1
|
Fongsaran C, Verhoeve VI, Jirakanwisal K, Harris EK, Macaluso KR. Identification and characterization of a Relish-type NF-κB, DvRelish, in Dermacentor variabilis in response to Rickettsia rickettsii infection. Front Cell Infect Microbiol 2024; 14:1494450. [PMID: 39735256 PMCID: PMC11682715 DOI: 10.3389/fcimb.2024.1494450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/14/2024] [Indexed: 12/31/2024] Open
Abstract
Ixodid ticks serve as hosts and transmission vectors for several obligate intracellular bacteria, including members of the spotted fever group (SFG) of Rickettsia. Although ticks generate an immune response to bacterial insults, many of the signaling molecules associated with the response and how they may contribute to vector competence for Rickettsia are undefined. In this study, we isolated a full-length dvrelish transcript from Dermacentor variabilis, which encoded a Relish-type NF-κB. The presence of a canonical Rel homology domain (RHD) consistent with NF-κB proteins suggested a role in tick immune response for DvRelish. The expression of DvRelish was confirmed in tick tissues and fluorescent microscopy of tick hemocytes indicated increased expression following infection with Rickettsia as compared to a non-tick-borne bacterial pathogen. To further determine the effect of dvRelish gene knockdown on rickettsial infection, we used RNA interference-mediated gene knockdown in D. variabilis and demonstrated that transcription of dvRelish was decreased after 24 h post-injection of siRNA. We then assessed the response of D. variabilis when exposed to Rickettsia rickettsii and determined that transcription of dvRelish was inversely associated with rickettsial loads at 48 h post-exposure. Further studies are required to broaden the understanding of differential immune responses in ticks to SFG Rickettsia infection and elucidate the role played by the arthropod immune system in vector competence.
Collapse
|
2
|
Jiménez-Florido P, Aquilino M, Buckley D, Bella JL, Planelló R. Differential gene expression in Chorthippus parallelus (Zetterstedt, 1821) (Orthoptera: Acrididae: Gomphocerinae) induced by Wolbachia infection. INSECT SCIENCE 2024. [PMID: 39614636 DOI: 10.1111/1744-7917.13481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/16/2024] [Accepted: 10/24/2024] [Indexed: 12/01/2024]
Abstract
Distinct lineages of the grasshopper Chorthippus parallelus (Orthoptera: Acrididae) form well-known hybrid zones (HZs) both in the Pyrenees and the Alps mountain ranges in South Europe. These HZs represent unique experimental systems to identify "key genes" that maintain genetic boundaries between emerging species. The Iberian endemism C. p. erythropus (Cpe) and the subspecies C. p. parallelus (Cpp), widely distributed throughout the rest of Europe, overlap and form the Pyrenean HZ. Both subspecies differ morphologically, as well as in behavioral, mitochondrial, nuclear, and chromosomal traits, and in the strains of the maternally transmitted bacterial endosymbiont Wolbachia infecting them. This results in either unidirectional and bidirectional cytoplasmic incompatibility between both grasshopper subspecies, pointing out that Wolbachia clearly affects gene expression in the infected individuals. Here we explore how Wolbachia may modify the expression of some major genes involved in relevant pathways in Cpp in the Pyrenean HZ. We have analyzed, through molecular biomarkers, the physiological responses in C. parallelus individuals infected by Wolbachia, with particular attention to the energy metabolism, the immune system response, and the reproduction. qPCR was used to evaluate the expression of selected genes in the gonads of infected and uninfected adults of both sexes, since this tissue constitutes the main target of Wolbachia infection. Transcriptional analyses also showed differential sex-dependent responses in most of the analyzed biomarkers in infected and noninfected individuals. We identified for the first time new sensitive biomarkers that might be involved in the reproductive barrier induced by Wolbachia in the hybrid zone.
Collapse
Affiliation(s)
- Patricia Jiménez-Florido
- Departamento de Biología (Genética), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación en Biodiversidad y Cambio Global (CIBC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Mónica Aquilino
- Grupo de Entomología Molecular, Biomarcadores y Estrés Ambiental, Facultad de Ciencias, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - David Buckley
- Departamento de Biología (Genética), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación en Biodiversidad y Cambio Global (CIBC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - José L Bella
- Departamento de Biología (Genética), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación en Biodiversidad y Cambio Global (CIBC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Rosario Planelló
- Grupo de Entomología Molecular, Biomarcadores y Estrés Ambiental, Facultad de Ciencias, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| |
Collapse
|
3
|
Hixson B, Chen R, Buchon N. Innate immunity in Aedes mosquitoes: from pathogen resistance to shaping the microbiota. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230063. [PMID: 38497256 PMCID: PMC10945403 DOI: 10.1098/rstb.2023.0063] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/12/2023] [Indexed: 03/19/2024] Open
Abstract
Discussions of host-microbe interactions in mosquito vectors are frequently dominated by a focus on the human pathogens they transmit (e.g. Plasmodium parasites and arboviruses). Underlying the interactions between a vector and its transmissible pathogens, however, is the physiology of an insect living and interacting with a world of bacteria and fungi including commensals, mutualists and primary and opportunistic pathogens. Here we review what is known about the bacteria and fungi associated with mosquitoes, with an emphasis on the members of the Aedes genus. We explore the reciprocal effects of microbe on mosquito, and mosquito on microbe. We analyse the roles of bacterial and fungal symbionts in mosquito development, their effects on vector competence, and their potential uses as biocontrol agents and vectors for paratransgenesis. We explore the compartments of the mosquito gut, uncovering the regionalization of immune effectors and modulators, which create the zones of resistance and immune tolerance with which the mosquito host controls and corrals its microbial symbionts. We examine the anatomical patterning of basally expressed antimicrobial peptides. Finally, we review the relationships between inducible antimicrobial peptides and canonical immune signalling pathways, comparing and contrasting current knowledge on each pathway in mosquitoes to the model insect Drosophila melanogaster. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
- Bretta Hixson
- Department of Entomology, Cornell University College of Agriculture and Life Sciences, Ithaca, 14853, NY, USA
| | - Robin Chen
- Department of Entomology, Cornell University College of Agriculture and Life Sciences, Ithaca, 14853, NY, USA
| | - Nicolas Buchon
- Department of Entomology, Cornell University College of Agriculture and Life Sciences, Ithaca, 14853, NY, USA
| |
Collapse
|
4
|
Mehta D, Chaudhary S, Sunil S. Oxidative stress governs mosquito innate immune signalling to reduce chikungunya virus infection in Aedes-derived cells. J Gen Virol 2024; 105. [PMID: 38488850 DOI: 10.1099/jgv.0.001966] [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: 03/19/2024] Open
Abstract
Arboviruses such as chikungunya, dengue and zika viruses cause debilitating diseases in humans. The principal vector species that transmits these viruses is the Aedes mosquito. Lack of substantial knowledge of the vector species hinders the advancement of strategies for controlling the spread of arboviruses. To supplement our information on mosquitoes' responses to virus infection, we utilized Aedes aegypti-derived Aag2 cells to study changes at the transcriptional level during infection with chikungunya virus (CHIKV). We observed that genes belonging to the redox pathway were significantly differentially regulated. Upon quantifying reactive oxygen species (ROS) in the cells during viral infection, we further discovered that ROS levels are considerably higher during the early hours of infection; however, as the infection progresses, an increase in antioxidant gene expression suppresses the oxidative stress in cells. Our study also suggests that ROS is a critical regulator of viral replication in cells and inhibits intracellular and extracellular viral replication by promoting the Rel2-mediated Imd immune signalling pathway. In conclusion, our study provides evidence for a regulatory role of oxidative stress in infected Aedes-derived cells.
Collapse
Affiliation(s)
- Divya Mehta
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sakshi Chaudhary
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sujatha Sunil
- Vector Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
5
|
Mehmood N, Hassan A, Zhong X, Zhu Y, Ouyang G, Huang Q. Entomopathogenic fungal infection following immune gene silencing decreased behavioral and physiological fitness in Aedes aegypti mosquitoes. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2023; 195:105535. [PMID: 37666588 DOI: 10.1016/j.pestbp.2023.105535] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 09/06/2023]
Abstract
Entomopathogenic fungi are a promising category of biocontrol agents with mosquitocidal properties. Prior studies have proved their potential to reduce fecundity, human biting and vector competence, all of them together determine vectorial capacity of the mosquitoes. Unfortunately, conventional vector control strategies are inadequate with growing problem of insecticide resistance and environmental deterioration. Therefore, alternate vector control measures are immediately needed and to accomplish that, an improved understanding of behavioral and physiological defense mechanisms of the mosquitoes against fungal infection is essential. In this study, fitness was considered with respect to different behavioral (self-grooming and flight), physiological (antifungal activity and antimicrobial peptides) parameters and survival rates as compared to the control group. We found a significant upregulation in CLSP2, TEP22, Rel1 and Rel2 genes at multiple time periods of fungal infection, which indicates the successful fungal infection and activation of Toll and IMD pathways in mosquitoes. RNAi-mediated silencing of Rel1 and Rel2 genes (transcription factors of Toll and IMD pathways, respectively) significantly reduced the survival, self-grooming frequencies and durations, and flight locomotor activity among adult Ae. aegypti female mosquitoes. Moreover, Rel1 and Rel2 knockdown significantly decreased antifungal activity and antimicrobial peptides expression levels in target mosquitoes. These results indicate an overall decrease in fitness of the mosquitoes after fungal challenge following Rel1 and Rel2 silencing. These findings provide an improved understanding of behavioral and physiological responses in mosquitoes with altered immunity against entomopathogenic fungal infections which can guide us towards the development of novel biocontrol strategies against mosquitoes.
Collapse
Affiliation(s)
- Nasir Mehmood
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Ali Hassan
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Xueshan Zhong
- Yuexiu Center for Disease Control and Prevention, Guangzhou 510055, Guangdong Province, China
| | - Yongzhang Zhu
- Guangzhou Yongliang Environmental Protection Technology Service CO., LTD, Guangzhou 510405, Guangdong Province, China
| | - Guang Ouyang
- Guangzhou Yongliang Environmental Protection Technology Service CO., LTD, Guangzhou 510405, Guangdong Province, China
| | - Qiuying Huang
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
6
|
Prince BC, Walsh E, Torres TZB, Rückert C. Recognition of Arboviruses by the Mosquito Immune System. Biomolecules 2023; 13:1159. [PMID: 37509194 PMCID: PMC10376960 DOI: 10.3390/biom13071159] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Arthropod-borne viruses (arboviruses) pose a significant threat to both human and animal health worldwide. These viruses are transmitted through the bites of mosquitoes, ticks, sandflies, or biting midges to humans or animals. In humans, arbovirus infection often results in mild flu-like symptoms, but severe disease and death also occur. There are few vaccines available, so control efforts focus on the mosquito population and virus transmission control. One area of research that may enable the development of new strategies to control arbovirus transmission is the field of vector immunology. Arthropod vectors, such as mosquitoes, have coevolved with arboviruses, resulting in a balance of virus replication and vector immune responses. If this balance were disrupted, virus transmission would likely be reduced, either through reduced replication, or even through enhanced replication, resulting in mosquito mortality. The first step in mounting any immune response is to recognize the presence of an invading pathogen. Recent research advances have been made to tease apart the mechanisms of arbovirus detection by mosquitoes. Here, we summarize what is known about arbovirus recognition by the mosquito immune system, try to generate a comprehensive picture, and highlight where there are still gaps in our current understanding.
Collapse
Affiliation(s)
- Brian C Prince
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Elizabeth Walsh
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Tran Zen B Torres
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| | - Claudia Rückert
- Department of Biochemistry and Molecular Biology, College of Agriculture, Biotechnology & Natural Resources, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
7
|
Novelo M, Dutra HLC, Metz HC, Jones MJ, Sigle LT, Frentiu FD, Allen SL, Chenoweth SF, McGraw EA. Dengue and chikungunya virus loads in the mosquito Aedes aegypti are determined by distinct genetic architectures. PLoS Pathog 2023; 19:e1011307. [PMID: 37043515 PMCID: PMC10124881 DOI: 10.1371/journal.ppat.1011307] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/24/2023] [Accepted: 03/19/2023] [Indexed: 04/13/2023] Open
Abstract
Aedes aegypti is the primary vector of the arboviruses dengue (DENV) and chikungunya (CHIKV). These viruses exhibit key differences in their vector interactions, the latter moving more quicky through the mosquito and triggering fewer standard antiviral pathways. As the global footprint of CHIKV continues to expand, we seek to better understand the mosquito's natural response to CHIKV-both to compare it to DENV:vector coevolutionary history and to identify potential targets in the mosquito for genetic modification. We used a modified full-sibling design to estimate the contribution of mosquito genetic variation to viral loads of both DENV and CHIKV. Heritabilities were significant, but higher for DENV (40%) than CHIKV (18%). Interestingly, there was no genetic correlation between DENV and CHIKV loads between siblings. These data suggest Ae. aegypti mosquitoes respond to the two viruses using distinct genetic mechanisms. We also examined genome-wide patterns of gene expression between High and Low CHIKV families representing the phenotypic extremes of viral load. Using RNAseq, we identified only two loci that consistently differentiated High and Low families: a long non-coding RNA that has been identified in mosquito screens post-infection and a distant member of a family of Salivary Gland Specific (SGS) genes. Interestingly, the latter gene is also associated with horizontal gene transfer between mosquitoes and the endosymbiotic bacterium Wolbachia. This work is the first to link the SGS gene to a mosquito phenotype. Understanding the molecular details of how this gene contributes to viral control in mosquitoes may, therefore, also shed light on its role in Wolbachia.
Collapse
Affiliation(s)
- Mario Novelo
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Heverton LC Dutra
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Hillery C. Metz
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Matthew J. Jones
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Leah T. Sigle
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Francesca D. Frentiu
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Herston, Queensland, Australia
| | - Scott L. Allen
- School of Biological Sciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Stephen F. Chenoweth
- School of Biological Sciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Elizabeth A. McGraw
- Center for Infectious Disease Dynamics, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| |
Collapse
|
8
|
Morejon B, Michel K. A zone-of-inhibition assay to screen for humoral antimicrobial activity in mosquito hemolymph. Front Cell Infect Microbiol 2023; 13:891577. [PMID: 36779191 PMCID: PMC9908765 DOI: 10.3389/fcimb.2023.891577] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
In insects, antibacterial immunity largely depends on the activation of downstream signaling and effector responses, leading to the synthesis and secretion of soluble effector molecules, such as antimicrobial peptides (AMPs). AMPs are acute infection response peptides secreted into the hemolymph upon bacterial stimulation. The transcription of innate immunity genes encoding for AMPs is highly dependent on several signaling cascade pathways, such as the Toll pathway. In the African malaria mosquito, Anopheles gambiae, AMPs hold a special interest as their upregulation have been shown to limit the growth of malaria parasites, bacteria, and fungi. Most of the current knowledge on the regulation of insect AMPs in microbial infection have been obtained from Drosophila. However, largely due to the lack of convenient assays, the regulation of antimicrobial activity in mosquito hemolymph is still not completely understood. In this study, we report a zone of inhibition assay to identify the contribution of AMPs and components of the Toll pathway to the antimicrobial activity of A. gambiae hemolymph. As a proof of principle, we demonstrate that Micrococcus luteus challenge induces antimicrobial activity in the adult female mosquito hemolymph, which is largely dependent on defensin 1. Moreover, by using RNAi to silence Cactus, REL1, and MyD88, we showed that Cactus kd induces antimicrobial activity in the mosquito hemolymph, whereas the antimicrobial activity in REL1 kd and MyD88 kd is reduced after challenge. Finally, while injection itself is not sufficient to induce antimicrobial activity, our results show that it primes the response to bacterial challenge. Our study provides information that increases our knowledge of the regulation of antimicrobial activity in response to microbial infections in mosquitoes. Furthermore, this assay represents an ex vivo medium throughput assay that can be used to determine the upstream regulatory elements of antimicrobial activity in A. gambiae hemolymph.
Collapse
Affiliation(s)
- Bianca Morejon
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | | |
Collapse
|
9
|
Bui M, Dalla Benetta E, Dong Y, Zhao Y, Yang T, Li M, Antoshechkin IA, Buchman A, Bottino-Rojas V, James AA, Perry MW, Dimopoulos G, Akbari OS. CRISPR mediated transactivation in the human disease vector Aedes aegypti. PLoS Pathog 2023; 19:e1010842. [PMID: 36656895 PMCID: PMC9888728 DOI: 10.1371/journal.ppat.1010842] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/31/2023] [Accepted: 12/28/2022] [Indexed: 01/20/2023] Open
Abstract
As a major insect vector of multiple arboviruses, Aedes aegypti poses a significant global health and economic burden. A number of genetic engineering tools have been exploited to understand its biology with the goal of reducing its impact. For example, current tools have focused on knocking-down RNA transcripts, inducing loss-of-function mutations, or expressing exogenous DNA. However, methods for transactivating endogenous genes have not been developed. To fill this void, here we developed a CRISPR activation (CRISPRa) system in Ae. aegypti to transactivate target gene expression. Gene expression is activated through pairing a catalytically-inactive ('dead') Cas9 (dCas9) with a highly-active tripartite activator, VP64-p65-Rta (VPR) and synthetic guide RNA (sgRNA) complementary to a user defined target-gene promoter region. As a proof of concept, we demonstrate that engineered Ae. aegypti mosquitoes harboring a binary CRISPRa system can be used to effectively overexpress two developmental genes, even-skipped (eve) and hedgehog (hh), resulting in observable morphological phenotypes. We also used this system to overexpress the positive transcriptional regulator of the Toll immune pathway known as AaRel1, which resulted in a significant suppression of dengue virus serotype 2 (DENV2) titers in the mosquito. This system provides a versatile tool for research pathways not previously possible in Ae. aegypti, such as programmed overexpression of endogenous genes, and may aid in gene characterization studies and the development of innovative vector control tools.
Collapse
Affiliation(s)
- Michelle Bui
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Elena Dalla Benetta
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Yunchong Zhao
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Ting Yang
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Ming Li
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Igor A. Antoshechkin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Anna Buchman
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - Vanessa Bottino-Rojas
- Department of Microbiology & Molecular Genetics, University of California Irvine, Irvine, California, United States of America
| | - Anthony A. James
- Department of Microbiology & Molecular Genetics, University of California Irvine, Irvine, California, United States of America
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, California, United States of America
| | - Michael W. Perry
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Omar S. Akbari
- Department of Cell and Developmental Biology, School of Biological Sciences, University of California San Diego, San Diego, California, United States of America
| |
Collapse
|
10
|
Shinzawa N, Kashima C, Aonuma H, Takahashi K, Shimojima M, Fukumoto S, Saiki E, Yamamoto DS, Yoshida S, Matsuoka H, Kawaoka Y, Kanuka H. Generation of Transgenic Mosquitoes Harboring a Replication-Restricted Virus. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2022.850111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Live microbe vaccines are designed to elicit strong cellular and antibody responses without developing the symptoms of the disease, and these are effective in preventing infectious diseases. A flying vaccinator (also known as a flying syringe) is a conceptual, genetically engineered hematophagous insect that is used to deliver vaccines such as an antigen from a parasite produced in mosquito saliva; bites from such insects may elicit antibody production by immunizing the host with an antigen through blood-feeding. In addition to a simple vaccine antigen, a flying vaccinator may potentially load a live attenuated microbe with an appropriate mechanism for sustaining its constitutive proliferation in the insect. In this study, a recombinant vesicular stomatitis virus (VSV) lacking the glycoprotein gene (VSV-G) was used to produce replication-restricted VSV (rrVSV) containing GFP. Transgenic Anopheles stephensi mosquitoes, in which the salivary glands expressed a VSV-G gene driven by an aapp salivary gland-specific promoter, were generated and injected intraperitoneally with rrVSV. The injected rrVSV entered the cells of the salivary gland and stimulated endogenous production of progeny rrVSV particles, as seen in rrVSV-infected Drosophila melanogaster expressing VSV-G. These data suggested the possibility of developing a valuable tool for delivering genetically attenuated virus vaccines via mosquito saliva, although efficient replication-restricted virus production is required.
Collapse
|
11
|
Alam I, Batool K, Idris AL, Tan W, Guan X, Zhang L. Role of Lectin in the Response of Aedes aegypti Against Bt Toxin. Front Immunol 2022; 13:898198. [PMID: 35634312 PMCID: PMC9136036 DOI: 10.3389/fimmu.2022.898198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/19/2022] [Indexed: 12/05/2022] Open
Abstract
Aedes aegypti is one of the world’s most dangerous mosquitoes, and a vector of diseases such as dengue fever, chikungunya virus, yellow fever, and Zika virus disease. Currently, a major global challenge is the scarcity of antiviral medicine and vaccine for arboviruses. Bacillus thuringiensis var israelensis (Bti) toxins are used as biological mosquito control agents. Endotoxins, including Cry4Aa, Cry4Ba, Cry10Aa, Cry11Aa, and Cyt1Aa, are toxic to mosquitoes. Insect eradication by Cry toxin relies primarily on the interaction of cry toxins with key toxin receptors, such as aminopeptidase (APN), alkaline phosphatase (ALP), cadherin (CAD), and ATP-binding cassette transporters. The carbohydrate recognition domains (CRDs) of lectins and domains II and III of Cry toxins share similar structural folds, suggesting that midgut proteins, such as C-type lectins (CTLs), may interfere with interactions among Cry toxins and receptors by binding to both and alter Cry toxicity. In the present review, we summarize the functional role of C-type lectins in Ae. aegypti mosquitoes and the mechanism underlying the alteration of Cry toxin activity by CTLs. Furthermore, we outline future research directions on elucidating the Bti resistance mechanism. This study provides a basis for understanding Bti resistance, which can be used to develop novel insecticides.
Collapse
Affiliation(s)
- Intikhab Alam
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Key Lab of Biopesticides and Chemical Biology, MOE, Fujian Agriculture and Forestry University, Fuzhou, China
- College of Life Sciences, South China Agricultural University, Guangzhou, China
| | - Khadija Batool
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Key Lab of Biopesticides and Chemical Biology, MOE, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Aisha Lawan Idris
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Key Lab of Biopesticides and Chemical Biology, MOE, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Weilong Tan
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, China
| | - Xiong Guan
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Key Lab of Biopesticides and Chemical Biology, MOE, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Lingling Zhang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Key Lab of Biopesticides and Chemical Biology, MOE, Fujian Agriculture and Forestry University, Fuzhou, China
- *Correspondence: Lingling Zhang,
| |
Collapse
|
12
|
Cheung YP, Park S, Pagtalunan J, Maringer K. The antiviral role of NF-κB-mediated immune responses and their antagonism by viruses in insects. J Gen Virol 2022; 103. [PMID: 35510990 DOI: 10.1099/jgv.0.001741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The antiviral role of innate immune responses mediated by the NF-κB family of transcription factors is well established in vertebrates but was for a long time less clear in insects. Insects encode two canonical NF-κB pathways, the Toll and Imd ('immunodeficiency') pathways, which are best characterised for their role in antibacterial and antifungal defence. An increasing body of evidence has also implicated NF-κB-mediated innate immunity in antiviral responses against some, but not all, viruses. Specific pattern recognition receptors (PRRs) and molecular events leading to NF-κB activation by viral pathogen-associated molecular patterns (PAMPs) have been elucidated for a number of viruses and insect species. Particularly interesting are recent findings indicating that the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway detects viral RNA to activate NF-κB-regulated gene expression. We summarise the literature on virus-NF-κB pathway interactions across the class Insecta, with a focus on the dipterans Drosophila melanogaster and Aedes aegypti. We discuss potential reasons for differences observed between different virus-host combinations, and highlight similarities and differences between cGAS-STING signalling in insects versus vertebrates. Finally, we summarise the increasing number of known molecular mechanisms by which viruses antagonise NF-κB responses, which suggest that NF-κB-mediated immunity exerts strong evolutionary pressures on viruses. These developments in our understanding of insect antiviral immunity have relevance to the large number of insect species that impact on humans through their transmission of human, livestock and plant diseases, exploitation as biotechnology platforms, and role as parasites, pollinators, livestock and pests.
Collapse
Affiliation(s)
- Yin P Cheung
- The Pirbright Institute, Pirbright, Surrey, GU24 0NF, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Sohyun Park
- The Pirbright Institute, Pirbright, Surrey, GU24 0NF, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Justine Pagtalunan
- The Pirbright Institute, Pirbright, Surrey, GU24 0NF, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK
| | - Kevin Maringer
- The Pirbright Institute, Pirbright, Surrey, GU24 0NF, UK
| |
Collapse
|
13
|
Wu S, He Y, Wei Y, Fan P, Ni W, Zhong D, Zhou G, Zheng X. Effects of Guangzhou seasonal climate change on the development of Aedes albopictus and its susceptibility to DENV-2. PLoS One 2022; 17:e0266128. [PMID: 35363810 PMCID: PMC8975156 DOI: 10.1371/journal.pone.0266128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
Abstract
The susceptibility of Asian tiger mosquitoes to DENV-2 in different seasons was observed in simulated field environments as a reference to design dengue fever control strategies in Guangzhou. The life table experiments of mosquitoes in four seasons were carried out in the field. The susceptibility of Ae. albopictus to dengue virus was observed in both environments in Guangzhou in summer and winter. Ae. albopictus was infected with dengue virus by oral feeding. On day 7 and 14 after infection, the viral load in the head, ovary, and midgut of the mosquito was detected using real-time fluorescent quantitative PCR. Immune-associated gene expression in infected mosquitoes was performed using quantitative real-time reverse transcriptase PCR. The hatching rate and pupation rate of Ae. albopictus larvae in different seasons differed significantly. The winter hatching rate of larvae was lower than that in summer, and the incubation time was longer than in summer. In the winter field environment, Ae. albopictus still underwent basic growth and development processes. Mosquitoes in the simulated field environment were more susceptible to DENV-2 than those in the simulated laboratory environment. In the midgut, viral RNA levels on day 7 in summer were higher than those on day 7 in winter (F = 14.459, P = 0.01); ovarian viral RNA levels on day 7 in summer were higher than those on day 7 in winter (F = 8.656, P < 0.001), but there was no significant difference in the viral load at other time points (P > 0.05). Dicer-2 mRNA expression on day 7 in winter was 4.071 times than that on day 7 in summer: the viral load and Dicer-2 expression correlated moderately. Ae. albopictus could still develop and transmit dengue virus in winter in Guangzhou. Mosquitoes under simulated field conditions were more susceptible to DENV-2 than those under simulated laboratory conditions.
Collapse
Affiliation(s)
- Shanshan Wu
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yulan He
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yong Wei
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Peiyang Fan
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Weigui Ni
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Daibin Zhong
- Program in Public Health, College of Health Sciences, University of California, Irvine, CA, United States of America
| | - Guofa Zhou
- Program in Public Health, College of Health Sciences, University of California, Irvine, CA, United States of America
| | - Xueli Zheng
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Zhu M, Su F, Leng J, Jian S, Yi P, Wen C, Hu B. Two NF-κB subunits are associated with antimicrobial immunity in Hyriopsis cumingii. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 129:104336. [PMID: 34921862 DOI: 10.1016/j.dci.2021.104336] [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: 09/04/2021] [Revised: 12/09/2021] [Accepted: 12/12/2021] [Indexed: 06/14/2023]
Abstract
The NF-κB pathway activated by bacteria and viruses produces a series of antimicrobial peptides that participate in the innate immune response. In this study, two NF-κB subunits were cloned and identified from Hyriopsis cumingii (named Hcp65 and Hcp105) using RT-PCR and RACE. The predicted Hcp65 protein possessed a N-terminal Rel homology domain (RHD) and an Ig-like/plexins/transcription factors domain (IPT); the Hcp105 contained an RHD, an IPT domain, 6 ankyrin (ANK) domain and a death domain. Quantitative reverse transcription PCR (qRT-PCR) showed that Hcp65 and Hcp105 were constitutively expressed in the detected tissues, and were significantly up-regulated in hemocytes, hepatopancreas and gill of mussels challenged with lipopolysaccharide (LPS), peptidoglycan (PGN) and polyinosinic-polycytidylic acid (poly I: C). The dsRNA-mediated silencing of Hcp65 and Hcp105 caused significant reduction of immune genes such as lysozyme (HcLyso), theromacin (Hcther), whey acid protein (HcWAP), LPS-binding protein/bactericidal permeability protein (HcLBP/BPI) 1 and 2. In addition, subcellular localization experiments showed that Hcp65 and Hcp105 proteins were expressed in both the nucleus and cytoplasm of HEK-293T cells, and Hcp50 proteins (mature peptide of Hcp105) were mainly localized in the nucleus. The recombinant Hcp65 and Hcp50 protein could form homodimer and heterodimer and bind κB site in vitro. These results provide useful information for understanding the role of NF-κB in mollusks.
Collapse
Affiliation(s)
- Mingxing Zhu
- Department of Aquatic Science, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China
| | - Feixiang Su
- Department of Aquatic Science, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China
| | - Jianghe Leng
- Department of Aquatic Science, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China
| | - Shaoqing Jian
- Department of Aquatic Science, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China
| | - Peipei Yi
- Zhejiang Normal University, 688 Yingbin Road, Jinhua, Zhejiang Province, 321001, China
| | - Chungen Wen
- Department of Aquatic Science, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China
| | - Baoqing Hu
- Department of Aquatic Science, College of Life Science, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi Province, 330031, China.
| |
Collapse
|
15
|
Transgenic Silkworms Overexpressing Relish and Expressing Drosomycin Confer Enhanced Immunity to Multiple Pathogens. Mol Biotechnol 2022; 64:711-724. [DOI: 10.1007/s12033-021-00438-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 12/09/2021] [Indexed: 10/19/2022]
|
16
|
Adelman ZN, Kojin BB. Malaria-Resistant Mosquitoes (Diptera: Culicidae); The Principle is Proven, But Will the Effectors Be Effective? JOURNAL OF MEDICAL ENTOMOLOGY 2021; 58:1997-2005. [PMID: 34018548 DOI: 10.1093/jme/tjab090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Indexed: 06/12/2023]
Abstract
Over the last few decades, a substantial number of anti-malarial effector genes have been evaluated for their ability to block parasite infection in the mosquito vector. While many of these approaches have yielded significant effects on either parasite intensity or prevalence of infection, just a few have been able to completely block transmission. Additionally, many approaches, while effective against the parasite, also disrupt or alter important aspects of mosquito physiology, leading to corresponding changes in lifespan, reproduction, and immunity. As the most promising approaches move towards field-based evaluation, questions of effector gene robustness and durability move to the forefront. In this forum piece, we critically evaluate past effector gene approaches with an eye towards developing a deeper pipeline to augment the current best candidates.
Collapse
Affiliation(s)
- Zach N Adelman
- Department of Entomology and AgriLife Research, Texas A&M University, College Station, TX, USA
| | - Bianca B Kojin
- Department of Entomology and AgriLife Research, Texas A&M University, College Station, TX, USA
| |
Collapse
|
17
|
Wei Y, Wang J, Wei YH, Song Z, Hu K, Chen Y, Zhou G, Zhong D, Zheng X. Vector Competence for DENV-2 Among Aedes albopictus (Diptera: Culicidae) Populations in China. Front Cell Infect Microbiol 2021; 11:649975. [PMID: 33834007 PMCID: PMC8021855 DOI: 10.3389/fcimb.2021.649975] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/08/2021] [Indexed: 11/21/2022] Open
Abstract
Aedes albopictus is a vector of over 20 arboviruses that has spread throughout the world, mainly in the second half of the twentieth century. Approximately 50–100 million people are infected with dengue virus (DENV) transmitted by Aedes mosquitoes each year, leading to heavy economic burdens for both governments and individuals, among countless other negative consequences. Understanding the vector competence of vector species is critical for effectively preventing and controlling vector-borne diseases. Accordingly, in this study, vector competence was evaluated by quantitative analysis of DENV-2 loads in mosquito tissues (midguts, heads, and salivary glands) and whole mosquitoes through real-time quantitative polymerase chain reaction (RT-qPCR) analysis. Wolbachia and the expression of immune-associated genes (Rel1, Rel2, Dicer2, and STAT) in mosquitoes were also detected by RT-qPCR to explore their impact on vector competence. The amount of DENV-2 in the mosquito midguts, heads, and salivary glands from southern-western China were found to be lower than those from eastern-central-northern China. The DENV-2 loads in whole mosquitoes showed a negative correlation with Rel1 gene (r = -0.285, P = 0.011) and STAT gene expression levels (r = -0.289, P = 0.009). In terms of Wolbachia strains, the density of the wAlbB strain was found to be significantly higher than that of the wAlbA strain in the eight Ae. albopictus populations, and the relative density of the wAlbB strain in mosquitoes from southern-western China was higher than those from eastern-central-northern China. The relative density of the wAlbB strain showed a negative correlation with the mean loads of DENV-2 in the heads (r = -0.729, P = 0.040), salivary glands (r = -0.785, P = 0.021), and whole mosquitoes (r = -0.909, P = 0.002). Thus, there are lower DENV-2 loads in the mosquitoes from southern-western China, which may be related to the innate immunity of mosquitoes as affected by Rel1 in the Toll pathway, STAT in the JAK-STAT pathway, and the relative density of the wAlbB strain.
Collapse
Affiliation(s)
- Yong Wei
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jiatian Wang
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuan-Huan Wei
- Department of Clinical Nutrition, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Zhangyao Song
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Ke Hu
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yulan Chen
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guofa Zhou
- Program in Public Health, College of Health Sciences, University of California, Irvine, Irvine, CA, United States
| | - Daibin Zhong
- Program in Public Health, College of Health Sciences, University of California, Irvine, Irvine, CA, United States
| | - Xueli Zheng
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
18
|
Rolandelli A, Nascimento AEC, Silva LS, Rivera-Pomar R, Guarneri AA. Modulation of IMD, Toll, and Jak/STAT Immune Pathways Genes in the Fat Body of Rhodnius prolixus During Trypanosoma rangeli Infection. Front Cell Infect Microbiol 2021; 10:598526. [PMID: 33537241 PMCID: PMC7848085 DOI: 10.3389/fcimb.2020.598526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/30/2020] [Indexed: 12/31/2022] Open
Abstract
Trypanosoma rangeli is the second most common American trypanosome that infects man. It is vectored by triatomines from the genus Rhodnius, in which it invades the hemolymph and infects the salivary glands, avoiding the bug immune responses. In insects, these responses are initiated by well conserved pathways, mainly the IMD, Toll, and Jak/STAT. We hypothesize that long-term infection with T. rangeli in the gut or hemolymph of Rhodnius prolixus triggers different systemic immune responses, which influence the number of parasites that survive inside the vector. Thus, we investigated groups of insects with infections in the gut and/or hemolymph, and evaluated the parasite load and the expression in the fat body of transcription factors (Rp-Relish, Rp-Dorsal, and Rp-STAT) and inhibitors (Rp-Cactus and Rp-Caspar) of the IMD, Toll, and Jak/STAT pathways. We detected lower parasite counts in the gut of insects without hemolymph infection, compared to hemolymph-infected groups. Besides, we measured higher parasite numbers in the gut of bugs that were first inoculated with T. rangeli and then fed on infected mice, compared with control insects, indicating that hemolymph infection increases parasite numbers in the gut. Interestingly, we observed that genes from the three immune pathways where differentially modulated, depending on the region parasites were present, as we found (1) Rp-Relish downregulated in gut-and/or-hemolymph-infected insects, compared with controls; (2) Rp-Cactus upregulated in gut-infected insect, compared with controls and gut-and-hemolymph-infected groups; and (3) Rp-STAT downregulated in all groups of hemolymph-infected insects. Finally, we uncovered negative correlations between parasite loads in the gut and Rp-Relish and Rp-Cactus expression, and between parasite counts in the hemolymph and Rp-Relish levels, suggesting an association between parasite numbers and the IMD and Toll pathways. Overall, our findings reveal new players in R. prolixus-T. rangeli interactions that could be key for the capacity of the bug to transmit the pathogen.
Collapse
Affiliation(s)
- Agustín Rolandelli
- Centro de Bioinvestigaciones (CeBio), Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT-NOBA), Universidad Nacional del Noroeste de la Provincia de Buenos Aires (UNNOBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Pergamino, Argentina
| | - Adeisa E C Nascimento
- Vector Behaviour and Pathogen Interaction Group, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| | - Leticia S Silva
- Vector Behaviour and Pathogen Interaction Group, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| | - Rolando Rivera-Pomar
- Centro de Bioinvestigaciones (CeBio), Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires (CIT-NOBA), Universidad Nacional del Noroeste de la Provincia de Buenos Aires (UNNOBA)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Pergamino, Argentina
| | - Alessandra A Guarneri
- Vector Behaviour and Pathogen Interaction Group, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Brazil
| |
Collapse
|
19
|
Two Roles for the Tenebrio molitor Relish in the Regulation of Antimicrobial Peptides and Autophagy-Related Genes in Response to Listeria monocytogenes. INSECTS 2020; 11:insects11030188. [PMID: 32188156 PMCID: PMC7142762 DOI: 10.3390/insects11030188] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/04/2020] [Accepted: 03/12/2020] [Indexed: 12/24/2022]
Abstract
Relish is a key NF-κB transcription factor of the immune-deficiency (Imd) pathway that combats infection by regulating antimicrobial peptides (AMPs). Understanding of the fundamental role of Tenebrio molitor Relish (TmRelish) in controlling of Listeria monocytogenes virulence through the regulation of both AMPs and autophagy-related (ATG) genes is unclear. Here, we show that TmRelish transcripts were highly abundant in the larval fat body and hemocytes compared to the gut upon L. monocytogenes infection. Furthermore, significant mortality was observed in TmRelish-silenced larvae after intracellular insult. To investigate the cause of this lethality, we measured the induction of AMPs and ATG genes in the TmRelish dsRNA-treated T. molitor larvae. The expression of TmTenecin-1, TmTenecin-4, TmColeptericin-1, TmAttacin-2, and TmCecropin-2 were suppressed in the fat body and hemocytes of dsTmRelish-injected larvae during L. monocytogenes infection. In addition, TmRelish knockdown led to a noticeable downregulation of TmATG1 (a serine-threonine protein kinase) in the fat body and hemocytes of young larvae 6 h post-infection (pi). The notable increase of autophagy genes in the early stage of infection (6 h pi), suggesting autophagy response is crucial for Listeria clearance. Taken together, these results suggest that TmRelish plays pivotal roles in not only regulation of AMP genes but also induction of autophagy genes in response to L. monocytogenes challenge in fat body and hemocytes of T. molitor larvae. Furthermore, negative regulation of several AMPs by TmRelish in the fat body, hemocytes, and gut leaves open the possibility of a crosstalk between Toll and Imd pathway.
Collapse
|
20
|
Keshavarz M, Jo YH, Patnaik BB, Park KB, Ko HJ, Kim CE, Edosa TT, Lee YS, Han YS. TmRelish is required for regulating the antimicrobial responses to Escherichia coli and Staphylococcus aureus in Tenebrio molitor. Sci Rep 2020; 10:4258. [PMID: 32144366 PMCID: PMC7060202 DOI: 10.1038/s41598-020-61157-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/18/2020] [Indexed: 11/09/2022] Open
Abstract
Relish, a transcription factor, is a critical downstream component of the immune deficiency (Imd) pathway and regulates host defense against bacterial infection by mediating antimicrobial peptide (AMP) synthesis. Understanding the immunological function of the mealworm beetle, Tenebrio molitor Relish (TmRelish) will be instructive in understanding insect immunity. In the present study, full-length ORF of TmRelish was retrieved from T. molitor-expressed sequence tags and RNA-seq database. The predicted TmRelish amino acid sequence contained an N-terminal Rel-homology domain; an Ig-like, plexin, and transcription factor domain; ankyrin repeat motifs; a nuclear localization signal; and a C-terminal death domain and shared the highly conserved structure of the Relish proteins of other insect species. TmRelish mRNA was detected in all developmental stages of the insect; however, the highest levels were detected in the larval gut tissue and adult hemocytes. TmRelish mRNA level was upregulated in the fat body, hemocyte, and gut tissue 9 h after infection of T. molitor larvae by the gram-negative bacteria, Escherichia coli. Furthermore, TmRelish knockdown led to significantly higher mortality of the E. coli-infected larvae, and significantly lower mortality of larvae infected with Staphylococcus aureus or Candida albicans. To elucidate the possible cause of mortality, we measured AMP transcription in the fat body, hemocytes, gut, and Malpighian tubules (MTs) of T. molitor larvae. TmRelish knockdown suppressed the expression of nine AMP genes in the larval fat body and gut tissue during E. coli infection, suggesting that TmRelish positively regulates AMP expression in both immune-related tissues, in response to E. coli challenge. Furthermore, negative regulation of some AMPs by TmRelish in the MTs, gut and hemocytes in response to C. albicans infection suggests a crosstalk between the Toll and Imd pathways.
Collapse
Affiliation(s)
- Maryam Keshavarz
- Department of Applied Biology, Institute of Environmentally Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yong Hun Jo
- Department of Applied Biology, Institute of Environmentally Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Bharat Bhusan Patnaik
- Department of Applied Biology, Institute of Environmentally Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea.,Department of Biotechnology, Trident Academy of Technology (TAT), F2-A, Chandaka Industrial Estate, Chandrasekharpur, Bhubaneswar, Odisha, 751024, India
| | - Ki Beom Park
- Department of Applied Biology, Institute of Environmentally Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hye Jin Ko
- Department of Applied Biology, Institute of Environmentally Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Chang Eun Kim
- Department of Applied Biology, Institute of Environmentally Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Tariku Tesfaye Edosa
- Department of Applied Biology, Institute of Environmentally Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yong Seok Lee
- School of Biotechnology and Life Sciences, College of Natural Sciences, Soonchunhyang University, 22 Soonchunhyangro, Shinchang-myeon, Asan, Chungchungnam-do, 31538, South Korea
| | - Yeon Soo Han
- Department of Applied Biology, Institute of Environmentally Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
21
|
Cabral S, de Paula A, Samuels R, da Fonseca R, Gomes S, Silva JR, Mury F. Aedes aegypti (Diptera: Culicidae) Immune Responses with Different Feeding Regimes Following Infection by the Entomopathogenic Fungus Metarhizium anisopliae. INSECTS 2020; 11:E95. [PMID: 32024202 PMCID: PMC7074208 DOI: 10.3390/insects11020095] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/17/2020] [Accepted: 01/30/2020] [Indexed: 12/21/2022]
Abstract
The mosquito Aedes aegypti is the most notorious vector of illness-causing viruses. The use of entomopathogenic fungi as bioinsecticides is a promising alternative for the development of novel mosquito control strategies. We investigate whether differences in immune responses could be responsible for modifications in survival rates of insects following different feeding regimes. Sucrose and blood-fed adult A. aegypti females were sprayed with M. anisopliae 1 × 106 conidia mL-1, and after 48 h, the midgut and fat body were dissected. We used RT-qPCR to monitor the expression of Cactus and REL1 (Toll pathway), IMD, REL2, and Caspar (IMD pathway), STAT and PIAS (JAK-STAT pathway), as well as the expression of antimicrobial peptides (Defensin A, Attacin and Cecropin G). REL1 and REL2 expression in both the midgut and fat body were higher in blood-fed fungus-challenged A. aegypti than in sucrose-fed counterparts. Interestingly, infection of sucrose-fed insects induced Cactus expression in the fat body, a negative regulator of the Toll pathway. The IMD gene was upregulated in the fat body in response to fungal infection after a blood meal. Additionally, we observed the induction of antimicrobial peptides in the blood-fed fungus-challenged insects. This study suggests that blood-fed A. aegypti are less susceptible to fungal infection due to the rapid induction of Toll and IMD immune pathways.
Collapse
Affiliation(s)
- Sara Cabral
- Laboratório Integrado de Bioquímica—Instituto de Biodiversidade e Sustentabilidade—NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ 27965-045, Brazil; (S.C.); (J.R.S.)
| | - Adriano de Paula
- Laboratório de Entomologia e Fitopatologia—CCTA, Universidade Estadual do Norte FluminenseDarcy Ribeiro, Campos dos Goytacazes, RJ 28013-603, Brazil; (A.d.P.); (S.G.)
| | - Richard Samuels
- Laboratório de Entomologia e Fitopatologia—CCTA, Universidade Estadual do Norte FluminenseDarcy Ribeiro, Campos dos Goytacazes, RJ 28013-603, Brazil; (A.d.P.); (S.G.)
| | - Rodrigo da Fonseca
- Laboratório Integrado de Ciências Morfofuncionais—Instituto de Biodiversidade e Sustentabilidade –NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ 27965-045, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular—INCT-EM, Rio de Janeiro 21941-590, Brazil
| | - Simone Gomes
- Laboratório de Entomologia e Fitopatologia—CCTA, Universidade Estadual do Norte FluminenseDarcy Ribeiro, Campos dos Goytacazes, RJ 28013-603, Brazil; (A.d.P.); (S.G.)
| | - José Roberto Silva
- Laboratório Integrado de Bioquímica—Instituto de Biodiversidade e Sustentabilidade—NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ 27965-045, Brazil; (S.C.); (J.R.S.)
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular—INCT-EM, Rio de Janeiro 21941-590, Brazil
| | - Flávia Mury
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular—INCT-EM, Rio de Janeiro 21941-590, Brazil
- Laboratório Integrado de Biociências Translacionais—Instituto de Biodiversidade e Sustentabilidade—NUPEM, Universidade Federal do Rio de Janeiro, Macaé, RJ 27965-045, Brazil
| |
Collapse
|
22
|
Tawidian P, Rhodes VL, Michel K. Mosquito-fungus interactions and antifungal immunity. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 111:103182. [PMID: 31265904 PMCID: PMC6639037 DOI: 10.1016/j.ibmb.2019.103182] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 06/28/2019] [Indexed: 05/14/2023]
Abstract
The mosquito immune system has evolved in the presence of continuous encounters with fungi that range from food to foes. Herein, we review the field of mosquito-fungal interactions, providing an overview of current knowledge and topics of interest. Mosquitoes encounter fungi in their aquatic and terrestrial habitats. Mosquito larvae are exposed to fungi on plant detritus, within the water column, and at the water surface. Adult mosquitoes are exposed to fungi during indoor and outdoor resting, blood and sugar feeding, mating, and oviposition. Fungi enter the mosquito body through different routes, including ingestion and through active or passive breaches in the cuticle. Oral uptake of fungi can be beneficial to mosquitoes, as yeasts hold nutritional value and support larval development. However, ingestion of or surface contact with fungal entomopathogens leads to colonization of the mosquito with often lethal consequences to the host. The mosquito immune system recognizes fungi and mounts cellular and humoral immune responses in the hemocoel, and possibly epithelial immune responses in the gut. These responses are regulated transcriptionally through multiple signal transduction pathways. Proteolytic protease cascades provide additional regulation of antifungal immunity. Together, these immune responses provide an efficient barrier to fungal infections, which need to be overcome by entomopathogens. Therefore, fungi constitute an excellent tool to examine the molecular underpinnings of mosquito immunity and to identify novel antifungal peptides. In addition, recent advances in mycobiome analyses can now be used to examine the contribution of fungi to various mosquito traits, including vector competence.
Collapse
Affiliation(s)
- P Tawidian
- Division of Biology, Kansas State University, 267 Chalmers Hall, Manhattan, KS, 66506, USA
| | - V L Rhodes
- Missouri Southern State University, Biology Department, Reynolds Hall 220, 3950 E. Newman Rd., Joplin, MO, 64801-1595, USA
| | - K Michel
- Division of Biology, Kansas State University, 267 Chalmers Hall, Manhattan, KS, 66506, USA.
| |
Collapse
|
23
|
Filosa JN, Berry CT, Ruthel G, Beverley SM, Warren WC, Tomlinson C, Myler PJ, Dudkin EA, Povelones ML, Povelones M. Dramatic changes in gene expression in different forms of Crithidia fasciculata reveal potential mechanisms for insect-specific adhesion in kinetoplastid parasites. PLoS Negl Trop Dis 2019; 13:e0007570. [PMID: 31356610 PMCID: PMC6687205 DOI: 10.1371/journal.pntd.0007570] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/08/2019] [Accepted: 06/22/2019] [Indexed: 01/08/2023] Open
Abstract
Kinetoplastids are a group of parasites that includes several medically-important species. These human-infective species are transmitted by insect vectors in which the parasites undergo specific developmental transformations. For each species, this includes a stage in which parasites adhere to insect tissue via a hemidesmosome-like structure. Although this structure has been described morphologically, it has never been molecularly characterized. We are using Crithidia fasciculata, an insect parasite that produces large numbers of adherent parasites inside its mosquito host, as a model kinetoplastid to investigate both the mechanism of adherence and the signals required for differentiation to an adherent form. An advantage of C. fasciculata is that adherent parasites can be generated both in vitro, allowing a direct comparison to cultured swimming forms, as well as in vivo within the mosquito. Using RNAseq, we identify genes associated with adherence in C. fasciculata. As almost all of these genes have orthologs in other kinetoplastid species, our findings may reveal shared mechanisms of adherence, allowing investigation of a crucial step in parasite development and disease transmission. In addition, dual-RNAseq allowed us to explore the interaction between the parasites and the mosquito. Although the infection is well-tolerated, anti-microbial peptides and other components of the mosquito innate immune system are upregulated. Our findings indicate that C. fasciculata is a powerful model system for probing kinetoplastid-insect interactions. Kinetoplastids are single-celled parasites that cause devastating human diseases worldwide. Although this group includes many species that infect a variety of hosts, they have a great deal of shared biology. One relatively unexplored aspect of the kinetoplastid life cycle is their ability to adhere to insect tissue. For pathogenic species, adherence is critical for transmission by insect vectors. We have used an insect parasite called Crithidia fasciculata as a model kinetoplastid to reveal shared mechanisms of insect adherence. We have compared gene expression profiles of motile, non-adherent C. fasciculata to those of C. fasciculata adhered to non-living substrates and those attached to the hindgut of mosquitoes. Through this analysis, we have identified a large number of candidate proteins that may mediate adhesion in these and related parasites. In addition, our findings suggest that the mosquito immune system is responding to the presence of parasites in the gut. These results establish a new, robust system to explore the interaction between kinetoplastids and their insect hosts.
Collapse
Affiliation(s)
- John N. Filosa
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Corbett T. Berry
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Gordon Ruthel
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Wesley C. Warren
- University of Missouri, Bond Life Sciences Center, Columbia, Missouri, United States of America
| | - Chad Tomlinson
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Peter J. Myler
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, Washington, United States of America
| | - Elizabeth A. Dudkin
- Department of Biology, Penn State Brandywine, Media, Pennsylvania, United States of America
| | - Megan L. Povelones
- Department of Biology, Penn State Brandywine, Media, Pennsylvania, United States of America
- * E-mail: (MLP); (MP)
| | - Michael Povelones
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (MLP); (MP)
| |
Collapse
|
24
|
Zhao L, Alto BW, Jiang Y, Yu F, Zhang Y. Transcriptomic Analysis of Aedes aegypti Innate Immune System in Response to Ingestion of Chikungunya Virus. Int J Mol Sci 2019; 20:ijms20133133. [PMID: 31252518 PMCID: PMC6651163 DOI: 10.3390/ijms20133133] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/21/2019] [Accepted: 06/23/2019] [Indexed: 01/07/2023] Open
Abstract
Aedes aegypti (L.) is the primary vector of emergent mosquito-borne viruses, including chikungunya, dengue, yellow fever, and Zika viruses. To understand how these viruses interact with their mosquito vectors, an analysis of the innate immune system response was conducted. The innate immune system is a conserved evolutionary defense strategy and is the dominant immune system response found in invertebrates and vertebrates, as well as plants. RNA-sequencing analysis was performed to compare target transcriptomes of two Florida Ae. aegypti strains in response to chikungunya virus infection. We analyzed a strain collected from a field population in Key West, Florida, and a laboratory strain originating from Orlando. A total of 1835 transcripts were significantly expressed at different levels between the two Florida strains of Ae. aegypti. Gene Ontology analysis placed these genes into 12 categories of biological processes, including 856 transcripts (up/down regulated) with more than 1.8-fold (p-adj (p-adjust value) ≤ 0.01). Transcriptomic analysis and q-PCR data indicated that the members of the AaeCECH genes are important for chikungunya infection response in Ae. aegypti. These immune-related enzymes that the chikungunya virus infection induces may inform molecular-based strategies for interruption of arbovirus transmission by mosquitoes.
Collapse
Affiliation(s)
- Liming Zhao
- Florida Medical Entomology Laboratory, University of Florida, 200 9th Street South East, Vero Beach, FL 32962, USA.
| | - Barry W Alto
- Florida Medical Entomology Laboratory, University of Florida, 200 9th Street South East, Vero Beach, FL 32962, USA
| | - Yongxing Jiang
- Mosquito Control Services, City of Gainesville, 405 NW 39th Avenue Gainesville, FL 32609, USA
| | - Fahong Yu
- Interdisciplinary Center for Biotechnology Research, University of Florida, 2033 Mowry Road, Gainesville, FL 32611, USA
| | - Yanping Zhang
- Interdisciplinary Center for Biotechnology Research, University of Florida, 2033 Mowry Road, Gainesville, FL 32611, USA
| |
Collapse
|
25
|
Chowdhury M, Zhang J, Xu XX, He Z, Lu Y, Liu XS, Wang YF, Yu XQ. An in vitro study of NF-κB factors cooperatively in regulation of Drosophila melanogaster antimicrobial peptide genes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 95:50-58. [PMID: 30735676 DOI: 10.1016/j.dci.2019.01.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 06/09/2023]
Abstract
An important innate immune response in Drosophila melanogaster is the production of antimicrobial peptides (AMPs). Expression of AMP genes is mediated by the Toll and immune deficiency (IMD) pathways via NF-κB transcription factors Dorsal, DIF and Relish. Dorsal and DIF act downstream of the Toll pathway, whereas Relish acts in the IMD pathway. Dorsal and DIF are held inactive in the cytoplasm by the IκB protein Cactus, while Relish contains an IκB-like inhibitory domain at the C-terminus. NF-κB factors normally form homodimers and heterodimers to regulate gene expression, but formation of heterodimers between Relish and DIF or Dorsal and the specificity and activity of the three NF-κB homodimers and heterodimers are not well understood. In this study, we compared the activity of Rel homology domains (RHDs) of Dorsal, DIF and Relish in activation of Drosophila AMP gene promoters, demonstrated that Relish-RHD (Rel-RHD) interacted with both Dorsal-RHD and DIF-RHD, Relish-N interacted with DIF and Dorsal, and overexpression of individual RHD and co-expression of any two RHDs activated the activity of AMP gene promoters to various levels, suggesting formation of homodimers and heterodimers among Dorsal, DIF and Relish. Rel-RHD homodimers were stronger activators than heterodimers of Rel-RHD with either DIF-RHD or Dorsal-RHD, while DIF-RHD-Dorsal-RHD heterodimers were stronger activators than either DIF-RHD or Dorsal-RHD homodimers in activation of AMP gene promoters. We also identified the nucleotides at the 6th and 8th positions of the 3' half-sites of the κB motifs that are important for the specificity and activity of NF-κB transcription factors.
Collapse
Affiliation(s)
- Munmun Chowdhury
- Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri - Kansas City, Kansas City, MO, 64110, USA
| | - Jie Zhang
- Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri - Kansas City, Kansas City, MO, 64110, USA
| | - Xiao-Xia Xu
- Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri - Kansas City, Kansas City, MO, 64110, USA; College of Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Zhen He
- Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri - Kansas City, Kansas City, MO, 64110, USA; School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Yuzhen Lu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, 510631, 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, 510631, China
| | - Xu-Sheng Liu
- School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Yu-Feng Wang
- School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Xiao-Qiang Yu
- Division of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri - Kansas City, Kansas City, MO, 64110, USA; School of Life Sciences, Central China Normal University, Wuhan, 430079, China; Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, 510631, 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, 510631, China.
| |
Collapse
|
26
|
Ramirez JL, Muturi EJ, Barletta ABF, Rooney AP. The Aedes aegypti IMD pathway is a critical component of the mosquito antifungal immune response. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 95:1-9. [PMID: 30582948 DOI: 10.1016/j.dci.2018.12.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 06/09/2023]
Abstract
Successful infection of the insect body by entomopathogenic fungi is the result of complex molecular interactions between the host and the invading pathogenic fungi. The mosquito antifungal response is multifaceted and is regulated in part by the Toll and Jak-STAT pathways. Here, we assessed the role of the IMD pathway in the mosquito Ae. aegypti antifungal immune response when challenged with one of two entomopathogenic fungi, Beauveria bassiana and Isaria javanica. IMD pathway components of the mosquito immune system were elicited in response to infection with both entomopathogenic fungi, primarily in the fat body of mosquitoes. Furthermore, we observed induction of antimicrobial peptides that in turn appear to be tissue and fungal strain-specific. IMD pathway impairment by RNAi gene silencing resulted in higher fungal proliferation and reduction in survival of fungi-infected mosquitoes. Collectively, these data indicates that the IMD pathway plays a more significant role in the antifungal immune response than previously recognized.
Collapse
Affiliation(s)
- José L Ramirez
- Crop Bioprotection Research Unit, National Center for Agricultural Utilization Research, Agricultural Research Service, United States Department of Agriculture, Peoria, IL, USA.
| | - Ephantus J Muturi
- Crop Bioprotection Research Unit, National Center for Agricultural Utilization Research, Agricultural Research Service, United States Department of Agriculture, Peoria, IL, USA
| | - Ana B F Barletta
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Alejandro P Rooney
- Crop Bioprotection Research Unit, National Center for Agricultural Utilization Research, Agricultural Research Service, United States Department of Agriculture, Peoria, IL, USA
| |
Collapse
|
27
|
Alonso-Palomares LA, Moreno-García M, Lanz-Mendoza H, Salazar MI. Molecular Basis for Arbovirus Transmission by Aedes aegypti Mosquitoes. Intervirology 2019; 61:255-264. [PMID: 31082816 DOI: 10.1159/000499128] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 02/17/2019] [Indexed: 11/19/2022] Open
Abstract
Mosquitoes are considered the most important vectors for the transmission of pathogens to humans. Aedes aegypti is a unique species, not only by its highly anthropophilic and peridomestic habits but also because it can transmit an important variety of pathogenic viruses. Examples are dengue, yellow fever, chikungunya, Zika, and Mayaro viruses. After ingesting viremic blood, a wide range of mechanisms are activated in the mosquito to counteract viral infection. Nevertheless, these arboviruses possess strategies to overcome barriers in the mosquito and eventually reach the salivary glands to continue the transmission cycle. However, the infection and eventual transmission of arbovirus depends on multiple factors. The current review focuses in detail on the anatomic, physiological, and molecular characteristics of the mosquito A. aegypti that participate in response to a viral infection. In the past decades, the awareness of the importance of this mosquito as a disease vector and its impact on human health was largely recognized. We need to improve our comprehension of molecular mechanisms that determine the outcome of successful virus replication or control of infection for each arbovirus in the vector; this could lead to the design of effective control strategies in the future.
Collapse
Affiliation(s)
- Luis A Alonso-Palomares
- Laboratorio de Virología e Inmunovirología, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional (ENCB-IPN), Unidad Profesional "Lázaro Cárdenas", Mexico City, Mexico.,Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública (CISEI-INSP), Cuernavaca, Mexico
| | - Miguel Moreno-García
- Centro Regional de Control de Vectores, Secretaría de Salud (CERECOVE-SS), Panchimalco, Mexico
| | - Humberto Lanz-Mendoza
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública (CISEI-INSP), Cuernavaca, Mexico
| | - Ma Isabel Salazar
- Laboratorio de Virología e Inmunovirología, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional (ENCB-IPN), Unidad Profesional "Lázaro Cárdenas", Mexico City, Mexico,
| |
Collapse
|
28
|
Lin CC, Chen YH, Guan TC, Chang SW, Pai H, Chou SJ, Tsai HP. Expression of foreign proteins by antimicrobial peptide gene promoters in mosquitoes. JOURNAL OF MEDICAL SCIENCES 2019. [DOI: 10.4103/jmedsci.jmedsci_194_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
29
|
Li TT, Ding ZF, Pan XT, Ma FT, Han KK, Wu L, Zhao LL, Ren Q, Zhang XW. Characterization of an immune deficiency (IMD) homolog from the oriental river prawn, Macrobrachium nipponense. FISH & SHELLFISH IMMUNOLOGY 2018; 83:115-122. [PMID: 30195908 DOI: 10.1016/j.fsi.2018.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 09/01/2018] [Accepted: 09/05/2018] [Indexed: 06/08/2023]
Abstract
The immune deficiency (IMD) signal pathway mediates innate immunity against Gram-negative bacteria in crustaceans. In the present study, an IMD homolog (MnIMD) from the oriental river prawn, Macrobrachium nipponense was identified. The full-length cDNA of MnIMD was 782bp with an open reading frame of 549 bp that encodes a putative protein of 182 amino acids including a death domain at the C-terminus. Multiple alignment analysis showed that IMDs in prawn M. nipponense and other crustaceans shared high similarity. The recombinant protein of MnIMD was expressed and purified for further functional analyses. Western blot analysis indicated that MnIMD was present in many tissues, but with the highest level in the gills, which was consistent with the qRT-PCR results. After Vibrio parahaemolyticus challenge, MnIMD was significantly induced in gills. RNA interference analysis showed that the IMD pathway was involved in regulating the expression of different antimicrobial peptide (AMP) genes, including Cru4 and Cru6. These results are helpful in promoting research on the innate immunity in M. nipponense.
Collapse
Affiliation(s)
- Ting-Ting Li
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China; College of Marine Science and Engineering, Nanjing Normal University, Nanjing, 210046, China
| | - Zheng-Feng Ding
- Jiangsu Key Laboratory for Biofunctional Molecules, College of Life Science and Chemistry, Jiangsu Second Normal University, Nanjing, 210013, China
| | - Xin-Tong Pan
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China
| | - Fu-Tong Ma
- College of Marine Science and Engineering, Nanjing Normal University, Nanjing, 210046, China
| | - Ke-Ke Han
- College of Marine Science and Engineering, Nanjing Normal University, Nanjing, 210046, China
| | - Lei Wu
- College of Marine Science and Engineering, Nanjing Normal University, Nanjing, 210046, China
| | - Ling-Ling Zhao
- College of Marine Science and Engineering, Nanjing Normal University, Nanjing, 210046, China
| | - Qian Ren
- College of Marine Science and Engineering, Nanjing Normal University, Nanjing, 210046, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, China.
| | - Xiao-Wen Zhang
- College of Life Science, Henan Normal University, Xinxiang, Henan, 453007, China.
| |
Collapse
|
30
|
Guo Z, Qin J, Zhou X, Zhang Y. Insect Transcription Factors: A Landscape of Their Structures and Biological Functions in Drosophila and beyond. Int J Mol Sci 2018; 19:ijms19113691. [PMID: 30469390 PMCID: PMC6274879 DOI: 10.3390/ijms19113691] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/17/2022] Open
Abstract
Transcription factors (TFs) play essential roles in the transcriptional regulation of functional genes, and are involved in diverse physiological processes in living organisms. The fruit fly Drosophila melanogaster, a simple and easily manipulated organismal model, has been extensively applied to study the biological functions of TFs and their related transcriptional regulation mechanisms. It is noteworthy that with the development of genetic tools such as CRISPR/Cas9 and the next-generation genome sequencing techniques in recent years, identification and dissection the complex genetic regulatory networks of TFs have also made great progress in other insects beyond Drosophila. However, unfortunately, there is no comprehensive review that systematically summarizes the structures and biological functions of TFs in both model and non-model insects. Here, we spend extensive effort in collecting vast related studies, and attempt to provide an impartial overview of the progress of the structure and biological functions of current documented TFs in insects, as well as the classical and emerging research methods for studying their regulatory functions. Consequently, considering the importance of versatile TFs in orchestrating diverse insect physiological processes, this review will assist a growing number of entomologists to interrogate this understudied field, and to propel the progress of their contributions to pest control and even human health.
Collapse
Affiliation(s)
- Zhaojiang Guo
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Jianying Qin
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
- Longping Branch, Graduate School of Hunan University, Changsha 410125, China.
| | - Xiaomao Zhou
- Longping Branch, Graduate School of Hunan University, Changsha 410125, China.
| | - Youjun Zhang
- Department of Plant Protection, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
31
|
Shaw WR, Catteruccia F. Vector biology meets disease control: using basic research to fight vector-borne diseases. Nat Microbiol 2018; 4:20-34. [PMID: 30150735 DOI: 10.1038/s41564-018-0214-7] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 06/29/2018] [Indexed: 12/11/2022]
Abstract
Human pathogens that are transmitted by insects are a global problem, particularly those vectored by mosquitoes; for example, malaria parasites transmitted by Anopheles species, and viruses such as dengue, Zika and chikungunya that are carried by Aedes mosquitoes. Over the past 15 years, the prevalence of malaria has been substantially reduced and virus outbreaks have been contained by controlling mosquito vectors using insecticide-based approaches. However, disease control is now threatened by alarming rates of insecticide resistance in insect populations, prompting the need to develop a new generation of specific strategies that can reduce vector-mediated transmission. Here, we review how increased knowledge in insect biology and insect-pathogen interactions is stimulating new concepts and tools for vector control. We focus on strategies that either interfere with the development of pathogens within their vectors or directly impact insect survival, including enhancement of vector-mediated immune control, manipulation of the insect microbiome, or use of powerful new genetic tools such as CRISPR-Cas systems to edit vector genomes. Finally, we offer a perspective on the implementation hurdles as well as the knowledge gaps that must be filled in the coming years to safely realize the potential of these novel strategies to eliminate the scourge of vector-borne disease.
Collapse
Affiliation(s)
- W Robert Shaw
- Harvard T. H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA, USA.
| | - Flaminia Catteruccia
- Harvard T. H. Chan School of Public Health, Department of Immunology and Infectious Diseases, Boston, MA, USA.
| |
Collapse
|
32
|
Kang S, Shin D, Noh MY, Peters JS, Smartt CT, Han YS, Hong YS. Optimization of double-stranded RNAi intrathoracic injection method in Aedes aegypti. ENTOMOLOGICAL RESEARCH 2018; 48:269-278. [PMID: 30505333 PMCID: PMC6261502 DOI: 10.1111/1748-5967.12300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
RNA interference is widely used to analyze gene functions via phenotypic knockdown of target transcripts in mosquitoes, which transmit numerous mosquito-borne diseases. Functional analysis of mosquito genes is indispensable to understand and reduce transmission of mosquito-borne diseases in mosquitoes. Intrathoracic injection of double-stranded RNA (dsRNA) remains the simplest and most customizable method in mosquitoes for functional analysis of the genes of interest. However, achieving consistent and effective knockdown by dsRNAi is often elusive and may require extensive optimization. We tested the effectiveness of gene silencing by intrathoracic injection of four different quantities of dsRNA targeting two Ae. aegypti genes, cysteine desulfurylase (Nfs1) and short-chain dehydrogenase (SDH). We found that Nfs1 gene has a lower expression level upon silencing than SDH gene. In the case of the gene that is easier to silence, Nfs1 gene expression was significantly silenced by all four tested quantities of dsRNA up to 21 d.p.i., but silencing of SDH, the gene that is difficult to silence, was less effective, with knockdown lasting up to 9 d.p.i. only when 1,000 ng of dsRNA was used. Based on our observation, intrathoracic injection of 500 ng of dsRNAs per mosquito is recommended to achieve effective knockdown for well-silenced transcripts such as Nfs1 for up to 3 weeks. This includes most in vivo bioassays involving arboviral infections in Ae. aegypti. The estimated quantities of dsRNA described in this study should be applicable to most Ae. aegypti dsRNAi studies and thus provide a guideline to develop efficient dsRNAi in other experimental investigations.
Collapse
Affiliation(s)
- Seokyoung Kang
- Dept. of Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Dongyoung Shin
- Dept. of Nematology and Entomology, University of Florida, Vero Beach, FL32962, USA
| | - Mi Young Noh
- Dept. of Agricultural Biology, Chonnam National University, Gwang-ju, 500-757, Republic of Korea
| | - Jill S. Peters
- Dept. of Tropical Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Chelsea T Smartt
- Dept. of Nematology and Entomology, University of Florida, Vero Beach, FL32962, USA
| | - Yeon Soo Han
- Dept. of Agricultural Biology, Chonnam National University, Gwang-ju, 500-757, Republic of Korea
| | | |
Collapse
|
33
|
Pan X, Pike A, Joshi D, Bian G, McFadden MJ, Lu P, Liang X, Zhang F, Raikhel AS, Xi Z. The bacterium Wolbachia exploits host innate immunity to establish a symbiotic relationship with the dengue vector mosquito Aedes aegypti. ISME JOURNAL 2017; 12:277-288. [PMID: 29099491 PMCID: PMC5739022 DOI: 10.1038/ismej.2017.174] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/30/2017] [Accepted: 09/06/2017] [Indexed: 01/15/2023]
Abstract
A host's immune system plays a central role in shaping the composition of the microbiota and, in return, resident microbes influence immune responses. Symbiotic associations of the maternally transmitted bacterium Wolbachia occur with a wide range of arthropods. It is, however, absent from the dengue and Zika vector mosquito Aedes aegypti in nature. When Wolbachia is artificially forced to form symbiosis with this new mosquito host, it boosts the basal immune response and enhances the mosquito's resistance to pathogens, including dengue, Zika virus and malaria parasites. The mechanisms involved in establishing a symbiotic relationship between Wolbachia and A. aegypti, and the long-term outcomes of this interaction, are not well understood. Here, we have demonstrated that both the immune deficiency (IMD) and Toll pathways are activated by the Wolbachia strain wAlbB upon its introduction into A. aegypti. Silencing the Toll and IMD pathways via RNA interference reduces the wAlbB load. Notably, wAlbB induces peptidoglycan recognition protein (PGRP)-LE expression in the carcass of A. aegypti, and its silencing results in a reduction of symbiont load. Using transgenic mosquitoes with stage-specific induction of the IMD and Toll pathways, we have shown that elevated wAlbB infection in these mosquitoes is maintained via maternal transmission. These results indicate that host innate immunity is utilized to establish and promote host-microbial symbiosis. Our results will facilitate a long-term projection of the stability of the Wolbachia-A. aegypti mosquito system that is being developed to control dengue and Zika virus transmission to humans.
Collapse
Affiliation(s)
- Xiaoling Pan
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.,School of Medicine, Hunan Normal University, Changsha, Hunan 410013, China
| | - Andrew Pike
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Deepak Joshi
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Guowu Bian
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Michael J McFadden
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Peng Lu
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Xiao Liang
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Fengrui Zhang
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Alexander S Raikhel
- Department of Entomology and Institute for Integrative Molecular Biology, University of California, Riverside, CA 92521, USA
| | - Zhiyong Xi
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA.,Sun Yat-sen University-Michigan State University Joint Center of Vector Control for Tropical Diseases, Guangzhou, Guangdong 510080, China
| |
Collapse
|
34
|
Zakovic S, Levashina EA. NF-κB-Like Signaling Pathway REL2 in Immune Defenses of the Malaria Vector Anopheles gambiae. Front Cell Infect Microbiol 2017; 7:258. [PMID: 28680852 PMCID: PMC5478692 DOI: 10.3389/fcimb.2017.00258] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 06/01/2017] [Indexed: 12/04/2022] Open
Abstract
The blood feeding requirements of insects are often exploited by pathogens for their transmission. This is also the case of the protozoan parasites of genus Plasmodium, the causative agents of malaria. Every year malaria claims the lives of a half million people, making its vector, the Anopheles mosquito, the deadliest animal in the world. However, mosquitoes mount powerful immune responses that efficiently limit parasite proliferation. Among the immune signaling pathways identified in the main malaria vector Anopheles gambiae, the NF-κB-like signaling cascades REL2 and REL1 are essential for eliciting proper immune reactions, but only REL2 has been implicated in the responses against the human malaria parasite Plasmodium falciparum. Instead, constitutive activation of REL1 causes massive killing of rodent malaria parasites. In this review, we summarize our present knowledge on the REL2 pathway in Anopheles mosquitoes and its role in mosquito immune responses to diverse pathogens, with a focus on Plasmodium. Mosquito-parasite interactions are crucial for malaria transmission and, therefore, represent a potential target for malaria control strategies.
Collapse
Affiliation(s)
- Suzana Zakovic
- Vector Biology, Max-Planck Institute for Infection BiologyBerlin, Germany
| | - Elena A Levashina
- Vector Biology, Max-Planck Institute for Infection BiologyBerlin, Germany
| |
Collapse
|
35
|
Shi Z, Liang H, Hou Y. Functional analysis of a NF-κB transcription factor in the immune defense of Oriental fruit fly, Bactrocera dorsalis Hendel (Diptera: Tephritidae). BULLETIN OF ENTOMOLOGICAL RESEARCH 2017; 107:251-260. [PMID: 27871341 DOI: 10.1017/s0007485316000845] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Although some novel antimicrobial peptides (AMP) have been successfully isolated from Bactrocera dorsalis Hendel, the mechanisms underlying the induction of these peptides are still elusive. The homolog of NF-κB transcription factor Relish, designated as BdRelish, was cloned from B. dorsalis. The full length cDNA of BdRelish is 3954 bp with an open reading frame that encodes 1013 amino acids. Similar to Drosophila Relish and the mammalian p100, it is a compound protein containing a conserved Rel homology domain, an IPT (Ig-like, plexins, transcription factors) domain and an IκB-like domain (four ankyrin repeats), the nuclear localization signal RKRRR is also detected at the residues 449-453, suggesting that it has homology to Relish and it is a member of the Rel family of transcription activator proteins. Reverse transcription quantitative polymerase chain reaction analysis reveals that BdRelish mRNAs are detected in different quantities from various tissues and the highest transcription level of BdRelish is determined in fat body. The injection challenge of Escherichia coli and Staphylococcus aureas significantly upregulated the expression of BdRelish. The injection of BdRelish dsRNA markedly reduced the expression of BdRelish and decreased the transcription magnitude of antimicrobial peptides. Individuals injected BdRelish dsRNA died at a significantly faster rate compared with the control groups. Therefore, BdRelish is vital for the transcription of AMPs to attack the invading bacteria.
Collapse
Affiliation(s)
- Z Shi
- State Key Laboratory of Ecological Pest Control of Fujian-Taiwan Crops,Fujian Agriculture and Forestry University,Fuzhou, 350002,China
| | - H Liang
- State Key Laboratory of Ecological Pest Control of Fujian-Taiwan Crops,Fujian Agriculture and Forestry University,Fuzhou, 350002,China
| | - Y Hou
- State Key Laboratory of Ecological Pest Control of Fujian-Taiwan Crops,Fujian Agriculture and Forestry University,Fuzhou, 350002,China
| |
Collapse
|
36
|
Barletta ABF, Nascimento-Silva MCL, Talyuli OAC, Oliveira JHM, Pereira LOR, Oliveira PL, Sorgine MHF. Microbiota activates IMD pathway and limits Sindbis infection in Aedes aegypti. Parasit Vectors 2017; 10:103. [PMID: 28231846 PMCID: PMC5324288 DOI: 10.1186/s13071-017-2040-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 02/16/2017] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Aedes aegypti is the main vector of important arboviruses such as dengue, Zika and chikungunya. During infections mosquitoes can activate the immune pathways Toll, IMD and JAK/STAT to limit pathogen replication. RESULTS Here, we evaluate the immune response profile of Ae. aegypti against Sindbis virus (SINV). We analyzed gene expression of components of Toll, IMD and JAK/STAT pathways and showed that a blood meal and virus infection upregulated aaREL2 in a microbiota-dependent fashion, since this induction was prevented by antibiotic. The presence of the microbiota activates IMD and impaired the replication of SINV in the midgut. Constitutive activation of the IMD pathway, by Caspar depletion, leads to a decrease in microbiota levels and an increase in SINV loads. CONCLUSION Together, these results suggest that a blood meal is able to activate innate immune pathways, through a nutrient induced growth of microbiota, leading to upregulation of aaREL2 and IMD activation. Microbiota levels seemed to have a reciprocal interaction, where the proliferation of the microbiota activates IMD pathway that in turn controls bacterial levels, allowing SINV replication in Ae. aegypti mosquitoes. The activation of the IMD pathway seems to have an indirect effect in SINV levels that is induced by the microbiota.
Collapse
Affiliation(s)
- Ana Beatriz Ferreira Barletta
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | | | - Octávio A C Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - José Henrique M Oliveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | | | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcos Henrique F Sorgine
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
37
|
Zhang H, Dong S, Chen X, Stanley D, Beerntsen B, Feng Q, Song Q. Relish2 mediates bursicon homodimer-induced prophylactic immunity in the mosquito Aedes aegypti. Sci Rep 2017; 7:43163. [PMID: 28225068 PMCID: PMC5320557 DOI: 10.1038/srep43163] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/20/2017] [Indexed: 01/12/2023] Open
Abstract
Bursicon is a neuropeptide hormone consisting of two cystine-knot proteins (burs α and burs β), responsible for cuticle tanning and other developmental processes in insects. Recent studies show that each bursicon subunit forms homodimers that induce prophylactic immunity in Drosophila melanogaster. Here, we investigated the hypothesis that bursicon homodimers act in prophylactic immunity in insects, and possibly arthropods, generally, using the mosquito, Aedes aegypti. We found that burs α and burs β are expressed in larvae, pupae and newly emerged adults. Treating newly emerged Ae. aegypti and D. melanogaster adults with recombinant bursicon (r-bursicon) heterodimer led to cuticle tanning in both species. Treating larvae and adults with r-bursicon homodimers led to up-regulation of five anti-microbial peptide (AMP) genes, noting the possibility that bursicon heterodimers also lead to up-regulation of these genes can not been excluded. The induced AMPs effectively suppressed the growth of bacteria in vitro. RNAi knock-down of the transcriptional factor Relish2 abolished the influence of r-bursicon homodimers on AMP production. We infer the bursicon homodimers induce expression of AMP genes via Relish2 in Ae. aegypti, as prophylactic immunity to protect mosquitoes during the vulnerable stages of each molt.
Collapse
Affiliation(s)
- Hongwei Zhang
- Division of Plant Sciences, University of Missouri, Columbia, Missouri, USA
| | - Shengzhang Dong
- Division of Plant Sciences, University of Missouri, Columbia, Missouri, USA
| | - Xi Chen
- Division of Plant Sciences, University of Missouri, Columbia, Missouri, USA
| | - David Stanley
- USDA/Agricultural Research Service, Biological Control of Insects Research Laboratory, Columbia, Missouri, USA
| | - Brenda Beerntsen
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, USA
| | - Qili Feng
- Guangzhou Key Laboratory of Insect Development Regulation and Application Research, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Qisheng Song
- Division of Plant Sciences, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
38
|
Simões ML, Dong Y, Hammond A, Hall A, Crisanti A, Nolan T, Dimopoulos G. The Anopheles FBN9 immune factor mediates Plasmodium species-specific defense through transgenic fat body expression. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 67:257-265. [PMID: 27667688 DOI: 10.1016/j.dci.2016.09.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 09/20/2016] [Accepted: 09/21/2016] [Indexed: 06/06/2023]
Abstract
Mosquitoes have a multifaceted innate immune system that is actively engaged in warding off various pathogens, including the protozoan malaria parasite Plasmodium. Various immune signaling pathways and effectors have been shown to mediate a certain degree of defense specificity against different Plasmodium species. A key pattern recognition receptor of the Anopheles gambiae immune system is the fibrinogen domain-containing immunolectin FBN9, which has been shown to be transcriptonally induced by Plasmodium infection, and to mediate defense against both rodent and human malaria parasites and bacteria. Here we have further studied the defense specificity of FBN9 using a transgenic approach, in which FBN9 is overexpressed in the fat body tissue after a blood meal through a vitellogenin promoter. Interestingly, the Vg-FBN9 transgenic mosquitoes showed increased resistance only to the rodent parasite P. berghei, and not to the human parasite P. falciparum, pointing to differences in the mosquito's defense mechanisms against the two parasite species. The Vg-FBN9 transgenic mosquitoes were also more resistant to infection with both Gram-positive and Gram-negative bacteria and showed increased longevity when infected with P. berghei. Our study points to the importance of both experimentally depleting and enriching candidate anti-Plasmodium effectors in functional studies in order to ascertain their suitability for the development of transgenic mosquito-based malaria control strategies.
Collapse
Affiliation(s)
- Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St., Baltimore, MD 21205, USA
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St., Baltimore, MD 21205, USA
| | - Andrew Hammond
- Department of Life Sciences, Imperial College London, SW7 2AZ, UK
| | - Ann Hall
- Department of Life Sciences, Imperial College London, SW7 2AZ, UK
| | - Andrea Crisanti
- Department of Life Sciences, Imperial College London, SW7 2AZ, UK
| | - Tony Nolan
- Department of Life Sciences, Imperial College London, SW7 2AZ, UK
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe St., Baltimore, MD 21205, USA.
| |
Collapse
|
39
|
de Mendonça Amarante A, Jupatanakul N, de Abreu da Silva IC, Carneiro VC, Vicentino ARR, Dimopolous G, Talyuli OAC, Fantappié MR. The DNA chaperone HMGB1 potentiates the transcriptional activity of Rel1A in the mosquito Aedes aegypti. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2017; 80:32-41. [PMID: 27867076 DOI: 10.1016/j.ibmb.2016.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 06/06/2023]
Abstract
High Mobility Group protein 1 (HMGB1) is a non-histone, chromatin-associated nuclear protein that functions in regulating eukaryotic gene expression. We investigated the influence and mechanism of action of Aedes aegypti HMGB1 (AaHMGB1) on mosquito Rel1A-mediated transcription from target gene promoters. The DNA-binding domain (RHD) of AaRel1A was bacterially expressed and purified, and AaHMGB1 dramatically enhanced RHD binding to consensus NF-kB/Rel DNA response elements. Luciferase reporter analyses using a cecropin gene promoter showed that AaHMGB1 potentiates the transcriptional activity of AaRel1A in Aag-2 cells. Moreover, overexpression of AaHMGB1 in Aag-2 cells led to an increase in mRNA levels of antimicrobial peptide genes. In vitro GST pull-down assays revealed that the presence of DNA is a pre-requisite for assembly of a possible ternary complex containing DNA, AaHMGB1 and AaRel1A. Notably, DNA bending by AaHMGB1 enhanced the binding of AaRel1A to a DNA fragment containing a putative NF-kB/Rel response element. Importantly, AaHMGB1 was identified as a potential immune modulator in A. aegypti through AaHMGB1 overexpression or RNAi silencing in Aag-2 cells followed by bacterial challenge or through AaHMGB1 RNAi knockdown in mosquitoes followed by Dengue virus (DENV) infection. We propose a model in which AaHMGB1 bends NF-kB/Rel target DNA to recruit and allow more efficient AaRel1A binding to activate transcription of effector genes, culminating in a stronger Toll pathway-mediated response against DENV infection.
Collapse
Affiliation(s)
- Anderson de Mendonça Amarante
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Brazil; Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Brazil
| | - Natapong Jupatanakul
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, United States
| | - Isabel Caetano de Abreu da Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Brazil; Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Brazil
| | - Vitor Coutinho Carneiro
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Brazil; Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Brazil
| | - Amanda Roberta Revoredo Vicentino
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Brazil; Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Brazil
| | - George Dimopolous
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Brazil; Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Brazil; W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, United States
| | - Octávio Augusto C Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Brazil; Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Brazil
| | - Marcelo Rosado Fantappié
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Brazil; Instituto Nacional de Entomologia Molecular, Universidade Federal do Rio de Janeiro, Brazil.
| |
Collapse
|
40
|
Khan MB, Liew JWK, Leong CS, Lau YL. Role of NF-kβ factor Rel2 during Plasmodium falciparum and bacterial infection in Anopheles dirus. Parasit Vectors 2016; 9:525. [PMID: 27688040 PMCID: PMC5041562 DOI: 10.1186/s13071-016-1810-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 09/21/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Anopheles mosquitoes transmit malaria which is one of the world's most threatening diseases. Anopheles dirus (sensu stricto) is among the main vectors of malaria in South East Asia. The mosquito innate immune response is the first line of defence against malaria parasites during its development. The immune deficiency (IMD) pathway, a conserved immune signaling pathway, influences anti-Plasmodium falciparum activity in Anopheles gambiae, An. stephensi and An. albimanus. The aim of the study was to determine the role of Rel2, an IMD pathway-controlled NF-kappaβ transcription factor, in An. dirus. METHODS RACE (Rapid amplification of cDNA ends) was performed on the Rel2 gene. Double-stranded Rel2 was constructed and injected into the thorax of female mosquitoes. The injected mosquitoes were fed on a P. falciparum gametocyte culture and dissected on day 7-9 post-feeding in order to count the oocysts. A survival analysis was conducted by exposing the dsRNA injected mosquitoes to Gram-positive and Gram-negative bacteria. RESULTS This study demonstrated that the Rel2 gene in An. dirus has two isoforms, short length and full length. RNA interference-mediated gene silencing of Rel2 showed that the latter is involved in protection against P. falciparum, Gram-positive bacteria (Micrococcus luteus) with Lys-type peptidoglycan and Gram-negative bacteria (Escherichia coli) with DAP-type peptidoglycan. CONCLUSION This study suggested that there are similarities in the splicing events and functionality of the Rel2 gene, between the Anopheles species. Among all the important anophelines, the immunity of only a few has been thoroughly investigated. In order to develop novel vector-based control strategies and restrict malaria transmission, the immune pathways of these important vectors should be thoroughly investigated.
Collapse
Affiliation(s)
- Mohammad Behram Khan
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Jonathan Wee Kent Liew
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Cherng Shii Leong
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yee-Ling Lau
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
41
|
Caljon G, De Muylder G, Durnez L, Jennes W, Vanaerschot M, Dujardin JC. Alice in microbes' land: adaptations and counter-adaptations of vector-borne parasitic protozoa and their hosts. FEMS Microbiol Rev 2016; 40:664-85. [PMID: 27400870 DOI: 10.1093/femsre/fuw018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2016] [Indexed: 12/24/2022] Open
Abstract
In the present review, we aim to provide a general introduction to different facets of the arms race between pathogens and their hosts/environment, emphasizing its evolutionary aspects. We focus on vector-borne parasitic protozoa, which have to adapt to both invertebrate and vertebrate hosts. Using Leishmania, Trypanosoma and Plasmodium as main models, we review successively (i) the adaptations and counter-adaptations of parasites and their invertebrate host, (ii) the adaptations and counter-adaptations of parasites and their vertebrate host and (iii) the impact of human interventions (chemotherapy, vaccination, vector control and environmental changes) on these adaptations. We conclude by discussing the practical impact this knowledge can have on translational research and public health.
Collapse
Affiliation(s)
- Guy Caljon
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Health, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Géraldine De Muylder
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium
| | - Lies Durnez
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium
| | - Wim Jennes
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium
| | - Manu Vanaerschot
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium Columbia University, College of Physicians and Surgeons, Department of Microbiology and Immunology, Fidock Lab, New York, NY 10032, USA
| | - Jean-Claude Dujardin
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Health, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| |
Collapse
|
42
|
Merga YJ, O'Hara A, Burkitt MD, Duckworth CA, Probert CS, Campbell BJ, Pritchard DM. Importance of the alternative NF-κB activation pathway in inflammation-associated gastrointestinal carcinogenesis. Am J Physiol Gastrointest Liver Physiol 2016; 310:G1081-90. [PMID: 27102559 DOI: 10.1152/ajpgi.00026.2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/17/2016] [Indexed: 02/07/2023]
Abstract
Chronic inflammation is a common factor in the development of many gastrointestinal malignancies. Examples include inflammatory bowel disease predisposing to colorectal cancer, Barrett's esophagus as a precursor of esophageal adenocarcinoma, and Helicobacter pylori-induced gastric cancer. The classical activation pathway of NF-κB signaling has been identified as regulating several sporadic and inflammation-associated gastrointestinal tract malignancies. Emerging evidence suggests that the alternative NF-κB signaling pathway also exerts a distinct influence on these processes. This review brings together current knowledge of the role of the alternative NF-κB signaling pathway in the gastrointestinal tract, with a particular emphasis on inflammation-associated cancer development.
Collapse
Affiliation(s)
- Yvette J Merga
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Adrian O'Hara
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Michael D Burkitt
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Carrie A Duckworth
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Christopher S Probert
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Barry J Campbell
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - D Mark Pritchard
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
43
|
Li X, Meng K, Qiao J, Liu H, Zhong C, Liu Q. Identification of Aadnr1, a novel gene related to innate immunity and apoptosis in Aedes albopictus. Gene 2016; 587:18-26. [PMID: 27045774 DOI: 10.1016/j.gene.2016.03.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 03/24/2016] [Accepted: 03/25/2016] [Indexed: 12/01/2022]
Abstract
Innate immunity and apoptosis play critical roles in defending pathogens in insects. In Drosophila, Dnr1 was reported as a negative regulator of apoptosis and immune deficiency (Imd) pathway which belongs to innate immunity. Aedes albopictus is an important kind of arbovirus vector and becoming a significant threat to public health due to its rapid global expansion. Here we identified an ortholog of dnr1 from A. albopictus, named as Aadnr1. Aadnr1 encoded a putative protein containing an N-terminal FERM domain and a C-terminal RING domain. AaDnr1 shared high identity with dipteran insects Dnr1 orthologs. Phylogenetic analyses showed that the closest relative of AaDnr1 was Aedes aegypti Dnr1. Real-time PCR proved that Aadnr1 mRNA was expressed ubiquitously during developmental and adult stages. Transcriptional levels of Aadnr1 were decreased drastically in C6/36 cells underwent apoptosis induced by Actinomycin D (Act D) treatment. Partial silence of Aadnr1 enhanced Act D-induced caspase activity. When challenged by heat-inactivated E. coli, transcriptional level of Aadnr1 was also decreased dramatically in C6/36 cells. While when C6/36 cells were infected with Sindbis virus TE/GFP, transcriptional level of Aadnr1 was reduced and recovered repeatedly, with an overall decreasing trend. It was also shown in this study that similar to Drosophila Dnr1, RING domain destabilized AaDnr1 protein. Taken together, the study identified an innate immunity and apoptosis related gene Aadnr1 in A. albopictus.
Collapse
Affiliation(s)
- Xiaomei Li
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Kun Meng
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Jialu Qiao
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Hao Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Chunyan Zhong
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Qingzhen Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China.
| |
Collapse
|
44
|
Sim S, Hibberd ML. Genomic approaches for understanding dengue: insights from the virus, vector, and host. Genome Biol 2016; 17:38. [PMID: 26931545 PMCID: PMC4774013 DOI: 10.1186/s13059-016-0907-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The incidence and geographic range of dengue have increased dramatically in recent decades. Climate change, rapid urbanization and increased global travel have facilitated the spread of both efficient mosquito vectors and the four dengue virus serotypes between population centers. At the same time, significant advances in genomics approaches have provided insights into host–pathogen interactions, immunogenetics, and viral evolution in both humans and mosquitoes. Here, we review these advances and the innovative treatment and control strategies that they are inspiring.
Collapse
Affiliation(s)
- Shuzhen Sim
- Infectious Diseases, Genome Institute of Singapore, Singapore, 138672, Singapore
| | - Martin L Hibberd
- Infectious Diseases, Genome Institute of Singapore, Singapore, 138672, Singapore. .,Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK.
| |
Collapse
|
45
|
Co-expression of Dorsal and Rel2 Negatively Regulates Antimicrobial Peptide Expression in the Tobacco Hornworm Manduca sexta. Sci Rep 2016; 6:20654. [PMID: 26847920 PMCID: PMC4742911 DOI: 10.1038/srep20654] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/30/2015] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor κB (NF-κB) plays an essential role in regulation of innate immunity. In mammals, NF-κB factors can form homodimers and heterodimers to activate gene expression. In insects, three NF-κB factors, Dorsal, Dif and Relish, have been identified to activate antimicrobial peptide (AMP) gene expression. However, it is not clear whether Dorsal (or Dif) and Relish can form heterodimers. Here we report the identification and functional analysis of a Dorsal homologue (MsDorsal) and two Relish short isoforms (MsRel2A and MsRel2B) from the tobacco hornworm, Manduca sexta. Both MsRel2A and MsRel2B contain only a Rel homology domain (RHD) and lack the ankyrin-repeat inhibitory domain. Overexpression of the RHD domains of MsDorsal and MsRel2 in Drosophila melanogaster S2 and Spodoptera frugiperda Sf9 cells can activate AMP gene promoters from M. sexta and D. melanogaster. We for the first time confirmed the interaction between MsDorsal-RHD and MsRel2-RHD, and suggesting that Dorsal and Rel2 may form heterodimers. More importantly, co-expression of MsDorsal-RHD with MsRel2-RHD suppressed activation of several M. sexta AMP gene promoters. Our results suggest that the short MsRel2 isoforms may form heterodimers with MsDorsal as a novel mechanism to prevent over-activation of antimicrobial peptides.
Collapse
|
46
|
Pietri JE, Pietri EJ, Potts R, Riehle MA, Luckhart S. Plasmodium falciparum suppresses the host immune response by inducing the synthesis of insulin-like peptides (ILPs) in the mosquito Anopheles stephensi. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 53:134-44. [PMID: 26165161 PMCID: PMC4536081 DOI: 10.1016/j.dci.2015.06.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 06/17/2015] [Accepted: 06/17/2015] [Indexed: 05/12/2023]
Abstract
The insulin-like peptides (ILPs) and their respective signaling and regulatory pathways are highly conserved across phyla. In invertebrates, ILPs regulate diverse physiological processes, including metabolism, reproduction, behavior, and immunity. We previously reported that blood feeding alone induced minimal changes in ILP expression in Anopheles stephensi. However, ingestion of a blood meal containing human insulin or Plasmodium falciparum, which can mimic insulin signaling, leads to significant increases in ILP expression in the head and midgut, suggesting a potential role for AsILPs in the regulation of P. falciparum sporogonic development. Here, we show that soluble P. falciparum products, but not LPS or zymosan, directly induced AsILP expression in immortalized A. stephensi cells in vitro. Further, AsILP expression is dependent on signaling by the mitogen-activated protein kinase kinase/extracellular signal-regulated kinase (MEK/ERK) and phosphatidylinositol 3'-kinase (PI3K)/Akt branches of the insulin/insulin-like growth factor signaling (IIS) pathway. Inhibition of P. falciparum-induced ILPs in vivo decreased parasite development through kinetically distinct effects on mosquito innate immune responses. Specifically, knockdown of AsILP4 induced early expression of immune effector genes (1-6 h after infection), a pattern associated with significantly reduced parasite abundance prior to invasion of the midgut epithelium. In contrast, knockdown of AsILP3 increased later expression of the same genes (24 h after infection), a pattern that was associated with significantly reduced oocyst development. These data suggest that P. falciparum parasites alter the expression of mosquito AsILPs to dampen the immune response and facilitate their development in the mosquito vector.
Collapse
Affiliation(s)
- Jose E Pietri
- Department of Medical Microbiology and Immunology, 3437 Tupper Hall, One Shields Avenue, School of Medicine, University of California, Davis, CA 95616, USA.
| | - Eduardo J Pietri
- Department of Medical Microbiology and Immunology, 3437 Tupper Hall, One Shields Avenue, School of Medicine, University of California, Davis, CA 95616, USA.
| | - Rashaun Potts
- Department of Medical Microbiology and Immunology, 3437 Tupper Hall, One Shields Avenue, School of Medicine, University of California, Davis, CA 95616, USA.
| | - Michael A Riehle
- Department of Entomology, 410 Forbes, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ 85721, USA.
| | - Shirley Luckhart
- Department of Medical Microbiology and Immunology, 3437 Tupper Hall, One Shields Avenue, School of Medicine, University of California, Davis, CA 95616, USA.
| |
Collapse
|
47
|
Kean J, Rainey SM, McFarlane M, Donald CL, Schnettler E, Kohl A, Pondeville E. Fighting Arbovirus Transmission: Natural and Engineered Control of Vector Competence in Aedes Mosquitoes. INSECTS 2015; 6:236-78. [PMID: 26463078 PMCID: PMC4553541 DOI: 10.3390/insects6010236] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/18/2015] [Accepted: 03/10/2015] [Indexed: 01/09/2023]
Abstract
Control of aedine mosquito vectors, either by mosquito population reduction or replacement with refractory mosquitoes, may play an essential role in the fight against arboviral diseases. In this review, we will focus on the development and application of biological approaches, both natural or engineered, to limit mosquito vector competence for arboviruses. The study of mosquito antiviral immunity has led to the identification of a number of host response mechanisms and proteins that are required to control arbovirus replication in mosquitoes, though more factors influencing vector competence are likely to be discovered. We will discuss key aspects of these pathways as targets either for selection of naturally resistant mosquito populations or for mosquito genetic manipulation. Moreover, we will consider the use of endosymbiotic bacteria such as Wolbachia, which in some cases have proven to be remarkably efficient in disrupting arbovirus transmission by mosquitoes, but also the use of naturally occurring insect-specific viruses that may interfere with arboviruses in mosquito vectors. Finally, we will discuss the use of paratransgenesis as well as entomopathogenic fungi, which are also proposed strategies to control vector competence.
Collapse
Affiliation(s)
- Joy Kean
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Stephanie M Rainey
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Melanie McFarlane
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Claire L Donald
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Esther Schnettler
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| | - Emilie Pondeville
- MRC-University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, Scotland, UK.
| |
Collapse
|
48
|
Mosquito immunity against arboviruses. Viruses 2014; 6:4479-504. [PMID: 25415198 PMCID: PMC4246235 DOI: 10.3390/v6114479] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 10/30/2014] [Accepted: 11/11/2014] [Indexed: 01/03/2023] Open
Abstract
Arthropod-borne viruses (arboviruses) pose a significant threat to global health, causing human disease with increasing geographic range and severity. The recent availability of the genome sequences of medically important mosquito species has kick-started investigations into the molecular basis of how mosquito vectors control arbovirus infection. Here, we discuss recent findings concerning the role of the mosquito immune system in antiviral defense, interactions between arboviruses and fundamental cellular processes such as apoptosis and autophagy, and arboviral suppression of mosquito defense mechanisms. This knowledge provides insights into co-evolutionary processes between vector and virus and also lays the groundwork for the development of novel arbovirus control strategies that target the mosquito vector.
Collapse
|
49
|
Wu RCC, Cho WL. Cloning and characterization of microbial activated Aedes aegypti MEK4 (AaMEK4): influences of noncatalytic domains on enzymatic activity. INSECT MOLECULAR BIOLOGY 2014; 23:644-655. [PMID: 25039995 DOI: 10.1111/imb.12116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Protein kinases are known to be involved in a number of signal transduction cascades. Both the stress-activated Jun N-terminal kinase (JNK) and mitogen-activated protein kinase (MAPK) p38 pathways have been shown to correlate with the insect immune response to microbial infection. MAP kinase kinase 4 (MEK4) is an upstream kinase of JNK and p38 kinase. The cDNA of AaMEK4 was cloned and characterized. AaMEK4 was activated by microbial lysates of Gram-positive, Gram-negative bacteria and yeast. The conserved lysine (K112 ) and the putative phosphorylation sites (S238 and T242 ) were shown to be important for kinase activity by site-directed mutagenesis. A common MAPK docking site (MAPK_dsA) was found and in addition, a new nearby docking site, MAPK_dsB, was identified in the N-terminal noncatalytic domain of AaMEK4. MAPK_dsB was shown to be a unique element in the MEK4 family. In this study, both MAPK_dsA and _dsB were demonstrated to be important to AaMEK4 enzymatic activity for the downstream protein kinase, Aap38.
Collapse
Affiliation(s)
- R C-C Wu
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei City, Taiwan
| | | |
Collapse
|
50
|
Geiser DL, Zhou G, Mayo JJ, Winzerling JJ. The effect of bacterial challenge on ferritin regulation in the yellow fever mosquito, Aedes aegypti. INSECT SCIENCE 2013; 20:601-19. [PMID: 23956079 PMCID: PMC4554699 DOI: 10.1111/j.1744-7917.2012.01581.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/03/2012] [Indexed: 05/02/2023]
Abstract
Secreted ferritin is the major iron storage and transport protein in insects. Here, we characterize the message and protein expression profiles of yellow fever mosquito (Aedes aegypti) ferritin heavy chain homologue (HCH) and light chain homologue (LCH) subunits in response to iron and bacterial challenge. In vivo experiments demonstrated tissue-specific regulation of HCH and LCH expression over time post-blood meal (PBM). Transcriptional regulation of HCH and LCH was treatment specific, with differences in regulation for naïve versus mosquitoes challenged with heat-killed bacteria (HKB). Translational regulation by iron regulatory protein (IRP) binding activity for the iron-responsive element (IRE) was tissue-specific and time-dependent PBM. However, mosquitoes challenged with HKB showed little change in IRP/IRE binding activity compared to naïve animals. The changes in ferritin regulation and expression in vivo were confirmed with in vitro studies. We challenged mosquitoes with HKB followed by a blood meal to determine the effects on ferritin expression, and demonstrate a synergistic, time-dependent regulation of expression for HCH and LCH.
Collapse
Affiliation(s)
- Dawn L Geiser
- Department of Nutritional Sciences, College of Agriculture and Life Sciences, the University of Arizona, Tucson, Arizona, 85721, USA
| | | | | | | |
Collapse
|