addition, mosquitoes from separate populations of the same city have different abilities to deal with the viruses in both cases, dengue and Zika infections. Our results show the diversity of responses that the mosquitoes may present to viral infections. These findings may better direct disease control strategies to combat dengue and Zika outbreaks in endemic regions.
Collapse
Affiliation(s)
| | - Luiza dos Santos Felix
- Instituto de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Biologia Celular, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Bárbara Aparecida Chaves
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | | | - Breno dos Anjos Costa
- Instituto de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
| | - Aline Silva Soares
- Instituto de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Biologia Celular, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Rafael Nacif-Pimenta
- Instituto de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
| | - Breno Mello Silva
- Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Ana Paula Duarte
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Marcus Vinicius Guimarães de Lacerda
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
- Instituto Leônidas e Maria Deane, FIOCRUZ, Manaus, Amazonas, Brazil
| | - Wuelton Marcelo Monteiro
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Nágila Francinete Costa Secundino
- Instituto de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Paulo Filemon Paolucci Pimenta
- Instituto de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
- Programa de Pós-Graduação em Biologia Celular, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
- * E-mail:
| |
Collapse
|
15
|
Eleftherianos I, Heryanto C, Bassal T, Zhang W, Tettamanti G, Mohamed A. Haemocyte-mediated immunity in insects: Cells, processes and associated components in the fight against pathogens and parasites. Immunology 2021; 164:401-432. [PMID: 34233014 PMCID: PMC8517599 DOI: 10.1111/imm.13390] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/28/2021] [Indexed: 12/27/2022] Open
Abstract
The host defence of insects includes a combination of cellular and humoral responses. The cellular arm of the insect innate immune system includes mechanisms that are directly mediated by haemocytes (e.g., phagocytosis, nodulation and encapsulation). In addition, melanization accompanying coagulation, clot formation and wound healing, nodulation and encapsulation processes leads to the formation of cytotoxic redox-cycling melanin precursors and reactive oxygen and nitrogen species. However, demarcation between cellular and humoral immune reactions as two distinct categories is not straightforward. This is because many humoral factors affect haemocyte functions and haemocytes themselves are an important source of many humoral molecules. There is also a considerable overlap between cellular and humoral immune functions that span from recognition of foreign intruders to clot formation. Here, we review these immune reactions starting with the cellular mechanisms that limit haemolymph loss and participate in wound healing and clot formation and advancing to cellular functions that are critical in restricting pathogen movement and replication. This information is important because it highlights that insect cellular immunity is controlled by a multilayered system, different components of which are activated by different pathogens or during the different stages of the infection.
Collapse
Affiliation(s)
- Ioannis Eleftherianos
- Infection and Innate Immunity LaboratoryDepartment of Biological SciencesInstitute for Biomedical SciencesThe George Washington UniversityWashingtonDCUSA
| | - Christa Heryanto
- Infection and Innate Immunity LaboratoryDepartment of Biological SciencesInstitute for Biomedical SciencesThe George Washington UniversityWashingtonDCUSA
| | - Taha Bassal
- Department of EntomologyFaculty of ScienceCairo UniversityGizaEgypt
| | - Wei Zhang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural BioengineeringKey Laboratory of Green Pesticide and Agricultural BioengineeringMinistry of EducationGuizhou UniversityGuiyangChina
| | - Gianluca Tettamanti
- Department of Biotechnology and Life SciencesUniversity of InsubriaVareseItaly
- BAT Center‐Interuniversity Center for Studies on Bioinspired Agro‐Environmental TechnologyUniversity of Napoli Federico IINapoliItaly
| | - Amr Mohamed
- Department of EntomologyFaculty of ScienceCairo UniversityGizaEgypt
| |
Collapse
|
16
|
Sheffield L, Sciambra N, Evans A, Hagedorn E, Goltz C, Delfeld M, Kuhns H, Fierst JL, Chtarbanova S. Age-dependent impairment of disease tolerance is associated with a robust transcriptional response following RNA virus infection in Drosophila. G3-GENES GENOMES GENETICS 2021; 11:6219303. [PMID: 33836060 PMCID: PMC8495950 DOI: 10.1093/g3journal/jkab116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/26/2021] [Indexed: 12/18/2022]
Abstract
Advanced age in humans is associated with greater susceptibility to and higher mortality rates from infections, including infections with some RNA viruses. The underlying innate immune mechanisms, which represent the first line of defense against pathogens, remain incompletely understood. Drosophila melanogaster is able to mount potent and evolutionarily conserved innate immune defenses against a variety of microorganisms including viruses and serves as an excellent model organism for studying host–pathogen interactions. With its relatively short lifespan, Drosophila also is an organism of choice for aging studies. Despite numerous advantages that this model offers, Drosophila has not been used to its full potential to investigate the response of the aged host to viral infection. Here, we show that, in comparison to younger flies, aged Drosophila succumb more rapidly to infection with the RNA-containing Flock House virus due to an age-dependent defect in disease tolerance. Relative to younger individuals, we find that older Drosophila mount transcriptional responses characterized by differential regulation of more genes and genes regulated to a greater extent. We show that loss of disease tolerance to Flock House virus with age associates with a stronger regulation of genes involved in apoptosis, some genes of the Drosophila immune deficiency NF-kB pathway, and genes whose products function in mitochondria and mitochondrial respiration. Our work shows that Drosophila can serve as a model to investigate host–virus interactions during aging and furthermore sets the stage for future analysis of the age-dependent mechanisms that govern survival and control of virus infections at older age.
Collapse
Affiliation(s)
- Lakbira Sheffield
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA.,Graduate Biomedical Sciences program, University of Alabama at Birmingham, Birmingham, AL- 35294, USA
| | - Noah Sciambra
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Alysa Evans
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Eli Hagedorn
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Casey Goltz
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Megan Delfeld
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Haley Kuhns
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Janna L Fierst
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| | - Stanislava Chtarbanova
- Department of Biological Sciences, University of Alabama, 300, Hackberry lane, Tuscaloosa, AL-35487, USA
| |
Collapse
|
17
|
Schneider J, Imler JL. Sensing and signalling viral infection in drosophila. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 117:103985. [PMID: 33358662 DOI: 10.1016/j.dci.2020.103985] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 06/12/2023]
Abstract
The fruitfly Drosophila melanogaster is a valuable model to unravel mechanisms of innate immunity, in particular in the context of viral infections. RNA interference, and more specifically the small interfering RNA pathway, is a major component of antiviral immunity in drosophila. In addition, the contribution of inducible transcriptional responses to the control of viruses in drosophila and other invertebrates is increasingly recognized. In particular, the recent discovery of a STING-IKKβ-Relish signalling cassette in drosophila has confirmed that NF-κB transcription factors play an important role in the control of viral infections, in addition to bacterial and fungal infections. Here, we review recent developments in the field, which begin to shed light on the mechanisms involved in sensing of viral infections and in signalling leading to production of antiviral effectors.
Collapse
Affiliation(s)
- Juliette Schneider
- Université de Strasbourg, CNRS UPR9022, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Jean-Luc Imler
- Université de Strasbourg, CNRS UPR9022, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
18
|
Wilson K, Grzywacz D, Cory JS, Donkersley P, Graham RI. Trans-generational viral transmission and immune priming are dose-dependent. J Anim Ecol 2021; 90:1560-1569. [PMID: 33724454 DOI: 10.1111/1365-2656.13476] [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] [Received: 02/02/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022]
Abstract
It is becoming increasingly apparent that trans-generational immune priming (i.e. the transfer of the parental immunological experience to its progeny resulting in offspring protection from pathogens that persist across generations) is a common phenomenon not only in vertebrates, but also invertebrates. Likewise, it is known that covert pathogenic infections may become 'triggered' into an overt infection by various stimuli, including exposure to heterologous infections. Yet, rarely have both phenomena been explored in parallel. Using as a model system the African armyworm Spodoptera exempta, an eruptive agricultural pest and its endemic dsDNA virus (Spodoptera exempta nucleopolyhedrovirus, SpexNPV), the aim of this study was to explore the impact of parental inoculating-dose on trans-generational pathogen transmission and immune priming (in its broadest sense). Larvae were orally challenged with one of five doses of SpexNPV and survivors from these treatments were mated and their offspring monitored for viral mortality. Offspring from parents challenged with low viral doses showed evidence of 'immune priming' (i.e. enhanced survival following SpexNPV challenge); in contrast, offspring from parents challenged with higher viral doses exhibited greater susceptibility to viral challenge. Most offspring larvae died of the virus they were orally challenged with; in contrast, most offspring from parents that had been challenged with the highest doses were killed by the vertically transmitted virus (90%) and not the challenge virus. These results demonstrate that the outcome of a potentially lethal virus challenge is critically dependent on the level of exposure to virus in the parental generation-either increasing resistance at very low parental viral doses (consistent with trans-generational immune priming) or increasing susceptibility at higher parental doses (consistent with virus triggering). We discuss the implications of these findings for understanding both natural epizootics of baculoviruses and for using them as biological control agents.
Collapse
Affiliation(s)
- Kenneth Wilson
- Lancaster Environment Centre, Lancaster University, Lancaster, UK
| | - David Grzywacz
- Department of Agriculture Health and Environment, Natural Resources Institute, University of Greenwich, Kent, UK
| | - Jenny S Cory
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | | | - Robert I Graham
- Lancaster Environment Centre, Lancaster University, Lancaster, UK
| |
Collapse
|
19
|
Wang F. Sending Out Alarms: A Perspective on Intercellular Communications in Insect Antiviral Immune Response. Front Immunol 2021; 12:613729. [PMID: 33708207 PMCID: PMC7940532 DOI: 10.3389/fimmu.2021.613729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/12/2021] [Indexed: 11/13/2022] Open
Abstract
Viral infection triggers insect immune response, including RNA interference, apoptosis and autophagy, and profoundly changes the gene expression profiles in infected cells. Although intracellular degradation is crucial for restricting viral infection, intercellular communication is required to mount a robust systemic immune response. This review focuses on recent advances in understanding the intercellular communications in insect antiviral immunity, including protein-based and virus-derived RNA based cell-cell communications, with emphasis on the signaling pathway that induces the production of the potential cytokines. The prospects and challenges of future work are also discussed.
Collapse
Affiliation(s)
- Fei Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China.,Biological Science Research Center, Southwest University, Chongqing, China
| |
Collapse
|
20
|
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
|
21
|
Harsh S, Eleftherianos I. Flavivirus Infection and Regulation of Host Immune and Tissue Homeostasis in Insects. Front Immunol 2020; 11:618801. [PMID: 33329613 PMCID: PMC7733989 DOI: 10.3389/fimmu.2020.618801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 11/16/2020] [Indexed: 11/28/2022] Open
Affiliation(s)
- Sneh Harsh
- Infection and Innate Immunity Lab, Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Washington, DC, United States.,Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, United States
| | - Ioannis Eleftherianos
- Infection and Innate Immunity Lab, Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Washington, DC, United States
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Samadder S. Drosophila melanogaster: A Robust Tool to Study Candidate Drug against Epidemic and Pandemic Diseases. ANIMAL MODELS IN MEDICINE AND BIOLOGY 2020. [DOI: 10.5772/intechopen.90073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
25
|
Abdeltawab MSA, Rifaie SA, Shoeib EY, El-Latif HAA, Badawi M, Salama WH, El-Aal AAA. Insights into the impact of Ivermectin on some protein aspects linked to Culex pipiens digestion and immunity. Parasitol Res 2019; 119:55-62. [DOI: 10.1007/s00436-019-06539-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 11/03/2019] [Indexed: 02/08/2023]
|
26
|
Caicedo PA, Serrato IM, Sim S, Dimopoulos G, Coatsworth H, Lowenberger C, Ocampo CB. Immune response-related genes associated to blocking midgut dengue virus infection in Aedes aegypti strains that differ in susceptibility. INSECT SCIENCE 2019; 26:635-648. [PMID: 29389079 DOI: 10.1111/1744-7917.12573] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/19/2017] [Accepted: 01/15/2018] [Indexed: 06/07/2023]
Abstract
Aedes (Stegomyia) aegypti, the principal global vector of dengue viruses, has differences in its susceptibility to dengue virus infection. We compared the global expression of genes in the midguts of Colombian Ae. aegypti dengue-susceptible (Cali-S) and dengue-refractory (Cali-MIB) field derived strains after ingesting either a sugarmeal, a bloodmeal, or a bloodmeal containing dengue virus serotype 2 (DENV-2). Microarray-based transcriptome analysis among treatments indicated a total of 4725 transcripts with differential expression between the two strains. Eleven genes were selected from different functional groups based on their significant up or down expression levels as well as reports in the literature suggesting they are associated with dengue virus elimination. We measured mRNA abundance of these 11 genes at 0, 8, 24, and 36 h postinfection using quantitative real time PCR (qPCR) to confirm the microarray results and assess any temporal patterns. Four genes were selected (Gram-negative binding protein-GNBP [AAEL009176], Niemann Pick Type-C2-NPC2 [AAEL015136], Keratinocyte lectin [AAEL009842], and Cathepsin-b [AAEL007585]) for knockdown experiments using RNA interference (RNAi) methodology to determine the phenotype (DENV-2 susceptible or refractory). Silencing GNBP, Cathepsin-b and Keratinocyte lectin reduced the percentage of mosquitoes with disseminated virus in the Cali-S strain to 8%, 20%, and 12% respectively compared with 96% in the controls. Silencing of NPC2 increased the percentage of mosquitos with disseminated virus infections in Cali-MIB to 66% compared with 35% in the controls. This study provides insight into genes that may contribute to the Cali-S susceptible and Cali-MIB refractory phenotypes in Ae. aegypti.
Collapse
Affiliation(s)
- Paola A Caicedo
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
| | - Idalba Mildred Serrato
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
| | - Shuzhen Sim
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Heather Coatsworth
- Department of Biological Sciences, Simon Fraser University, Burnaby, Canada
| | - Carl Lowenberger
- Department of Biological Sciences, Simon Fraser University, Burnaby, Canada
| | - Clara B Ocampo
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
| |
Collapse
|
27
|
Homologs of Human Dengue-Resistance Genes, FKBP1B and ATCAY, Confer Antiviral Resistance in Aedes aegypti Mosquitoes. INSECTS 2019; 10:insects10020046. [PMID: 30717390 PMCID: PMC6409984 DOI: 10.3390/insects10020046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 01/26/2019] [Accepted: 01/29/2019] [Indexed: 02/06/2023]
Abstract
Dengue virus (DENV) is transmitted by mosquitoes and is a major public health concern. The study of innate mosquito defense mechanisms against DENV have revealed crucial roles for the Toll, Imd, JAK-STAT, and RNAi pathways in mediating DENV in the mosquito. Often overlooked in such studies is the role of intrinsic cellular defense mechanisms that we hypothesize to work in concert with the classical immune pathways to affect organismal defense. Our understanding of the molecular interaction of DENV with mosquito host cells is limited, and we propose to expand upon the recent results from a genome-scale, small interfering RNA (siRNA)-based study that identified mammalian host proteins associated with resistance to dengue/West Nile virus (DENV/WNV) infection. The study identified 22 human DENV/WNV resistance genes (DVR), and we hypothesized that a subset would be functionally conserved in Aedes aegypti mosquitoes, imparting cellular defense against flaviviruses in this species. We identified 12 homologs of 22 human DVR genes in the Ae. aegypti genome. To evaluate their possible role in cellular resistance/antiviral defense against DENV, we used siRNA silencing targeted against each of the 12 homologs in an Ae. aegypti cell line (Aag2) infected with DENV2 and identified that silencing of the two candidates, AeFKBP1 and AeATCAY, homologs of human FKBP1B and ATCAY, were associated with a viral increase. We then used dsRNA to silence each of the two genes in adult mosquitoes to validate the observed antiviral functions in vivo. Depletion of AeFKBP1 or AeATCAY increased viral dissemination through the mosquito at 14 days post-infection. Our results demonstrated that AeFKBP1 and AeATCAY mediate resistance to DENV akin to what has been described for their homologs in humans. AeFKBP1 and AeATCAY provide a rare opportunity to elucidate a DENV-resistance mechanism that may be evolutionarily conserved between humans and Ae. aegypti.
Collapse
|
28
|
Zhou L. P53 and Apoptosis in the Drosophila Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1167:105-112. [PMID: 31520351 DOI: 10.1007/978-3-030-23629-8_6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Human P53 (HsP53) is the most frequently mutated gene associated with cancers. Despite heightened research interest over the last four decades, a clear picture of how wild type HsP53 functions as the guardian against malignant transformation remains elusive. Studying the ortholog of P53 in the genetic model organism Drosophila melanogaster (DmP53) has revealed many interesting insights. This chapter focuses on recent findings that have shed light on how DmP53 -mediated apoptosis plays an important role in maintaining genome integrity, and how the immediate output of activated DmP53 is determined by the epigenetic landscape of individual cells.
Collapse
Affiliation(s)
- Lei Zhou
- Department of Molecular Genetics and Microbiology, UF Health Cancer Center, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
29
|
Erekat NS, Al-Jarrah MD. Association of Parkinson Disease Induction with Cardiac Upregulation of Apoptotic Mediators P53 and Active Caspase-3: An Immunohistochemistry Study. Med Sci Monit Basic Res 2018; 24:120-126. [PMID: 30135418 PMCID: PMC6118164 DOI: 10.12659/msmbr.910307] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/22/2018] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Apoptosis plays a key role in the pathogenesis of Parkinson disease (PD). Active caspase-3, which is a proapoptotic factor, has been shown to reduce cardiac contractility, causing cardiac dysfunction in many pathological diseases. Reduced cardiac contractility and cardiac autonomic dysfunction have been reported in PD patients and PD mice treated with MPTP. The aim of this study was to show the impact of PD induction on the expression of the apoptotic mediators p53 and active caspase-3 in the heart. MATERIAL AND METHODS Equal control and PD groups were formed by 20 randomly selected normal albino mice. We used 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (25 mg/kg) and probenecid (250 mg/kg) (MPTP/p) to induce chronic Parkinsonism in the PD group. Immunohistochemistry was performed to investigate the expression of p53, active caspase-3, and β-adrenergic receptor in hearts from the 2 animal groups. RESULTS P53 and active caspase-3 expression was significantly higher in PD hearts than in the control hearts (p value <0.01). β-adrenergic receptor expression was significantly lower in PD hearts than in control hearts (p value <0.01). CONCLUSIONS Our results show an association of PD with p53 and active caspase-3 overexpression and β-adrenergic receptor underexpression in the heart, potentially promoting the cardiac autonomic dysfunction frequently observed in PD.
Collapse
Affiliation(s)
- Nour S. Erekat
- Department of Anatomy, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Muhammed D. Al-Jarrah
- Department of Rehabilitation Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
30
|
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
|
31
|
Chen TH, Wu YJ, Hou JN, Chiang YH, Cheng CC, Sifiyatun E, Chiu CH, Wang LC, Chen WJ. A novel p53 paralogue mediates antioxidant defense of mosquito cells to survive dengue virus replication. Virology 2018; 519:156-169. [PMID: 29727815 DOI: 10.1016/j.virol.2018.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 03/31/2018] [Accepted: 04/16/2018] [Indexed: 01/22/2023]
Abstract
Mosquito cells allow dengue viruses (DENVs) to undergo replication without causing serious deleterious effects on the cells, leading to advantages for dissemination to other cells. Despite this, increased accumulation of reactive oxygen species (ROS) is usually detected in C6/36 cells with DENV2 infection as shown in mammalian cells. Uniquely, oxidative stress caused by the ROS is alleviated by eliciting antioxidant defense which leads to protection of mosquito cells from the infection. In the present study, a novel p53 paralogue (p53-2) was identified and proved to be regulated in C6/36 cells with DENV2 infection. With a gene-knockdown technique, p53-2 was demonstrated to transcribe catalase which plays a critical role in reducing ROS accumulation and the death rate of infected cells. Ecologically, a higher survival rate of mosquito cells is a prerequisite for prosperous production of viral progeny, allowing infected mosquitoes to remain healthy and active for virus transmission.
Collapse
Affiliation(s)
- Tien-Huang Chen
- Departments of Public Health and Parasitology, Chang Gung University, Kwei-San, Tao-Yuan 33332, Taiwan
| | - Yi-Jun Wu
- Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-San, Tao-Yuan 33332, Taiwan
| | - Jiun-Nan Hou
- Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-San, Tao-Yuan 33332, Taiwan
| | - Yi-Hsuan Chiang
- Departments of Public Health and Parasitology, Chang Gung University, Kwei-San, Tao-Yuan 33332, Taiwan
| | - Chih-Chieh Cheng
- Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-San, Tao-Yuan 33332, Taiwan
| | - Eny Sifiyatun
- Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-San, Tao-Yuan 33332, Taiwan; Program in Tropical Medical Science, Gadjah Mada University, Yogyakartan, Indonesia
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Kwei-San, Tao-Yuan, Taiwan; Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Children's Hospital, Chang Gung University College of Medicine, Kwei-San, Tao-Yuan, Taiwan
| | - Lian-Chen Wang
- Departments of Public Health and Parasitology, Chang Gung University, Kwei-San, Tao-Yuan 33332, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-San, Tao-Yuan 33332, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Kwei-San, Tao-Yuan, Taiwan.
| | - Wei-June Chen
- Departments of Public Health and Parasitology, Chang Gung University, Kwei-San, Tao-Yuan 33332, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Kwei-San, Tao-Yuan 33332, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Kwei-San, Tao-Yuan, Taiwan.
| |
Collapse
|
32
|
Abstract
The power and ease of Drosophila genetics and the medical relevance of mosquito-transmitted viruses have made dipterans important model organisms in antiviral immunology. Studies of virus-host interactions at the molecular and population levels have illuminated determinants of resistance to virus infection. Here, we review the sources and nature of variation in antiviral immunity and virus susceptibility in model dipteran insects, specifically the fruit fly Drosophila melanogaster and vector mosquitoes of the genera Aedes and Culex. We first discuss antiviral immune mechanisms and describe the virus-specificity of these responses. In the following sections, we review genetic and microbiota-dependent variation in antiviral immunity. In the final sections, we explore less well-studied sources of variation, including abiotic factors, sexual dimorphism, infection history, and endogenous viral elements. We borrow from work on other pathogen types and non-dipteran species when it parallels or complements studies in dipterans. Understanding natural variation in virus-host interactions may lead to the identification of novel restriction factors and immune mechanisms and shed light on the molecular determinants of vector competence.
Collapse
Affiliation(s)
- William H Palmer
- Institute of Evolutionary Biology and Centre for Infection, Evolution and Immunity, University of Edinburgh, Edinburgh EH9 3FL UK.
| | - Finny S Varghese
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, Nijmegen 6500 HB, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands.
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, Nijmegen 6500 HB, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands.
| |
Collapse
|
33
|
Bartholomay LC, Michel K. Mosquito Immunobiology: The Intersection of Vector Health and Vector Competence. ANNUAL REVIEW OF ENTOMOLOGY 2018; 63:145-167. [PMID: 29324042 DOI: 10.1146/annurev-ento-010715-023530] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
As holometabolous insects that occupy distinct aquatic and terrestrial environments in larval and adult stages and utilize hematophagy for nutrient acquisition, mosquitoes are subjected to a wide variety of symbiotic interactions. Indeed, mosquitoes play host to endosymbiotic, entomopathogenic, and mosquito-borne organisms, including protozoa, viruses, bacteria, fungi, fungal-like organisms, and metazoans, all of which trigger and shape innate infection-response capacity. Depending on the infection or interaction, the mosquito may employ, for example, cellular and humoral immune effectors for septic infections in the hemocoel, humoral infection responses in the midgut lumen, and RNA interference and programmed cell death for intracellular pathogens. These responses often function in concert, regardless of the infection type, and provide a robust front to combat infection. Mosquito-borne pathogens and entomopathogens overcome these immune responses, employing avoidance or suppression strategies. Burgeoning methodologies are capitalizing on this concerted deployment of immune responses to control mosquito-borne disease.
Collapse
Affiliation(s)
- Lyric C Bartholomay
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Wisconsin 53706;
| | - Kristin Michel
- Division of Biology, Kansas State University, Manhattan, Kansas 66506;
| |
Collapse
|
34
|
Wang Z, Xia X, Yang X, Zhang X, Liu Y, Wu D, Fang Y, Liu Y, Xu J, Qiu Y, Zhou X. A picorna-like virus suppresses the N-end rule pathway to inhibit apoptosis. eLife 2017; 6:30590. [PMID: 29231806 PMCID: PMC5739542 DOI: 10.7554/elife.30590] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/11/2017] [Indexed: 12/21/2022] Open
Abstract
The N-end rule pathway is an evolutionarily conserved proteolytic system that degrades proteins containing N-terminal degradation signals called N-degrons, and has emerged as a key regulator of various processes. Viruses manipulate diverse host pathways to facilitate viral replication and evade antiviral defenses. However, it remains unclear if viral infection has any impact on the N-end rule pathway. Here, using a picorna-like virus as a model, we found that viral infection promoted the accumulation of caspase-cleaved Drosophila inhibitor of apoptosis 1 (DIAP1) by inducing the degradation of N-terminal amidohydrolase 1 (NTAN1), a key N-end rule component that identifies N-degron to initiate the process. The virus-induced NTAN1 degradation is independent of polyubiquitylation but dependent on proteasome. Furthermore, the virus-induced N-end rule pathway suppression inhibits apoptosis and benefits viral replication. Thus, our findings demonstrate that a virus can suppress the N-end rule pathway, and uncover a new mechanism for virus to evade apoptosis.
Collapse
Affiliation(s)
- Zhaowei Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xiaoling Xia
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology & School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xueli Yang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xueyi Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yongxiang Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Di Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yuan Fang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yujie Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jiuyue Xu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yang Qiu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xi Zhou
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China.,State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
35
|
Mondotte JA, Saleh MC. Antiviral Immune Response and the Route of Infection in Drosophila melanogaster. Adv Virus Res 2017; 100:247-278. [PMID: 29551139 DOI: 10.1016/bs.aivir.2017.10.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The use of Drosophila as a model organism has made an important contribution to our understanding of the function and regulation of innate immunity in insects. Indeed, insects can discriminate between different types of pathogens and mount specific and effective responses. Strikingly, the same pathogen can trigger a different immune response in the same organism, depending solely on the route of infection by which the pathogen is delivered. In this review, we recapitulate what is known about antiviral responses in Drosophila, and how they are triggered depending on the route and the mode used for the virus to infect its host.
Collapse
Affiliation(s)
- Juan A Mondotte
- Institut Pasteur, Viruses and RNA Interference Unit, CNRS Unité Mixte de Recherche 3569, Paris, France
| | - Maria-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference Unit, CNRS Unité Mixte de Recherche 3569, Paris, France.
| |
Collapse
|
36
|
Vogels CB, Göertz GP, Pijlman GP, Koenraadt CJ. Vector competence of European mosquitoes for West Nile virus. Emerg Microbes Infect 2017; 6:e96. [PMID: 29116220 PMCID: PMC5717085 DOI: 10.1038/emi.2017.82] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/23/2017] [Accepted: 08/27/2017] [Indexed: 01/02/2023]
Abstract
West Nile virus (WNV) is an arthropod-borne flavivirus of high medical and veterinary importance. The main vectors for WNV are mosquito species of the Culex genus that transmit WNV among birds, and occasionally to humans and horses, which are ‘dead-end’ hosts. Recently, several studies have been published that aimed to identify the mosquito species that serve as vectors for WNV in Europe. These studies provide insight in factors that can influence vector competence of European mosquito species for WNV. Here, we review the current knowledge on vector competence of European mosquitoes for WNV, and the molecular knowledge on physical barriers, anti-viral pathways and microbes that influence vector competence based on studies with other flaviviruses. By comparing the 12 available WNV vector competence studies with European mosquitoes we evaluate the effect of factors such as temperature, mosquito origin and mosquito biotype on vector competence. In addition, we propose a standardised methodology to allow for comparative studies across Europe. Finally, we identify knowledge gaps regarding vector competence that, once addressed, will provide important insights into WNV transmission and ultimately contribute to effective strategies to control WNV.
Collapse
Affiliation(s)
- Chantal Bf Vogels
- Laboratory of Entomology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| | - Giel P Göertz
- Laboratory of Virology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| | - Gorben P Pijlman
- Laboratory of Virology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| | - Constantianus Jm Koenraadt
- Laboratory of Entomology, Wageningen University & Research, P.O. Box 16, 6700 AA, Wageningen, The Netherlands
| |
Collapse
|
37
|
Nainu F, Shiratsuchi A, Nakanishi Y. Induction of Apoptosis and Subsequent Phagocytosis of Virus-Infected Cells As an Antiviral Mechanism. Front Immunol 2017; 8:1220. [PMID: 29033939 PMCID: PMC5624992 DOI: 10.3389/fimmu.2017.01220] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/14/2017] [Indexed: 01/14/2023] Open
Abstract
Viruses are infectious entities that hijack host replication machineries to produce their progeny, resulting, in most cases, in disease and, sometimes, in death in infected host organisms. Hosts are equipped with an array of defense mechanisms that span from innate to adaptive as well as from humoral to cellular immune responses. We previously demonstrated that mouse cells underwent apoptosis in response to influenza virus infection. These apoptotic, virus-infected cells were then targeted for engulfment by macrophages and neutrophils. We more recently reported similar findings in the fruit fly Drosophila melanogaster, which lacks adaptive immunity, after an infection with Drosophila C virus. In these experiments, the inhibition of phagocytosis led to severe influenza pathologies in mice and early death in Drosophila. Therefore, the induction of apoptosis and subsequent phagocytosis of virus-infected cells appear to be an antiviral innate immune mechanism that is conserved among multicellular organisms. We herein discuss the underlying mechanisms and significance of the apoptosis-dependent phagocytosis of virus-infected cells. Investigations on the molecular and cellular features responsible for this underrepresented virus–host interaction may provide a promising avenue for the discovery of novel substances that are targeted in medical treatments against virus-induced intractable diseases.
Collapse
Affiliation(s)
- Firzan Nainu
- Laboratory of Pharmacology and Toxicology, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia.,Laboratory of Host Defense and Responses, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Akiko Shiratsuchi
- Laboratory of Host Defense and Responses, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshinobu Nakanishi
- Laboratory of Host Defense and Responses, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
38
|
Mussabekova A, Daeffler L, Imler JL. Innate and intrinsic antiviral immunity in Drosophila. Cell Mol Life Sci 2017; 74:2039-2054. [PMID: 28102430 PMCID: PMC5419870 DOI: 10.1007/s00018-017-2453-9] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/11/2016] [Accepted: 01/03/2017] [Indexed: 02/07/2023]
Abstract
The fruit fly Drosophila melanogaster has been a valuable model to investigate the genetic mechanisms of innate immunity. Initially focused on the resistance to bacteria and fungi, these studies have been extended to include antiviral immunity over the last decade. Like all living organisms, insects are continually exposed to viruses and have developed efficient defense mechanisms. We review here our current understanding on antiviral host defense in fruit flies. A major antiviral defense in Drosophila is RNA interference, in particular the small interfering (si) RNA pathway. In addition, complex inducible responses and restriction factors contribute to the control of infections. Some of the genes involved in these pathways have been conserved through evolution, highlighting loci that may account for susceptibility to viral infections in humans. Other genes are not conserved and represent species-specific innovations.
Collapse
Affiliation(s)
- Assel Mussabekova
- Institut de Biologie Moléculaire et Cellulaire, CNRS UPR9022, Université de Strasbourg, 15 rue René Descartes, 67000, Strasbourg, France.
| | - Laurent Daeffler
- Institut de Biologie Moléculaire et Cellulaire, CNRS UPR9022, Université de Strasbourg, 15 rue René Descartes, 67000, Strasbourg, France
| | - Jean-Luc Imler
- Institut de Biologie Moléculaire et Cellulaire, CNRS UPR9022, Université de Strasbourg, 15 rue René Descartes, 67000, Strasbourg, France
- Faculté des Sciences de la Vie, Université de Strasbourg, 28 rue Goethe, 67000, Strasbourg, France
| |
Collapse
|
39
|
El Chamy L, Matt N, Reichhart JM. Advances in Myeloid-Like Cell Origins and Functions in the Model Organism Drosophila melanogaster. Microbiol Spectr 2017; 5:10.1128/microbiolspec.mchd-0038-2016. [PMID: 28102122 PMCID: PMC11687447 DOI: 10.1128/microbiolspec.mchd-0038-2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Indexed: 12/22/2022] Open
Abstract
Drosophila has long served as a valuable model for deciphering many biological processes, including immune responses. Indeed, the genetic tractability of this organism is particularly suited for large-scale analyses. Studies performed during the last 3 decades have proven that the signaling pathways that regulate the innate immune response are conserved between Drosophila and mammals. This review summarizes the recent advances on Drosophila hematopoiesis and immune cellular responses, with a particular emphasis on phagocytosis.
Collapse
Affiliation(s)
- Laure El Chamy
- Laboratoire de Génétique de la drosophile et virulence microbienne, UR. EGFEM, Faculté des Sciences, Université Saint-Joseph de Beyrouth, B.P. 17-5208 Mar Mikhaël Beyrouth 1104 2020, Liban
| | - Nicolas Matt
- Université de Strasbourg, UPR 9022 du CNRS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg Cedex 67084, France
| | - Jean-Marc Reichhart
- Université de Strasbourg, UPR 9022 du CNRS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg Cedex 67084, France
| |
Collapse
|
40
|
Saraiva RG, Kang S, Simões ML, Angleró-Rodríguez YI, Dimopoulos G. Mosquito gut antiparasitic and antiviral immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 64:53-64. [PMID: 26827888 DOI: 10.1016/j.dci.2016.01.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/16/2016] [Accepted: 01/26/2016] [Indexed: 06/05/2023]
Abstract
Mosquitoes are responsible for the transmission of diseases with a serious impact on global human health, such as malaria and dengue. All mosquito-transmitted pathogens complete part of their life cycle in the insect gut, where they are exposed to mosquito-encoded barriers and active factors that can limit their development. Here we present the current understanding of mosquito gut immunity against malaria parasites, filarial worms, and viruses such as dengue, Chikungunya, and West Nile. The most recently proposed immune mediators involved in intestinal defenses are discussed, as well as the synergies identified between the recognition of gut microbiota and the mounting of the immune response.
Collapse
Affiliation(s)
- Raúl G Saraiva
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Seokyoung Kang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Yesseinia I Angleró-Rodríguez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
41
|
Marques JT, Imler JL. The diversity of insect antiviral immunity: insights from viruses. Curr Opin Microbiol 2016; 32:71-76. [PMID: 27232381 DOI: 10.1016/j.mib.2016.05.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/03/2016] [Indexed: 11/30/2022]
Abstract
Insects represent over 70% of all animal species. Recent virome analyses reveal unprecedented genetic diversity of insect viruses, which appears to match that of their hosts. Thus, insect-virus interactions may provide information on a vast repertoire of antiviral immune mechanisms. Tapping into this diversity is challenging because of several constraints imposed by the uniqueness of each insect model. Nevertheless, it is clear that many conserved and divergent pathways participate in the control of viral infection in insects. Co-evolution between hosts and viruses favors the development of immune evasion mechanisms by the pathogen. Viral suppressors can offer unique perspective on host pathways and emphasize the importance of RNA interference, apoptosis, but also NF-κB pathways and translation control in insect antiviral immunity.
Collapse
Affiliation(s)
- João T Marques
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil; CNRS UPR9022, IBMC, Strasbourg, France.
| | - Jean-Luc Imler
- CNRS UPR9022, IBMC, Strasbourg, France; Faculté des Sciences de la Vie, Université de Strasbourg, Strasbourg, France.
| |
Collapse
|
42
|
Analysis of the Contribution of Hemocytes and Autophagy to Drosophila Antiviral Immunity. J Virol 2016; 90:5415-5426. [PMID: 27009948 DOI: 10.1128/jvi.00238-16] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/16/2016] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Antiviral immunity in the model organism Drosophila melanogaster involves the broadly active intrinsic mechanism of RNA interference (RNAi) and virus-specific inducible responses. Here, using a panel of six viruses, we investigated the role of hemocytes and autophagy in the control of viral infections. Injection of latex beads to saturate phagocytosis, or genetic depletion of hemocytes, resulted in decreased survival and increased viral titers following infection with Cricket paralysis virus (CrPV), Flock House virus (FHV), and vesicular stomatitis virus (VSV) but had no impact on Drosophila C virus (DCV), Sindbis virus (SINV), and Invertebrate iridescent virus 6 (IIV6) infection. In the cases of CrPV and FHV, apoptosis was induced in infected cells, which were phagocytosed by hemocytes. In contrast, VSV did not trigger any significant apoptosis but we confirmed that the autophagy gene Atg7 was required for full virus resistance, suggesting that hemocytes use autophagy to recognize the virus. However, this recognition does not depend on the Toll-7 receptor. Autophagy had no impact on DCV, CrPV, SINV, or IIV6 infection and was required for replication of the sixth virus, FHV. Even in the case of VSV, the increases in titers were modest in Atg7 mutant flies, suggesting that autophagy does not play a major role in antiviral immunity in Drosophila Altogether, our results indicate that, while autophagy plays a minor role, phagocytosis contributes to virus-specific immune responses in insects. IMPORTANCE Phagocytosis and autophagy are two cellular processes that involve lysosomal degradation and participate in Drosophila immunity. Using a panel of RNA and DNA viruses, we have addressed the contribution of phagocytosis and autophagy in the control of viral infections in this model organism. We show that, while autophagy plays a minor role, phagocytosis contributes to virus-specific immune responses in Drosophila This work brings to the front a novel facet of antiviral host defense in insects, which may have relevance in the control of virus transmission by vector insects or in the resistance of beneficial insects to viral pathogens.
Collapse
|
43
|
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
|
44
|
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
|
45
|
Cytokine Diedel and a viral homologue suppress the IMD pathway in Drosophila. Proc Natl Acad Sci U S A 2016; 113:698-703. [PMID: 26739560 DOI: 10.1073/pnas.1516122113] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Viruses are obligatory intracellular parasites that suffer strong evolutionary pressure from the host immune system. Rapidly evolving viral genomes can adapt to this pressure by acquiring genes that counteract host defense mechanisms. For example, many vertebrate DNA viruses have hijacked cellular genes encoding cytokines or cytokine receptors to disrupt host cell communication. Insect viruses express suppressors of RNA interference or apoptosis, highlighting the importance of these cell intrinsic antiviral mechanisms in invertebrates. Here, we report the identification and characterization of a family of proteins encoded by insect DNA viruses that are homologous to a 12-kDa circulating protein encoded by the virus-induced Drosophila gene diedel (die). We show that die mutant flies have shortened lifespan and succumb more rapidly than controls when infected with Sindbis virus. This reduced viability is associated with deregulated activation of the immune deficiency (IMD) pathway of host defense and can be rescued by mutations in the genes encoding the homolog of IKKγ or IMD itself. Our results reveal an endogenous pathway that is exploited by insect viruses to modulate NF-κB signaling and promote fly survival during the antiviral response.
Collapse
|
46
|
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
|
47
|
Wang L, Yin Z, Wang Y, Lu Y, Zhang D, Srivastava A, Ling C, Aslanidi GV, Ling C. Productive life cycle of adeno-associated virus serotype 2 in the complete absence of a conventional polyadenylation signal. J Gen Virol 2015; 96:2780-2787. [PMID: 26297494 DOI: 10.1099/jgv.0.000229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We showed that WT adeno-associated virus serotype 2 (AAV2) genome devoid of a conventional polyadenylation [poly(A)] signal underwent complete genome replication, encapsidation and progeny virion production in the presence of adenovirus. The infectivity of the progeny virion was also retained. Using recombinant AAV2 vectors devoid of a human growth hormone poly(A) signal, we also demonstrated that a subset of mRNA transcripts contained the inverted terminal repeat (ITR) sequence at the 3' end, which we designated ITR in RNA (ITRR). Furthermore, AAV replication (Rep) proteins were able to interact with the ITRR. Taken together, our studies suggest a new function of the AAV2 ITR as an RNA element to mediate transgene expression from poly(A)-deleted mRNA.
Collapse
Affiliation(s)
- Lina Wang
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA.,Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, PR China.,Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Zifei Yin
- Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, PR China.,Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA.,Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Yuan Wang
- Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, PR China.,Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA.,Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Yuan Lu
- Department of Orthopedics and Rehabilitation, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Daniel Zhang
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Arun Srivastava
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL, 32611, USA.,Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA.,Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL 32611, USA.,Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA.,Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Changquan Ling
- Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, PR China.,E-Institute of TCM Internal Medicine, Shanghai Municipal Education Commission, Shanghai 201203, PR China
| | - George V Aslanidi
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA.,Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA.,Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA.,Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA.,Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL, 32611, USA
| |
Collapse
|
48
|
Erekat NS. Apoptotic Mediators are Upregulated in the Skeletal Muscle of Chronic/Progressive Mouse Model of Parkinson's Disease. Anat Rec (Hoboken) 2015; 298:1472-8. [DOI: 10.1002/ar.23124] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 01/10/2023]
Affiliation(s)
- Nour S. Erekat
- Department of Anatomy; Faculty of Medicine; Jordan University of Science and Technology (JUST); Irbid Jordan
| |
Collapse
|
49
|
Zhang YH, Wang Y, Yusufali AH, Ashby F, Zhang D, Yin ZF, Aslanidi GV, Srivastava A, Ling CQ, Ling C. Cytotoxic genes from traditional Chinese medicine inhibit tumor growth both in vitro and in vivo. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2015; 12:483-94. [PMID: 25412666 DOI: 10.1016/s2095-4964(14)60057-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Little effort has been made to study the protein-encoding genes isolated from traditional Chinese medicine (TCM) drugs, and the delivery of these genes into malignant cells through recombinant adeno-associated virus (rAAV) vectors has not been attempted. METHODS We synthesized the cDNAs of five known cytotoxic proteins isolated from TCM drugs and the FLAG epitope-tagged cDNAs were subcloned into a rAAV plasmid vector. The protein expression was confirmed by Western blot assay. Various cancer cell lines were transfected with the above plasmids and cell growth was monitored both in vitro and in vivo. The best cytotoxic gene was further packaged into rAAV vectors, under the control of a liver cancer-specific promoter. The liver tumor growth was then monitored following intratumor administration of the rAAV vectors. RESULTS The expression plasmids, encoding individual potential cytotoxic genes tagged with FLAG epitope, were successfully generated and sequenced. Among these genes, trichosanthin (TCS) gene yielded the most promising results for the inhibition of cancer cell growth in vitro. The over-expressed TCS functioned as a type I ribosome-inactivating protein, followed by inducing apoptosis that is associated with the Bcl-PARP signaling pathway. Furthermore, intratumor injection of rAAV vectors containing the TCS gene significantly inhibited the growth of human hepatocellular carcinoma tumors in a murine xenograft model. CONCLUSION Our studies suggest that the use of TCM cytotoxic genes is a useful therapeutic strategy for treating human cancers in general, and liver tumors in particular.
Collapse
Affiliation(s)
- Yuan-hui Zhang
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai 200433, China
| | - Yuan Wang
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai 200433, China
| | - Ali Hussein Yusufali
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32611, USA
| | - Frederick Ashby
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32611, USA
| | - Daniel Zhang
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32611, USA
| | - Zi-fei Yin
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai 200433, China
| | - George V Aslanidi
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32611, USA
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32611, USA
| | - Chang-quan Ling
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai 200433, China; E-mail:
| | - Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida 32611, USA; E-mail:
| |
Collapse
|
50
|
Rapid selection against arbovirus-induced apoptosis during infection of a mosquito vector. Proc Natl Acad Sci U S A 2015; 112:E1152-61. [PMID: 25713358 DOI: 10.1073/pnas.1424469112] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Millions of people are infected each year by arboviruses (arthropod-borne viruses) such as chikungunya, dengue, and West Nile viruses, yet for reasons that are largely unknown, only a relatively small number of mosquito species are able to transmit arboviruses. Understanding the complex factors that determine vector competence could facilitate strategies for controlling arbovirus infections. Apoptosis is a potential antiviral defense response that has been shown to be important in other virus-host systems. However, apoptosis is rarely seen in arbovirus-infected mosquito cells, raising questions about its importance as an antiviral defense in mosquitoes. We tested the effect of stimulating apoptosis during arbovirus infection by infecting Aedes aegypti mosquitoes with a Sindbis virus (SINV) clone called MRE/Rpr, in which the MRE-16 strain of SINV was engineered to express the proapoptotic gene reaper from Drosophila. MRE/Rpr exhibited an impaired infection phenotype that included delayed midgut infection, delayed virus replication, and reduced virus accumulation in saliva. Nucleotide sequencing of the reaper insert in virus populations isolated from individual mosquitoes revealed evidence of rapid and strong selection against maintenance of Reaper expression in MRE/Rpr-infected mosquitoes. The impaired phenotype of MRE/Rpr, coupled with the observed negative selection against Reaper expression, indicates that apoptosis is a powerful defense against arbovirus infection in mosquitoes and suggests that arboviruses have evolved mechanisms to avoid stimulating apoptosis in mosquitoes that serve as vectors.
Collapse
|