Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 260
Filtrar
1.
Curr Biol ; 34(13): R618-R620, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38981424

RESUMO

Viral infection causes an increase in age-related intestinal pathologies. New research finds that oral viral infection leads to intestinal stem-cell proliferation and a decrease in lifespan in Drosophila melanogaster that depends on Sting-NF-κB signaling.


Assuntos
Envelhecimento , Drosophila melanogaster , NF-kappa B , Transdução de Sinais , Animais , NF-kappa B/metabolismo , Drosophila melanogaster/virologia , Drosophila melanogaster/fisiologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Intestinos/virologia , Viroses/metabolismo , Viroses/virologia , Viroses/imunologia
2.
Dev Comp Immunol ; 159: 105224, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38969190

RESUMO

Stimulator of interferon genes (STING) mediates innate immune response upon binding to cyclic GMP-AMP (cGAMP). It recruits tank-binding kinase 1 (TBK1) and transcription factor interferon regulatory factor 3 (IRF3) through its C-terminal tail and facilitates TBK1-dependent phosphorylation of IRF3 via forming STING polymers in mammalian cells. However, the mechanism behind STING-mediated activation of NF-κB transcription factor, Relish, in insect cells is unknown. Our study revealed that insect STING formed oligomers and the cryptic RIP homotypic interaction motif (cRHIM) was required for its oligomerization and its anti-viral functions. Cells expressing cRHIM-deficient mutants exhibited lower levels of anti-viral molecules, higher viral load after viral infection and weak activation of Relish. Moreover, we observed that under cGAMP stimulation, insect STING interacted with IMD, and deletion of the cRHIM motif on either protein prevented this interaction. Finally, we demonstrated that cGAMP enhanced the amyloid-like property of insect STING aggregates by ThT staining. In summary, our research showed that insect STING employed a homotypic motif to form intermolecular interactions that are essential for its antiviral signaling.


Assuntos
Imunidade Inata , Fator Regulador 3 de Interferon , Proteínas de Membrana , Transdução de Sinais , Animais , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Transdução de Sinais/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Nucleotídeos Cíclicos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Motivos de Aminoácidos/genética , Humanos , Linhagem Celular , Ligação Proteica , Fosforilação , Multimerização Proteica , Drosophila melanogaster/imunologia , Drosophila melanogaster/virologia
3.
Curr Biol ; 34(13): 2785-2800.e7, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38823381

RESUMO

Host-microbe interactions influence intestinal stem cell (ISC) activity to modulate epithelial turnover and composition. Here, we investigated the functional impacts of viral infection on intestinal homeostasis and the mechanisms by which viral infection alters ISC activity. We report that Drosophila A virus (DAV) infection disrupts intestinal homeostasis in Drosophila by inducing sustained ISC proliferation, resulting in intestinal dysplasia, loss of gut barrier function, and reduced lifespan. We found that additional viruses common in laboratory-reared Drosophila also promote ISC proliferation. The mechanism of DAV-induced ISC proliferation involves progenitor-autonomous epidermal growth factor receptor (EGFR) signaling, c-Jun N-terminal kinase (JNK) activity in enterocytes, and requires Sting-dependent nuclear factor κB (NF-κB) (Relish) activity. We further demonstrate that activating Sting-Relish signaling is sufficient to induce ISC proliferation, promote intestinal dysplasia, and reduce lifespan in the absence of infection. Our results reveal that viral infection can significantly disrupt intestinal physiology, highlight a novel role for Sting-Relish signaling, and support a role for viral infection in aging.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Homeostase , Intestinos , Proteínas de Membrana , NF-kappa B , Transdução de Sinais , Animais , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , NF-kappa B/metabolismo , Drosophila melanogaster/virologia , Drosophila melanogaster/fisiologia , Intestinos/virologia , Células-Tronco/virologia , Células-Tronco/metabolismo , Proliferação de Células , Fatores de Transcrição
4.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167324, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38925484

RESUMO

Responsible for COVID-19, SARS-CoV-2 is a coronavirus in which contagious variants continue to appear. Therefore, some population groups have demonstrated greater susceptibility to contagion and disease progression. For these reasons, several researchers have been studying the SARS-CoV-2/human interactome to understand the pathophysiology of COVID-19 and develop new pharmacological strategies. D. melanogaster is a versatile animal model with approximately 90 % human protein orthology related to SARS-CoV-2/human interactome and is widely used in metabolic studies. In this context, our work assessed the potential interaction between human proteins (ZNF10, NUP88, BCL2L1, UBC9, and RBX1) and their orthologous proteins in D. melanogaster (gl, Nup88, Buffy, ubc9, and Rbx1a) with proteins from SARS-CoV-2 (nsp3, nsp9, E, ORF7a, N, and ORF10) using computational approaches. Our results demonstrated that all the proteins have the potential to interact, and we compared the binding sites between humans and fruit flies. The stability and consistency in the structure of the gl_nsp3 complex, specifically, could be crucial for its specific biological functions. Lastly, to enhance the understanding of the influence of host factors on coronavirus infection, we also analyse the mRNA expression of the five genes (mbo, gl, lwr, Buffy, and Roc1a) responsible for encoding the fruit fly proteins. Briefly, we demonstrated that those genes were differentially regulated according to diets, sex, and age. Two groups showed higher positive gene regulation than others: females in the HSD group and males in the aging group, which could imply a higher virus-host susceptibility. Overall, while preliminary, our work contributes to the understanding of host defense mechanisms and potentially identifies candidate proteins and genes for in vivo viral studies against SARS-CoV-2.


Assuntos
COVID-19 , Drosophila melanogaster , RNA Mensageiro , SARS-CoV-2 , SARS-CoV-2/metabolismo , SARS-CoV-2/genética , Animais , Humanos , COVID-19/virologia , COVID-19/metabolismo , COVID-19/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/virologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Feminino , Masculino , Interações Hospedeiro-Patógeno/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética
5.
Nat Rev Microbiol ; 22(8): 455, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38877320
6.
Proc Biol Sci ; 291(2023): 20240518, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38747703

RESUMO

Drosophila remains a pre-eminent insect model system for host-virus interaction, but the host range and fitness consequences of the drosophilid virome are poorly understood. Metagenomic studies have reported approximately 200 viruses associated with Drosophilidae, but few isolates are available to characterize the Drosophila immune response, and most characterization has relied on injection and systemic infection. Here, we use a more natural infection route to characterize the fitness effects of infection and to study a wider range of viruses. We exposed laboratory Drosophila melanogaster to 23 naturally occurring viruses from wild-collected drosophilids. We recorded transmission rates along with two components of female fitness: survival and the lifetime number of adult offspring produced. Nine different viruses transmitted during contact with laboratory D. melanogaster, although for the majority, rates of transmission were less than 20%. Five virus infections led to a significant decrease in lifespan (D. melanogaster Nora virus, D. immigrans Nora virus, Muthill virus, galbut virus and Prestney Burn virus), and three led to a reduction in the total number of offspring. Our findings demonstrate the utility of the Drosophila model for community-level studies of host-virus interactions, and suggest that viral infection could be a substantial fitness burden on wild flies.


Assuntos
Drosophila melanogaster , Longevidade , Animais , Drosophila melanogaster/virologia , Drosophila melanogaster/fisiologia , Feminino , Vírus de Insetos/fisiologia , Interações Hospedeiro-Patógeno
7.
Viruses ; 16(4)2024 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-38675904

RESUMO

Currently, insecticides that target nicotinic acetylcholine receptors (nAChR) are widely used. Studies on the sublethal effects of insecticides have found that they can affect the amount of virus in insects. The mechanism by which insecticides affect insect virus load remain unclear. Here, we show that nAChR targeting insecticide can affect viral replication through the immune deficiency (IMD) pathway. We demonstrate that a low dose of spinosad (6.8 ng/mL), acting as an antagonist to Drosophila melanogaster nicotinic acetylcholine receptor α6 (Dα6), significantly elevates Drosophila melanogaster sigmavirus (DMelSV) virus titers in adults of Drosophila melanogaster. Conversely, a high dose of spinosad (50 ng/mL), acting as an agonist to Dα6, substantially decreases viral load. This bidirectional regulation of virus levels is absent in Dα6-knockout flies, signifying the specificity of spinosad's action through Dα6. Furthermore, the knockdown of Dα6 results in decreased expression of genes in the IMD pathway, including dredd, imd, relish, and downstream antimicrobial peptide genes AttA and AttB, indicating a reduced innate immune response. Subsequent investigations reveal no significant difference in viral titers between relish mutant flies and Dα6-relish double mutants, suggesting that the IMD pathway's role in antiviral defense is dependent on Dα6. Collectively, our findings shed light on the intricate interplay between nAChR signaling and the IMD pathway in mediating antiviral immunity, highlighting the potential for nAChR-targeting compounds to inadvertently influence viral dynamics in insect hosts. This knowledge may inform the development of integrated pest management strategies that consider the broader ecological impact of insecticide use.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Receptores Nicotínicos , Animais , Drosophila melanogaster/imunologia , Drosophila melanogaster/virologia , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Combinação de Medicamentos , Macrolídeos/farmacologia , Replicação Viral/efeitos dos fármacos , Imunidade Inata , Inseticidas/farmacologia , Carga Viral/efeitos dos fármacos , Transdução de Sinais
8.
J Virol ; 98(5): e0017724, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38563731

RESUMO

Cactin, a highly conserved protein, plays a crucial role in various physiological processes in eukaryotes, including innate immunity. Recently, the function of Cactin in the innate immunity of Drosophila has been explored, revealing that Cactin regulates a non-canonical signaling pathway associated with the Toll and Imd pathways via the Cactin-Deaf1 axis. In addition, Cactin exhibits specific antiviral activity against the Drosophila C virus (DCV) in Drosophila, with an unknown mechanism. During DCV infection, it has been confirmed that the protein level and antiviral activity of Cactin are regulated by ubiquitination. However, the precise ubiquitination and deubiquitination mechanisms of Cactin in Drosophila remain unexplored. In this study, we identified ubiquitin-specific protease 14 (Usp14) as a major deubiquitinase for Cactin through comprehensive deubiquitinase screening. Our results demonstrate that Usp14 interacts with the C_Cactus domain of Cactin via its USP domain. Usp14 efficiently removes K48- and K63-linked polyubiquitin chains from Cactin, thereby preventing its degradation through the ubiquitin-proteasome pathway. Usp14 significantly inhibits DCV replication in Drosophila cells by stabilizing Cactin. Moreover, Usp14-deficient fruit flies exhibit increased susceptibility to DCV infection compared to wild-type flies. Collectively, our findings reveal the regulation of ubiquitination and antiviral activity of Cactin by the deubiquitinase Usp14, providing valuable insights into the modulation of Cactin-mediated antiviral activity in Drosophila.IMPORTANCEViral infections pose a severe threat to human health, marked by high pathogenicity and mortality rates. Innate antiviral pathways, such as Toll, Imd, and JAK-STAT, are generally conserved across insects and mammals. Recently, the multi-functionality of Cactin in innate immunity has been identified in Drosophila. In addition to regulating a non-canonical signaling pathway through the Cactin-Deaf1 axis, Cactin exhibits specialized antiviral activity against the Drosophila C virus (DCV) with an unknown mechanism. A previous study emphasized the significance of the Cactin level, regulated by the ubiquitin-proteasome pathway, in modulating antiviral signaling. However, the regulatory mechanisms governing Cactin remain unexplored. In this study, we demonstrate that Usp14 stabilizes Cactin by preventing its ubiquitination and subsequent degradation. Furthermore, Usp14 plays a crucial role in regulating the antiviral function mediated by Cactin. Therefore, our findings elucidate the regulatory mechanism of Cactin in Drosophila, offering a potential target for the prevention and treatment of viral infections.


Assuntos
Proteínas de Drosophila , Imunidade Inata , Ubiquitinação , Animais , Dicistroviridae/metabolismo , Drosophila/metabolismo , Drosophila melanogaster/virologia , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Transdução de Sinais , Ubiquitina Tiolesterase/metabolismo , Ubiquitina Tiolesterase/genética , Replicação Viral
9.
J Virol ; 97(10): e0111223, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37796127

RESUMO

IMPORTANCE: Arboviruses, particularly those transmitted by mosquitoes, pose a significant threat to humans and are an increasing concern because of climate change, human activity, and expanding vector-competent populations. West Nile virus is of significant concern as the most frequent mosquito-borne disease transmitted annually within the continental United States. Here, we identify a previously uncharacterized signaling pathway that impacts West Nile virus infection, namely endothelin signaling. Additionally, we demonstrate that we can successfully translate results obtained from D. melanogaster into the more relevant human system. Our results add to the growing field of insulin-mediated antiviral immunity and identify potential biomarkers or intervention targets to better address West Nile virus infection and severe disease.


Assuntos
Endotelinas , Insulina , Febre do Nilo Ocidental , Animais , Humanos , Drosophila melanogaster/imunologia , Drosophila melanogaster/metabolismo , Drosophila melanogaster/virologia , Insulina/metabolismo , Transdução de Sinais , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/metabolismo , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/fisiologia , Endotelinas/imunologia , Endotelinas/metabolismo
10.
Nat Commun ; 14(1): 1357, 2023 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-36914655

RESUMO

In most eukaryotes, biparentally inherited nuclear genomes and maternally inherited cytoplasmic genomes have different evolutionary interests. Strongly female-biased sex ratios that are repeatedly observed in various arthropods often result from the male-specific lethality (male-killing) induced by maternally inherited symbiotic bacteria such as Spiroplasma and Wolbachia. However, despite some plausible case reports wherein viruses are raised as male-killers, it is not well understood how viruses, having much smaller genomes than bacteria, are capable of inducing male-killing. Here we show that a maternally inherited double-stranded RNA (dsRNA) virus belonging to the family Partitiviridae (designated DbMKPV1) induces male-killing in Drosophila. DbMKPV1 localizes in the cytoplasm and possesses only four genes, i.e., one gene in each of the four genomic segments (dsRNA1-dsRNA4), in contrast to ca. 1000 or more genes possessed by Spiroplasma or Wolbachia. We also show that a protein (designated PVMKp1; 330 amino acids in size), encoded by a gene on the dsRNA4 segment, is necessary and sufficient for inducing male-killing. Our results imply that male-killing genes can be easily acquired by symbiotic viruses through reassortment and that symbiotic viruses are hidden players in arthropod evolution. We anticipate that host-manipulating genes possessed by symbiotic viruses can be utilized for controlling arthropods.


Assuntos
Drosophila melanogaster , Genes Virais , Vírus de Insetos , Razão de Masculinidade , Simbiose , Drosophila melanogaster/embriologia , Drosophila melanogaster/virologia , Vírus de Insetos/genética , Genes Virais/fisiologia , Masculino , Animais , Desenvolvimento Embrionário , RNA Viral/fisiologia , RNA de Cadeia Dupla/fisiologia , Evolução Molecular , Fases de Leitura Aberta/genética , Caracteres Sexuais , Feminino
11.
Front Immunol ; 13: 903860, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35844546

RESUMO

Zika is a member of the Flaviviridae virus family that poses some of the most significant global health risks, causing neurologic complications that range from sensory neuropathy and seizures to congenital Zika syndrome (microcephaly) in infants born to mothers infected during pregnancy. The recent outbreak of Zika virus (ZIKV) and its serious health threats calls for the characterization and understanding of Zika pathogenesis, as well as host antiviral immune functions. Although ZIKV has been associated with activating the RNA interference (RNAi) immune pathway and altering host metabolism, in-depth studies are still required to uncover the specifics of the complex host-virus interactions and provide additional insights into the molecular components that determine the outcome of this disease. Previous research establishes the fruit fly Drosophila melanogaster as a reliable model for studying viral pathogens, as it shares significant similarities with that of vertebrate animal systems. Here, we have developed an in vivo Drosophila model to investigate ZIKV-mediated perturbed metabolism in correlation to the RNAi central mediator Dicer-2. We report that ZIKV infection reprograms glucose and glycogen metabolism in Dicer-2 mutants to maintain efficient replication and successful propagation. Flies that exhibit these metabolic effects also show reduced food intake, which highlights the complicated neurological defects associated with ZIKV. We show that ZIKV infection significantly reduces insulin gene expression in Dicer-2 mutants, suggesting an insulin antiviral role against ZIKV and a direct connection to RNAi immunity. Moreover, we find that the insulin receptor substrate chico is crucial to the survival of ZIKV-infected flies. These observations are remarkably more severe in adult female flies compared to males, indicating possible sex differences in the rates of infection and susceptibility to the development of disease. Such findings not only demonstrate that metabolic alterations can be potentially exploited for developing immune therapeutic strategies but also that preventive measures for disease development may require sex-specific approaches. Therefore, further studies are urgently needed to explore the molecular factors that could be considered as targets to inhibit ZIKV manipulation of host cell metabolism in females and males.


Assuntos
Drosophila melanogaster/virologia , Insulinas , Infecção por Zika virus , Zika virus , Animais , Antivirais/uso terapêutico , Drosophila melanogaster/imunologia , Feminino , Humanos , Insulinas/metabolismo , Insulinas/farmacologia , Insulinas/uso terapêutico , Masculino , Fatores Sexuais , Replicação Viral
12.
Proc Natl Acad Sci U S A ; 119(29): e2122026119, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35858337

RESUMO

Hosts are continually selected to evolve new defenses against an ever-changing array of pathogens. To understand this process, we examined the genetic basis of resistance to the Drosophila A virus in Drosophila melanogaster. In a natural population, we identified a polymorphic transposable element (TE) insertion that was associated with an ∼19,000-fold reduction in viral titers, allowing flies to largely escape the harmful effects of infection by this virulent pathogen. The insertion occurs in the protein-coding sequence of the gene Veneno, which encodes a Tudor domain protein. By mutating Veneno with CRISPR-Cas9 in flies and expressing it in cultured cells, we show that the ancestral allele of the gene has no effect on viral replication. Instead, the TE insertion is a gain-of-function mutation that creates a gene encoding a novel resistance factor. Viral titers remained reduced when we deleted the TE sequence from the transcript, indicating that resistance results from the TE truncating the Veneno protein. This is a novel mechanism of virus resistance and a new way by which TEs can contribute to adaptation.


Assuntos
Elementos de DNA Transponíveis , Dicistroviridae , Drosophila melanogaster , Interações Hospedeiro-Patógeno , Domínio Tudor , Animais , Elementos de DNA Transponíveis/genética , Drosophila melanogaster/genética , Drosophila melanogaster/virologia , Mutação com Ganho de Função , Interações Hospedeiro-Patógeno/genética , Deleção de Sequência
13.
J Virol ; 96(12): e0021522, 2022 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-35608346

RESUMO

The baculovirus envelope protein GP64 is an essential component of the budded virus and is necessary for efficient virion assembly. Little is known regarding intracellular trafficking of GP64 to the plasma membrane, where it is incorporated into budding virions during egress. To identify host proteins and potential cellular trafficking pathways that are involved in delivery of GP64 to the plasma membrane, we developed and characterized a stable Drosophila cell line that inducibly expresses the AcMNPV GP64 protein and used that cell line in combination with a targeted RNA interference (RNAi) screen of vesicular protein trafficking pathway genes. Of the 37 initial hits from the screen, we validated and examined six host genes that were important for trafficking of GP64 to the cell surface. Validated hits included Rab GTPases Rab1 and Rab4, Clathrin heavy chain, clathrin adaptor protein genes AP-1-2ß and AP-2µ, and Snap29. Two gene knockdowns (Rab5 and Exo84) caused substantial increases (up to 2.5-fold) of GP64 on the plasma membrane. We found that a small amount of GP64 is released from cells in exosomes and that some portion of cell surface GP64 is endocytosed, suggesting that recycling helps to maintain GP64 at the cell surface. IMPORTANCE While much is known regarding trafficking of viral envelope proteins in mammalian cells, little is known about this process in insect cells. To begin to understand which factors and pathways are needed for trafficking of insect virus envelope proteins, we engineered a Drosophila melanogaster cell line and implemented an RNAi screen to identify cellular proteins that aid transport of the model baculovirus envelope protein (GP64) to the cell surface. For this we developed an experimental system that leverages the large array of tools available for Drosophila and performed a targeted RNAi screen to identify cellular proteins involved in GP64 trafficking to the cell surface. Since viral envelope proteins are often critical for production of infectious progeny virions, these studies lay the foundation for understanding how either pathogenic insect viruses (baculoviruses) or insect-vectored viruses (e.g., flaviviruses, alphaviruses) egress from cells in tissues such as the midgut to enable systemic virus infection.


Assuntos
Baculoviridae , Membrana Celular , Proteínas de Insetos , Proteínas do Envelope Viral , Animais , Baculoviridae/metabolismo , Linhagem Celular , Membrana Celular/virologia , Drosophila melanogaster/virologia , Proteínas de Insetos/genética , Interferência de RNA , Proteínas do Envelope Viral/metabolismo
14.
Viruses ; 15(1)2022 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-36680096

RESUMO

Polydnavirus (PDV) is a parasitic factor of endoparasitic wasps and contributes greatly to overcoming the immune response of parasitized hosts. Protein tyrosine phosphatases (PTPs) regulate a wide variety of biological processes at the post-transcriptional level in mammals, but knowledge of PDV PTP action during a parasitoid−host interaction is limited. In this study, we characterized a PTP gene, CvBV_12-6, derived from Cotesia vestalis bracovirus (CvBV), and explored its possible regulatory role in the immune response of the host Plutella xylostella. Our results from qPCR show that CvBV_12-6 was highly expressed in hemocytes at an early stage of parasitization. To explore CvBV_12-6 function, we specifically expressed CvBV_12-6 in Drosophila melanogaster hemocytes. The results show that Hml-Gal4 > CvBV_12-6 suppressed the phenoloxidase activity of hemolymph in D. melanogaster, but exerted no effect on the total count or the viability of the hemocytes. In addition, the Hml-Gal4 > CvBV_12-6 flies exhibited decreased antibacterial abilities against Staphylococcus aureus. Similarly, we found that CvBV_12-6 significantly suppressed the melanization of the host P. xylostella 24 h post parasitization and reduced the viability, but not the number, of hemocytes. In conclusion, CvBV_12-6 negatively regulated both cellular and humoral immunity in P. xylostella, and the related molecular mechanism may be universal to insects.


Assuntos
Mariposas , Polydnaviridae , Animais , Sequência de Aminoácidos , Drosophila melanogaster/virologia , Monofenol Mono-Oxigenase/metabolismo , Mariposas/virologia , Polydnaviridae/genética , Polydnaviridae/metabolismo , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Interações Hospedeiro-Patógeno
15.
J Mol Biol ; 434(6): 167308, 2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-34678301

RESUMO

Quantification of viral replication underlies investigations into host-virus interactions. In Drosophila melanogaster, persistent infections with Drosophila C virus, Drosophila A virus, and Nora virus are commonly observed in nature and in laboratory fly stocks. However, traditional endpoint dilution assays to quantify infectious titers are not compatible with persistently infecting isolates of these viruses that do not cause cytopathic effects in cell culture. Here we present a novel assay based on immunological detection of Drosophila C virus infection that allows quantification of infectious titers for a wider range of Drosophila C virus isolates. We also describe strand specific RT-qPCR assays for quantification of viral negative strand RNA produced during Drosophila C virus, Drosophila A virus, and Nora virus infection. Finally, we demonstrate the utility of these assays for quantification of viral replication during oral infections and persistent infections with each virus.


Assuntos
Dicistroviridae , Drosophila melanogaster , Imunoensaio , Reação em Cadeia da Polimerase em Tempo Real , Animais , Dicistroviridae/isolamento & purificação , Dicistroviridae/fisiologia , Drosophila melanogaster/virologia , RNA Viral/genética , Replicação Viral
16.
Commun Biol ; 4(1): 1327, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34824354

RESUMO

Wolbachia is a maternally transmitted bacterial symbiont that is estimated to infect approximately half of arthropod species. In the laboratory it can increase the resistance of insects to viral infection, but its effect on viruses in nature is unknown. Here we report that in a natural population of Drosophila melanogaster, individuals that are infected with Wolbachia are less likely to be infected by viruses. By characterising the virome by metagenomic sequencing and then testing individual flies for infection, we found the protective effect of Wolbachia was virus-specific, with the prevalence of infection being up to 15% greater in Wolbachia-free flies. The antiviral effects of Wolbachia may contribute to its extraordinary ecological success, and in nature the symbiont may be an important component of the antiviral defences of insects.


Assuntos
Drosophila melanogaster/microbiologia , Drosophila melanogaster/virologia , Wolbachia/fisiologia , Animais , Connecticut , Masculino , Simbiose
17.
Viruses ; 13(11)2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34835089

RESUMO

Single-cell RNA sequencing (scRNA-seq) offers the possibility to monitor both host and pathogens transcriptomes at the cellular level. Here, public scRNA-seq datasets from Drosophila melanogaster midgut cells were used to compare the differences in replication strategy and cellular responses between two fly picorna-like viruses, Thika virus (TV) and D. melanogaster Nora virus (DMelNV). TV exhibited lower levels of viral RNA accumulation but infected a higher number of cells compared to DMelNV. In both cases, viral RNA accumulation varied according to cell subtype. The cellular heat shock response to TV and DMelNV infection was cell-subtype- and virus-specific. Disruption of bottleneck genes at later stages of infection in the systemic response, as well as of translation-related genes in the cellular response to DMelNV in two cell subtypes, may affect the virus replication.


Assuntos
Drosophila melanogaster/virologia , Vírus de RNA/classificação , Vírus de RNA/fisiologia , Animais , Heterogeneidade Genética , Filogenia , Vírus de RNA/isolamento & purificação , RNA Viral/química , RNA Viral/classificação , RNA Viral/genética , Viroses/veterinária , Replicação Viral
18.
mBio ; 12(5): e0292320, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34488458

RESUMO

Wolbachia is a maternally transmitted bacterium that is widespread in arthropods and filarial nematodes and confers strong antiviral protection in Drosophila melanogaster and other arthropods. Wolbachia-transinfected Aedes aegypti mosquitoes are currently being deployed to fight transmission of dengue and Zika viruses. However, the mechanism of antiviral protection and the factors influencing are still not fully understood. Here, we show that temperature modulates Wolbachia-conferred protection in Drosophila melanogaster. Temperature after infection directly impacts Drosophila C virus (DCV) replication and modulates Wolbachia protection. At higher temperatures, viruses proliferate more and are more lethal, while Wolbachia confers lower protection. Strikingly, host developmental temperature is a determinant of Wolbachia-conferred antiviral protection. While there is strong protection when flies develop from egg to adult at 25°C, the protection is highly reduced or abolished when flies develop at 18°C. However, Wolbachia-induced changes during development are not sufficient to limit virus-induced mortality, as Wolbachia is still required to be present in adults at the time of infection. This developmental effect is general, since it was present in different host genotypes, Wolbachia variants, and upon infection with different viruses. Overall, we show that Wolbachia-conferred antiviral protection is temperature dependent, being present or absent depending on the environmental conditions. This interaction likely impacts Wolbachia-host interactions in nature and, as a result, frequencies of host and symbionts in different climates. Dependence of Wolbachia-mediated pathogen blocking on developmental temperature could be used to dissect the mechanistic bases of protection and influence the deployment of Wolbachia to prevent transmission of arboviruses. IMPORTANCE Insects are often infected with beneficial intracellular bacteria. The bacterium Wolbachia is extremely common in insects and can protect them from pathogenic viruses. This effect is being used to prevent transmission of dengue and Zika viruses by Wolbachia-infected mosquitoes. To understand the biology of insects in the wild, we need to discover which factors affect Wolbachia-conferred antiviral protection. Here, we show that the temperature at which insects develop from eggs to adults can determine the presence or absence of antiviral protection. The environment, therefore, strongly influences this insect-bacterium interaction. Our work may help to provide insights into the mechanism of viral blocking by Wolbachia, deepen our understanding of the geographical distribution of host and symbiont, and incentivize further research on the temperature dependence of Wolbachia-conferred protection for control of mosquito-borne disease.


Assuntos
Dicistroviridae/fisiologia , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/microbiologia , Interações Hospedeiro-Patógeno/fisiologia , Temperatura , Wolbachia/patogenicidade , Animais , Drosophila melanogaster/virologia , Feminino , Masculino , Carga Viral , Viroses/prevenção & controle , Replicação Viral
19.
Insect Biochem Mol Biol ; 138: 103648, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34536505

RESUMO

Baculoviruses are double-stranded DNA entomopathogenic viruses that infect predominantly insects of the order Lepidoptera. Research in the last decade has started to disentangle the mechanisms underlying the insect-virus interaction, particularly focusing on the effects of the baculovirus infection in the host's physiology. Among crucial physiological functions, olfaction has a key role in reproductive tasks, food source detection and enemy avoidance. In this work, we describe that Spodoptera exigua multiple nucleopolyhedrovirus (SeMNPV) induces expression changes in some odorant receptors (ORs) - the centrepiece of insect's olfaction - when infecting larvae from its natural host Spodoptera exigua (Lepidoptera: Noctuidae). Different ORs are up-regulated in larvae after SeMNPV infection, and two of them, SexiOR35 and SexiOR23, were selected for further functional characterization by heterologous expression in empty neurons of Drosophila melanogaster coupled to single-sensillum recordings. SexiOR35 appears to be a broadly tuned receptor able to recognise multiple and different chemical compounds. SexiOR23, although correctly expressed in Drosophila neurons, did not display any significant response to a panel of 58 stimuli. Behavioural experiments revealed that larvae infected by SeMNPV exhibit altered olfactory-driven behaviour to diet when it is supplemented with the plant volatiles linalool or estragole, two of the main SexiOR35 ligands, supporting the hypothesis that viral infection triggers changes in host perception through changes in the expression level of specific ORs.


Assuntos
Proteínas de Insetos/fisiologia , Nucleopoliedrovírus/fisiologia , Receptores Odorantes/fisiologia , Spodoptera/fisiologia , Animais , Drosophila melanogaster/fisiologia , Drosophila melanogaster/virologia , Larva/crescimento & desenvolvimento , Larva/fisiologia , Larva/virologia , Neurônios/fisiologia , Neurônios/virologia , Spodoptera/crescimento & desenvolvimento , Spodoptera/virologia
20.
Nature ; 597(7874): 109-113, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34261127

RESUMO

Cyclic GMP-AMP synthase (cGAS) is a cytosolic DNA sensor that produces the second messenger cG[2'-5']pA[3'-5']p (2'3'-cGAMP) and controls activation of innate immunity in mammalian cells1-5. Animal genomes typically encode multiple proteins with predicted homology to cGAS6-10, but the function of these uncharacterized enzymes is unknown. Here we show that cGAS-like receptors (cGLRs) are innate immune sensors that are capable of recognizing divergent molecular patterns and catalysing synthesis of distinct nucleotide second messenger signals. Crystal structures of human and insect cGLRs reveal a nucleotidyltransferase signalling core shared with cGAS and a diversified primary ligand-binding surface modified with notable insertions and deletions. We demonstrate that surface remodelling of cGLRs enables altered ligand specificity and used a forward biochemical screen to identify cGLR1 as a double-stranded RNA sensor in the model organism Drosophila melanogaster. We show that RNA recognition activates Drosophila cGLR1 to synthesize the novel product cG[3'-5']pA[2'-5']p (3'2'-cGAMP). A crystal structure of Drosophila stimulator of interferon genes (dSTING) in complex with 3'2'-cGAMP explains selective isomer recognition, and 3'2'-cGAMP induces an enhanced antiviral state in vivo that protects from viral infection. Similar to radiation of Toll-like receptors in pathogen immunity, our results establish cGLRs as a diverse family of metazoan pattern recognition receptors.


Assuntos
Drosophila melanogaster/metabolismo , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/metabolismo , RNA de Cadeia Dupla/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Sistemas do Segundo Mensageiro , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/imunologia , Drosophila melanogaster/virologia , Feminino , Humanos , Imunidade Inata , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Modelos Moleculares , Nucleotidiltransferases/química , Nucleotidiltransferases/imunologia , RNA de Cadeia Dupla/análise , RNA de Cadeia Dupla/imunologia , Receptores de Reconhecimento de Padrão/química , Receptores de Reconhecimento de Padrão/imunologia , Vírus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA