Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Biol ; 22(3): e3002573, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38547237

RESUMO

The rising interest and success in deploying inherited microorganisms and cytoplasmic incompatibility (CI) for vector control strategies necessitate an explanation of the CI mechanism. Wolbachia-induced CI manifests in the form of embryonic lethality when sperm from Wolbachia-bearing testes fertilize eggs from uninfected females. Embryos from infected females however survive to sustain the maternally inherited symbiont. Previously in Drosophila melanogaster flies, we demonstrated that CI modifies chromatin integrity in developing sperm to bestow the embryonic lethality. Here, we validate these findings using wMel-transinfected Aedes aegypti mosquitoes released to control vector-borne diseases. Once again, the prophage WO CI proteins, CifA and CifB, target male gametic nuclei to modify chromatin integrity via an aberrant histone-to-protamine transition. Cifs are not detected in the embryo, and thus elicit CI via the nucleoprotein modifications established pre-fertilization. The rescue protein CifA in oogenesis localizes to stem cell, nurse cell, and oocyte nuclei, as well as embryonic DNA during embryogenesis. Discovery of the nuclear targeting Cifs and altered histone-to-protamine transition in both Aedes aegypti mosquitoes and D. melanogaster flies affirm the Host Modification Model of CI is conserved across these host species. The study also newly uncovers the cell biology of Cif proteins in the ovaries, CifA localization in the embryos, and an impaired histone-to-protamine transition during spermiogenesis of any mosquito species. Overall, these sperm modification findings may enable future optimization of CI efficacy in vectors or pests that are refractory to Wolbachia transinfections.


Assuntos
Aedes , Arbovírus , Wolbachia , Animais , Feminino , Masculino , Drosophila melanogaster/genética , Histonas/genética , Mosquitos Vetores , Sêmen , Drosophila/genética , Cromatina , Protaminas/genética
2.
PLoS Pathog ; 19(4): e1011307, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37043515

RESUMO

Aedes aegypti is the primary vector of the arboviruses dengue (DENV) and chikungunya (CHIKV). These viruses exhibit key differences in their vector interactions, the latter moving more quicky through the mosquito and triggering fewer standard antiviral pathways. As the global footprint of CHIKV continues to expand, we seek to better understand the mosquito's natural response to CHIKV-both to compare it to DENV:vector coevolutionary history and to identify potential targets in the mosquito for genetic modification. We used a modified full-sibling design to estimate the contribution of mosquito genetic variation to viral loads of both DENV and CHIKV. Heritabilities were significant, but higher for DENV (40%) than CHIKV (18%). Interestingly, there was no genetic correlation between DENV and CHIKV loads between siblings. These data suggest Ae. aegypti mosquitoes respond to the two viruses using distinct genetic mechanisms. We also examined genome-wide patterns of gene expression between High and Low CHIKV families representing the phenotypic extremes of viral load. Using RNAseq, we identified only two loci that consistently differentiated High and Low families: a long non-coding RNA that has been identified in mosquito screens post-infection and a distant member of a family of Salivary Gland Specific (SGS) genes. Interestingly, the latter gene is also associated with horizontal gene transfer between mosquitoes and the endosymbiotic bacterium Wolbachia. This work is the first to link the SGS gene to a mosquito phenotype. Understanding the molecular details of how this gene contributes to viral control in mosquitoes may, therefore, also shed light on its role in Wolbachia.


Assuntos
Aedes , Febre de Chikungunya , Vírus Chikungunya , Dengue , Animais , Vírus Chikungunya/fisiologia , Mosquitos Vetores
3.
Insect Mol Biol ; 33(4): 362-371, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38450861

RESUMO

Multiple Wolbachia strains can block pathogen infection, replication and/or transmission in Aedes aegypti mosquitoes under both laboratory and field conditions. However, Wolbachia effects on pathogens can be highly variable across systems and the factors governing this variability are not well understood. It is increasingly clear that the mosquito host is not a passive player in which Wolbachia governs pathogen transmission phenotypes; rather, the genetics of the host can significantly modulate Wolbachia-mediated pathogen blocking. Specifically, previous work linked variation in Wolbachia pathogen blocking to polymorphisms in the mosquito alpha-mannosidase-2 (αMan2) gene. Here we use CRISPR-Cas9 mutagenesis to functionally test this association. We developed αMan2 knockouts and examined effects on both Wolbachia and virus levels, using dengue virus (DENV; Flaviviridae) and Mayaro virus (MAYV; Togaviridae). Wolbachia titres were significantly elevated in αMan2 knockout (KO) mosquitoes, but there were complex interactions with virus infection and replication. In Wolbachia-uninfected mosquitoes, the αMan2 KO mutation was associated with decreased DENV titres, but in a Wolbachia-infected background, the αMan2 KO mutation significantly increased virus titres. In contrast, the αMan2 KO mutation significantly increased MAYV replication in Wolbachia-uninfected mosquitoes and did not affect Wolbachia-mediated virus blocking. These results demonstrate that αMan2 modulates arbovirus infection in A. aegypti mosquitoes in a pathogen- and Wolbachia-specific manner, and that Wolbachia-mediated pathogen blocking is a complex phenotype dependent on the mosquito host genotype and the pathogen. These results have a significant impact for the design and use of Wolbachia-based strategies to control vector-borne pathogens.


Assuntos
Aedes , Wolbachia , alfa-Manosidase , Animais , Aedes/microbiologia , Aedes/virologia , Aedes/genética , Wolbachia/fisiologia , alfa-Manosidase/metabolismo , alfa-Manosidase/genética , Vírus da Dengue/fisiologia , Arbovírus/fisiologia , Mosquitos Vetores/microbiologia , Mosquitos Vetores/virologia , Mosquitos Vetores/genética , Feminino , Infecções por Arbovirus/transmissão , Proteínas de Insetos/metabolismo , Proteínas de Insetos/genética , Sistemas CRISPR-Cas
4.
Glob Chang Biol ; 30(1): e17041, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38273521

RESUMO

Most models exploring the effects of climate change on mosquito-borne disease ignore thermal adaptation. However, if local adaptation leads to changes in mosquito thermal responses, "one size fits all" models could fail to capture current variation between populations and future adaptive responses to changes in temperature. Here, we assess phenotypic adaptation to temperature in Aedes aegypti, the primary vector of dengue, Zika, and chikungunya viruses. First, to explore whether there is any difference in existing thermal response of mosquitoes between populations, we used a thermal knockdown assay to examine five populations of Ae. aegypti collected from climatically diverse locations in Mexico, together with a long-standing laboratory strain. We identified significant phenotypic variation in thermal tolerance between populations. Next, to explore whether such variation can be generated by differences in temperature, we conducted an experimental passage study by establishing six replicate lines from a single field-derived population of Ae. aegypti from Mexico, maintaining half at 27°C and the other half at 31°C. After 10 generations, we found a significant difference in mosquito performance, with the lines maintained under elevated temperatures showing greater thermal tolerance. Moreover, these differences in thermal tolerance translated to shifts in the thermal performance curves for multiple life-history traits, leading to differences in overall fitness. Together, these novel findings provide compelling evidence that Ae. aegypti populations can and do differ in thermal response, suggesting that simplified thermal performance models might be insufficient for predicting the effects of climate on vector-borne disease transmission.


Assuntos
Aedes , Infecção por Zika virus , Zika virus , Animais , Mosquitos Vetores/fisiologia , Aedes/fisiologia , Temperatura
5.
Cell ; 139(7): 1268-78, 2009 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-20064373

RESUMO

Wolbachia are maternally inherited intracellular bacterial symbionts that are estimated to infect more than 60% of all insect species. While Wolbachia is commonly found in many mosquitoes it is absent from the species that are considered to be of major importance for the transmission of human pathogens. The successful introduction of a life-shortening strain of Wolbachia into the dengue vector Aedes aegypti that halves adult lifespan has recently been reported. Here we show that this same Wolbachia infection also directly inhibits the ability of a range of pathogens to infect this mosquito species. The effect is Wolbachia strain specific and relates to Wolbachia priming of the mosquito innate immune system and potentially competition for limiting cellular resources required for pathogen replication. We suggest that this Wolbachia-mediated pathogen interference may work synergistically with the life-shortening strategy proposed previously to provide a powerful approach for the control of insect transmitted diseases.


Assuntos
Aedes/microbiologia , Vírus Chikungunya/fisiologia , Vírus da Dengue/fisiologia , Plasmodium gallinaceum/fisiologia , Wolbachia/fisiologia , Aedes/parasitologia , Aedes/fisiologia , Aedes/virologia , Animais , Interações Hospedeiro-Parasita , Simbiose
6.
Glob Chang Biol ; 29(19): 5540-5551, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37560790

RESUMO

By 2100, greenhouse gases are predicted to reduce ozone and cloud cover over the tropics causing increased exposure of organisms to harmful ultraviolet-B radiation (UVBR). UVBR damages DNA and is an important modulator of immune function and disease susceptibility in humans and other vertebrates. The effect of UVBR on invertebrate immune function is largely unknown, but UVBR together with ultraviolet-A radiation impairs an insect immune response that utilizes melanin, a pigment that also protects against UVBR-induced DNA damage. If UVBR weakens insect immunity, then it may make insect disease vectors more susceptible to infection with pathogens of socioeconomic and public health importance. In the tropics, where UVBR is predicted to increase, the mosquito-borne dengue virus (DENV), is prevalent and a growing threat to humans. We therefore examined the effect of UVBR on the mosquito Aedes aegypti, the primary vector for DENV, to better understand the potential implications of increased tropical UVBR for mosquito-borne disease risk. We found that exposure to a UVBR dose that caused significant larval mortality approximately doubled the probability that surviving females would become infected with DENV, despite this UVBR dose having no effect on the expression of an effector gene involved in antiviral immunity. We also found that females exposed to a lower UVBR dose were more likely to have low fecundity even though this UVBR dose had no effect on larval size or activity, pupal cuticular melanin content, or adult mass, metabolic rate, or flight capacity. We conclude that future increases in tropical UVBR associated with anthropogenic global change may have the benefit of reducing mosquito-borne disease risk for humans by reducing mosquito fitness, but this benefit may be eroded if it also makes mosquitoes more likely to be infected with deadly pathogens.


Assuntos
Aedes , Vírus da Dengue , Dengue , Humanos , Animais , Feminino , Vírus da Dengue/genética , Vírus da Dengue/metabolismo , Mosquitos Vetores , Melaninas/metabolismo , Aedes/genética , Aedes/metabolismo , Larva
7.
J Virol ; 95(13): e0223220, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33827954

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has initiated a global pandemic, and several vaccines have now received emergency use authorization. Using the reference strain SARS-CoV-2 USA-WA1/2020, we evaluated modes of transmission and the ability of prior infection or vaccine-induced immunity to protect against infection in ferrets. Ferrets were semipermissive to infection with the USA-WA1/2020 isolate. When transmission was assessed via the detection of viral RNA (vRNA) at multiple time points, direct contact transmission was efficient to 3/3 and 3/4 contact animals in 2 respective studies, while respiratory droplet transmission was poor to only 1/4 contact animals. To determine if previously infected ferrets were protected against reinfection, ferrets were rechallenged 28 or 56 days postinfection. Following viral challenge, no infectious virus was recovered in nasal wash samples. In addition, levels of vRNA in the nasal wash were several orders of magnitude lower than during primary infection, and vRNA was rapidly cleared. To determine if intramuscular vaccination protected ferrets, ferrets were vaccinated using a prime-boost strategy with the S protein receptor-binding domain formulated with an oil-in-water adjuvant. Upon viral challenge, none of the mock or vaccinated animals were protected against infection, and there were no significant differences in vRNA or infectious virus titers in the nasal wash. Combined, these studies demonstrate direct contact is the predominant mode of transmission of the USA-WA1/2020 isolate in ferrets and that immunity to SARS-CoV-2 is maintained for at least 56 days. Our studies also indicate protection of the upper respiratory tract against SARS-CoV-2 will require vaccine strategies that mimic natural infection or induce site-specific immunity. IMPORTANCE The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) USA-WA1/2020 strain is a CDC reference strain used by multiple research laboratories. Here, we show that the predominant mode of transmission of this isolate in ferrets is by direct contact. We further demonstrate ferrets are protected against reinfection for at least 56 days even when levels of neutralizing antibodies are low or undetectable. Last, we show that when ferrets were vaccinated by the intramuscular route to induce antibodies against SARS-CoV-2, ferrets remain susceptible to infection of the upper respiratory tract. Collectively, these studies suggest that protection of the upper respiratory tract will require vaccine approaches that mimic natural infection.


Assuntos
COVID-19/transmissão , Modelos Animais de Doenças , Reinfecção/prevenção & controle , SARS-CoV-2/fisiologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , Vacinas contra COVID-19/administração & dosagem , Furões , Injeções Intramusculares , Nariz/virologia , Reinfecção/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/isolamento & purificação , Glicoproteína da Espícula de Coronavírus/administração & dosagem , Carga Viral
8.
Insect Mol Biol ; 31(3): 356-368, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35112745

RESUMO

One approach to control dengue virus transmission is the symbiont Wolbachia, which limits viral infection in mosquitoes. Despite plans for its widespread use in Aedes aegypti, Wolbachia's mode of action remains poorly understood. Many studies suggest that the mechanism is likely multifaceted, involving aspects of immunity, cellular stress and nutritional competition. A previous study from our group used artificial selection to identify a new mosquito candidate gene related to viral blocking; alpha-mannosidase-2a (alpha-Mann-2a) with a predicted role in protein glycosylation. Protein glycosylation pathways tend to be involved in complex host-viral interactions; however, the function of alpha-mannosidases has not been described in mosquito-virus interactions. We examined alpha-Mann-2a expression in response to virus and Wolbachia infections and whether reduced gene expression, caused by RNA interference, affected viral loads. We show that dengue virus (DENV) infection affects the expression of alpha-Mann-2a in a tissue- and time-dependent manner, whereas Wolbachia infection had no effect. In the midgut, DENV prevalence increased following knockdown of alpha-Mann-2a expression in Wolbachia-free mosquitoes, suggesting that alpha-Mann-2a interferes with infection. Expression knockdown had the same effect on the togavirus chikungunya virus, indicating that alpha-Mann-2a may have broad antivirus effects in the midgut. Interestingly, we were unable to knockdown the expression in Wolbachia-infected mosquitoes. We also provide evidence that alpha-Mann-2a may affect the transcriptional level of another gene predicted to be involved in viral blocking and cell adhesion; cadherin87a. These data support the hypothesis that glycosylation and adhesion pathways may broadly be involved in viral infection in Ae. aegypti.


Assuntos
Aedes , Vírus Chikungunya , Vírus da Dengue , Viroses , Wolbachia , Aedes/genética , Animais , Vírus da Dengue/genética , Mosquitos Vetores/genética , Wolbachia/fisiologia
9.
PLoS Pathog ; 15(12): e1008218, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31790509

RESUMO

Dengue virus (DENV) transmission by mosquitoes is a time-dependent process that begins with the consumption of an infectious blood-meal. DENV infection then proceeds stepwise through the mosquito from the midgut to the carcass, and ultimately to the salivary glands, where it is secreted into saliva and then transmitted anew on a subsequent bite. We examined viral kinetics in tissues of the Aedes aegypti mosquito over a finely graded time course, and as per previous studies, found that initial viral dose and serotype strain diversity control infectivity. We also found that a threshold level of virus is required to establish body-wide infections and that replication kinetics in the early and intermediate tissues do not predict those of the salivary glands. Our findings have implications for mosquito GMO design, modeling the contribution of transmission to vector competence and the role of mosquito kinetics in the overall DENV epidemiological landscape.


Assuntos
Vírus da Dengue , Dengue/virologia , Interações Hospedeiro-Parasita/fisiologia , Mosquitos Vetores/virologia , Aedes , Animais , Cinética , Replicação Viral
10.
BMC Infect Dis ; 19(1): 580, 2019 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-31272403

RESUMO

BACKGROUND: Transcriptomic profiling has generated extensive lists of genes that respond to viral infection in mosquitoes. These gene lists contain two types of genes; (1) those that are responsible for the insect's natural antiviral defense mechanisms, including some known innate immunity genes, and (2) genes whose change in expression may occur simply as a result of infection. As genetic modification tools for mosquitoes continue to improve, the opportunities to make refractory insects via allelic replacement or delivery of small RNAs that alter gene expression are expanding. Therefore, the ability to identify which genes in transcriptional profiles may have immune function has increasing value. Arboviruses encounter a range of mosquito tissues and physiologies as they traverse from the midgut to the salivary glands. While the midgut is well-studied as the primary tissue barrier, antiviral genes expressed in the subsequent tissues of the carcass offer additional candidates for second stage intervention in the mosquito body. METHODS: Mosquito lines collected recently from field populations exhibit natural genetic variation for dengue virus susceptibility. We sought to use a modified full-sib breeding design to identify mosquito families that varied in their dengue viral load in their bodies post infection. RESULTS: By delivering virus intrathoracically, we bypassed the midgut and focused on whole body responses in order to evaluate carcass-associated refractoriness. We tested 25 candidate genes selected for their appearance in multiple published transcriptional profiles and were able to identify 12 whose expression varied with susceptibility in the genetic families. CONCLUSIONS: This method, using natural genetic variation, offers a simple means to screen and reduce candidate gene lists prior to carrying out more labor-intensive functional studies. The extracted RNA from the females across the families represents a storable resource that can be used to screen subsequent candidate genes in the future. The aspect of vector competence being assessed could be varied by focusing on different tissues or time points post infection.


Assuntos
Aedes/virologia , Vírus da Dengue/genética , Dengue/virologia , Variação Genética , Imunidade Inata/genética , Mosquitos Vetores/virologia , Animais , Vírus da Dengue/isolamento & purificação , Feminino , Perfilação da Expressão Gênica , Masculino , Carga Viral
11.
J Gen Virol ; 99(6): 832-836, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29741476

RESUMO

A novel negative-sense RNA virus, Aedes aegypti anphevirus, was recently identified in wild Aedes aegypti mosquitoes. We show that this virus is also present in the Aag2 Aedes aegypti cell line and characterize its complete genome and evolutionary history. The Aedes aegypti anphevirus genome is estimated to be 12 916 nucleotides in length, contains four genes and has a genome structure similar to that of other anpheviruses. Phylogenetically, Aedes aegypti anphevirus falls within an unclassified group of insect-specific viruses in the order Mononegavirales that form a sister-group to the chuviruses. Notably, the Aag2 cell line used here was also experimentally infected with dengue virus and naturally contained a Phasi Charoen-like virus and cell-fusing agent virus. All four viruses were at relatively high abundance, with 0.5 % of sequence reads assigned to Aedes aegypti anphevirus. The Aag2 cell line is therefore permissive to efficient co-infection with dengue virus and multiple insect-specific viruses.


Assuntos
Aedes/virologia , Genoma Viral , Vírus de Insetos/genética , Animais , Linhagem Celular , Vírus da Dengue/genética , Insetos Vetores , Vírus de Insetos/fisiologia , Vírus de RNA/genética , Vírus de RNA/fisiologia , Replicação Viral
12.
Mol Ecol ; 27(1): 297-309, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29165845

RESUMO

Insect-symbiont interactions are known to play key roles in host functions and fitness. The common insect endosymbiont Wolbachia can reduce the ability of several human pathogens, including arboviruses and the malaria parasite, to replicate in insect hosts. Wolbachia does not naturally infect Aedes aegypti, the primary vector of dengue virus, but transinfected Ae. aegypti have antidengue virus properties and are currently being trialled as a dengue biocontrol strategy. Here, we assess the impact of Wolbachia infection of Ae. aegypti on the microbiome of wild mosquito populations (adults and larvae) collected from release sites in Cairns, Australia, by profiling the 16S rRNA gene using next-generation sequencing. Our data indicate that Wolbachia reduces the relative abundance of a large proportion of bacterial taxa in Ae. aegypti adults, that is in accordance with the known pathogen-blocking effects of Wolbachia on a variety of bacteria and viruses. In adults, several of the most abundant bacterial genera were found to undergo significant shifts in relative abundance. However, the genera showing the greatest changes in relative abundance in Wolbachia-infected adults represented a low proportion of the total microbiome. In addition, there was little effect of Wolbachia infection on the relative abundance of bacterial taxa in larvae, or on species diversity (accounting for species richness and evenness together) detected in adults or larvae. These results offer insight into the effects of Wolbachia on the Ae. aegypti microbiome in a native setting, an important consideration for field releases of Wolbachia into the population.


Assuntos
Aedes/microbiologia , Wolbachia/fisiologia , Animais , Austrália , Biodiversidade , Feminino , Genes Bacterianos , Sequenciamento de Nucleotídeos em Larga Escala , Larva/microbiologia , Microbiota/genética , Filogenia , RNA Ribossômico 16S/genética , Wolbachia/classificação
13.
J Invertebr Pathol ; 143: 18-25, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27871813

RESUMO

Animals experience near constant infection with microorganisms. A significant proportion of these microbiota reside in the alimentary tract. There is a growing appreciation for the roles gut microbiota play in host biology. The gut microbiota of insects, for example, have been shown to help the host overcome pathogen infection either through direct competition or indirectly by stimulating host immunity. These defenses may also be supplemented by coinfecting maternally inherited microbes such as Wolbachia. The presence of Wolbachia in a host can delay and/or reduce death caused by RNA viruses. Whether the gut microbiota of the host interacts with Wolbachia, or vice versa, the precise role of Wolbachia in antiviral protection is not known. In this study, we used 16S rDNA sequencing to characterise changes in gut microbiota composition in Drosophila melanogaster associated with Wolbachia infection and antibiotic treatment. We subsequently tested whether changes in gut composition via antibiotic treatment altered Wolbachia-mediated antiviral properties. We found that both antibiotics and Wolbachia significantly reduced the biodiversity of the gut microbiota without changing the total microbial load. We also showed that changing the gut microbiota composition with antibiotic treatment enhanced Wolbachia density but did not confer greater antiviral protection against Drosophila C virus to the host. We concluded there are significant interactions between Wolbachia and gut microbiota, but changing gut microbiota composition is not likely to be a means through which Wolbachia conveys antiviral protection to its host.


Assuntos
Drosophila melanogaster/microbiologia , Microbioma Gastrointestinal , Interações Hospedeiro-Patógeno , Viroses/microbiologia , Wolbachia , Animais , Antibacterianos/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Vírus de Insetos , Vírus de RNA
14.
PLoS Pathog ; 9(6): e1003459, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23825950

RESUMO

The bacterial endosymbiont Wolbachia pipientis protects its hosts from a range of pathogens by limiting their ability to form infections inside the insect. This "pathogen blocking" could be explained by innate immune priming by the symbiont, competition for host-derived resources between pathogens and Wolbachia, or the direct modification of the cell or cellular environment by Wolbachia. Recent comparative work in Drosophila and the mosquito Aedes aegypti has shown that an immune response is not required for pathogen blocking, implying that there must be an additional component to the mechanism. Here we have examined the involvement of cholesterol in pathogen blocking using a system of dietary manipulation in Drosophila melanogaster in combination with challenge by Drosophila C virus (DCV), a common fly pathogen. We observed that flies reared on cholesterol-enriched diets infected with the Wolbachia strains wMelPop and wMelCS exhibited reduced pathogen blocking, with viral-induced mortality occurring 2-5 days earlier than flies reared on Standard diet. This shift toward greater virulence in the presence of cholesterol also corresponded to higher viral copy numbers in the host. Interestingly, an increase in dietary cholesterol did not have an effect on Wolbachia density except in one case, but this did not directly affect the strength of pathogen blocking. Our results indicate that host cholesterol levels are involved with the ability of Wolbachia-infected flies to resist DCV infections, suggesting that cholesterol contributes to the underlying mechanism of pathogen blocking.


Assuntos
Aedes , Colesterol/farmacologia , Dicistroviridae/metabolismo , Gorduras na Dieta/farmacologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Wolbachia/fisiologia , Aedes/metabolismo , Aedes/microbiologia , Aedes/virologia , Animais , Colesterol/metabolismo , Gorduras na Dieta/metabolismo , Drosophila melanogaster , Interações Hospedeiro-Patógeno/fisiologia
15.
PLoS Pathog ; 8(2): e1002548, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22383881

RESUMO

The non-virulent Wolbachia strain wMel and the life-shortening strain wMelPop-CLA, both originally from Drosophila melanogaster, have been stably introduced into the mosquito vector of dengue fever, Aedes aegypti. Each of these Wolbachia strains interferes with viral pathogenicity and/or dissemination in both their natural Drosophila host and in their new mosquito host, and it has been suggested that this virus interference may be due to host immune priming by Wolbachia. In order to identify aspects of the mosquito immune response that might underpin virus interference, we used whole-genome microarrays to analyse the transcriptional response of A. aegypti to the wMel and wMelPop-CLA Wolbachia strains. While wMel affected the transcription of far fewer host genes than wMelPop-CLA, both strains activated the expression of some immune genes including anti-microbial peptides, Toll pathway genes and genes involved in melanization. Because the induction of these immune genes might be associated with the very recent introduction of Wolbachia into the mosquito, we also examined the same Wolbachia strains in their original host D. melanogaster. First we demonstrated that when dengue viruses were injected into D. melanogaster, virus accumulation was significantly reduced in the presence of Wolbachia, just as in A. aegypti. Second, when we carried out transcriptional analyses of the same immune genes up-regulated in the new heterologous mosquito host in response to Wolbachia we found no over-expression of these genes in D. melanogaster, infected with either wMel or wMelPop. These results reinforce the idea that the fundamental mechanism involved in viral interference in Drosophila and Aedes is not dependent on the up-regulation of the immune effectors examined, although it cannot be excluded that immune priming in the heterologous mosquito host might enhance the virus interference trait.


Assuntos
Antibiose/imunologia , Vírus da Dengue/imunologia , Dengue/imunologia , Imunidade Inata/fisiologia , Wolbachia/fisiologia , Aedes/genética , Aedes/imunologia , Aedes/microbiologia , Aedes/virologia , Animais , Antibiose/genética , Dengue/genética , Dengue/microbiologia , Vírus da Dengue/fisiologia , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Drosophila melanogaster/microbiologia , Drosophila melanogaster/virologia , Feminino , Perfilação da Expressão Gênica , Imunidade Inata/genética , Análise em Microsséries , Infecções por Rickettsiaceae/genética , Infecções por Rickettsiaceae/imunologia , Infecções por Rickettsiaceae/microbiologia , Infecções por Rickettsiaceae/virologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Wolbachia/imunologia
16.
Microb Ecol ; 67(1): 205-18, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24337107

RESUMO

The endosymbiont Wolbachia represents a promising method of dengue control, as it reduces the ability of the primary vector, the mosquito Aedes aegypti, to transmit viruses. When mosquitoes infected with the virulent Wolbachia strain wMelPop are fed non-human blood, there is a drastic reduction in mosquito fecundity and egg viability. Wolbachia has a reduced genome and is clearly dependent on its host for a wide range of nutritional needs. The fitness defects seen in wMelPop-infected A. aegypti could be explained by competition between the mosquito and the symbiont for essential blood meal nutrients, the profiles of which are suboptimal in non-human blood. Here, we examine cholesterol and amino acids as candidate molecules for competition, as they have critical roles in egg structural development and are known to vary between blood sources. We found that Wolbachia infection reduces total cholesterol levels in mosquitoes by 15-25%. We then showed that cholesterol supplementation of a rat blood meal did not improve fecundity or egg viability deficits. Conversely, amino acid supplementation of sucrose before and after a sheep blood meal led to statistically significant increases in fecundity of approximately 15-20 eggs per female and egg viability of 30-40%. This mosquito system provides the first empirical evidence of competition between Wolbachia and a host over amino acids and may suggest a general feature of Wolbachia-insect associations. These competitive processes could affect many aspects of host physiology and potentially mosquito fitness, a key concern for Wolbachia-based mosquito biocontrol.


Assuntos
Aedes/microbiologia , Aedes/fisiologia , Aminoácidos/metabolismo , Wolbachia/fisiologia , Animais , Colesterol/metabolismo , Feminino , Fertilidade , Humanos , Óvulo/fisiologia , Ratos , Ratos Endogâmicos Lew , Ovinos , Simbiose
17.
PLoS Negl Trop Dis ; 18(7): e0012305, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38976758

RESUMO

As Wolbachia pipientis is more widely being released into field populations of Aedes aegypti for disease control, the ability to select the appropriate strain for differing environments is increasingly important. A previous study revealed that longer-term quiescence in the egg phase reduced the fertility of mosquitoes, especially those harboring the wAlbB Wolbachia strain. This infertility was also associated with a greater biting rate. Here, we attempt to quantify the effect of this heightened biting behavior on the transmission potential of the dengue virus using a combination of assays for fitness, probing behavior, and vector competence, allowing repeat feeding, and incorporate these effects in a model of R0. We show that Wolbachia-infected infertile mosquitoes are more interested in feeding almost immediately after an initial blood meal relative to wild type and Wolbachia-infected fertile mosquitoes and that these differences continue for up to 8 days over the period we measured. As a result, the infertile Wolbachia mosquitoes have higher virus prevalence and loads than Wolbachia-fertile mosquitoes. We saw limited evidence of Wolbachia-mediated blocking in the disseminated tissue (legs) in terms of prevalence but did see reduced viral loads. Using a previously published estimate of the extrinsic incubation period, we demonstrate that the effect of repeat feeding/infertility is insufficient to overcome the effects of Wolbachia-mediated blocking on R0. These estimates are very conservative, however, and we posit that future studies should empirically measure EIP under a repeat feeding model. Our findings echo previous work where periods of extensive egg quiescence affected the reproductive success of Wolbachia-infected Ae. aegypti. Additionally, we show that increased biting behavior in association with this infertility in Wolbachia-infected mosquitoes may drive greater vector competence. These relationships require further exploration, given their ability to affect the success of field releases of Wolbachia for human disease reduction in drier climates where longer egg quiescence periods are expected.


Assuntos
Aedes , Vírus da Dengue , Dengue , Comportamento Alimentar , Mosquitos Vetores , Wolbachia , Aedes/microbiologia , Aedes/virologia , Aedes/fisiologia , Animais , Wolbachia/fisiologia , Vírus da Dengue/fisiologia , Mosquitos Vetores/microbiologia , Mosquitos Vetores/virologia , Mosquitos Vetores/fisiologia , Dengue/transmissão , Feminino , Carga Viral , Óvulo/virologia , Óvulo/microbiologia
18.
BMC Genomics ; 14: 20, 2013 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-23324387

RESUMO

BACKGROUND: The endosymbiont Wolbachia pipientis causes diverse and sometimes dramatic phenotypes in its invertebrate hosts. Four Wolbachia strains sequenced to date indicate that the constitution of the genome is dynamic, but these strains are quite divergent and do not allow resolution of genome diversification over shorter time periods. We have sequenced the genome of the strain wBol1-b, found in the butterfly Hypolimnas bolina, which kills the male offspring of infected hosts during embyronic development and is closely related to the non-male-killing strain wPip from Culex pipiens. RESULTS: The genomes of wBol1-b and wPip are similar in genomic organisation, sequence and gene content, but show substantial differences at some rapidly evolving regions of the genome, primarily associated with prophage and repetitive elements. We identified 44 genes in wBol1-b that do not have homologs in any previously sequenced strains, indicating that Wolbachia's non-core genome diversifies rapidly. These wBol1-b specific genes include a number that have been recently horizontally transferred from phylogenetically distant bacterial taxa. We further report a second possible case of horizontal gene transfer from a eukaryote into Wolbachia. CONCLUSIONS: Our analyses support the developing view that many endosymbiotic genomes are highly dynamic, and are exposed and receptive to exogenous genetic material from a wide range of sources. These data also suggest either that this bacterial species is particularly permissive for eukaryote-to-prokaryote gene transfers, or that these transfers may be more common than previously believed. The wBol1-b-specific genes we have identified provide candidates for further investigations of the genomic bases of phenotypic differences between closely-related Wolbachia strains.


Assuntos
Transferência Genética Horizontal , Genoma Bacteriano/genética , Wolbachia/genética , Adenosina Trifosfatases/genética , Animais , Proteínas de Bactérias/genética , Borboletas/microbiologia , Masculino , Proteínas de Membrana Transportadoras/genética , Anotação de Sequência Molecular , Filogenia , Canais de Translocação SEC , Proteínas SecA , Simbiose/genética
19.
Philos Trans R Soc Lond B Biol Sci ; 378(1873): 20220011, 2023 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-36744557

RESUMO

The geographical range of the mosquito vector for many human disease-causing viruses, Aedes aegypti, is expanding, in part owing to changing climate. The capacity of this species to adapt to thermal stress will affect its future distributions. It is unclear how much heritable genetic variation may affect the upper thermal limits of mosquito populations over the long term. Nor are the genetic pathways that confer thermal tolerance fully understood. In the short term, cells induce a plastic, protective response known as 'heat shock'. Using a physiological 'knockdown' assay, we investigated mosquito thermal tolerance to characterize the genetic architecture of the trait. While families representing the extreme ends of the distribution for knockdown time differed from one another, the trait exhibited low but non-zero broad-sense heritability. We then explored whether families representing thermal performance extremes differed in their heat shock response by measuring gene expression of heat shock protein-encoding genes Hsp26, Hsp83 and Hsp70. Contrary to prediction, the families with higher thermal tolerance demonstrated less Hsp expression. This pattern may indicate that other mechanisms of heat tolerance, rather than heat shock, may underpin the stress response, and the costly production of HSPs may instead signal poor adaptation. This article is part of the theme issue 'Infectious disease ecology and evolution in a changing world'.


Assuntos
Aedes , Proteínas de Choque Térmico , Proteínas de Insetos , Termotolerância , Animais , Aedes/genética , Expressão Gênica , Resposta ao Choque Térmico , Proteínas de Insetos/genética , Proteínas de Choque Térmico/genética
20.
PLoS Negl Trop Dis ; 17(9): e0011616, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37669272

RESUMO

Jamestown Canyon virus (JCV), a negative-sense arbovirus, is increasingly common in the upper Midwest of the USA. Transmitted by a range of mosquito genera, JCV's primary amplifying host is white-tailed deer. Aedes aegypti is responsible for transmitting various positive-sense viruses globally including dengue (DENV), Zika, chikungunya, and Yellow Fever. Ae. aegypti's distribution, once confined to the tropics, is expanding, in part due to climate change. Wolbachia, an insect endosymbiont, limits the replication of co-infecting viruses inside insects. The release and spread of the symbiont into Ae. aegypti populations have been effective in reducing transmission of DENV to humans, although the mechanism of Wolbachia-mediated viral blocking is still poorly understood. Here we explored JCV infection potential in Ae. aegypti, the nature of the vector's immune response, and interactions with Wolbachia infection. We show that Ae. aegypti is highly competent for JCV, which grows to high loads and rapidly reaches the saliva after an infectious blood meal. The mosquito immune system responds with strong induction of RNAi and JAK/STAT. Neither the direct effect of viral infection nor the energetic investment in immunity appears to affect mosquito longevity. Wolbachia infection blocked JCV only in the early stages of infection. Wolbachia-induced immunity was small compared to that of JCV, suggesting innate immune priming does not likely explain blocking. We propose two models to explain why Wolbachia's blocking of negative-sense viruses like JCV may be less than that of positive-sense viruses, relating to the slowdown of host protein synthesis and the triggering of interferon-like factors like Vago. In conclusion, we highlight the risk for increased human disease with the predicted future overlap of Ae. aegypti and JCV ranges. We suggest that with moderate Wolbachia-mediated blocking and distinct biology, negative-sense viruses represent a fruitful comparator model to other viruses for understanding blocking mechanisms in mosquitoes.


Assuntos
Aedes , Coinfecção , Cervos , Vírus da Encefalite da Califórnia , Wolbachia , Infecção por Zika virus , Zika virus , Animais , Humanos , Mosquitos Vetores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA