Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 388
Filter
Add more filters

Publication year range
1.
PLoS Pathog ; 20(6): e1012318, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38865374

ABSTRACT

Many plant arboviruses are persistently transmitted by piercing-sucking insect vectors. However, it remains largely unknown how conserved insect Toll immune response exerts antiviral activity and how plant viruses antagonize it to facilitate persistent viral transmission. Here, we discover that southern rice black-streaked dwarf virus (SRBSDV), a devastating planthopper-transmitted rice reovirus, activates the upstream Toll receptors expression but suppresses the downstream MyD88-Dorsal-defensin cascade, resulting in the attenuation of insect Toll immune response. Toll pathway-induced the small antibacterial peptide defensin directly interacts with viral major outer capsid protein P10 and thus binds to viral particles, finally blocking effective viral infection in planthopper vector. Furthermore, viral tubular protein P7-1 directly interacts with and promotes RING E3 ubiquitin ligase-mediated ubiquitinated degradation of Toll pathway adaptor protein MyD88 through the 26 proteasome pathway, finally suppressing antiviral defensin production. This virus-mediated attenuation of Toll antiviral immune response to express antiviral defensin ensures persistent virus infection without causing evident fitness costs for the insects. E3 ubiquitin ligase also is directly involved in the assembly of virus-induced tubules constructed by P7-1 to facilitate viral spread in planthopper vector, thereby acting as a pro-viral factor. Together, we uncover a previously unknown mechanism used by plant arboviruses to suppress Toll immune response through the ubiquitinated degradation of the conserved adaptor protein MyD88, thereby facilitating the coexistence of arboviruses with their vectors in nature.


Subject(s)
Arboviruses , Insect Vectors , Signal Transduction , Toll-Like Receptors , Animals , Arboviruses/immunology , Toll-Like Receptors/metabolism , Insect Vectors/virology , Insect Vectors/immunology , Plant Diseases/virology , Plant Diseases/immunology , Reoviridae/physiology , Reoviridae/immunology , Hemiptera/virology , Hemiptera/immunology , Oryza/virology , Oryza/immunology , Insect Proteins/metabolism , Immunity, Innate
2.
J Gen Virol ; 105(5)2024 May.
Article in English | MEDLINE | ID: mdl-38717918

ABSTRACT

The tomato spotted wilt virus (TSWV) is a member of the Tospoviridae family and has an negative/ambisense single-stranded RNA genome. Frankliniella occidentalis and F. intonsa are known to be dominant pests in Capsicum annuum (hot pepper) and can cause damage to the plant either directly by feeding, or indirectly by transmitting TSWV in a persistent and propagative manner, resulting in serious economic damage. This study compared the immune responses of two different thrips species against TSWV infection by transcriptome analysis, which then allowed the assessment of antiviral responses using RNA interference (RNAi). Both adult thrips shared about 90 % of the transcripts in non-viruliferous conditions. Most signal components of the immune pathways were shared by these two thrips species, and their expression levels fluctuated differentially in response to TSWV infection at early immature stages. The functional assays using RNAi treatments indicated that the Toll and JAK/STAT pathways were associated with the antiviral responses, but the IMD pathway was not. The upregulation of dorsal switch protein one supported its physiological role in recognizing TSWV infection and triggering the eicosanoid biosynthetic pathway, which mediates melanization and apoptosis in thrips. In addition, the signal components of the RNAi pathways fluctuated highly after TSWV infection. Individual RNAi treatments specific to the antiviral signalling and response components led to significant increases in the TSWV amount in the thrips, causing virus-induced mortality. These findings suggest that immune signalling pathways leading to antiviral responses are operating in the thrips to regulate TSWV litres to prevent a fatal viral overload. This study also indicates the differential antiviral responses between the TSWV-transmitting F. occidentalis and F. intonsa.


Subject(s)
Plant Diseases , Thysanoptera , Tospovirus , Tospovirus/immunology , Tospovirus/physiology , Tospovirus/genetics , Animals , Thysanoptera/virology , Thysanoptera/immunology , Plant Diseases/virology , Plant Diseases/immunology , Capsicum/virology , Capsicum/immunology , Virus Replication , RNA Interference , Insect Vectors/virology , Insect Vectors/immunology , Gene Expression Profiling , Signal Transduction
3.
PLoS Pathog ; 17(9): e1009870, 2021 09.
Article in English | MEDLINE | ID: mdl-34473801

ABSTRACT

As mosquito females require a blood meal to reproduce, they can act as vectors of numerous pathogens, such as arboviruses (e.g. Zika, dengue and chikungunya viruses), which constitute a substantial worldwide public health burden. In addition to blood meals, mosquito females can also take sugar meals to get carbohydrates for their energy reserves. It is now recognised that diet is a key regulator of health and disease outcome through interactions with the immune system. However, this has been mostly studied in humans and model organisms. So far, the impact of sugar feeding on mosquito immunity and in turn, how this could affect vector competence for arboviruses has not been explored. Here, we show that sugar feeding increases and maintains antiviral immunity in the digestive tract of the main arbovirus vector Aedes aegypti. Our data demonstrate that the gut microbiota does not mediate the sugar-induced immunity but partly inhibits it. Importantly, sugar intake prior to an arbovirus-infected blood meal further protects females against infection with arboviruses from different families. Sugar feeding blocks arbovirus initial infection and dissemination from the gut and lowers infection prevalence and intensity, thereby decreasing the transmission potential of female mosquitoes. Finally, we show that the antiviral role of sugar is mediated by sugar-induced immunity. Overall, our findings uncover a crucial role of sugar feeding in mosquito antiviral immunity which in turn decreases vector competence for arboviruses. Since Ae. aegypti almost exclusively feed on blood in some natural settings, our findings suggest that this lack of sugar intake could increase the spread of mosquito-borne arboviral diseases.


Subject(s)
Aedes/virology , Arbovirus Infections , Diet , Insect Vectors/virology , Intestines/immunology , Aedes/immunology , Animals , Arboviruses , Insect Vectors/immunology , Sugars
4.
PLoS Pathog ; 17(3): e1009347, 2021 03.
Article in English | MEDLINE | ID: mdl-33647067

ABSTRACT

In the field, many insect-borne crop viral diseases are more suitable for maintenance and spread in hot-temperature areas, but the mechanism remains poorly understood. The epidemic of a planthopper (Sogatella furcifera)-transmitted rice reovirus (southern rice black-streaked dwarf virus, SRBSDV) is geographically restricted to southern China and northern Vietnam with year-round hot temperatures. Here, we reported that two factors of endoplasmic reticulum-associated degradation (ERAD) machinery, the heat shock protein DnaJB11 and ER membrane protein BAP31, were activated by viral infection to mediate the adaptation of S. furcifera to high temperatures. Infection and transmission efficiencies of SRBSDV by S. furcifera increased with the elevated temperatures. We observed that high temperature (35°C) was beneficial for the assembly of virus-containing tubular structures formed by nonstructural protein P7-1 of SRBSDV, which facilitates efficient viral transmission by S. furcifera. Both DnaJB11 and BAP31 competed to directly bind to the tubule protein P7-1 of SRBSDV; however, DnaJB11 promoted whereas BAP31 inhibited P7-1 tubule assembly at the ER membrane. Furthermore, the binding affinity of DnaJB11 with P7-1 was stronger than that of BAP31 with P7-1. We also revealed that BAP31 negatively regulated DnaJB11 expression through their direct interaction. High temperatures could significantly upregulate DnaJB11 expression but inhibit BAP31 expression, thereby strongly facilitating the assembly of abundant P7-1 tubules. Taken together, we showed that a new temperature-dependent protein quality control pathway in the ERAD machinery has evolved for strong activation of DnaJB11 for benefiting P7-1 tubules assembly to support efficient transmission of SRBSDV in high temperatures. We thus deduced that ERAD machinery has been hitchhiked by insect-borne crop viruses to enhance their transmission in tropical climates.


Subject(s)
Hot Temperature/adverse effects , Insect Vectors/virology , Plant Diseases/virology , Reoviridae/immunology , Animals , Endoplasmic Reticulum-Associated Degradation/immunology , Insect Vectors/immunology , Orthoreovirus/pathogenicity
5.
PLoS Pathog ; 16(8): e1008754, 2020 08.
Article in English | MEDLINE | ID: mdl-32776975

ABSTRACT

Arbovirus infection of Aedes aegypti salivary glands (SGs) determines transmission. However, there is a dearth of knowledge on SG immunity. Here, we characterized SG immune response to dengue, Zika and chikungunya viruses using high-throughput transcriptomics. We also describe a transcriptomic response associated to apoptosis, blood-feeding and lipid metabolism. The three viruses differentially regulate components of Toll, Immune deficiency (IMD) and c-Jun N- terminal Kinase (JNK) pathways. However, silencing of the Toll and IMD pathway components showed variable effects on SG infection by each virus. In contrast, regulation of the JNK pathway produced consistent responses in both SGs and midgut. Infection by the three viruses increased with depletion of the activator Kayak and decreased with depletion of the negative regulator Puckered. Virus-induced JNK pathway regulates the complement factor, Thioester containing protein-20 (TEP20), and the apoptosis activator, Dronc, in SGs. Individual and co-silencing of these genes demonstrate their antiviral effects and that both may function together. Co-silencing either TEP20 or Dronc with Puckered annihilates JNK pathway antiviral effect. Upon infection in SGs, TEP20 induces antimicrobial peptides (AMPs), while Dronc is required for apoptosis independently of TEP20. In conclusion, we revealed the broad antiviral function of JNK pathway in SGs and showed that it is mediated by a TEP20 complement and Dronc-induced apoptosis response. These results expand our understanding of the immune arsenal that blocks arbovirus transmission.


Subject(s)
Aedes/immunology , Apoptosis , Chikungunya Fever/immunology , Complement System Proteins/immunology , Dengue/immunology , MAP Kinase Signaling System , Salivary Glands/immunology , Zika Virus Infection/immunology , Aedes/virology , Animals , Chikungunya Fever/metabolism , Chikungunya Fever/prevention & control , Chikungunya Fever/virology , Chikungunya virus/immunology , Complement System Proteins/metabolism , Dengue/metabolism , Dengue/prevention & control , Dengue/virology , Dengue Virus/immunology , Female , Host-Pathogen Interactions , Insect Proteins/genetics , Insect Proteins/metabolism , Insect Vectors/immunology , Insect Vectors/virology , Salivary Glands/virology , Transcriptome , Virus Replication , Zika Virus/immunology , Zika Virus Infection/metabolism , Zika Virus Infection/prevention & control , Zika Virus Infection/virology
6.
Methods ; 183: 38-42, 2020 11 01.
Article in English | MEDLINE | ID: mdl-31654749

ABSTRACT

During infection, RNA viruses can produce two types of virus-derived small RNAs (vsRNAs), small interfering RNA (siRNA) and microRNA (miRNA), that play a key role in RNA silencing-mediated antiviral mechanisms in various hosts by associating with different Argonaute (Ago) proteins. Ago1 has been widely identified as an essential part of the miRNA pathway, while Ago2 is required for the siRNA pathway. Thus, analysis of the interaction between vsRNAs and Ago proteins can provide a clue about which pathway the vsRNA may be involved in. In this study, using rice stripe virus (RSV)-small brown planthoppers (Laodelphax striatellus, Fallen) as an infection model, the interactions of eight vsRNAs derived from four viral genomic RNA fragments and Ago1 or Ago2 were detected via the RNA immunoprecipitation (RIP) method. vsRNA4-1 and vsRNA4-2 derived from RSV RNA4 were significantly enriched in Ago1-immunoprecipitated complexes, whereas vsRNA2-1 and vsRNA3-2 seemed enriched in Ago2-immunoprecipitated complexes. vsRNA1-2 and vsRNA2-2 were detected in both of the two Ago-immunoprecipitated complexes. In contrast, vsRNA1-1 and vsRNA3-1 did not accumulate in either Ago1- or Ago2-immunoprecipitated complexes, indicating that regulatory pathways other than miRNA or siRNA pathways might be employed. In addition, two conserved L. striatellus miRNAs were analysed via the RIP method. Both miRNAs accumulated in Ago1-immunoprecipitated complexes, which was consistent with previous studies, suggesting that our experimental system can be widely used. In conclusion, our study provides an accurate and convenient detection system to determine the potential pathway of vsRNAs, and this method may also be suitable for studying other sRNAs.


Subject(s)
Argonaute Proteins/isolation & purification , Hemiptera/genetics , Immunoprecipitation/methods , Insect Vectors/genetics , RNA, Viral/isolation & purification , Animals , Argonaute Proteins/immunology , Argonaute Proteins/metabolism , Hemiptera/immunology , Hemiptera/metabolism , Hemiptera/virology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Insect Vectors/immunology , Insect Vectors/metabolism , Insect Vectors/virology , MicroRNAs/genetics , MicroRNAs/immunology , MicroRNAs/metabolism , Oryza , Plant Diseases/genetics , Plant Diseases/virology , RNA, Small Interfering/immunology , RNA, Small Interfering/isolation & purification , RNA, Small Interfering/metabolism , RNA, Viral/immunology , RNA, Viral/metabolism , Tenuivirus/genetics , Tenuivirus/immunology , Tenuivirus/pathogenicity
7.
PLoS Pathog ; 14(3): e1006879, 2018 03.
Article in English | MEDLINE | ID: mdl-29494679

ABSTRACT

Wolbachia is currently being developed as a novel tool to block the transmission of dengue viruses (DENV) by Aedes aegypti. A number of mechanisms have been proposed to explain the DENV-blocking phenotype in mosquitoes, including competition for fatty acids like cholesterol, manipulation of host miRNAs and upregulation of innate immune pathways in the mosquito. We examined the various stages in the DENV infection process to better understand the mechanism of Wolbachia-mediated virus blocking (WMVB). Our results suggest that infection with Wolbachia does not inhibit DENV binding or cell entry, but reduces virus replication. In contrast to a previous report, we also observed a similar reduction in replication of West Nile virus (WNV). This reduced replication is associated with rapid viral RNA degradation in the cytoplasm. We didn't find a role for host miRNAs in WMVB. Further analysis showed that the 3' end of the virus subgenomic RNA was protected and accumulated over time suggesting that the degradation is XRN1-mediated. We also found that sub genomic flavivirus RNA accumulation inactivated XRN1 in mosquito cells in the absence of Wolbachia and led to enhancement of RNA degradation in its presence. Depletion of XRN1 decreased WMVB which was associated with a significant increase in DENV RNA. We also observed that WMVB is influenced by virus MOI and rate of virus replication. A comparatively elevated blocking was observed for slowly replicating DENV, compared to WNV. Similar results were obtained while analysing different DENV serotypes.


Subject(s)
Aedes/immunology , Dengue/prevention & control , Exoribonucleases/metabolism , Microtubule-Associated Proteins/metabolism , RNA, Viral/metabolism , Virus Replication , West Nile Fever/prevention & control , Wolbachia/physiology , Aedes/metabolism , Aedes/microbiology , Aedes/virology , Animals , Dengue/immunology , Dengue/virology , Dengue Virus/physiology , Exoribonucleases/genetics , Humans , Insect Vectors/immunology , Insect Vectors/microbiology , Insect Vectors/virology , MicroRNAs , Microtubule-Associated Proteins/genetics , RNA, Viral/genetics , Symbiosis , West Nile Fever/immunology , West Nile Fever/virology , West Nile virus/physiology
8.
Int J Mol Sci ; 21(15)2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32751625

ABSTRACT

Many pathogens are transmitted by tick bites, including Anaplasma spp., Ehrlichia spp., Rickettsia spp., Babesia and Theileria sensu stricto species. These pathogens cause infectious diseases both in animals and humans. Different types of immune effector mechanisms could be induced in hosts by these microorganisms, triggered either directly by pathogen-derived antigens or indirectly by molecules released by host cells binding to these antigens. The components of innate immunity, such as natural killer cells, complement proteins, macrophages, dendritic cells and tumor necrosis factor alpha, cause a rapid and intense protection for the acute phase of infectious diseases. Moreover, the onset of a pro-inflammatory state occurs upon the activation of the inflammasome, a protein scaffold with a key-role in host defense mechanism, regulating the action of caspase-1 and the maturation of interleukin-1ß and IL-18 into bioactive molecules. During the infection caused by different microbial agents, very similar profiles of the human innate immune response are observed including secretion of IL-1α, IL-8, and IFN-α, and suppression of superoxide dismutase, IL-1Ra and IL-17A release. Innate immunity is activated immediately after the infection and inflammasome-mediated changes in the pro-inflammatory cytokines at systemic and intracellular levels can be detected as early as on days 2-5 after tick bite. The ongoing research field of "inflammasome biology" focuses on the interactions among molecules and cells of innate immune response that could be responsible for triggering a protective adaptive immunity. The knowledge of the innate immunity mechanisms, as well as the new targets of investigation arising by bioinformatics analysis, could lead to the development of new methods of emergency diagnosis and prevention of tick-borne infections.


Subject(s)
Immunity, Innate , Insect Vectors/immunology , Tick-Borne Diseases/immunology , Ticks/pathogenicity , Anaplasma/pathogenicity , Animals , Babesia/pathogenicity , Ehrlichia/pathogenicity , Humans , Insect Vectors/pathogenicity , Rickettsia/pathogenicity , Theileria/pathogenicity , Tick-Borne Diseases/transmission , Ticks/microbiology
9.
PLoS Pathog ; 13(7): e1006535, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28753642

ABSTRACT

Globally re-emerging dengue viruses are transmitted from human-to-human by Aedes mosquitoes. While viral determinants of human pathogenicity have been defined, there is a lack of knowledge of how dengue viruses influence mosquito transmission. Identification of viral determinants of transmission can help identify isolates with high epidemiological potential. Additionally, mechanistic understanding of transmission will lead to better understanding of how dengue viruses harness evolution to cycle between the two hosts. Here, we identified viral determinants of transmission and characterized mechanisms that enhance production of infectious saliva by inhibiting immunity specifically in salivary glands. Combining oral infection of Aedes aegypti mosquitoes and reverse genetics, we identified two 3' UTR substitutions in epidemic isolates that increased subgenomic flaviviral RNA (sfRNA) quantity, infectious particles in salivary glands and infection rate of saliva, which represents a measure of transmission. We also demonstrated that various 3'UTR modifications similarly affect sfRNA quantity in both whole mosquitoes and human cells, suggesting a shared determinism of sfRNA quantity. Furthermore, higher relative quantity of sfRNA in salivary glands compared to midgut and carcass pointed to sfRNA function in salivary glands. We showed that the Toll innate immune response was preferentially inhibited in salivary glands by viruses with the 3'UTR substitutions associated to high epidemiological fitness and high sfRNA quantity, pointing to a mechanism for higher saliva infection rate. By determining that sfRNA is an immune suppressor in a tissue relevant to mosquito transmission, we propose that 3'UTR/sfRNA sequence evolution shapes dengue epidemiology not only by influencing human pathogenicity but also by increasing mosquito transmission, thereby revealing a viral determinant of epidemiological fitness that is shared between the two hosts.


Subject(s)
Aedes/immunology , Aedes/virology , Dengue Virus/physiology , Dengue/transmission , Insect Vectors/immunology , Insect Vectors/virology , Animals , Dengue/virology , Dengue Virus/genetics , Humans , RNA, Viral/genetics , RNA, Viral/metabolism , Salivary Glands/immunology , Salivary Glands/virology , Virus Replication
10.
FASEB J ; 32(9): 4862-4877, 2018 09.
Article in English | MEDLINE | ID: mdl-29608332

ABSTRACT

Immune response of insect vectors to transmitted pathogens or insect hosts against parasites are well studied, whereas the mechanism of tripartite interactions remains elusive. In this study, we investigated the immune interactions of the vector beetle Monochamus alternatus ( Ma) to the devastating plant parasitic nematode Bursaphelenchus xylophilus ( Bx) and the insect parasitic nematode Howardula phyllotretae ( Hp). We report the unique immune mechanism by which the vector beetle tolerates many devastating Bx in its trachea, yet that immune tolerance is compromised by the parasitic nematode Hp. Contact with either nematode species triggers epithelial reactive oxygen species (ROS) production in Ma. Only the entry of Bx, not Hp, infection, induces increased expression of antioxidative genes, through which the ROS levels are balanced in the trachea of beetles. Furthermore, we found that up-regulation of antioxidative genes was induced by the interaction of Toll receptors. In contrast, beetles infected by Hp retain high levels of oxidative stress and melanization in trachea, and as a result, decrease Bx loading. This study highlights the role of Toll receptors in mediating the activation of antioxidative genes in immune tolerance to plant parasitic nematodes, and suggests the use of insect parasites as a biologic control.-Zhou, J., Zhao, L.-L., Yu, H.-Y., Wang, Y.-H., Zhang, W., Hu, S.-N., Zou, Z., Sun, J.-H. Immune tolerance of vector beetle to its partner plant parasitic nematode modulated by its insect parasitic nematode.


Subject(s)
Immune Tolerance/immunology , Insect Vectors/immunology , Larva/immunology , Nematoda/metabolism , Tylenchida/metabolism , Animals , Coleoptera , Nematoda/immunology , Toll-Like Receptors/immunology , Tylenchida/immunology
11.
J Proteome Res ; 17(9): 2995-3011, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30106293

ABSTRACT

Huanglongbing (HLB), also known as citrus greening disease, is the most serious disease of citrus plants. It is associated with the Gram-negative bacterium ' Candidatus Liberibacter asiaticus' ( CLas), which is transmitted between host plants by the hemipteran insect vector Diaphorina citri in a circulative, propagative manner involving specific interactions with various insect tissues including the hemolymph, fluid that occupies the body cavity akin to insect blood. High resolution quantitative mass spectrometry was performed to investigate the effect of CLas exposure on D. citri hemolymph at the proteome level. In contrast to the broad proteome effects on hundreds of proteins and a diverse array of metabolic pathways previously reported in gut and whole insect proteome analyses, the effect of CLas on the hemolymph was observed to be highly specific, restricted to key immunity and metabolism pathways, and lower in magnitude than that previously observed in the whole insect body and gut. Vitellogenins were abundantly expressed and CLas-responsive. Gene-specific RNA expression analysis suggests that these proteins are expressed in both male and female insects and may have roles outside of reproductive vitellogenesis. Proteins for fatty acid synthesis were found to be up-regulated, along with metabolic proteins associated with energy production, supported at the organismal level by the previously published observation that D. citri individuals experience a higher level of hunger when reared on CLas-infected plants. Prediction of post-translational modifications identified hemolymph proteins with phosphorylation and acetylation upon CLas exposure. Proteins derived from the three most prominent bacterial endosymbionts of the psyllid were also detected in the hemolymph, and several of these have predicted secretion signals. A DNAK protein, the bacterial HSP70, detected in the hemolymph expressed from Wolbachia pipientis was predicted to encode a eukaryotic nuclear localization signal. Taken together, these data show specific changes to immunity and metabolism in D. citri hemolymph involving host and endosymbiont proteins. These data provide a novel context for proteomic changes seen in other D. citri tissues in response to CLas and align with organismal data on the effects of CLas on D. citri metabolism and reproduction.


Subject(s)
Bacterial Proteins/metabolism , Hemiptera/metabolism , Hemolymph/chemistry , Insect Proteins/metabolism , Protein Processing, Post-Translational , Proteome/metabolism , Rhizobiaceae/metabolism , Acetylation , Animals , Bacterial Proteins/classification , Bacterial Proteins/genetics , Citrus/parasitology , Energy Metabolism , Fatty Acids , Gene Ontology , Hemiptera/genetics , Hemiptera/immunology , Hemiptera/microbiology , Hemolymph/immunology , Hemolymph/metabolism , Hemolymph/microbiology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Insect Proteins/classification , Insect Proteins/genetics , Insect Proteins/immunology , Insect Vectors/genetics , Insect Vectors/immunology , Insect Vectors/metabolism , Insect Vectors/microbiology , Lipid Metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Molecular Sequence Annotation , Phosphorylation , Plant Diseases/parasitology , Proteome/classification , Proteome/genetics , Proteome/immunology , Proteomics/methods , Rhizobiaceae/genetics , Symbiosis/genetics , Symbiosis/immunology , Vitellogenins , Wolbachia/genetics , Wolbachia/metabolism
12.
Infect Immun ; 86(5)2018 05.
Article in English | MEDLINE | ID: mdl-29531134

ABSTRACT

Phytoplasmas are plant-pathogenic bacteria transmitted by hemipteran insects. The leafhopper Euscelidius variegatus is a natural vector of chrysanthemum yellows phytoplasma (CYp) and a laboratory vector of flavescence dorée phytoplasma (FDp). The two phytoplasmas induce different effects on this species: CYp slightly improves whereas FDp negatively affects insect fitness. To investigate the molecular bases of these different responses, transcriptome sequencing (RNA-seq) analysis of E. variegatus infected with either CYp or FDp was performed. The sequencing provided the first de novo transcriptome assembly for a phytoplasma vector and a starting point for further analyses on differentially regulated genes, mainly related to immune system and energy metabolism. Insect phenoloxidase activity, immunocompetence, and body pigmentation were measured to investigate the immune response, while respiration and movement rates were quantified to confirm the effects on energy metabolism. The activation of the insect immune response upon infection with FDp, which is not naturally transmitted by E. variegatus, confirmed that this bacterium is mostly perceived as a potential pathogen. Conversely, the acquisition of CYp, which is naturally transmitted by E. variegatus, seems to increase the insect fitness by inducing a prompt response to stress. This long-term relationship is likely to improve survival and dispersal of the infected insect, thus enhancing the opportunity of phytoplasma transmission.


Subject(s)
Chrysanthemum/microbiology , Hemiptera/immunology , Hemiptera/microbiology , Insect Vectors/immunology , Insect Vectors/microbiology , Phytoplasma/immunology , Phytoplasma/pathogenicity , Animals , Host-Pathogen Interactions
13.
Infect Immun ; 86(1)2018 01.
Article in English | MEDLINE | ID: mdl-29084898

ABSTRACT

Rickettsia species are obligate intracellular bacteria with both conserved and lineage-specific strategies for invading and surviving within eukaryotic cells. One variable component of Rickettsia biology involves arthropod vectors: for instance, typhus group rickettsiae are principally vectored by insects (i.e., lice and fleas), whereas spotted fever group rickettsiae are exclusively vectored by ticks. For flea-borne Rickettsia typhi, the etiological agent of murine typhus, research on vertebrate host biology is facilitated using cell lines and animal models. However, due to the lack of any stable flea cell line or a published flea genome sequence, little is known regarding R. typhi biology in flea vectors that, importantly, do not suffer lethality due to R. typhi infection. To address if fleas combat rickettsial infection, we characterized the cat flea (Ctenocephalides felis) innate immune response to R. typhi Initially, we determined that R. typhi infects Drosophila cells and increases antimicrobial peptide (AMP) gene expression, indicating immune pathway activation. While bioinformatics analysis of the C. felis transcriptome identified homologs to all of the Drosophila immune deficiency (IMD) and Toll pathway components, an AMP gene expression profile in Drosophila cells indicated IMD pathway activation upon rickettsial infection. Accordingly, we assessed R. typhi-mediated flea IMD pathway activation in vivo using small interfering RNA (siRNA)-mediated knockdown. Knockdown of Relish and Imd increased R. typhi infection levels, implicating the IMD pathway as a critical regulator of R. typhi burden in C. felis These data suggest that targeting the IMD pathway could minimize the spread of R. typhi, and potentially other human pathogens, vectored by fleas.


Subject(s)
Ctenocephalides/immunology , Flea Infestations/immunology , Rickettsia Infections/immunology , Rickettsia typhi/immunology , Signal Transduction/immunology , Siphonaptera/immunology , Adenosine Monophosphate/metabolism , Animals , Cats , Cell Line , Chlorocebus aethiops , Ctenocephalides/microbiology , Drosophila/microbiology , Flea Infestations/microbiology , Gene Expression/immunology , Immunity, Innate/immunology , Insect Vectors/immunology , Insect Vectors/microbiology , Siphonaptera/microbiology , Typhus, Endemic Flea-Borne/immunology , Typhus, Endemic Flea-Borne/microbiology , Vero Cells
14.
PLoS Pathog ; 12(2): e1005434, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26891349

ABSTRACT

Wolbachia pipientis is an endosymbiotic bacterium estimated to chronically infect between 40-75% of all arthropod species. Aedes aegypti, the principle mosquito vector of dengue virus (DENV), is not a natural host of Wolbachia. The transinfection of Wolbachia strains such as wAlbB, wMel and wMelPop-CLA into Ae. aegypti has been shown to significantly reduce the vector competence of this mosquito for a range of human pathogens in the laboratory. This has led to wMel-transinfected Ae. aegypti currently being released in five countries to evaluate its effectiveness to control dengue disease in human populations. Here we describe the generation of a superinfected Ae. aegypti mosquito line simultaneously infected with two avirulent Wolbachia strains, wMel and wAlbB. The line carries a high overall Wolbachia density and tissue localisation of the individual strains is very similar to each respective single infected parental line. The superinfected line induces unidirectional cytoplasmic incompatibility (CI) when crossed to each single infected parental line, suggesting that the superinfection would have the capacity to replace either of the single constituent infections already present in a mosquito population. No significant differences in fitness parameters were observed between the superinfected line and the parental lines under the experimental conditions tested. Finally, the superinfected line blocks DENV replication more efficiently than the single wMel strain when challenged with blood meals from viremic dengue patients. These results suggest that the deployment of superinfections could be used to replace single infections and may represent an effective strategy to help manage potential resistance by DENV to field deployments of single infected strains.


Subject(s)
Aedes/microbiology , Insect Vectors/immunology , Pest Control, Biological , Rickettsiaceae Infections/microbiology , Superinfection/microbiology , Wolbachia , Animals , Dengue/virology , Dengue Virus , Female , Humans , Pest Control, Biological/methods , Saliva/microbiology , Virus Replication
15.
PLoS Pathog ; 12(6): e1005676, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27310141

ABSTRACT

Dengue remains the most prevalent arthropod-borne viral disease in humans. While probing for blood vessels, Aedes aegypti and Ae. albopictus mosquitoes transmit the four serotypes of dengue virus (DENV1-4) by injecting virus-containing saliva into the skin. Even though arthropod saliva is known to facilitate transmission and modulate host responses to other pathogens, the full impact of mosquito saliva on dengue pathogenesis is still not well understood. Inoculating mice lacking the interferon-α/ß receptor intradermally with DENV revealed that mosquito salivary gland extract (SGE) exacerbates dengue pathogenesis specifically in the presence of enhancing serotype-cross-reactive antibodies-when individuals already carry an increased risk for severe disease. We further establish that SGE increases viral titers in the skin, boosts antibody-enhanced DENV infection of dendritic cells and macrophages in the dermis, and amplifies dendritic cell migration to skin-draining lymph nodes. We demonstrate that SGE directly disrupts endothelial barrier function in vitro and induces endothelial permeability in vivo in the skin. Finally, we show that surgically removing the site of DENV transmission in the skin after 4 hours rescued mice from disease in the absence of SGE, but no longer prevented lethal antibody-enhanced disease when SGE was present. These results indicate that SGE accelerates the dynamics of dengue pathogenesis after virus transmission in the skin and induces severe antibody-enhanced disease systemically. Our study reveals novel aspects of dengue pathogenesis and suggests that animal models of dengue and pre-clinical testing of dengue vaccines should consider mosquito-derived factors as well as enhancing antibodies.


Subject(s)
Antibody-Dependent Enhancement/immunology , Cell Movement , Culicidae/virology , Dengue/transmission , Endothelial Cells/virology , Insect Vectors/pathogenicity , Saliva/metabolism , Animals , Capillary Permeability , Chemotaxis, Leukocyte/immunology , Culicidae/metabolism , Dengue/immunology , Dengue Virus/immunology , Dengue Virus/pathogenicity , Disease Models, Animal , Endothelial Cells/immunology , Flow Cytometry , Insect Vectors/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Saliva/immunology , Saliva/virology , Skin/blood supply , Skin/immunology
16.
PLoS Biol ; 13(9): e1002256, 2015.
Article in English | MEDLINE | ID: mdl-26394127

ABSTRACT

A study of Anopheles gambiae mosquitoes shows that a molecule involved in defense against the malaria parasite also plays a role in male fertility, identifying a potential evolutionary trade-off between immunity and reproductive fitness. Read the Research Article.


Subject(s)
Anopheles/metabolism , Insect Proteins/metabolism , Spermatogenesis , Animals , Anopheles/immunology , Female , Fertility , Host-Parasite Interactions , Insect Vectors/immunology , Insect Vectors/metabolism , Male , Plasmodium/immunology , Spermatozoa/metabolism , Testis/metabolism
17.
PLoS Biol ; 13(9): e1002255, 2015.
Article in English | MEDLINE | ID: mdl-26394016

ABSTRACT

Thioester-containing protein 1 (TEP1) is a key immune factor that determines mosquito resistance to a wide range of pathogens, including malaria parasites. Here we report a new allele-specific function of TEP1 in male fertility. We demonstrate that during spermatogenesis TEP1 binds to and removes damaged cells through the same complement-like cascade that kills malaria parasites in the mosquito midgut. Further, higher fertility rates are mediated by an allele that renders the mosquito susceptible to Plasmodium. By elucidating the molecular and genetic mechanisms underlying TEP1 function in spermatogenesis, our study suggests that pleiotropic antagonism between reproduction and immunity may shape resistance of mosquito populations to malaria parasites.


Subject(s)
Anopheles/metabolism , Insect Proteins/metabolism , Spermatogenesis , Alleles , Animals , Animals, Genetically Modified , Anopheles/immunology , Female , Fertility , Gamma Rays , Genetic Pleiotropy , Host-Parasite Interactions , Insect Vectors/immunology , Insect Vectors/metabolism , Male , Plasmodium/immunology , Spermatozoa/metabolism , Testis/metabolism
18.
Parasitology ; 145(10): 1336-1353, 2018 09.
Article in English | MEDLINE | ID: mdl-29921334

ABSTRACT

In this review, we explore the state-of-the-art of sand fly relationships with microbiota, viruses and Leishmania, with particular emphasis on the vector immune responses. Insect-borne diseases are a major public health problem in the world. Phlebotomine sand flies are proven vectors of several aetiological agents including viruses, bacteria and the trypanosomatid Leishmania, which are responsible for diseases such as viral encephalitis, bartonellosis and leishmaniasis, respectively. All metazoans in nature coexist intimately with a community of commensal microorganisms known as microbiota. The microbiota has a fundamental role in the induction, maturation and function of the host immune system, which can modulate host protection from pathogens and infectious diseases. We briefly review viruses of public health importance present in sand flies and revisit studies done on bacterial and fungal gut contents of these vectors. We bring this information into the context of sand fly development and immune responses. We highlight the immunity mechanisms that the insect utilizes to survive the potential threats involved in these interactions and discuss the recently discovered complex interactions among microbiota, sand fly, Leishmania and virus. Additionally, some of the alternative control strategies that could benefit from the current knowledge are considered.


Subject(s)
Insect Vectors/immunology , Leishmania/physiology , Microbiota/immunology , Psychodidae/immunology , Animals , Humans , Insect Control/methods , Insect Vectors/microbiology , Insect Vectors/parasitology , Insect Vectors/virology , Leishmania/immunology , Leishmania/pathogenicity , Mice , Psychodidae/microbiology , Psychodidae/parasitology , Psychodidae/virology , Public Health
19.
Proc Natl Acad Sci U S A ; 112(2): E176-85, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25548172

ABSTRACT

Arboviruses are transmitted by mosquitoes and other arthropods to humans and animals. The risk associated with these viruses is increasing worldwide, including new emergence in Europe and the Americas. Anopheline mosquitoes are vectors of human malaria but are believed to transmit one known arbovirus, o'nyong-nyong virus, whereas Aedes mosquitoes transmit many. Anopheles interactions with viruses have been little studied, and the initial antiviral response in the midgut has not been examined. Here, we determine the antiviral immune pathways of the Anopheles gambiae midgut, the initial site of viral infection after an infective blood meal. We compare them with the responses of the post-midgut systemic compartment, which is the site of the subsequent disseminated viral infection. Normal viral infection of the midgut requires bacterial flora and is inhibited by the activities of immune deficiency (Imd), JAK/STAT, and Leu-rich repeat immune factors. We show that the exogenous siRNA pathway, thought of as the canonical mosquito antiviral pathway, plays no detectable role in antiviral defense in the midgut but only protects later in the systemic compartment. These results alter the prevailing antiviral paradigm by describing distinct protective mechanisms in different body compartments and infection stages. Importantly, the presence of the midgut bacterial flora is required for full viral infectivity to Anopheles, in contrast to malaria infection, where the presence of the midgut bacterial flora is required for protection against infection. Thus, the enteric flora controls a reciprocal protection tradeoff in the vector for resistance to different human pathogens.


Subject(s)
Anopheles/immunology , Anopheles/virology , Arboviruses/immunology , Arboviruses/pathogenicity , Alphavirus Infections/immunology , Alphavirus Infections/transmission , Animals , Anopheles/genetics , Arbovirus Infections/immunology , Arbovirus Infections/transmission , Arboviruses/genetics , Digestive System/immunology , Digestive System/microbiology , Digestive System/virology , Female , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Insect Vectors/genetics , Insect Vectors/immunology , Insect Vectors/virology , Janus Kinases/immunology , Microbiota , O'nyong-nyong Virus/genetics , O'nyong-nyong Virus/immunology , O'nyong-nyong Virus/pathogenicity , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , RNA Interference , RNA, Small Interfering/genetics , STAT Transcription Factors/immunology , Signal Transduction/immunology
20.
PLoS Pathog ; 11(12): e1005306, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26633695

ABSTRACT

Nucleotide variation patterns across species are shaped by the processes of natural selection, including exposure to environmental pathogens. We examined patterns of genetic variation in two sister species, Anopheles gambiae and Anopheles coluzzii, both efficient natural vectors of human malaria in West Africa. We used the differentiation signature displayed by a known coordinate selective sweep of immune genes APL1 and TEP1 in A. coluzzii to design a population genetic screen trained on the sweep, classified a panel of 26 potential immune genes for concordance with the signature, and functionally tested their immune phenotypes. The screen results were strongly predictive for genes with protective immune phenotypes: genes meeting the screen criteria were significantly more likely to display a functional phenotype against malaria infection than genes not meeting the criteria (p = 0.0005). Thus, an evolution-based screen can efficiently prioritize candidate genes for labor-intensive downstream functional testing, and safely allow the elimination of genes not meeting the screen criteria. The suite of immune genes with characteristics similar to the APL1-TEP1 selective sweep appears to be more widespread in the A. coluzzii genome than previously recognized. The immune gene differentiation may be a consequence of adaptation of A. coluzzii to new pathogens encountered in its niche expansion during the separation from A. gambiae, although the role, if any of natural selection by Plasmodium is unknown. Application of the screen allowed identification of new functional immune factors, and assignment of new functions to known factors. We describe biochemical binding interactions between immune proteins that underlie functional activity for malaria infection, which highlights the interplay between pathogen specificity and the structure of immune complexes. We also find that most malaria-protective immune factors display phenotypes for either human or rodent malaria, with broad specificity a rarity.


Subject(s)
Anopheles/genetics , Anopheles/immunology , Insect Vectors/genetics , Insect Vectors/immunology , Animals , Base Sequence , Evolution, Molecular , Genes, Insect/immunology , Genetic Variation , Insect Proteins/genetics , Insect Proteins/immunology , Malaria/transmission , Mice , Molecular Sequence Data , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL