51
|
Vivero-Gomez RJ, Mesa GB, Higuita-Castro J, Robledo SM, Moreno-Herrera CX, Cadavid-Restrepo G. Detection of Quorum Sensing Signal Molecules, Particularly N-Acyl Homoserine Lactones, 2-Alky-4-Quinolones, and Diketopiperazines, in Gram-Negative Bacteria Isolated From Insect Vector of Leishmaniasis. FRONTIERS IN TROPICAL DISEASES 2021. [DOI: 10.3389/fitd.2021.760228] [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
Gram-negative bacteria are known to use a quorum sensing system to facilitate and stimulate cell to cell communication, mediated via regulation of specific genes. This system is further involved in the modulation of cell density and metabolic and physiological processes that putatively either affect the survival of insect vectors or the establishment of pathogens transmitted by them. The process of quorum sensing generally involves N-acyl homoserine lactones and 2-alkyl-4-quinolones signaling molecules. The present study aimed to detect and identify quorum sensing signaling molecules of AHLs and AHQs type that are secreted by intestinal bacteria, and link their production to their extracellular milieu and intracellular content. Isolates for assessment were obtained from the intestinal tract of Pintomyia evansi (Leishmania insect vector). AHLs and AHQs molecules were detected using chromatography (TLC) assays, with the aid of specific and sensitive biosensors. For identity confirmation, ultra-high-performance liquid chromatography coupled with mass spectrometry was used. TLC assays detected quorum sensing molecules (QSM) in the supernatant of the bacterial isolates and intracellular content. Interestingly, Pseudomonas otitidis, Enterobacter aerogenes, Enterobacter cloacae, and Pantoea ananatis isolates showed a migration pattern similar to the synthetic molecule 3-oxo-C6-HSL (OHHL), which was used as a control. Enterobacter cancerogenus secreted C6-HSL, a related molecules to N-hexanoyl homoserine lactone (HHL), while Acinetobacter gyllenbergii exhibited a migration pattern similar to 2-heptyl-4-quinolone (HHQ) molecules. In comparison to this, 3-oxo-C12-HSL (OdDHL) type molecules were produced by Lysobacter soli, Pseudomonas putida, A. gyllenbergii, Acinetobacter calcoaceticus, and Pseudomonas aeruginosa, while Enterobacter cloacae produced molecules similar to 2-heptyl-3-hydroxy-4-quinolone (PQS). For Pseudomonas putida, Enterobacter aerogenes, P. ananatis, and Pseudomonas otitidis extracts, peak chromatograms with distinct retention times and areas, consistent with the molecules described in case of TLC, were obtained using HPLC. Importantly, P. ananatis produced a greater variety of high QSM concentration, and thus served as a reference for confirmation and identification by UHPLC-MRM-MS/MS. The molecules that were identified included N-hexanoyl-L-homoserine lactone [HHL, C10H18NO3, (M + H)], N-(3-oxohexanoyl)-L-homoserine lactone [OHHL, C10H16NO4, (M + H)], N-(3-oxododecanoyl)-L-homoserine lactone [OdDHL, C16H28NO4, (M + H)], and 2-heptyl-3-hydroxy-4(1H)-quinolone [PQS, C16H22NO2, (M + H)]. Besides this, the detection of diketopiperazines, namely L-Pro-L-Tyr and ΔAla-L-Val cyclopeptides was reported for P. ananatis. These molecules might be potentially associated with the regulation of QSM system, and might represent another small molecule-mediated bacterial sensing system. This study presents the first report regarding the detection and identification of QSM and diketopiperazines in the gut sand fly bacteria. The possible effect of QSM on the establishment of Leishmania must be explored to determine its role in the modulation of intestinal microbiome and the life cycle of Pi. evansi.
Collapse
|
52
|
Díaz S, Camargo C, Avila FW. Characterization of the reproductive tract bacterial microbiota of virgin, mated, and blood-fed Aedes aegypti and Aedes albopictus females. Parasit Vectors 2021; 14:592. [PMID: 34852835 PMCID: PMC8638121 DOI: 10.1186/s13071-021-05093-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022] Open
Abstract
Background Aedes aegypti and Ae. albopictus are vectors of numerous arboviruses that adversely affect human health. In mosquito vectors of disease, the bacterial microbiota influence several physiological processes, including fertility and vector competence, making manipulation of the bacterial community a promising method to control mosquito vectors. In this study, we describe the reproductive tract tissue microbiota of lab-reared virgin Ae. aegypti and Ae. albopictus males, and virgin, mated, and mated + blood-fed females of each species, comparing the bacterial composition found there to the well-described gut microbiota. Methods We performed metabarcoding of the 16S rRNA isolated from the gut, upper reproductive tract (URT; testes or ovaries), and lower reproductive tract (LRT; males: seminal vesicles and accessory glands; females: oviduct, spermathecae, and bursa) for each species, and evaluated the influence of host species, tissue, nutritional status, and reproductive status on microbiota composition. Finally, based on the identified taxonomic profiles of the tissues assessed, bacterial metabolic pathway abundance was predicted. Results The community structure of the reproductive tract is unique compared to the gut. Asaia is the most prevalent OTU in the LRTs of both Ae. aegypti and Ae. albopictus. In the URT, we observed differences between species, with Wolbachia OTUs being dominant in the Ae. albopictus URT, while Enterobacter and Serratia were dominant in Ae. aegypti URT. Host species and tissue were the best predictors of the community composition compared to reproductive status (i.e., virgin or mated) and nutritional status (i.e., sugar or blood-fed). The predicted functional profile shows changes in the abundance of specific microbial pathways that are associated with mating and blood-feeding, like energy production in mated tissues and siderophore synthesis in blood-fed female tissues. Conclusions Aedes aegypti and Ae. albopictus have distinct differences in the composition of microbiota found in the reproductive tract. The distribution of the bacterial taxonomic groups indicates that some bacteria have tissue-specific tropism for reproductive tract tissue, such as Asaia and Wolbachia. No significant differences in the taxonomic composition were observed in the reproductive tract between virgin, mated, and mated + blood-fed females, but changes in the abundance of specific metabolic pathways were found in the predicted microbial functional profiles in mated and blood-fed females. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-05093-7.
Collapse
Affiliation(s)
- Sebastián Díaz
- Max Planck Tandem Group in Mosquito Reproductive Biology, Universidad de Antioquia, Medellín, 050010, Antioquia, Colombia
| | - Carolina Camargo
- Max Planck Tandem Group in Mosquito Reproductive Biology, Universidad de Antioquia, Medellín, 050010, Antioquia, Colombia
| | - Frank W Avila
- Max Planck Tandem Group in Mosquito Reproductive Biology, Universidad de Antioquia, Medellín, 050010, Antioquia, Colombia.
| |
Collapse
|
53
|
de Melo Lara L, Pereira-Filho AA, Mateus Pereira RH, Ferreira Malta LG, D'Ávila Pessoa GC, Koerich LB, Pereira MH, Araujo RN, de Figueiredo Gontijo N, Viana Sant'Anna MR. Adaptations to haematophagy: Investigations on how male and female Culex quinquefasciatus (Diptera: Culicidae) deal with human complement activation after a blood meal. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 139:103650. [PMID: 34571142 DOI: 10.1016/j.ibmb.2021.103650] [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: 02/03/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 06/13/2023]
Abstract
Culex quinquefasciatus is a mosquito species with an anthropophilic habit, often associated with areas with poor sanitation in tropical and urban regions. Adult males and females feed on sugars but only females feed on blood in natural conditions for egg maturation. During haematophagy, female C. quinquefasciatus transmit pathogens such as the West Nile virus, Oropouche virus, various encephalitis viruses, and Wuchereria bancrofti to human hosts. It has been observed in laboratory conditions that male C. quinquefasciatus may feed on blood during an artificial feed. Experiments were carried out to understand how males and females of this species deal with human complement activation. Our results showed that female C. quinquefasciatus, but not males, withstand the stress caused by the ingestion of normal human serum. It was observed that the salivary gland extracts from female mosquitoes were able to inhibit the classical and lectin pathways, whereas male salivary gland extracts only inhibited the lectin pathway. The male and female intestinal contents inhibited the classical and lectin pathways. Neither the salivary glands nor the intestinal contents from males and females showed inhibitory activity towards the alternative pathway. However, the guts of male and female C. quinquefasciatus captured factor H from the human serum, permitting C3b inactivation to its inactive form iC3b, and preventing the formation of the C3 convertase. The activity of the antioxidant enzyme catalase is similar in C. quinquefasciatus females and males. This article shows for the first time that males from a haematophagous arthropod species present human anti-complement activity in their salivary gland extracts and gut contents. The finding of an activity that helps to protect the damage caused by blood ingestion in sugar-feeding male mosquitoes suggests that this may be a pre-adaptation to blood-feeding.
Collapse
Affiliation(s)
- Luisa de Melo Lara
- Laboratório de Fisiologia de Insetos Hematófagos, Universidade Federal de Minas Gerais, Instituto de Ciencias Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Adalberto Alves Pereira-Filho
- Laboratório de Fisiologia de Insetos Hematófagos, Universidade Federal de Minas Gerais, Instituto de Ciencias Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Rafael Henrique Mateus Pereira
- Laboratório de Fisiologia de Insetos Hematófagos, Universidade Federal de Minas Gerais, Instituto de Ciencias Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Luccas Gabriel Ferreira Malta
- Laboratório de Fisiologia de Insetos Hematófagos, Universidade Federal de Minas Gerais, Instituto de Ciencias Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Grasielle Caldas D'Ávila Pessoa
- Laboratório de Fisiologia de Insetos Hematófagos, Universidade Federal de Minas Gerais, Instituto de Ciencias Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Leonardo Barbosa Koerich
- Laboratório de Fisiologia de Insetos Hematófagos, Universidade Federal de Minas Gerais, Instituto de Ciencias Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Marcos Horácio Pereira
- Laboratório de Fisiologia de Insetos Hematófagos, Universidade Federal de Minas Gerais, Instituto de Ciencias Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo Nascimento Araujo
- Laboratório de Fisiologia de Insetos Hematófagos, Universidade Federal de Minas Gerais, Instituto de Ciencias Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Nelder de Figueiredo Gontijo
- Laboratório de Fisiologia de Insetos Hematófagos, Universidade Federal de Minas Gerais, Instituto de Ciencias Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Mauricio Roberto Viana Sant'Anna
- Laboratório de Fisiologia de Insetos Hematófagos, Universidade Federal de Minas Gerais, Instituto de Ciencias Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
54
|
Singh S, Singh A, Baweja V, Roy A, Chakraborty A, Singh IK. Molecular Rationale of Insect-Microbes Symbiosis-From Insect Behaviour to Mechanism. Microorganisms 2021; 9:microorganisms9122422. [PMID: 34946024 PMCID: PMC8707026 DOI: 10.3390/microorganisms9122422] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 12/27/2022] Open
Abstract
Insects nurture a panoply of microbial populations that are often obligatory and exist mutually with their hosts. Symbionts not only impact their host fitness but also shape the trajectory of their phenotype. This co-constructed niche successfully evolved long in the past to mark advanced ecological specialization. The resident microbes regulate insect nutrition by controlling their host plant specialization and immunity. It enhances the host fitness and performance by detoxifying toxins secreted by the predators and abstains them. The profound effect of a microbial population on insect physiology and behaviour is exploited to understand the host–microbial system in diverse taxa. Emergent research of insect-associated microbes has revealed their potential to modulate insect brain functions and, ultimately, control their behaviours, including social interactions. The revelation of the gut microbiota–brain axis has now unravelled insects as a cost-effective potential model to study neurodegenerative disorders and behavioural dysfunctions in humans. This article reviewed our knowledge about the insect–microbial system, an exquisite network of interactions operating between insects and microbes, its mechanistic insight that holds intricate multi-organismal systems in harmony, and its future perspectives. The demystification of molecular networks governing insect–microbial symbiosis will reveal the perplexing behaviours of insects that could be utilized in managing insect pests.
Collapse
Affiliation(s)
- Sujata Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India; (S.S.); (V.B.)
- Department of Botany, Hansraj College, University of Delhi, New Delhi 110007, India;
| | - Archana Singh
- Department of Botany, Hansraj College, University of Delhi, New Delhi 110007, India;
| | - Varsha Baweja
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India; (S.S.); (V.B.)
- DBC i4 Center, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India
| | - Amit Roy
- EVA 4.0 Unit, Faculty of Forestry and Wood Sciences, Czech University of Life Sciences, Kamýcká 129, Suchdol, 16521 Prague 6, Czech Republic;
- Excelentní Tým pro Mitigaci (ETM), Faculty of Forestry and Wood Sciences, Czech University of Life Sciences, Kamýcká 129, Suchdol, 16521 Prague 6, Czech Republic
| | - Amrita Chakraborty
- EVA 4.0 Unit, Faculty of Forestry and Wood Sciences, Czech University of Life Sciences, Kamýcká 129, Suchdol, 16521 Prague 6, Czech Republic;
- Correspondence: (A.C.); (I.K.S.)
| | - Indrakant Kumar Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India; (S.S.); (V.B.)
- DBC i4 Center, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India
- Correspondence: (A.C.); (I.K.S.)
| |
Collapse
|
55
|
Ramirez JL, Ramos I, Gomes FM. Editorial: Systemic Coordination of Invertebrate Homeostasis. Front Physiol 2021; 12:736185. [PMID: 34557114 PMCID: PMC8452844 DOI: 10.3389/fphys.2021.736185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/12/2021] [Indexed: 11/22/2022] Open
Affiliation(s)
- José Luis Ramirez
- Crop Bioprotection Research Unit, United States Department of Agriculture, National Center for Agricultural Utilization Research, Agricultural Research Service, Peoria, IL, United States
| | - Isabela Ramos
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio M Gomes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
56
|
Calle-Tobón A, Holguin-Rocha AF, Moore C, Rippee-Brooks M, Rozo-Lopez P, Harrod J, Fatehi S, Rua-Uribe GL, Park Y, Londoño-Rentería B. Blood Meals With Active and Heat-Inactivated Serum Modifies the Gene Expression and Microbiome of Aedes albopictus. Front Microbiol 2021; 12:724345. [PMID: 34566927 PMCID: PMC8458951 DOI: 10.3389/fmicb.2021.724345] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/12/2021] [Indexed: 11/13/2022] Open
Abstract
The Asian "tiger mosquito" Aedes albopictus is currently the most widely distributed disease-transmitting mosquito in the world. Its geographical expansion has also allowed the expansion of multiple arboviruses like dengue, Zika, and chikungunya, to higher latitudes. Due to the enormous risk to global public health caused by mosquitoes species vectors of human disease, and the challenges in slowing their expansion, it is necessary to develop new and environmentally friendly vector control strategies. Among these, host-associated microbiome-based strategies have emerged as promising options. In this study, we performed an RNA-seq analysis on dissected abdomens of Ae. albopictus females from Manhattan, KS, United States fed with sugar and human blood containing either normal or heat-inactivated serum, to evaluate the effect of heat inactivation on gene expression, the bacteriome transcripts and the RNA virome of this mosquito species. Our results showed at least 600 genes with modified expression profile when mosquitoes were fed with normal vs. heat-inactivated-containing blood. These genes were mainly involved in immunity, oxidative stress, lipid metabolism, and oogenesis. Also, we observed bacteriome changes with an increase in transcripts of Actinobacteria, Rhodospirillaceae, and Anaplasmataceae at 6 h post-feeding. We also found that feeding with normal blood seems to particularly influence Wolbachia metabolism, demonstrated by a significant increase in transcripts of this bacteria in mosquitoes fed with blood containing normal serum. However, no differences were observed in the virome core of this mosquito population. These results suggest that heat and further inactivation of complement proteins in human serum may have profound effect on mosquito and microbiome metabolism, which could influence interpretation of the pathogen-host interaction findings when using this type of reagents specially when measuring the effect of Wolbachia in vector competence.
Collapse
Affiliation(s)
- Arley Calle-Tobón
- Department of Entomology, Kansas State University, Manhattan, KS, United States
- Grupo Entomología Médica, Universidad de Antioquia, Medellín, Colombia
| | | | - Celois Moore
- Department of Entomology, Kansas State University, Manhattan, KS, United States
| | - Meagan Rippee-Brooks
- Department of Biology, Missouri State University, Springfield, MO, United States
| | - Paula Rozo-Lopez
- Department of Entomology, Kansas State University, Manhattan, KS, United States
| | - Jania Harrod
- Department of Entomology, Kansas State University, Manhattan, KS, United States
| | - Soheila Fatehi
- Department of Entomology, Kansas State University, Manhattan, KS, United States
| | | | - Yoonseong Park
- Department of Entomology, Kansas State University, Manhattan, KS, United States
| | | |
Collapse
|
57
|
Bombaça ACS, Gandara ACP, Ennes-Vidal V, Bottino-Rojas V, Dias FA, Farnesi LC, Sorgine MH, Bahia AC, Bruno RV, Menna-Barreto RFS. Aedes aegypti Infection With Trypanosomatid Strigomonas culicis Alters Midgut Redox Metabolism and Reduces Mosquito Reproductive Fitness. Front Cell Infect Microbiol 2021; 11:732925. [PMID: 34485182 PMCID: PMC8414984 DOI: 10.3389/fcimb.2021.732925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/26/2021] [Indexed: 11/29/2022] Open
Abstract
Aedes aegypti mosquitoes transmit arboviruses of important global health impact, and their intestinal microbiota can influence vector competence by stimulating the innate immune system. Midgut epithelial cells also produce toxic reactive oxygen species (ROS) by dual oxidases (DUOXs) that are essential players in insect immunity. Strigomonas culicis is a monoxenous trypanosomatid that naturally inhabits mosquitoes; it hosts an endosymbiotic bacterium that completes essential biosynthetic pathways of the parasite and influences its oxidative metabolism. Our group previously showed that S. culicis hydrogen peroxide (H2O2)-resistant (WTR) strain is more infectious to A. aegypti mosquitoes than the wild-type (WT) strain. Here, we investigated the influence of both strains on the midgut oxidative environment and the effect of infection on mosquito fitness and immunity. WT stimulated the production of superoxide by mitochondrial metabolism of midgut epithelial cells after 4 days post-infection, while WTR exacerbated H2O2 production mediated by increased DUOX activity and impairment of antioxidant system. The infection with both strains also disrupted the fecundity and fertility of the females, with a greater impact on reproductive fitness of WTR-infected mosquitoes. The presence of these parasites induced specific transcriptional modulation of immune-related genes, such as attacin and defensin A during WTR infection (11.8- and 6.4-fold, respectively) and defensin C in WT infection (7.1-fold). Thus, we propose that A. aegypti oxidative response starts in early infection time and does not affect the survival of the H2O2-resistant strain, which has a more efficient antioxidant system. Our data provide new biological aspects of A. aegypti–S. culicis relationship that can be used later in alternative vector control strategies.
Collapse
Affiliation(s)
- Ana Cristina S Bombaça
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Ana Caroline P Gandara
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vitor Ennes-Vidal
- Laboratório de Estudos Integrados em Protozoologia, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Vanessa Bottino-Rojas
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe A Dias
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luana C Farnesi
- Laboratório de Biologia Molecular de Insetos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Marcos H Sorgine
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Cristina Bahia
- Laboratório de Bioquímica de Insetos e Parasitos, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafaela V Bruno
- Laboratório de Biologia Molecular de Insetos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM/CNPq), Rio de Janeiro, Brazil
| | | |
Collapse
|
58
|
Wang J, Mason CJ, Ju X, Xue R, Tong L, Peiffer M, Song Y, Zeng R, Felton GW. Parasitoid Causes Cascading Effects on Plant-Induced Defenses Mediated Through the Gut Bacteria of Host Caterpillars. Front Microbiol 2021; 12:708990. [PMID: 34552570 PMCID: PMC8452159 DOI: 10.3389/fmicb.2021.708990] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/09/2021] [Indexed: 11/18/2022] Open
Abstract
Koinobiont endoparasitoid wasps whose larvae develop inside a host insect alter several important facets of host physiology, potentially causing cascading effects across multiple trophic levels. For instance, the hijacking of the host immune responses may have effects on how insects interact with host plants and microbial associates. However, the parasitoid regulation of insect-plant-microbiome interactions is still understudied. In this study, we used the fall armyworm (FAW), Spodoptera frugiperda, and the braconid parasitoid Cotesia marginiventris to evaluate impacts of parasitism on the gut microbiome of FAW larvae, and respective maize plant defense responses. The level of reactive oxygen species and the microbial community in larval gut underwent significant changes in response to parasitism, leading to a significant reduction of Enterococcus, while elevating the relative abundance of Pseudomonas. FAW with parasitism had lower glucose oxidase (GOX) activity in salivary glands and triggered lower defense responses in maize plants. These changes corresponded to effects on plants, as Pseudomonas inoculated larvae had lower activity of salivary GOX and triggered lower defense responses in maize plants. Our results demonstrated that parasitism had cascading effects on microbial associates across trophic levels and also highlighted that insect gut bacteria may contribute to complex interrelationships among parasitoids, herbivores, and plants.
Collapse
Affiliation(s)
- Jie Wang
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
- Department of Entomology, Pennsylvania State University, University Park, PA, United States
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Charles J. Mason
- Department of Entomology, Pennsylvania State University, University Park, PA, United States
| | - Xueyang Ju
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Rongrong Xue
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Lu Tong
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Michelle Peiffer
- Department of Entomology, Pennsylvania State University, University Park, PA, United States
| | - Yuanyuan Song
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Rensen Zeng
- Key Laboratory of Ministry of Education for Genetics, Breeding and Multiple Utilization of Crops, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, China
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Gary W. Felton
- Department of Entomology, Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
59
|
Brown LD, Maness R, Hall C, Gibson JD. Reactive oxygen species-mediated immunity against bacterial infection in the gut of cat fleas (Ctenocephalides felis). INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 136:103620. [PMID: 34216781 DOI: 10.1016/j.ibmb.2021.103620] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/21/2021] [Accepted: 06/29/2021] [Indexed: 06/13/2023]
Abstract
Fleas (Order Siphonaptera) transmit numerous bacterial pathogens that cause severe human diseases (e.g., cat scratch disease, flea-borne spotted fever, murine typhus, plague). Because initial entry of these infectious agents occurs while blood feeding, the immune response in the flea gut is considered to be the first line of defense against invading microbes. However, relatively few studies have identified the flea immune molecules that effectively resist or limit infection in the gut. In other hematophagous insects, an immediate immune response to imbibed pathogens is the generation of reactive oxygen species (ROS). In this study, we utilized cat fleas (Ctenocephalides felis) to investigate whether oral infection with a well-known insect bacterial pathogen (Serratia marcescens) induces ROS synthesis in the flea gut, and whether production of ROS provides a defense mechanism against microbial colonization. Specifically, we treated fleas with an antioxidant to limit the number of free radicals in the digestive tract prior to infection, and then measured the following: S. marcescens infection loads, hydrogen peroxide (ROS) levels, and mRNA abundance of ROS signaling pathway genes. Overall, our data shows that ROS levels increase in response to infection in the flea gut, and that this increase helps to strengthen the flea immune response through the microbicidal activity of ROS.
Collapse
Affiliation(s)
- Lisa D Brown
- Department of Biology, Georgia Southern University, 4324 Old Register Rd., Statesboro, GA, 30460, USA.
| | - Ryne Maness
- Department of Biology, Georgia Southern University, 4324 Old Register Rd., Statesboro, GA, 30460, USA
| | - Clark Hall
- Department of Biology, Georgia Southern University, 4324 Old Register Rd., Statesboro, GA, 30460, USA
| | - Joshua D Gibson
- Department of Biology, Georgia Southern University, 4324 Old Register Rd., Statesboro, GA, 30460, USA
| |
Collapse
|
60
|
Ranasinghe K, Gunathilaka N, Amarasinghe D, Rodrigo W, Udayanga L. Diversity of midgut bacteria in larvae and females of Aedes aegypti and Aedes albopictus from Gampaha District, Sri Lanka. Parasit Vectors 2021; 14:433. [PMID: 34454583 PMCID: PMC8400895 DOI: 10.1186/s13071-021-04900-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 07/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The midgut microbiota of mosquitoes maintain basal immune activity and immune priming. In recent years, scientists have focused on the use of microbial communities for vector control interventions. In the present study, the midgut bacteria of larvae and adults of Aedes aegypti and Ae. albopictus were assessed using both field-collected and laboratory-reared mosquitoes from Sri Lanka. METHODS Adults and larvae of Ae. aegypti and Ae. albopictus were collected from three selected areas in Gampaha Medical Officer of Health area, Gampaha District, Western Province, Sri Lanka. Bacterial colonies isolated from mosquito midgut dissections were identified by PCR amplification and sequencing of partial 16S rRNA gene fragments. RESULTS Adults and larvae of Ae. aegypti and Ae. albopictus harbored 25 bacterial species. Bacillus endophyticus and Pantoea dispersa were found more frequently in field-collected Ae. aegypti and Ae. albopictus adults, respectively. The midgut bacteria of Ae. aegypti and Ae. albopictus adults (X2 = 556.167, df = 72, P < 0.001) and larvae (X2 = 633.11, df = 66, P < 0.001) were significantly different. There was a significant difference among the bacterial communities between field-collected adults (X2 = 48.974, df = 10, P < 0.001) and larvae (X2 = 84.981, df = 10, P < 0.001). Lysinibacillus sphaericus was a common species in adults and larvae of laboratory-reared Ae. aegypti. Only P. dispersa occurred in the field-collected adults of Ae. aegypti and Ae. albopictus. Species belonging to genera Terribacillus, Lysinibacillus, Agromyces and Kocuria were recorded from Aedes mosquitoes, in accordance with previously reported results. CONCLUSIONS This study generated a comprehensive database on the culturable bacterial community found in the midgut of field-collected (Ae. aegypti and Ae. albopictus) and laboratory-reared (Ae. aegypti) mosquito larvae and adults from Sri Lanka. Data confirm that the midgut bacterial diversity in the studied mosquitoes varies according to species, developmental stage and strain (field vs laboratory).
Collapse
Affiliation(s)
- Koshila Ranasinghe
- Department of Zoology and Environmental Management, Faculty of Science, University of Kelaniya, Dalugama, Sri Lanka
| | - Nayana Gunathilaka
- Department of Parasitology, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka.
| | - Deepika Amarasinghe
- Department of Zoology and Environmental Management, Faculty of Science, University of Kelaniya, Dalugama, Sri Lanka
| | - Wasana Rodrigo
- Department of Zoology, Faculty of Natural Sciences, The Open University of Sri Lanka, Nawala, Nugegoda, Sri Lanka
| | - Lahiru Udayanga
- Department of Bio-Systems Engineering, Faculty of Agriculture and Plantation Management, Wayamba University of Sri Lanka, Makadura, Sri Lanka
| |
Collapse
|
61
|
MacLeod HJ, Dimopoulos G, Short SM. Larval Diet Abundance Influences Size and Composition of the Midgut Microbiota of Aedes aegypti Mosquitoes. Front Microbiol 2021; 12:645362. [PMID: 34220739 PMCID: PMC8249813 DOI: 10.3389/fmicb.2021.645362] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/19/2021] [Indexed: 11/13/2022] Open
Abstract
The midgut microbiota of the yellow fever mosquito Aedes aegypti impacts pathogen susceptibility and transmission by this important vector species. However, factors influencing the composition and size of the microbiome in mosquitoes are poorly understood. We investigated the impact of larval diet abundance during development on the composition and size of the larval and adult microbiota by rearing Aedes aegypti under four larval food regimens, ranging from nutrient deprivation to nutrient excess. We assessed the persistent impacts of larval diet availability on the microbiota of the larval breeding water, larval mosquitoes, and adult mosquitoes under sugar and blood fed conditions using qPCR and high-throughput 16S amplicon sequencing to determine bacterial load and microbiota composition. Bacterial loads in breeding water increased with increasing larval diet. Larvae reared with the lowest diet abundance had significantly fewer bacteria than larvae from two higher diet treatments, but not from the highest diet abundance. Adults from the lowest diet abundance treatment had significantly fewer bacteria in their midguts compared to all higher diet abundance treatments. Larval diet amount also had a significant impact on microbiota composition, primarily within larval breeding water and larvae. Increasing diet correlated with increased relative levels of Enterobacteriaceae and Flavobacteriaceae and decreased relative levels of Sphingomonadaceae. Multiple individual OTUs were significantly impacted by diet including one mapping to the genus Cedecea, which increased with higher diet amounts. This was consistent across all sample types, including sugar fed and blood fed adults. Taken together, these data suggest that availability of diet during development can cause lasting shifts in the size and composition of the microbiota in the disease vector Aedes aegypti.
Collapse
Affiliation(s)
- Hannah J MacLeod
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Sarah M Short
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
62
|
De Boeck I, van den Broek MFL, Allonsius CN, Spacova I, Wittouck S, Martens K, Wuyts S, Cauwenberghs E, Jokicevic K, Vandenheuvel D, Eilers T, Lemarcq M, De Rudder C, Thys S, Timmermans JP, Vroegop AV, Verplaetse A, Van de Wiele T, Kiekens F, Hellings PW, Vanderveken OM, Lebeer S. Lactobacilli Have a Niche in the Human Nose. Cell Rep 2021; 31:107674. [PMID: 32460009 DOI: 10.1016/j.celrep.2020.107674] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 02/13/2020] [Accepted: 04/30/2020] [Indexed: 12/14/2022] Open
Abstract
Although an increasing number of beneficial microbiome members are characterized for the human gut and vagina, beneficial microbes are underexplored for the human upper respiratory tract (URT). In this study, we demonstrate that taxa from the beneficial Lactobacillus genus complex are more prevalent in the healthy URT than in patients with chronic rhinosinusitis (CRS). Several URT-specific isolates are cultured, characterized, and further explored for their genetic and functional properties related to adaptation to the URT. Catalase genes are found in the identified lactobacilli, which is a unique feature within this mostly facultative anaerobic genus. Moreover, one of our isolated strains, Lactobacillus casei AMBR2, contains fimbriae that enable strong adherence to URT epithelium, inhibit the growth and virulence of several URT pathogens, and successfully colonize nasal epithelium of healthy volunteers. This study thus demonstrates that specific lactobacilli are adapted to the URT and could have a beneficial keystone function in this habitat.
Collapse
Affiliation(s)
- Ilke De Boeck
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Marianne F L van den Broek
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Camille N Allonsius
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Irina Spacova
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Stijn Wittouck
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Katleen Martens
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium; Department of Microbiology and Immunology, Clinical Immunology, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Sander Wuyts
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Eline Cauwenberghs
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Katarina Jokicevic
- Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Pharmaceutical Technology and Biopharmacy, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Dieter Vandenheuvel
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Tom Eilers
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Michelle Lemarcq
- Department of Microbial and Molecular Systems, KU Leuven, Gebroeders De Smetstraat 1, 9000 Ghent, Belgium
| | - Charlotte De Rudder
- Center for Microbial Ecology and Technology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Sofie Thys
- Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Jean-Pierre Timmermans
- Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Anneclaire V Vroegop
- ENT, Head and Neck Surgery and Communication Disorders, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Alex Verplaetse
- Department of Microbial and Molecular Systems, KU Leuven, Gebroeders De Smetstraat 1, 9000 Ghent, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - Filip Kiekens
- Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Pharmaceutical Technology and Biopharmacy, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Peter W Hellings
- Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Pharmaceutical Technology and Biopharmacy, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Clinical Department of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Olivier M Vanderveken
- ENT, Head and Neck Surgery and Communication Disorders, Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium; Faculty of Medicine and Health Sciences, Translational Neurosciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium.
| |
Collapse
|
63
|
Cansado-Utrilla C, Zhao SY, McCall PJ, Coon KL, Hughes GL. The microbiome and mosquito vectorial capacity: rich potential for discovery and translation. MICROBIOME 2021; 9:111. [PMID: 34006334 PMCID: PMC8132434 DOI: 10.1186/s40168-021-01073-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/07/2021] [Indexed: 05/09/2023]
Abstract
Microbiome research has gained considerable interest due to the emerging evidence of its impact on human and animal health. As in other animals, the gut-associated microbiota of mosquitoes affect host fitness and other phenotypes. It is now well established that microbes can alter pathogen transmission in mosquitoes, either positively or negatively, and avenues are being explored to exploit microbes for vector control. However, less attention has been paid to how microbiota affect phenotypes that impact vectorial capacity. Several mosquito and pathogen components, such as vector density, biting rate, survival, vector competence, and the pathogen extrinsic incubation period all influence pathogen transmission. Recent studies also indicate that mosquito gut-associated microbes can impact each of these components, and therefore ultimately modulate vectorial capacity. Promisingly, this expands the options available to exploit microbes for vector control by also targeting parameters that affect vectorial capacity. However, there are still many knowledge gaps regarding mosquito-microbe interactions that need to be addressed in order to exploit them efficiently. Here, we review current evidence of impacts of the microbiome on aspects of vectorial capacity, and we highlight likely opportunities for novel vector control strategies and areas where further studies are required. Video abstract.
Collapse
Affiliation(s)
- Cintia Cansado-Utrilla
- Departments of Vector Biology and Tropical Disease Biology, Centre for Neglected Tropical Disease, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Serena Y Zhao
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Philip J McCall
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Kerri L Coon
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA.
| | - Grant L Hughes
- Departments of Vector Biology and Tropical Disease Biology, Centre for Neglected Tropical Disease, Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
64
|
Gabrieli P, Caccia S, Varotto-Boccazzi I, Arnoldi I, Barbieri G, Comandatore F, Epis S. Mosquito Trilogy: Microbiota, Immunity and Pathogens, and Their Implications for the Control of Disease Transmission. Front Microbiol 2021; 12:630438. [PMID: 33889137 PMCID: PMC8056039 DOI: 10.3389/fmicb.2021.630438] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/02/2021] [Indexed: 11/16/2022] Open
Abstract
In mosquitoes, the interaction between the gut microbiota, the immune system, and the pathogens that these insects transmit to humans and animals is regarded as a key component toward the development of control strategies, aimed at reducing the burden of severe diseases, such as malaria and dengue fever. Indeed, different microorganisms from the mosquito microbiota have been investigated for their ability to affect important traits of the biology of the host insect, related with its survival, development and reproduction. Furthermore, some microorganisms have been shown to modulate the immune response of mosquito females, significantly shaping their vector competence. Here, we will review current knowledge in this field, focusing on i) the complex interaction between the intestinal microbiota and mosquito females defenses, both in the gut and at humoral level; ii) how knowledge on these issues contributes to the development of novel and targeted strategies for the control of mosquito-borne diseases such as the use of paratransgenesis or taking advantage of the relationship between Wolbachia and mosquito hosts. We conclude by providing a brief overview of available knowledge on microbiota-immune system interplay in major insect vectors.
Collapse
Affiliation(s)
- Paolo Gabrieli
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Silvia Caccia
- Department of Agricultural Sciences, University of Naples "Federico II", Naples, Italy.,Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| | - Ilaria Varotto-Boccazzi
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Irene Arnoldi
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Giulia Barbieri
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Francesco Comandatore
- "L. Sacco" Department of Biomedical and Clinical Sciences, Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Sara Epis
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| |
Collapse
|
65
|
Gandara ACP, Dias FA, de Lemos PC, Stiebler R, Bombaça ACS, Menna-Barreto R, Oliveira PL. "Urate and NOX5 Control Blood Digestion in the Hematophagous Insect Rhodnius prolixus". Front Physiol 2021; 12:633093. [PMID: 33716782 PMCID: PMC7947236 DOI: 10.3389/fphys.2021.633093] [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: 11/24/2020] [Accepted: 01/27/2021] [Indexed: 11/16/2022] Open
Abstract
Low levels of reactive oxygen species (ROS) are now recognized as essential players in cell signaling. Here, we studied the role of two conserved enzymes involved in redox regulation that play a critical role in the control of ROS in the digestive physiology of a blood-sucking insect, the kissing bug Rhodnius prolixus. RNAi-mediated silencing of RpNOX5 and RpXDH induced early mortality in adult females after a blood meal. Recently, a role for RpNOX5 in gut motility was reported, and here, we show that midgut peristalsis is also under the control of RpXDH. Together with impaired peristalsis, silencing either genes impaired egg production and hemoglobin digestion, and decreased hemolymph urate titers. Ultrastructurally, the silencing of RpNOX5 or RpXDH affected midgut cells, changing the cells of blood-fed insects to a phenotype resembling the cells of unfed insects, suggesting that these genes work together in the control of blood digestion. Injection of either allopurinol (an XDH inhibitor) or uricase recapitulated the gene silencing effects, suggesting that urate itself is involved in the control of blood digestion. The silencing of each of these genes influenced the expression of the other gene in a complex way both in the unfed state and after a blood meal, revealing signaling crosstalk between them that influences redox metabolism and nitrogen excretion and plays a central role in the control of digestive physiology.
Collapse
Affiliation(s)
- Ana Caroline P Gandara
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe A Dias
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula C de Lemos
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata Stiebler
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| |
Collapse
|
66
|
Talyuli OAC, Bottino-Rojas V, Polycarpo CR, Oliveira PL, Paiva-Silva GO. Non-immune Traits Triggered by Blood Intake Impact Vectorial Competence. Front Physiol 2021; 12:638033. [PMID: 33737885 PMCID: PMC7960658 DOI: 10.3389/fphys.2021.638033] [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: 12/04/2020] [Accepted: 02/08/2021] [Indexed: 11/13/2022] Open
Abstract
Blood-feeding arthropods are considered an enormous public health threat. They are vectors of a plethora of infectious agents that cause potentially fatal diseases like Malaria, Dengue fever, Leishmaniasis, and Lyme disease. These vectors shine due to their own physiological idiosyncrasies, but one biological aspect brings them all together: the requirement of blood intake for development and reproduction. It is through blood-feeding that they acquire pathogens and during blood digestion that they summon a collection of multisystemic events critical for vector competence. The literature is focused on how classical immune pathways (Toll, IMD, and JAK/Stat) are elicited throughout the course of vector infection. Still, they are not the sole determinants of host permissiveness. The dramatic changes that are the hallmark of the insect physiology after a blood meal intake are the landscape where a successful infection takes place. Dominant processes that occur in response to a blood meal are not canonical immunological traits yet are critical in establishing vector competence. These include hormonal circuitries and reproductive physiology, midgut permeability barriers, midgut homeostasis, energy metabolism, and proteolytic activity. On the other hand, the parasites themselves have a role in the outcome of these blood triggered physiological events, consistently using them in their favor. Here, to enlighten the knowledge on vector-pathogen interaction beyond the immune pathways, we will explore different aspects of the vector physiology, discussing how they give support to these long-dated host-parasite relationships.
Collapse
Affiliation(s)
- Octavio A C Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Bottino-Rojas
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carla R Polycarpo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| | - Gabriela O Paiva-Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| |
Collapse
|
67
|
Fogaça AC, Sousa G, Pavanelo DB, Esteves E, Martins LA, Urbanová V, Kopáček P, Daffre S. Tick Immune System: What Is Known, the Interconnections, the Gaps, and the Challenges. Front Immunol 2021; 12:628054. [PMID: 33737931 PMCID: PMC7962413 DOI: 10.3389/fimmu.2021.628054] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Ticks are ectoparasitic arthropods that necessarily feed on the blood of their vertebrate hosts. The success of blood acquisition depends on the pharmacological properties of tick saliva, which is injected into the host during tick feeding. Saliva is also used as a vehicle by several types of pathogens to be transmitted to the host, making ticks versatile vectors of several diseases for humans and other animals. When a tick feeds on an infected host, the pathogen reaches the gut of the tick and must migrate to its salivary glands via hemolymph to be successfully transmitted to a subsequent host during the next stage of feeding. In addition, some pathogens can colonize the ovaries of the tick and be transovarially transmitted to progeny. The tick immune system, as well as the immune system of other invertebrates, is more rudimentary than the immune system of vertebrates, presenting only innate immune responses. Although simpler, the large number of tick species evidences the efficiency of their immune system. The factors of their immune system act in each tick organ that interacts with pathogens; therefore, these factors are potential targets for the development of new strategies for the control of ticks and tick-borne diseases. The objective of this review is to present the prevailing knowledge on the tick immune system and to discuss the challenges of studying tick immunity, especially regarding the gaps and interconnections. To this end, we use a comparative approach of the tick immune system with the immune system of other invertebrates, focusing on various components of humoral and cellular immunity, such as signaling pathways, antimicrobial peptides, redox metabolism, complement-like molecules and regulated cell death. In addition, the role of tick microbiota in vector competence is also discussed.
Collapse
Affiliation(s)
- Andréa C. Fogaça
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Géssica Sousa
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniel B. Pavanelo
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Eliane Esteves
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Larissa A. Martins
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
- Laboratory of Bacteriology, Tick-Pathogen Transmission Unit, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Veronika Urbanová
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Petr Kopáček
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Sirlei Daffre
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
68
|
Romoli O, Schönbeck JC, Hapfelmeier S, Gendrin M. Production of germ-free mosquitoes via transient colonisation allows stage-specific investigation of host-microbiota interactions. Nat Commun 2021; 12:942. [PMID: 33574256 PMCID: PMC7878806 DOI: 10.1038/s41467-021-21195-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
The mosquito microbiota impacts the physiology of its host and is essential for normal larval development, thereby influencing transmission of vector-borne pathogens. Germ-free mosquitoes generated with current methods show larval stunting and developmental deficits. Therefore, functional studies of the mosquito microbiota have so far mostly been limited to antibiotic treatments of emerging adults. In this study, we introduce a method to produce germ-free Aedes aegypti mosquitoes. It is based on reversible colonisation with bacteria genetically modified to allow complete decolonisation at any developmental stage. We show that, unlike germ-free mosquitoes previously produced using sterile diets, reversibly colonised mosquitoes show no developmental retardation and reach the same size as control adults. This allows us to uncouple the study of the microbiota in larvae and adults. In adults, we detect no impact of bacterial colonisation on mosquito fecundity or longevity. In larvae, data from our transcriptome analysis and diet supplementation experiments following decolonisation suggest that bacteria support larval development by contributing to folate biosynthesis and by enhancing energy storage. Our study establishes a tool to study the microbiota in insects and deepens our knowledge on the metabolic contribution of bacteria to mosquito development.
Collapse
Affiliation(s)
- Ottavia Romoli
- grid.418525.f0000 0001 2206 8813Microbiota of Insect Vectors Group, Institut Pasteur de la Guyane, Cayenne, French Guiana France
| | - Johan Claes Schönbeck
- grid.418525.f0000 0001 2206 8813Microbiota of Insect Vectors Group, Institut Pasteur de la Guyane, Cayenne, French Guiana France
| | - Siegfried Hapfelmeier
- grid.5734.50000 0001 0726 5157Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Mathilde Gendrin
- grid.418525.f0000 0001 2206 8813Microbiota of Insect Vectors Group, Institut Pasteur de la Guyane, Cayenne, French Guiana France ,grid.428999.70000 0001 2353 6535Parasites and Insect Vectors Department, Institut Pasteur, Paris, France
| |
Collapse
|
69
|
Gumiel M, de Mattos DP, Vieira CS, Moraes CS, Moreira CJDC, Gonzalez MS, Teixeira-Ferreira A, Waghabi M, Azambuja P, Carels N. Proteome of the Triatomine Digestive Tract: From Catalytic to Immune Pathways; Focusing on Annexin Expression. Front Mol Biosci 2020; 7:589435. [PMID: 33363206 PMCID: PMC7755933 DOI: 10.3389/fmolb.2020.589435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022] Open
Abstract
Rhodnius prolixus, Panstrongylus megistus, Triatoma infestans, and Dipetalogaster maxima are all triatomines and potential vectors of the protozoan Trypanosoma cruzi responsible for human Chagas' disease. Considering that the T. cruzi's cycle occurs inside the triatomine digestive tract (TDT), the analysis of the TDT protein profile is an essential step to understand TDT physiology during T. cruzi infection. To characterize the protein profile of TDT of D. maxima, P. megistus, R. prolixus, and T. infestans, a shotgun liquid chromatography-tandem mass spectrometry (LC-MS/MS) approach was applied in this report. Most proteins were found to be closely related to metabolic pathways such as gluconeogenesis/glycolysis, citrate cycle, fatty acid metabolism, oxidative phosphorylation, but also to the immune system. We annotated this new proteome contribution gathering it with those previously published in accordance with Gene Ontology and KEGG. Enzymes were classified in terms of class, acceptor, and function, while the proteins from the immune system were annotated by reference to the pathways of humoral response, cell cycle regulation, Toll, IMD, JNK, Jak-STAT, and MAPK, as available from the Insect Innate Immunity Database (IIID). These pathways were further subclassified in recognition, signaling, response, coagulation, melanization and none. Finally, phylogenetic affinities and gene expression of annexins were investigated for understanding their role in the protection and homeostasis of intestinal epithelial cells against the inflammation.
Collapse
Affiliation(s)
- Marcia Gumiel
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
- Research Department, Universidad Privada Franz Tamayo (UNIFRANZ), La Paz, Bolivia
| | - Debora Passos de Mattos
- Laboratório de Biologia de Insetos, Departamento de Biologia Geral, Universidade Federal Fluminense, Niterói, Brazil
- Programa de Pós-Graduação em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Cecília Stahl Vieira
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
- Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Caroline Silva Moraes
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
| | | | - Marcelo Salabert Gonzalez
- Laboratório de Biologia de Insetos, Departamento de Biologia Geral, Universidade Federal Fluminense, Niterói, Brazil
- Programa de Pós-Graduação em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
- Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | | | - Mariana Waghabi
- Laboratório de Genômica Funcional e Bioinformática, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Patricia Azambuja
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
- Laboratório de Biologia de Insetos, Departamento de Biologia Geral, Universidade Federal Fluminense, Niterói, Brazil
- Programa de Pós-Graduação em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
- Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Nicolas Carels
- Laboratório de Modelagem de Sistemas Biológicos, National Institute for Science and Technology on Innovation in Neglected Diseases (INCT-IDN), Centro de Desenvolvimento Tecnológico em Saúde (CDTS), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| |
Collapse
|
70
|
Rothman JA, Russell KA, Leger L, McFrederick QS, Graystock P. The direct and indirect effects of environmental toxicants on the health of bumblebees and their microbiomes. Proc Biol Sci 2020; 287:20200980. [PMID: 33109012 PMCID: PMC7661295 DOI: 10.1098/rspb.2020.0980] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/05/2020] [Indexed: 01/14/2023] Open
Abstract
Bumblebees (Bombus spp.) are important and widespread insect pollinators, but the act of foraging on flowers can expose them to harmful pesticides and chemicals such as oxidizers and heavy metals. How these compounds directly influence bee survival and indirectly affect bee health via the gut microbiome is largely unknown. As toxicants in floral nectar and pollen take many forms, we explored the genomes of bee-associated microbes for their potential to detoxify cadmium, copper, selenate, the neonicotinoid pesticide imidacloprid, and hydrogen peroxide-which have all been identified in floral nectar and pollen. We then exposed Bombus impatiens workers to varying concentrations of these chemicals via their diet and assayed direct effects on bee survival. Using field-realistic doses, we further explored the indirect effects on bee microbiomes. We found multiple putative genes in core gut microbes that may aid in detoxifying harmful chemicals. We also found that while the chemicals are largely toxic at levels within and above field-realistic concentrations, the field-realistic concentrations-except for imidacloprid-altered the composition of the bee microbiome, potentially causing gut dysbiosis. Overall, our study shows that chemicals found in floral nectar and pollen can cause bee mortality, and likely have indirect, deleterious effects on bee health via their influence on the bee microbiome.
Collapse
Affiliation(s)
- Jason A. Rothman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
- Department of Entomology, University of California, Riverside, CA 92521, USA
| | - Kaleigh A. Russell
- Department of Entomology, University of California, Riverside, CA 92521, USA
| | - Laura Leger
- Department of Entomology, University of California, Riverside, CA 92521, USA
| | | | - Peter Graystock
- Department of Entomology, University of California, Riverside, CA 92521, USA
- Department of Life Sciences, Imperial College London, Silwood Park Campus, Ascot SL5 7PY, UK
| |
Collapse
|
71
|
Tirloni L, Braz G, Nunes RD, Gandara ACP, Vieira LR, Assumpcao TC, Sabadin GA, da Silva RM, Guizzo MG, Machado JA, Costa EP, Santos D, Gomes HF, Moraes J, dos Santos Mota MB, Mesquita RD, de Souza Leite M, Alvarenga PH, Lara FA, Seixas A, da Fonseca RN, Fogaça AC, Logullo C, Tanaka AS, Daffre S, Oliveira PL, da Silva Vaz I, Ribeiro JMC. A physiologic overview of the organ-specific transcriptome of the cattle tick Rhipicephalus microplus. Sci Rep 2020. [DOI: 10.1246/nikkashi.1979.101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AbstractTo further obtain insights into the Rhipicephalus microplus transcriptome, we used RNA-seq to carry out a study of expression in (i) embryos; (ii) ovaries from partially and fully engorged females; (iii) salivary glands from partially engorged females; (iv) fat body from partially and fully engorged females; and (v) digestive cells from partially, and (vi) fully engorged females. We obtained > 500 million Illumina reads which were assembled de novo, producing > 190,000 contigs, identifying 18,857 coding sequences (CDS). Reads from each library were mapped back into the assembled transcriptome giving a view of gene expression in different tissues. Transcriptomic expression and pathway analysis showed that several genes related in blood digestion and host-parasite interaction were overexpressed in digestive cells compared with other tissues. Furthermore, essential genes for the cell development and embryogenesis were overexpressed in ovaries. Taken altogether, these data offer novel insights into the physiology of production and role of saliva, blood digestion, energy metabolism, and development with submission of 10,932 novel tissue/cell specific CDS to the NCBI database for this important tick species.
Collapse
|
72
|
A physiologic overview of the organ-specific transcriptome of the cattle tick Rhipicephalus microplus. Sci Rep 2020; 10:18296. [PMID: 33106528 PMCID: PMC7588415 DOI: 10.1038/s41598-020-75341-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
To further obtain insights into the Rhipicephalus microplus transcriptome, we used RNA-seq to carry out a study of expression in (i) embryos; (ii) ovaries from partially and fully engorged females; (iii) salivary glands from partially engorged females; (iv) fat body from partially and fully engorged females; and (v) digestive cells from partially, and (vi) fully engorged females. We obtained > 500 million Illumina reads which were assembled de novo, producing > 190,000 contigs, identifying 18,857 coding sequences (CDS). Reads from each library were mapped back into the assembled transcriptome giving a view of gene expression in different tissues. Transcriptomic expression and pathway analysis showed that several genes related in blood digestion and host-parasite interaction were overexpressed in digestive cells compared with other tissues. Furthermore, essential genes for the cell development and embryogenesis were overexpressed in ovaries. Taken altogether, these data offer novel insights into the physiology of production and role of saliva, blood digestion, energy metabolism, and development with submission of 10,932 novel tissue/cell specific CDS to the NCBI database for this important tick species.
Collapse
|
73
|
Barletta ABF, Alves e Silva TL, Talyuli OAC, Luna-Gomes T, Sim S, Angleró-Rodríguez Y, Dimopoulos G, Bandeira-Melo C, Sorgine MHF. Prostaglandins regulate humoral immune responses in Aedes aegypti. PLoS Negl Trop Dis 2020; 14:e0008706. [PMID: 33095767 PMCID: PMC7584201 DOI: 10.1371/journal.pntd.0008706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 08/12/2020] [Indexed: 02/01/2023] Open
Abstract
Prostaglandins (PGs) are immuno-active lipids that mediate the immune response in invertebrates and vertebrates. In insects, PGs play a role on different physiological processes such as reproduction, ion transport and regulation of cellular immunity. However, it is unclear whether PGs play a role in invertebrate's humoral immunity, and, if so, which immune signaling pathways would be modulated by PGs. Here, we show that Aedes aegypti gut microbiota and Gram-negative bacteria challenge induces prostaglandin production sensitive to an irreversible inhibitor of the vertebrate cyclooxygenase, acetylsalicylic acid (ASA). ASA treatment reduced PG synthesis and is associated with decreased expression of components of the Toll and IMD immune pathways, thereby rendering mosquitoes more susceptible to both bacterial and viral infections. We also shown that a cytosolic phospholipase (PLAc), one of the upstream regulators of PG synthesis, is induced by the microbiota in the midgut after blood feeding. The knockdown of the PLAc decreased prostaglandin production and enhanced the replication of Dengue in the midgut. We conclude that in Ae. aegypti, PGs control the amplitude of the immune response to guarantee an efficient pathogen clearance.
Collapse
Affiliation(s)
- Ana Beatriz Ferreira Barletta
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo De Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Brasil
| | - Thiago Luiz Alves e Silva
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo De Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Brasil
| | - Octavio A. C. Talyuli
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo De Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Tatiana Luna-Gomes
- Departamento de Ciências da Natureza, Instituto de Aplicação Fernando Rodrigues da Silveira (CAp-UERJ), Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Shuzhen Sim
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Yesseinia Angleró-Rodríguez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 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
| | - Christianne Bandeira-Melo
- Laboratório de Inflamação, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marcos H. Ferreira Sorgine
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo De Meis, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Brasil
| |
Collapse
|
74
|
Giraldo-Calderón GI, Calle-Tobón A, Rozo-López P, Colpitts TM, Park Y, Rua-Uribe GL, Londono-Renteria B. Transcriptome of the Aedes aegypti Mosquito in Response to Human Complement Proteins. Int J Mol Sci 2020; 21:ijms21186584. [PMID: 32916828 PMCID: PMC7555780 DOI: 10.3390/ijms21186584] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/04/2020] [Accepted: 09/05/2020] [Indexed: 12/17/2022] Open
Abstract
Aedes aegypti is the primary mosquito vector of several human arboviruses, including the dengue virus (DENV). Vector control is the principal intervention to decrease the transmission of these viruses. The characterization of molecules involved in the mosquito physiological responses to blood-feeding may help identify novel targets useful in designing effective control strategies. In this study, we evaluated the in vivo effect of feeding adult female mosquitoes with human red blood cells reconstituted with either heat-inactivated (IB) or normal plasma (NB). The RNA-seq based transcript expression of IB and NB mosquitoes was compared against sugar-fed (SF) mosquitoes. In in vitro experiments, we treated Aag2 cells with a recombinant version of complement proteins (hC3 or hC5a) and compared transcript expression to untreated control cells after 24 h. The transcript expression analysis revealed that human complement proteins modulate approximately 2300 transcripts involved in multiple biological functions, including immunity. We also found 161 upregulated and 168 downregulated transcripts differentially expressed when human complement protein C3 (hC3) and human complement protein C5a (hC5a) treated cells were compared to the control untreated cells. We conclude that active human complement induces significant changes to the transcriptome of Ae. aegypti mosquitoes, which may influence the physiology of these arthropods.
Collapse
Affiliation(s)
- Gloria I. Giraldo-Calderón
- VectorBase, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA;
- Departamento de Ciencias Biológicasy, Universidad Icesi, Calle 18 No. 122–135, 760020 Cali, Colombia
- Departamento de Ciencias Básicas Médicas, Universidad Icesi, Calle 18 No. 122–135, 760020 Cali, Colombia
| | - Arley Calle-Tobón
- Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (A.C.-T.); (P.R.-L.); (Y.P.)
- Grupo Entomología Médica, Universidad de Antioquia, 050001 Medellín, Colombia;
| | - Paula Rozo-López
- Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (A.C.-T.); (P.R.-L.); (Y.P.)
| | - Tonya M. Colpitts
- Department of Microbiology & National Emerging Infectious Diseases Laboratories, Boston University School of Medicine; Boston, MA 02118, USA;
| | - Yoonseong Park
- Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (A.C.-T.); (P.R.-L.); (Y.P.)
| | | | - Berlin Londono-Renteria
- Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (A.C.-T.); (P.R.-L.); (Y.P.)
- Correspondence:
| |
Collapse
|
75
|
Kakani P, Gupta L, Kumar S. Heme-Peroxidase 2, a Peroxinectin-Like Gene, Regulates Bacterial Homeostasis in Anopheles stephensi Midgut. Front Physiol 2020; 11:572340. [PMID: 33013485 PMCID: PMC7506126 DOI: 10.3389/fphys.2020.572340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/17/2020] [Indexed: 12/31/2022] Open
Abstract
The dynamic nature of mosquito gut microbiome is associated with different stages of development and feeding behaviors. Therefore, mosquito gut harbors a wide range of endogenous microbes that promote numerous life processes such as, nutrition, reproduction and immunity. In addition, gut microbiota also play an important role in the regulation of Plasmodium (malaria parasite) development. Thus, understanding the mechanism of microbial homeostasis in mosquito gut might be one of the strategies to manipulate malaria parasite development. In the present study, we characterized a 692 amino acids long secreted midgut heme-peroxidase 2 (AsHPX2) in Anopheles stephensi, the major Indian malaria vector. The presence of putative integrin binding motifs, LDV (Leu-Asp-Val), indicated its peroxinectin-like nature. Our phylogenetic analysis revealed that AsHPX2 is a Culicinae lineage-specific gene. RNA interference (RNAi)-mediated silencing of AsHPX2 gene significantly enhanced the growth of midgut bacteria in sugar-fed mosquitoes against sham-treated controls. Interestingly, blood-feeding drastically reduced AsHPX2 gene expression and enhanced the growth of midgut bacteria. These results revealed a negative correlation between the expression of AsHPX2 gene and gut bacterial growth. We proposed that AsHPX2, being a mosquito-specific gene, might serve as a "potent target" to manipulate midgut microbiota and vector competence.
Collapse
Affiliation(s)
- Parik Kakani
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani, India.,Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, India
| | - Lalita Gupta
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani, India.,Department of Zoology, Chaudhary Bansi Lal University, Bhiwani, India
| | - Sanjeev Kumar
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani, India.,Department of Biotechnology, Chaudhary Bansi Lal University, Bhiwani, India
| |
Collapse
|
76
|
Jia N, Wang J, Shi W, Du L, Sun Y, Zhan W, Jiang JF, Wang Q, Zhang B, Ji P, Bell-Sakyi L, Cui XM, Yuan TT, Jiang BG, Yang WF, Lam TTY, Chang QC, Ding SJ, Wang XJ, Zhu JG, Ruan XD, Zhao L, Wei JT, Ye RZ, Que TC, Du CH, Zhou YH, Cheng JX, Dai PF, Guo WB, Han XH, Huang EJ, Li LF, Wei W, Gao YC, Liu JZ, Shao HZ, Wang X, Wang CC, Yang TC, Huo QB, Li W, Chen HY, Chen SE, Zhou LG, Ni XB, Tian JH, Sheng Y, Liu T, Pan YS, Xia LY, Li J, Zhao F, Cao WC. Large-Scale Comparative Analyses of Tick Genomes Elucidate Their Genetic Diversity and Vector Capacities. Cell 2020; 182:1328-1340.e13. [PMID: 32814014 DOI: 10.1016/j.cell.2020.07.023] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/01/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022]
Abstract
Among arthropod vectors, ticks transmit the most diverse human and animal pathogens, leading to an increasing number of new challenges worldwide. Here we sequenced and assembled high-quality genomes of six ixodid tick species and further resequenced 678 tick specimens to understand three key aspects of ticks: genetic diversity, population structure, and pathogen distribution. We explored the genetic basis common to ticks, including heme and hemoglobin digestion, iron metabolism, and reactive oxygen species, and unveiled for the first time that genetic structure and pathogen composition in different tick species are mainly shaped by ecological and geographic factors. We further identified species-specific determinants associated with different host ranges, life cycles, and distributions. The findings of this study are an invaluable resource for research and control of ticks and tick-borne diseases.
Collapse
Affiliation(s)
- Na Jia
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, P.R. China
| | - Jinfeng Wang
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, P.R. China; State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Wenqiang Shi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Lifeng Du
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China
| | - Yi Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Wei Zhan
- Annoroad Gene Technology (Beijing) Company Limited, Beijing 100176, P.R. China
| | - Jia-Fu Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, P.R. China
| | - Qian Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China
| | - Bing Zhang
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Peifeng Ji
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Lesley Bell-Sakyi
- Department of Infection Biology and Microbiomes, Institute of Infection, Ecological and Veterinary Sciences, University of Liverpool, Liverpool L3 5RF, UK
| | - Xiao-Ming Cui
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, P.R. China
| | - Ting-Ting Yuan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Bao-Gui Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Wei-Fei Yang
- Annoroad Gene Technology (Beijing) Company Limited, Beijing 100176, P.R. China
| | - Tommy Tsan-Yuk Lam
- State Key Laboratory of Emerging Infectious Diseases and Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Qiao-Cheng Chang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, P.R. China
| | - Shu-Jun Ding
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Communicable Disease Control and Prevention, Jinan 250014, Shandong, P.R. China
| | - Xian-Jun Wang
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Communicable Disease Control and Prevention, Jinan 250014, Shandong, P.R. China
| | - Jin-Guo Zhu
- ManZhouLi Customs District, Manzhouli 021400, Inner Mongolia, P.R. China
| | - Xiang-Dong Ruan
- Academy of Forest Inventory and Planning, State Forestry and Grassland Administration, Beijing 100714, P.R. China
| | - Lin Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China
| | - Jia-Te Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China
| | - Run-Ze Ye
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China
| | - Teng Cheng Que
- Guangxi Zhuang Autonomous Region Terrestrial Wildlife Medical-aid and Monitoring Epidemic Diseases Research Center, Nanjing 530028, Guangxi, P.R. China
| | - Chun-Hong Du
- Yunnan Institute for Endemic Diseases Control and Prevention, Dali 671000, Yunnan, P.R. China
| | - Yu-Hao Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Jing Xia Cheng
- Shanxi Provence Center for Disease Control and Prevention, Xian 030012, Shanxi, P.R. China
| | - Pei-Fang Dai
- Shanxi Provence Center for Disease Control and Prevention, Xian 030012, Shanxi, P.R. China
| | - Wen-Bin Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Xiao-Hu Han
- Shenyang Agriculture University, Shenyang 110866, Liaoning, P.R. China
| | - En-Jiong Huang
- Fuzhou International Travel Healthcare Center, Fuzhou 350001, Fujian, P.R. China
| | - Lian-Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Wei Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Yu-Chi Gao
- Annoroad Gene Technology (Beijing) Company Limited, Beijing 100176, P.R. China
| | - Jing-Ze Liu
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, P.R. China
| | - Hong-Ze Shao
- Animal Husbandry and Veterinary Science Research Institute of Jilin Province, Changchun 130062, Jilin, P.R. China
| | - Xin Wang
- Qingjiangpu District Center for Disease Control and Prevention, Huai'an 223001, Jiangsu, P.R. China
| | - Chong-Cai Wang
- Hainan International Travel Healthcare Center, Haikou 570311, Hainan, P.R. China
| | - Tian-Ci Yang
- State Key Lab of Mosquito-borne Diseases, Hangzhou International Tourism Healthcare Center, Hangzhou Customs of China, Hangzhou 310012, Zhejiang, P.R. China
| | - Qiu-Bo Huo
- Mudanjiang Forestry Central Hospital, Mudanjiang 157000, Heilongjiang, P.R. China
| | - Wei Li
- Xinjiang Center for Disease Control and Prevention, Urumqi 830002, Xinjiang, P.R. China
| | - Hai-Ying Chen
- Collaboration Unit for Field Epidemiology of the State Key Laboratory for Infectious Disease Prevention and Control, Nanchang Center for Disease Control and Prevention. Nanchang 330038, Jiangxi, P.R. China
| | - Shen-En Chen
- Collaboration Unit for Field Epidemiology of the State Key Laboratory for Infectious Disease Prevention and Control, Nanchang Center for Disease Control and Prevention. Nanchang 330038, Jiangxi, P.R. China
| | - Ling-Guo Zhou
- Shaanxi Natural Reserve and Wildlife Administration Station, Xi'an 710082, Shaanxi, P.R. China
| | - Xue-Bing Ni
- State Key Laboratory of Emerging Infectious Diseases and Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Jun-Hua Tian
- Wuhan Center for Disease Control and Prevention, Wuhan 430015, Hubei, P.R. China
| | - Yue Sheng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Tao Liu
- Annoroad Gene Technology (Beijing) Company Limited, Beijing 100176, P.R. China
| | - Yu-Sheng Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Luo-Yuan Xia
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Jie Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Fangqing Zhao
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, P.R. China; State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, P.R. China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, Yunan, P.R. China; Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, Zhejiang, P.R. China; University of the Chinese Academy of Sciences, Beijing 100049, P.R. China.
| | - Wu-Chun Cao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, P.R. China.
| |
Collapse
|
77
|
Ma L, Liu L, Zhao Y, Yang L, Chen C, Li Z, Lu Z. JNK pathway plays a key role in the immune system of the pea aphid and is regulated by microRNA-184. PLoS Pathog 2020; 16:e1008627. [PMID: 32584915 PMCID: PMC7343183 DOI: 10.1371/journal.ppat.1008627] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/08/2020] [Accepted: 05/13/2020] [Indexed: 12/21/2022] Open
Abstract
Different from holometabolous insects, the hemipteran species such as pea aphid Acyrthosiphon pisum exhibit reduced immune responses with the absence of the genes coding for antimicrobial peptide (AMP), immune deficiency (IMD), peptidoglycan recognition proteins (PGRPs), and other immune-related molecules. Prior studies have proved that phenoloxidase (PO)-mediated melanization, hemocyte-mediated phagocytosis, and reactive oxygen species (ROS) participate in pea aphid defense against bacterial infection. Also, the conserved signaling, Jun N-terminal kinase (JNK) pathway, has been suggested to be involved in pea aphid immune defense. However, the precise role of the JNK signaling, its interplay with other immune responses and its regulation in pea aphid are largely unknown. In this study, using in vitro biochemical assays and in vivo bioassays, we demonstrated that the JNK pathway regulated hemolymph PO activity, hydrogen peroxide concentration and hemocyte phagocytosis in bacteria infected pea aphids, suggesting that the JNK pathway plays a central role in regulating immune responses in pea aphid. We further revealed the JNK pathway is regulated by microRNA-184 in response to bacterial infection. It is possible that in common the JNK pathway plays a key role in immune system of hemipteran insects and microRNA-184 regulates the JNK pathway in animals.
Collapse
Affiliation(s)
- Li Ma
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Lu Liu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Yujie Zhao
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Lei Yang
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Caihua Chen
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhaofei Li
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail:
| |
Collapse
|
78
|
Ferreira PG, Tesla B, Horácio ECA, Nahum LA, Brindley MA, de Oliveira Mendes TA, Murdock CC. Temperature Dramatically Shapes Mosquito Gene Expression With Consequences for Mosquito-Zika Virus Interactions. Front Microbiol 2020; 11:901. [PMID: 32595607 PMCID: PMC7303344 DOI: 10.3389/fmicb.2020.00901] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
Vector-borne flaviviruses are emerging threats to human health. For successful transmission, the virus needs to efficiently enter mosquito cells and replicate within and escape several tissue barriers while mosquitoes elicit major transcriptional responses to flavivirus infection. This process will be affected not only by the specific mosquito-pathogen pairing but also by variation in key environmental variables such as temperature. Thus far, few studies have examined the molecular responses triggered by temperature and how these responses modify infection outcomes, despite substantial evidence showing strong relationships between temperature and transmission in a diversity of systems. To define the host transcriptional changes associated with temperature variation during the early infection process, we compared the transcriptome of mosquito midgut samples from mosquitoes exposed to Zika virus (ZIKV) and non-exposed mosquitoes housed at three different temperatures (20, 28, and 36°C). While the high-temperature samples did not show significant changes from those with standard rearing conditions (28°C) 48 h post-exposure, the transcriptome profile of mosquitoes housed at 20°C was dramatically different. The expression of genes most altered by the cooler temperature involved aspects of blood-meal digestion, ROS metabolism, and mosquito innate immunity. Further, we did not find significant differences in the viral RNA copy number between 24 and 48 h post-exposure at 20°C, suggesting that ZIKV replication is limited by cold-induced changes to the mosquito midgut environment. In ZIKV-exposed mosquitoes, vitellogenin, a lipid carrier protein, was most up-regulated at 20°C. Our results provide a deeper understanding of the temperature-triggered transcriptional changes in Aedes aegypti and can be used to further define the molecular mechanisms driven by environmental temperature variation.
Collapse
Affiliation(s)
| | - Blanka Tesla
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Elvira Cynthia Alves Horácio
- René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Brazil.,Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Laila Alves Nahum
- René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Brazil.,Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Promove College of Technology, Belo Horizonte, Brazil
| | - Melinda Ann Brindley
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| | | | - Courtney Cuinn Murdock
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States.,Odum School of Ecology, University of Georgia, Athens, GA, United States.,Center for the Ecology of Infectious Diseases, University of Georgia, Athens, GA, United States.,Center for Emerging and Global Tropical Diseases, University of Georgia, Athens, GA, United States.,River Basin Center, University of Georgia, Athens, GA, United States.,Department of Entomology, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
79
|
Duan DY, Zhou HM, Cheng TY. Comparative analysis of microbial community in the whole body and midgut from fully engorged and unfed female adult Melophagus ovinus. MEDICAL AND VETERINARY ENTOMOLOGY 2020; 34:215-224. [PMID: 31840281 DOI: 10.1111/mve.12424] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 06/10/2023]
Abstract
Melophagus ovinus is a type of ectoparasite infesting sheep. Data regarding the comprehensive bacterial community associated with the whole body and midgut of M. ovinus under different engorged statuses are required. Melophagus ovinus were collected from the city of Jiuquan, China. Bacterial DNA was extracted from the whole body and midgut of fully engorged female adults, or newly hatched and unfed adult female M. ovinus. The 16S rRNA gene V3-V4 hypervariable regions were sequenced using the IonS5™XL platform (Thermo Fisher Scientific, Waltham, MA, U.S.A.). The whole body bacterial diversity of the newly hatched, unfed adult females was greater compared with that of the other three samples. Proteobacteria was the dominant bacterial phylum in all of the samples. Of the 42 total bacterial genera present in all of the experimental samples, Arsenophonus, Bartonella and Wolbachia were the dominant genera. The relative abundance of Arsenophonus in midgut was greater than that in the whole body. The relative abundance of Bartonella in fully engorged adults was far greater than those in newly hatched, unfed adults. The relative abundance of Wolbachia was highest in the whole body of newly hatched, unfed adults. Seventeen bacterial species were identified in all experimental samples. Bartonella chomelii, Streptococcus hyointestinalis and Escherichia coli were the first species reported in M. ovinus.
Collapse
Affiliation(s)
- D-Y Duan
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, China
| | - H-M Zhou
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, China
| | - T-Y Cheng
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, China
| |
Collapse
|
80
|
Martin E, Tang W, Briggs C, Hopson H, Juarez JG, Garcia-Luna SM, de Valdez MW, Badillo-Vargas IE, Borucki MK, Frank M, Hamer GL. Cell fusing agent virus (Flavivirus) infection in Aedes aegypti in Texas: seasonality, comparison by trap type, and individual viral loads. Arch Virol 2020; 165:1769-1776. [PMID: 32440701 PMCID: PMC7351801 DOI: 10.1007/s00705-020-04652-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 04/10/2020] [Indexed: 11/05/2022]
Abstract
South Texas has experienced local transmission of Zika virus and of other mosquito-borne viruses such as chikungunya virus and dengue virus in the last decades. Using a mosquito surveillance program in the Lower Rio Grande Valley (LRGV) and San Antonio, TX, from 2016 to 2018, we detected the presence of an insect-specific virus, cell fusing agent virus (CFAV), in the Aedes aegypti mosquito population. We tested 6,326 females and 1,249 males from the LRGV and 659 females from San Antonio for CFAV by RT-PCR using specific primers. Infection rates varied from 0 to 261 per 1,000 mosquitoes in the LRGV and 115 to 208 per 1,000 in San Antonio depending on the month of collection. Infection rates per 1,000 individuals appeared higher in females collected from BG Sentinel 2 traps compared to Autocidal Gravid Ovitraps, but the ratio of the percentage of infected pools did not differ by trap type. The natural viral load in individual males ranged from 1.25 x 102 to 5.50 x 106 RNA copies and in unfed females from 5.42 x 103 to 8.70 x 106 RNA copies. Gravid females were found to harbor fewer viral particles than males and unfed females.
Collapse
Affiliation(s)
- Estelle Martin
- Department of Entomology, Texas A&M University, College Station, TX, USA. .,Department of Entomology and Nematology, University of Florida, Gainesville, FL, USA.
| | - Wendy Tang
- Department of Entomology, Texas A&M University, College Station, TX, USA
| | - Cierra Briggs
- Department of Entomology, Texas A&M University, College Station, TX, USA
| | - Helena Hopson
- Department of Entomology, Texas A&M University, College Station, TX, USA
| | - Jose G Juarez
- Department of Entomology, Texas A&M University, College Station, TX, USA
| | | | - Megan Wise de Valdez
- Department of Science and Mathematics, Texas A&M University-San Antonio, San Antonio, TX, USA
| | | | | | - Matthias Frank
- Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Gabriel L Hamer
- Department of Entomology, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
81
|
Guizzo MG, Neupane S, Kucera M, Perner J, Frantová H, da Silva Vaz I, de Oliveira PL, Kopacek P, Zurek L. Poor Unstable Midgut Microbiome of Hard Ticks Contrasts With Abundant and Stable Monospecific Microbiome in Ovaries. Front Cell Infect Microbiol 2020; 10:211. [PMID: 32457850 PMCID: PMC7225584 DOI: 10.3389/fcimb.2020.00211] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/17/2020] [Indexed: 01/17/2023] Open
Abstract
Culture-independent metagenomic methodologies have enabled detection and identification of microorganisms in various biological systems and often revealed complex and unknown microbiomes. In many organisms, the microbiome outnumbers the host cells and greatly affects the host biology and fitness. Ticks are hematophagous ectoparasites with a wide host range. They vector a number of human and animal pathogens and also directly cause major economic losses in livestock. Although several reports on a tick midgut microbiota show a diverse bacterial community, in most cases the size of the bacterial population has not been determined. In this study, the microbiome was quantified in the midgut and ovaries of the ticks Ixodes ricinus and Rhipicephalus microplus before, during, and after blood feeding. Although the size of bacterial community in the midgut fluctuated with blood feeding, it was overall extremely low in comparison to that of other hematophagous arthropods. In addition, the tick ovarian microbiome of both tick species exceeded the midgut 16S rDNA copy numbers by several orders of magnitude. This indicates that the ratio of a tick midgut/ovary microbiome represents an exception to the general biology of other metazoans. In addition to the very low abundance, the tick midgut diversity in I. ricinus was variable and that is in contrast to that found in the tick ovary. The ovary of I. ricinus had a very low bacterial diversity and a very high and stable bacterial abundance with the dominant endosymbiont, Midichloria sp. The elucidation of this aspect of tick biology highlights a unique tissue-specific microbial-invertebrate host interaction.
Collapse
Affiliation(s)
- Melina Garcia Guizzo
- Central European Institute of Technology (CEITEC), Center for Zoonoses, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia.,Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Saraswoti Neupane
- Department of Entomology, Kansas State University, Manhattan, KS, United States
| | - Matej Kucera
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Ceske Budejovice, Czechia.,Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia
| | - Jan Perner
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Helena Frantová
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Itabajara da Silva Vaz
- Laboratório de Imunologia Aplicada a Sanidade Animal, Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Pedro L de Oliveira
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Petr Kopacek
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Ludek Zurek
- Central European Institute of Technology (CEITEC), Center for Zoonoses, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia.,Department of Chemistry and Biochemistry, Mendel University, Brno, Czechia
| |
Collapse
|
82
|
Chen S, Johnson BK, Yu T, Nelson BN, Walker ED. Elizabethkingia anophelis: Physiologic and Transcriptomic Responses to Iron Stress. Front Microbiol 2020; 11:804. [PMID: 32457715 PMCID: PMC7221216 DOI: 10.3389/fmicb.2020.00804] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 04/03/2020] [Indexed: 12/12/2022] Open
Abstract
In this study, we investigated the global gene expression responses of Elizabethkingia anophelis to iron fluxes in the midgut of female Anopheles stephensi mosquitoes fed sucrose or blood, and in iron-poor or iron-rich culture conditions. Of 3,686 transcripts revealed by RNAseq technology, 218 were upregulated while 112 were down-regulated under iron-poor conditions. Hemolysin gene expression was significantly repressed when cells were grown under iron-rich or high temperature (37°C) conditions. Furthermore, hemolysin gene expression was down-regulated after a blood meal, indicating that E. anophelis cells responded to excess iron and its associated physiological stress by limiting iron loading. By contrast, genes encoding respiratory chain proteins were up-regulated under iron-rich conditions, allowing these iron-containing proteins to chelate intracellular free iron. In vivo studies showed that growth of E. anophelis cells increased 3-fold in blood-fed mosquitoes over those in sucrose-fed ones. Deletion of siderophore synthesis genes led to impaired cell growth in both iron-rich and iron-poor media. Mutants showed more susceptibility to H2O2 toxicity and less biofilm formation than did wild-type cells. Mosquitoes with E. anophelis experimentally colonized in their guts produced more eggs than did those treated with erythromycin or left unmanipulated, as controls. Results reveal that E. anophelis bacteria respond to varying iron concentration in the mosquito gut, harvest iron while fending off iron-associated stress, contribute to lysis of red blood cells, and positively influence mosquito host fecundity.
Collapse
Affiliation(s)
- Shicheng Chen
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Benjamin K. Johnson
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Ting Yu
- Agro-Biological Gene Research Center, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Brooke N. Nelson
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Edward D. Walker
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
- Department of Entomology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
83
|
Yin C, Sun P, Yu X, Wang P, Cheng G. Roles of Symbiotic Microorganisms in Arboviral Infection of Arthropod Vectors. Trends Parasitol 2020; 36:607-615. [PMID: 32386795 DOI: 10.1016/j.pt.2020.04.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/10/2020] [Accepted: 04/10/2020] [Indexed: 12/30/2022]
Abstract
Arthropod vectors serve as native reservoirs and transmitters of hundreds of arboviruses. In arthropod vectors, symbiotic microorganisms residing in the gut lumen and/or hemocoelic tissues maintain complicated relationships with their host and influence multiple aspects of vector physiology. Recently, accumulating evidence has established an important role for symbiotic microorganisms in vector-virus interactions which could potentially be used to control viral transmission. Herein, we review recent progress on symbiotic microbe-arbovirus interactions and summarize the molecular mechanisms by which commensal microbes act on hosts and arboviruses. Understanding the sophisticated interactions among arthropod vectors, microbiota, and arboviruses may offer new strategies for the prevention of arboviral diseases in the future.
Collapse
Affiliation(s)
- Chunhong Yin
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China, 100084; Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China, 518055
| | - Peng Sun
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China, 100084; Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China, 518055
| | - Xi Yu
- School of Life Sciences, Tsinghua University, Beijing, China, 100084
| | - Penghua Wang
- Department of Immunology, School of Medicine, the University of Connecticut Health Center, Farmington, CT, USA, 06030
| | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China, 100084; Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong, China, 518055.
| |
Collapse
|
84
|
Huang Y, Yu Y, Zhan S, Tomberlin JK, Huang D, Cai M, Zheng L, Yu Z, Zhang J. Dual oxidase Duox and Toll-like receptor 3 TLR3 in the Toll pathway suppress zoonotic pathogens through regulating the intestinal bacterial community homeostasis in Hermetia illucens L. PLoS One 2020; 15:e0225873. [PMID: 32352968 PMCID: PMC7192390 DOI: 10.1371/journal.pone.0225873] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/26/2020] [Indexed: 01/26/2023] Open
Abstract
Black soldier fly (BSF; Hermetia illucens L.) larvae can convert fresh pig manure into protein and fat-rich biomass, which can then be used as aquafeed for select species. Currently, BSF is the only approved insect for such purposes in Canada, USA, and the European Union. Pig manure could serve as a feed substrate for BSF; however, it is contaminated with zoonotic pathogens (e.g., Staphylococcus aureus and Salmonella spp.). Fortunately, BSF larvae inhibit many of these zoonotic pathogens; however, the mechanisms employed are unclear. We employed RNAi, qRT-PCR, and Illumina MiSeq 16S rDNA high-throughput sequencing to examine the interaction between two immune genes (Duox in Duox-reactive oxygen species [ROS] immune system and TLR3 in the Toll signaling pathway) and select pathogens common in pig manure to decipher the mechanisms resulting in pathogen suppression. Results indicate Bsf Duox-TLR3 RNAi increased bacterial load but decreased relative abundance of Providencia and Dysgonomonas, which are thought to be commensals in the BSF larval gut. Bsf Duox-TLR3 RNAi also inactivated the NF-κB signaling pathway, downregulated the expression of antimicrobial peptides, and diminished inhibitory effects on zoonotic pathogen. The resulting dysbiosis stimulated an immune response by activating BsfDuox and promoting ROS, which regulated the composition and structure of the gut bacterial community. Thus, BsfDuox and BsfTLR3 are important factors in regulating these key gut microbes, while inhibiting target zoonotic pathogens.
Collapse
Affiliation(s)
- Yaqing Huang
- State Key Laboratory of Agricultural Microbiology, National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yongqiang Yu
- State Key Laboratory of Agricultural Microbiology, National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Shuai Zhan
- Institute of Plant Physiology & Ecology, SIBS, CAS, Shanghai, China
| | | | - Dian Huang
- State Key Laboratory of Agricultural Microbiology, National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Minmin Cai
- State Key Laboratory of Agricultural Microbiology, National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Longyu Zheng
- State Key Laboratory of Agricultural Microbiology, National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ziniu Yu
- State Key Laboratory of Agricultural Microbiology, National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jibin Zhang
- State Key Laboratory of Agricultural Microbiology, National Engineering Research Center of Microbial Pesticides, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- * E-mail:
| |
Collapse
|
85
|
Freitas MN, Marten AD, Moore GA, Tree MO, McBrayer SP, Conway MJ. Extracellular vesicles restrict dengue virus fusion in Aedes aegypti cells. Virology 2020; 541:141-149. [PMID: 32056712 DOI: 10.1016/j.virol.2019.12.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/23/2019] [Accepted: 12/27/2019] [Indexed: 01/24/2023]
Abstract
Aedes aegypti is the primary vector of dengue virus (DENV), and acquires this virus from a vertebrate host during blood feeding. Previous literature has shown that vertebrate blood factors such as complement protein C5a and low-density lipoprotein (LDL) influence DENV acquisition in the mosquito. Here, we show that extracellular vesicles in cell culture medium inhibit DENV infection in mosquito cells. Specifically, extracellular vesicles enter into mosquito cells and inhibit an early stage of infection. Extracellular vesicles had no effect on virus cell attachment or entry. Instead, extracellular vesicles restricted virus membrane fusion. Extracellular vesicles only inhibited DENV infection in mosquito cells and not vertebrate cells. These data highlight a novel virus-vector-host interaction that limits virus infection in mosquito cells by restricting virus membrane fusion.
Collapse
Affiliation(s)
- Megan N Freitas
- Central Michigan University College of Medicine, Foundational Sciences, Mount Pleasant, MI, 48859, USA
| | - Andrew D Marten
- Central Michigan University College of Medicine, Foundational Sciences, Mount Pleasant, MI, 48859, USA
| | - Gavin A Moore
- Central Michigan University College of Medicine, Foundational Sciences, Mount Pleasant, MI, 48859, USA
| | - Maya O Tree
- Central Michigan University College of Medicine, Foundational Sciences, Mount Pleasant, MI, 48859, USA
| | - Sean P McBrayer
- Central Michigan University College of Medicine, Foundational Sciences, Mount Pleasant, MI, 48859, USA
| | - Michael J Conway
- Central Michigan University College of Medicine, Foundational Sciences, Mount Pleasant, MI, 48859, USA.
| |
Collapse
|
86
|
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
|
87
|
Influences of a Prolific Gut Fungus ( Zancudomyces culisetae) on Larval and Adult Mosquito (Aedes aegypti)-Associated Microbiota. Appl Environ Microbiol 2020; 86:AEM.02334-19. [PMID: 31757825 DOI: 10.1128/aem.02334-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/12/2019] [Indexed: 11/20/2022] Open
Abstract
Adult mosquitoes inherit a bacterial community from larvae via transstadial transmission, an understudied process that may influence host-microbe interactions. Microbes contribute to important host life history traits, and analyzing transmitted microbial communities, the interrelationship between larval and adult-associated microbiota, and factors influencing host-microbe relationships provides targets for research. During its larval stage, the yellow fever mosquito (Aedes aegypti) hosts the trichomycete gut fungus Zancudomyces culisetae, and fungal colonization coincides with environmental perturbations in the digestive tract microecosystem. Natural populations are differentially exposed to fungi, thereby potentially harboring distinct microbiota and experiencing disparate host-microbe interactions. This study's objectives were to characterize larval and initial adult microbiomes, investigate variation in diversity and distribution of microbial communities across individuals, and assess whether larval fungal colonization impacted microbiomes at these developmental stages. Laboratory-based fungal infestation assays, sequencing of 16S rRNA gene amplicons, and bacterial load quantification protocols revealed that initial adult microbiomes varied in diversity and distribution. Larval fungal colonization had downstream effects on initial adult microbiomes, significantly reducing microbial community variation, shifting relative abundances of certain bacterial families, and influencing transstadial transmission outcomes of particular genera. Further, abundances of several families consistently decreased in adults relative to levels in larvae, possibly reflecting impacts of host development on specific bacterial taxa. These findings demonstrated that a prolific gut fungus impacted mosquito-associated microbiota at two developmental stages in an insect connected with global human health.IMPORTANCE Mosquitoes are widespread vectors of numerous human pathogens and harbor microbiota known to affect host phenotypic traits. However, little research has directly investigated how bacterial communities associated with larvae and adults are connected. We characterized whole-body bacterial communities in mosquito larvae preceding pupation and in newly emerged adults, and investigated whether a significant biotic factor, fungal colonization of the larval hindgut, impacted these microbiomes. Results showed that fungal colonization reduced microbial community variation across individuals and differentially impacted the outcomes of transstadial transmission for certain bacterial genera, revealing downstream effects of the fungus on initial adult microbiomes. The importance of our research is in providing a thorough comparative analysis of whole-body microbiota harbored in larvae and adults of the yellow fever mosquito (Aedes aegypti) and in demonstrating the important role a widespread gut fungus played in a host-associated microbiome.
Collapse
|
88
|
Mota MBS, Carvalho MA, Monteiro ANA, Mesquita RD. DNA damage response and repair in perspective: Aedes aegypti, Drosophila melanogaster and Homo sapiens. Parasit Vectors 2019; 12:533. [PMID: 31711518 PMCID: PMC6849265 DOI: 10.1186/s13071-019-3792-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/05/2019] [Indexed: 01/18/2023] Open
Abstract
Background The maintenance of genomic integrity is the responsibility of a complex network, denominated the DNA damage response (DDR), which controls the lesion detection and DNA repair. The main repair pathways are base excision repair (BER), nucleotide excision repair (NER), mismatch repair (MMR), homologous recombination repair (HR) and non-homologous end joining repair (NHEJ). They correct double-strand breaks (DSB), single-strand breaks, mismatches and others, or when the damage is quite extensive and repair insufficient, apoptosis is activated. Methods In this study we used the BLAST reciprocal best-hit methodology to search for DDR orthologs proteins in Aedes aegypti. We also provided a comparison between Ae. aegypti, D. melanogaster and human DDR network. Results Our analysis revealed the presence of ATR and ATM signaling, including the H2AX ortholog, in Ae. aegypti. Key DDR proteins (orthologs to RAD51, Ku and MRN complexes, XP-components, MutS and MutL) were also identified in this insect. Other proteins were not identified in both Ae. aegypti and D. melanogaster, including BRCA1 and its partners from BRCA1-A complex, TP53BP1, PALB2, POLk, CSA, CSB and POLβ. In humans, their absence affects DSB signaling, HR and sub-pathways of NER and BER. Seven orthologs not known in D. melanogaster were found in Ae. aegypti (RNF168, RIF1, WRN, RAD54B, RMI1, DNAPKcs, ARTEMIS). Conclusions The presence of key DDR proteins in Ae. aegypti suggests that the main DDR pathways are functional in this insect, and the identification of proteins not known in D. melanogaster can help fill gaps in the DDR network. The mapping of the DDR network in Ae. aegypti can support mosquito biology studies and inform genetic manipulation approaches applied to this vector.
Collapse
Affiliation(s)
- Maria Beatriz S Mota
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcelo Alex Carvalho
- Instituto Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,Instituto Nacional de Câncer, Divisão de Pesquisa Clínica, Rio de Janeiro, RJ, Brazil
| | - Alvaro N A Monteiro
- Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Rafael D Mesquita
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil. .,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
89
|
Bottino-Rojas V, Pereira LOR, Silva G, Talyuli OAC, Dunkov BC, Oliveira PL, Paiva-Silva GO. Non-canonical transcriptional regulation of heme oxygenase in Aedes aegypti. Sci Rep 2019; 9:13726. [PMID: 31551499 PMCID: PMC6760526 DOI: 10.1038/s41598-019-49396-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022] Open
Abstract
Heme oxygenase (HO) is a ubiquitous enzyme responsible for heme breakdown, which yields carbon monoxide (CO), biliverdin (BV) and ferrous ion. Here we show that the Aedes aegypti heme oxygenase gene (AeHO - AAEL008136) is expressed in different developmental stages and tissues. AeHO expression increases after a blood meal in the midgut, and its maximal transcription levels overlaps with the maximal rate of the further modified A. aegypti biglutaminyl-biliverdin (AeBV) pigment production. HO is a classical component of stress response in eukaryotic cells, being activated under oxidative stress or increased heme levels. Indeed, the final product of HO activity in the mosquito midgut, AeBV, exerts a protective antioxidant activity. AeHO, however, does not seem to be under a classical redox-sensitive transcriptional regulation, being unresponsive to heme itself, and even down regulated when insects face a pro-oxidant insult. In contrast, AeHO gene expression responds to nutrient sensing mechanisms, through the target of rapamycin (TOR) pathway. This unusual transcriptional control of AeHO, together with the antioxidant properties of AeBV, suggests that heme degradation by HO, in addition to its important role in protection of Aedes aegypti against heme exposure, also acts as a digestive feature, being an essential adaptation to blood feeding.
Collapse
Affiliation(s)
- Vanessa Bottino-Rojas
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Luiza O R Pereira
- Laboratório de Pesquisas em Leishmaniose, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, 21040-360, Brazil
| | - Gabriela Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Octavio A C Talyuli
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Boris C Dunkov
- Center for Insect Science, The University of Arizona, Tucson, AZ, 85721-0106, USA
| | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Gabriela O Paiva-Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| |
Collapse
|
90
|
Scolari F, Casiraghi M, Bonizzoni M. Aedes spp. and Their Microbiota: A Review. Front Microbiol 2019; 10:2036. [PMID: 31551973 PMCID: PMC6738348 DOI: 10.3389/fmicb.2019.02036] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/19/2019] [Indexed: 12/21/2022] Open
Abstract
Aedes spp. are a major public health concern due to their ability to be efficient vectors of dengue, Chikungunya, Zika, and other arboviruses. With limited vaccines available and no effective therapeutic treatments against arboviruses, the control of Aedes spp. populations is currently the only strategy to prevent disease transmission. Host-associated microbes (i.e., microbiota) recently emerged as a promising field to be explored for novel environmentally friendly vector control strategies. In particular, gut microbiota is revealing its impact on multiple aspects of Aedes spp. biology, including vector competence, thus being a promising target for manipulation. Here we describe the technological advances, which are currently expanding our understanding of microbiota composition, abundance, variability, and function in the two main arboviral vectors, the mosquitoes Aedes aegypti and Aedes albopictus. Aedes spp. microbiota is described in light of its tight connections with the environment, with which mosquitoes interact during their various developmental stages. Unraveling the dynamic interactions among the ecology of the habitat, the mosquito and the microbiota have the potential to uncover novel physiological interdependencies and provide a novel perspective for mosquito control.
Collapse
Affiliation(s)
- Francesca Scolari
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Maurizio Casiraghi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | | |
Collapse
|
91
|
Caragata EP, Tikhe CV, Dimopoulos G. Curious entanglements: interactions between mosquitoes, their microbiota, and arboviruses. Curr Opin Virol 2019; 37:26-36. [PMID: 31176069 PMCID: PMC6768729 DOI: 10.1016/j.coviro.2019.05.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 11/22/2022]
Abstract
Mosquitoes naturally harbor a diverse community of microorganisms that play a crucial role in their biology. Mosquito-microbiota interactions are abundant and complex. They can dramatically alter the mosquito immune response, and impede or enhance a mosquito's ability to transmit medically important arboviral pathogens. Yet critically, given the massive public health impact of arboviral disease, few such interactions have been well characterized. In this review, we describe the current state of knowledge of the role of microorganisms in mosquito biology, how microbial-induced changes to mosquito immunity moderate infection with arboviruses, cases of mosquito-microbial-virus interactions with a defined mechanism, and the molecular interactions that underlie the endosymbiotic bacterium Wolbachia's ability to block virus infection in mosquitoes.
Collapse
Affiliation(s)
- Eric P Caragata
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Chinmay V Tikhe
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
92
|
Nouzova M, Clifton ME, Noriega FG. Mosquito adaptations to hematophagia impact pathogen transmission. CURRENT OPINION IN INSECT SCIENCE 2019; 34:21-26. [PMID: 31247413 DOI: 10.1016/j.cois.2019.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/28/2019] [Accepted: 02/04/2019] [Indexed: 06/09/2023]
Abstract
Mosquito-borne diseases such as Dengue fever, Chikungunya, and Malaria are critical threats to public health in many parts of the world. Female mosquitoes have evolved multiple adaptive mechanisms to hematophagy, including the ability to efficiently draw and digest blood, as well as the ability to eliminate excess fluids and toxic by-products of blood digestion. Pathogenic agents enter the mosquito digestive tract with the blood meal and need to travel through the midgut and into the hemocele in order to reach the salivary glands and infect a new host. Pathogens need to adjust to these hostile gut, hemocele, and salivary gland environments, and when possible influence the physiology and behavior of their hosts to enhance transmission.
Collapse
Affiliation(s)
- Marcela Nouzova
- Department of Biological Sciences and Biomolecular Science Institute, Florida International University, Miami, FL, USA; Institute of Parasitology, Biology Centre CAS, Ceske Budejovice, Czech Republic
| | - Mark E Clifton
- North Shore Mosquito Abatement District, Northfield, IL, USA
| | - Fernando G Noriega
- Department of Biological Sciences and Biomolecular Science Institute, Florida International University, Miami, FL, USA.
| |
Collapse
|
93
|
O’Donnell AJ, Rund SSC, Reece SE. Time-of-day of blood-feeding: effects on mosquito life history and malaria transmission. Parasit Vectors 2019; 12:301. [PMID: 31262362 PMCID: PMC6604169 DOI: 10.1186/s13071-019-3513-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/17/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Biological rhythms allow organisms to compartmentalise and coordinate behaviours, physiologies, and cellular processes with the predictable daily rhythms of their environment. There is increasing recognition that the biological rhythms of mosquitoes that vector parasites are important for global health. For example, whether perturbations in blood foraging rhythms as a consequence of vector control measures can undermine disease control. To address this, we explore the impacts of altered timing of blood-feeding on mosquito life history traits and malaria transmission. METHODS We present three experiments in which Anopheles stephensi mosquitoes were fed in the morning or evening on blood that had different qualities, including: (i) chemical-induced or (ii) Plasmodium chabaudi infection-induced anaemia; (iii) Plasmodium berghei infection but no anaemia; or (iv) stemming from hosts at different times of day. We then compared whether time-of-day variation in blood meal characteristics influences mosquito fitness proxies relating to survival and reproduction, and malaria transmission proxies. RESULTS Mosquito lifespan is not influenced by the time-of-day they received a blood meal, but several reproductive metrics are affected, depending on other blood characteristics. Overall, our data suggest that receiving a blood meal in the morning makes mosquitoes more likely to lay eggs, lay slightly sooner and have a larger clutch size. In keeping with previous work, P. berghei infection reduces mosquito lifespan and the likelihood of laying eggs, but time-of-day of blood-feeding does not impact upon these metrics nor on transmission of this parasite. CONCLUSION The time-of-day of blood-feeding does not appear to have major consequences for mosquito fitness or transmission of asynchronous malaria species. If our results from a laboratory colony of mosquitoes living in benign conditions hold for wild mosquitoes, it suggests that mosquitoes have sufficient flexibility in their physiology to cope with changes in biting time induced by evading insecticide-treated bed nets. Future work should consider the impact of multiple feeding cycles and the abiotic stresses imposed by the need to forage for blood during times of day when hosts are not protected by bed nets.
Collapse
Affiliation(s)
- Aidan J. O’Donnell
- Institute of Evolutionary Biology, and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Samuel S. C. Rund
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556 USA
| | - Sarah E. Reece
- Institute of Evolutionary Biology, and Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
94
|
Zhang J, Ahmad S, Wang LY, Han Q, Zhang JC, Luo YP. Cell death induced by α-terthienyl via reactive oxygen species-mediated mitochondrial dysfunction and oxidative stress in the midgut of Aedes aegypti larvae. Free Radic Biol Med 2019; 137:87-98. [PMID: 31022448 DOI: 10.1016/j.freeradbiomed.2019.04.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 04/14/2019] [Accepted: 04/16/2019] [Indexed: 12/13/2022]
Abstract
α-Terthienyl (α-T) is a photosensitizer that produces many reactive oxygen species (ROS) under ultraviolet light. Here, we aimed to evaluate the oxidation mechanism of the 25%, 50%, and 75% lethal concentrations in Aedes aegypti larvae; the lethal concentration of α-T was used as the test value. The effects on mitochondria, oxidative stress, and cell death patterns caused by ROS were evaluated. The results showed that α-T mainly produced large amounts of ROS in the midgut of larvae. Moreover, mitochondrial ROS were increased in midgut cells, and the production of ROS sites, such as complex enzymes, was inhibited, resulting in enhanced production of ROS. Ultrastructural analysis of mitochondria revealed significant vacuolation, decreased activity of tricarboxylic acid cycle enzymes, and reduced ATP content and mitochondrial membrane potential in the high concentration group compared with those in the control group. Additionally, mitochondrial biosynthesis was blocked in the high concentration group. Thus, exposure to α-T disrupted mitochondrial function, although the mitochondrial DNA content may have increased because of mitochondrial self-protection mechanisms against oxidative stress. Furthermore, high concentrations of α-T aggravated oxidative stress and increased the number of intracellular oxidative damage products. Reverse transcription polymerase chain reaction and fluorescence staining showed that ROS induced by low α-T concentrations upregulated apoptotic genes, including Dronc (P < 0.05), thereby promoting apoptosis. Moderate concentrations of α-T promoted autophagy through induction of ROS, inhibited apoptosis, and induced necrosis. In contrast, high α-T concentrations induced high levels of ROS, which caused mitochondrial dysfunction and increased cytoplasmic Ca2+ concentration, directly inducing cell necrosis. We also found that α-T may disrupt the permeability of the peritrophic membrane, leading to intestinal barrier dysfunction. These results provided insights into the mode of action of α-T in Aedes aegypti.
Collapse
Affiliation(s)
- Jie Zhang
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Hainan University, Ministry of Education, Haikou, Hainan 570228, PR China
| | - Shakil Ahmad
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Hainan University, Ministry of Education, Haikou, Hainan 570228, PR China
| | - Lan-Ying Wang
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Hainan University, Ministry of Education, Haikou, Hainan 570228, PR China
| | - Qian Han
- The Laboratory of Tropical Animal Medicine and Vector Biology, Hainan University, Haikou, Hainan 570228, PR China
| | - Jian-Chun Zhang
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Hainan University, Ministry of Education, Haikou, Hainan 570228, PR China
| | - Yan-Ping Luo
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests, Hainan University, Ministry of Education, Haikou, Hainan 570228, PR China.
| |
Collapse
|
95
|
Aedes aegypti HPX8C modulates immune responses against viral infection. PLoS Negl Trop Dis 2019; 13:e0007287. [PMID: 30986216 PMCID: PMC6464178 DOI: 10.1371/journal.pntd.0007287] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 03/08/2019] [Indexed: 12/26/2022] Open
Abstract
Mosquitoes act as vectors of numerous pathogens that cause human diseases. Dengue virus (DENV) transmitted by mosquito, Aedes aegypti, is responsible for dengue fever epidemics worldwide with a serious impact on human health. Currently, disease control mainly relies on vector targeted intervention strategies. Therefore, it is imperative to understand the molecular mechanisms underlying the innate immune response of mosquitoes against pathogens. In the present study, the expression profiles of immunity-related genes in the midgut responding to DENV infection by feeding were analyzed by transcriptome and quantitative real-time PCR. The level of Antimicrobial peptides (AMPs) increased seven days post-infection (d.p.i.), which could be induced by the Toll immune pathway. The expression of reactive oxygen species (ROS) genes, including antioxidant genes, such as HPX7, HPX8A, HPX8B, HPX8C were induced at one d.p.i. and peaked again at ten d.p.i. in the midgut. Interestingly, down-regulation of the antioxidant gene HPX8C by RNA interference led to reduction in the virus titer in the mosquito, probably due to the elevated levels of ROS. Application of a ROS inhibitor and scavenger molecules further established the role of oxygen free radicals in the modulation of the immune response to DENV infection. Overall, our comparative transcriptome analyses provide valuable information about the regulation of immunity related genes in the transmission vector in response to DENV infection. It further allows us to identify novel molecular mechanisms underlying the host-virus interaction, which might aid in the development of novel strategies to control mosquito-borne diseases. HPX8C is a heme-containing peroxidase, which can move reactive oxygen species (ROS) damage to the organism by reducing H2O2 to H2O. Previously, the peroxidase gene has been shown to modulate midgut immunity and regulate anti-malarial response in mosquitoes. In this study, the classical immune signaling pathways, Toll and IMD genes might be late responses against the viruses. HPX8C was demonstrated here to play a role in antiviral immunity against DENV infection in Ae. Aegypti mosquitoes. HPX8C expression was induced by DENV infection and continued to increase with an elevated virus titer. In HPX8C-depleted mosquitoes, the ROS level was found to be increased with a corresponding decrease in the DENV and ZIKV virus titer. Therefore, it was speculated that HPX8C mediated immune responses against the DENV in the mosquito in the late stage of viral infection, which could be controlled by Toll pathway.
Collapse
|
96
|
Heu K, Gendrin M. [Mosquito microbiota and its influence on disease vectorial transmission]. Biol Aujourdhui 2019; 212:119-136. [PMID: 30973141 DOI: 10.1051/jbio/2019003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Indexed: 01/23/2023]
Abstract
Mosquitoes (Diptera: Culicidae) are found worldwide. Around 100 among 3500 mosquito species are known to be vectors of parasites and viruses, responsible for infectious diseases including malaria and dengue. Mosquitoes host diverse microbial communities that influence disease transmission, either by direct interference or via affecting host immunity and physiology. These microbial communities are present within diverse tissues, including the digestive tract, and vary depending on the sex of the mosquito, its developmental stage, and ecological factors. This review summarizes the current knowledge about the mosquito microbiota, defined as a community of commensal, symbiotic or pathogenic microbes harboured by a host. We first describe the current knowledge on the diversity of the microbiota, that includes bacteria, fungi, parasites and viruses and on its modes of acquisition throughout the mosquito life cycle. We then focus on microbial interactions within the mosquito gut, which notably affect vector competence, and on host-microbe interactions affecting mosquito fitness. Finally, we discuss current or potential methods based on the use of microbes or microbial products to interfere with pathogen transmission or to reduce mosquito lifespan and reproduction.
Collapse
Affiliation(s)
- Katy Heu
- Groupe « Microbiote des Insectes Vecteurs », Institut Pasteur de la Guyane, Cayenne, Guyane, France
| | - Mathilde Gendrin
- Groupe « Microbiote des Insectes Vecteurs », Institut Pasteur de la Guyane, Cayenne, Guyane, France - Département « Parasites et Insectes Vecteurs », Institut Pasteur, Paris, France
| |
Collapse
|
97
|
Kakani P, Kajla M, Choudhury TP, Gupta L, Kumar S. Anopheles stephensi Dual Oxidase Silencing Activates the Thioester-Containing Protein 1 Pathway to Suppress Plasmodium Development. J Innate Immun 2019; 11:496-505. [PMID: 30928970 DOI: 10.1159/000497417] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/25/2019] [Indexed: 12/29/2022] Open
Abstract
We characterized the dual oxidase (Duox) gene in the major Indian malaria vector Anopheles stephensi, which regulates the generation of reactive oxygen species. The AsDuox gene encodes for a 1,475-amino-acid transmembrane protein that contains an N-terminal noncytoplasmic heme peroxidase domain, a calcium-binding domain, seven transmembrane domains, and a C-terminal cytoplasmic NADPH domain. Phylogenetic analyses revealed that A. stephensi Duox protein is highly conserved and shares 97-100% amino acid identity with other anopheline Duoxes. AsDuox is expressed in all the developmental stages of A. stephensi and the pupal stages revealed relatively higher expressions. The Duox gene is induced in Plasmodium-infected mosquito midguts, and RNA interference-mediated silencing of this gene suppressed parasite development through activation of the thioester-containing protein 1 pathway. We propose that this highly conserved anopheline Duox, being a Plasmodium agonist, is an excellent target to control malaria parasite development inside the insect host.
Collapse
Affiliation(s)
- Parik Kakani
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science, Vidya Vihar Campus, Pilani, India
| | - Mithilesh Kajla
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science, Vidya Vihar Campus, Pilani, India.,National Institute of Malaria Research, New Delhi, India
| | - Tania Pal Choudhury
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science, Vidya Vihar Campus, Pilani, India
| | - Lalita Gupta
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science, Vidya Vihar Campus, Pilani, India.,Department of Zoology, Chaudhary Bansi Lal University, Bhiwani, India
| | - Sanjeev Kumar
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science, Vidya Vihar Campus, Pilani, India, .,Department of Biotechnology, Chaudhary Bansi Lal University, Bhiwani, India,
| |
Collapse
|
98
|
Haem Biology in Metazoan Parasites - 'The Bright Side of Haem'. Trends Parasitol 2019; 35:213-225. [PMID: 30686614 DOI: 10.1016/j.pt.2019.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/04/2019] [Accepted: 01/04/2019] [Indexed: 12/12/2022]
Abstract
Traditionally, host haem has been recognized as a cytotoxic molecule that parasites need to eliminate or detoxify in order to survive. However, recent evidence indicates that some lineages of parasites have lost genes that encode enzymes involved specifically in endogenous haem biosynthesis. Such lineages thus need to acquire and utilize haem originating from their host animal, making it an indispensable molecule for their survival and reproduction. In multicellular parasites, host haem needs to be systemically distributed throughout their bodies to meet the haem demands in all cell and tissue types. Host haem also gets deposited in parasite eggs, enabling embryogenesis and reproduction. Clearly, a better understanding of haem biology in multicellular parasites should elucidate organismal adaptations to obligatory blood-feeding.
Collapse
|
99
|
Sousa G, Gandara ACP, Oliveira PL, Gomes FM, Bahia AC, Machado EA. The relationship between oxidant levels and gut physiology in a litter-feeding termite. Sci Rep 2019; 9:670. [PMID: 30679618 PMCID: PMC6345907 DOI: 10.1038/s41598-018-37043-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 11/28/2018] [Indexed: 11/09/2022] Open
Abstract
The termite gut is an efficient decomposer of polyphenol-rich diets, such as lignocellulosic biomasses, and it has been proposed that non-enzymatic oxidative mechanisms could be involved with the digestive process in these animals. However, oxidant levels are completely unknown in termites, as well as protective mechanisms against oxidative damage to the termite gut and its microbiota. As the first step in investigating the role oxidants plays in termite gut physiology, this work presents oxidant levels, antioxidant enzymatic defenses, cell renewal and microbiota abundance along the litter-feeding termite Cornitermes cumulans gut compartments (foregut, midgut, mixed segment and hindgut p1, p3, p4, and p5 segments) and salivary glands. The results show variable levels of oxidants along the C. cumulans gut, the production of antioxidant enzymes, gut cell renewal as potential defenses against oxidative injuries and the profile of microbiota distribution (being predominantly inverse to oxidant levels). In this fashion, the oxidative challenges imposed by polyphenol-rich diet seem to be circumvented by the C. cumulans gut, ensuring efficiency of the digestive process together with preservation of tissue homoeostasis and microbiota growth. These results present new insights into the physicochemical properties of the gut in a litter-feeding termite, expanding our view in relation to termites’ digestive physiology.
Collapse
Affiliation(s)
- Gessica Sousa
- Laboratório de Bioquímica de Insetos e Parasitos (Labip), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Caroline P Gandara
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L Oliveira
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabio M Gomes
- Laboratory of Malaria and Vector Research, National Institute of Health, Bethesda, United States of America
| | - Ana Cristina Bahia
- Laboratório de Bioquímica de Insetos e Parasitos (Labip), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ednildo A Machado
- Laboratório de Bioquímica de Insetos e Parasitos (Labip), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil. .,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil.
| |
Collapse
|
100
|
Souza-Neto JA, Powell JR, Bonizzoni M. Aedes aegypti vector competence studies: A review. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2019; 67:191-209. [PMID: 30465912 PMCID: PMC8135908 DOI: 10.1016/j.meegid.2018.11.009] [Citation(s) in RCA: 254] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023]
Abstract
Aedes aegypti is the primary transmitter of the four viruses that have had the greatest impact on human health, the viruses causing yellow fever, dengue fever, chikungunya, and Zika fever. Because this mosquito is easy to rear in the laboratory and these viruses grow in laboratory tissue culture cells, many studies have been performed testing the relative competence of different populations of the mosquito to transmit many different strains of viruses. We review here this large literature including studies on the effect of the mosquito microbiota on competence. Because of the heterogeneity of both mosquito populations and virus strains used, as well as methods measuring potential to transmit, it is very difficult to perform detailed meta-analysis of the studies. However, a few conclusions can be drawn: (1) almost no population of Ae. aegypti is 100% naturally refractory to virus infection. Complete susceptibility to infection has been observed for Zika (ZIKV), dengue (DENV) and chikungunya (CHIKV), but not yellow fever viruses (YFV); (2) the dose of virus used is directly correlated to the rate of infection; (3) Brazilian populations of mosquito are particularly susceptible to DENV-2 infections; (4) the Asian lineage of ZIKV is less infective to Ae. aegypti populations from the American continent than is the African ZIKV lineage; (5) virus adaptation to different species of mosquitoes has been demonstrated with CHIKV; (6) co-infection with more than one virus sometimes causes displacement while in other cases has little effect; (7) the microbiota in the mosquito also has important effects on level of susceptibility to arboviral infection; (8) resistance to virus infection due to the microbiota may be direct (e.g., bacteria producing antiviral proteins) or indirect in activating the mosquito host innate immune system; (9) non-pathogenic insect specific viruses (ISVs) are also common in mosquitoes including genome insertions. These too have been shown to have an impact on the susceptibility of mosquitoes to pathogenic viruses. One clear conclusion is that it would be a great advance in this type of research to implement standardized procedures in order to obtain comparable and reproducible results.
Collapse
Affiliation(s)
- Jayme A Souza-Neto
- São Paulo State University (UNESP), School of Agricultural Sciences, Department of Bioprocesses and Biotechnology, Multiuser Central Laboratory, Botucatu, Brazil; São Paulo State University (UNESP), Institute of Biotechnology, Botucatu, Brazil
| | | | | |
Collapse
|