Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 56(9): 1991-2005.e9, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37659413

RESUMEN

In mammals, the enzyme cGAS senses the presence of cytosolic DNA and synthesizes the cyclic dinucleotide (CDN) 2'3'-cGAMP, which triggers STING-dependent immunity. In Drosophila melanogaster, two cGAS-like receptors (cGLRs) produce 3'2'-cGAMP and 2'3'-cGAMP to activate STING. We explored CDN-mediated immunity in 14 Drosophila species covering 50 million years of evolution and found that 2'3'-cGAMP and 3'2'-cGAMP failed to control infection by Drosophila C virus in D. serrata and two other species. We discovered diverse CDNs produced in a cGLR-dependent manner in response to viral infection in D. melanogaster, including 2'3'-c-di-GMP. This CDN was a more potent STING agonist than cGAMP in D. melanogaster and it also activated a strong antiviral transcriptional response in D. serrata. Our results shed light on the evolution of cGLRs in flies and provide a basis for understanding the function and regulation of this emerging family of pattern recognition receptors in animal innate immunity.


Asunto(s)
Antivirales , Drosophila , Animales , Drosophila melanogaster , GMP Cíclico , Mamíferos
3.
Cell ; 159(5): 1086-1095, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25416947

RESUMEN

Fighting viral infections is hampered by the scarcity of viral targets and their variability, resulting in development of resistance. Viruses depend on cellular molecules-which are attractive alternative targets-for their life cycle, provided that they are dispensable for normal cell functions. Using the model organism Drosophila melanogaster, we identify the ribosomal protein RACK1 as a cellular factor required for infection by internal ribosome entry site (IRES)-containing viruses. We further show that RACK1 is an essential determinant for hepatitis C virus translation and infection, indicating that its function is conserved for distantly related human and fly viruses. Inhibition of RACK1 does not affect Drosophila or human cell viability and proliferation, and RACK1-silenced adult flies are viable, indicating that this protein is not essential for general translation. Our findings demonstrate a specific function for RACK1 in selective mRNA translation and uncover a target for the development of broad antiviral intervention.


Asunto(s)
Dicistroviridae/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/virología , Proteínas de Unión al GTP/metabolismo , Hepatocitos/virología , Virus de Insectos/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Línea Celular Tumoral , Drosophila melanogaster/metabolismo , Hepacivirus/metabolismo , Hepatocitos/metabolismo , Humanos , Modelos Moleculares , Factores de Iniciación de Péptidos/metabolismo , Biosíntesis de Proteínas , Receptores de Cinasa C Activada , Secuencias Reguladoras de Ácido Ribonucleico , Replicación Viral
4.
Immunity ; 49(2): 225-234.e4, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30119996

RESUMEN

Antiviral immunity in Drosophila involves RNA interference and poorly characterized inducible responses. Here, we showed that two components of the IMD pathway, the kinase dIKKß and the transcription factor Relish, were required to control infection by two picorna-like viruses. We identified a set of genes induced by viral infection and regulated by dIKKß and Relish, which included an ortholog of STING. We showed that dSTING participated in the control of infection by picorna-like viruses, acting upstream of dIKKß to regulate expression of Nazo, an antiviral factor. Our data reveal an antiviral function for STING in an animal model devoid of interferons and suggest an evolutionarily ancient role for this molecule in antiviral immunity.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/inmunología , Drosophila melanogaster/virología , Quinasa I-kappa B/metabolismo , Proteínas de la Membrana/metabolismo , Factores de Iniciación de Péptidos/metabolismo , Infecciones por Picornaviridae/inmunología , Animales , Línea Celular , Dicistroviridae/inmunología , Proteínas de Drosophila/genética , Quinasa I-kappa B/genética , Proteínas de la Membrana/genética , Factores de Iniciación de Péptidos/genética , Interferencia de ARN , Factores de Transcripción/metabolismo
5.
Nature ; 597(7874): 109-113, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34261127

RESUMEN

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.


Asunto(s)
Drosophila melanogaster/metabolismo , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/metabolismo , ARN Bicatenario/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Sistemas de Mensajero Secundario , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/inmunología , Drosophila melanogaster/virología , Femenino , Humanos , Inmunidad Innata , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Nucleotidiltransferasas/química , Nucleotidiltransferasas/inmunología , ARN Bicatenario/análisis , ARN Bicatenario/inmunología , Receptores de Reconocimiento de Patrones/química , Receptores de Reconocimiento de Patrones/inmunología , Virus/inmunología
6.
Nature ; 597(7874): 114-118, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34261128

RESUMEN

In mammals, cyclic GMP-AMP (cGAMP) synthase (cGAS) produces the cyclic dinucleotide 2'3'-cGAMP in response to cytosolic DNA and this triggers an antiviral immune response. cGAS belongs to a large family of cGAS/DncV-like nucleotidyltransferases that is present in both prokaryotes1 and eukaryotes2-5. In bacteria, these enzymes synthesize a range of cyclic oligonucleotides and have recently emerged as important regulators of phage infections6-8. Here we identify two cGAS-like receptors (cGLRs) in the insect Drosophila melanogaster. We show that cGLR1 and cGLR2 activate Sting- and NF-κB-dependent antiviral immunity in response to infection with RNA or DNA viruses. cGLR1 is activated by double-stranded RNA to produce the cyclic dinucleotide 3'2'-cGAMP, whereas cGLR2 produces a combination of 2'3'-cGAMP and 3'2'-cGAMP in response to an as-yet-unidentified stimulus. Our data establish cGAS as the founding member of a family of receptors that sense different types of nucleic acids and trigger immunity through the production of cyclic dinucleotides beyond 2'3'-cGAMP.


Asunto(s)
Drosophila melanogaster/inmunología , Nucleotidiltransferasas/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Virus/inmunología , Secuencia de Aminoácidos , Animales , Línea Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/virología , Femenino , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Ligandos , Masculino , Proteínas de la Membrana/metabolismo , Modelos Moleculares , FN-kappa B/metabolismo , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/clasificación , Nucleotidiltransferasas/deficiencia , Nucleotidiltransferasas/metabolismo , ARN Bicatenario/análisis , ARN Bicatenario/inmunología , ARN Bicatenario/metabolismo , Receptores de Reconocimiento de Patrones/clasificación , Receptores de Reconocimiento de Patrones/deficiencia , Receptores de Reconocimiento de Patrones/inmunología
7.
Mol Biol Evol ; 41(3)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38377349

RESUMEN

Viruses represent a major threat to all animals, which defend themselves through induction of a large set of virus-stimulated genes that collectively control the infection. In vertebrates, these genes include interferons that play a critical role in the amplification of the response to infection. Virus- and interferon-stimulated genes include restriction factors targeting the different steps of the viral replication cycle, in addition to molecules associated with inflammation and adaptive immunity. Predictably, antiviral genes evolve dynamically in response to viral pressure. As a result, each animal has a unique arsenal of antiviral genes. Here, we exploit the capacity to experimentally activate the evolutionarily conserved stimulator of IFN genes (STING) signaling pathway by injection of the cyclic dinucleotide 2'3'-cyclic guanosine monophosphate-adenosine monophosphate into flies to define the repertoire of STING-regulated genes in 10 Drosophila species, spanning 40 million years of evolution. Our data reveal a set of conserved STING-regulated factors, including STING itself, a cGAS-like-receptor, the restriction factor pastel, and the antiviral protein Vago, but also 2 key components of the antiviral RNA interference pathway, Dicer-2, and Argonaute2. In addition, we identify unknown species- or lineage-specific genes that have not been previously associated with resistance to viruses. Our data provide insight into the core antiviral response in Drosophila flies and pave the way for the characterization of previously unknown antiviral effectors.


Asunto(s)
Drosophila , Inmunidad Innata , Animales , Nucleótidos Cíclicos , Antivirales/farmacología
9.
RNA ; 26(5): 581-594, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31996404

RESUMEN

Endogenous viral elements (EVEs) are found in many eukaryotic genomes. Despite considerable knowledge about genomic elements such as transposons (TEs) and retroviruses, we still lack information about nonretroviral EVEs. Aedes aegypti mosquitoes have a highly repetitive genome that is covered with EVEs. Here, we identified 129 nonretroviral EVEs in the AaegL5 version of the A. aegypti genome. These EVEs were significantly associated with TEs and preferentially located in repeat-rich clusters within intergenic regions. Genome-wide transcriptome analysis showed that most EVEs generated transcripts although only around 1.4% were sense RNAs. The majority of EVE transcription was antisense and correlated with the generation of EVE-derived small RNAs. A single genomic cluster of EVEs located in a 143 kb repetitive region in chromosome 2 contributed with 42% of antisense transcription and 45% of small RNAs derived from viral elements. This region was enriched for TE-EVE hybrids organized in the same coding strand. These generated a single long antisense transcript that correlated with the generation of phased primary PIWI-interacting RNAs (piRNAs). The putative promoter of this region had a conserved binding site for the transcription factor Cubitus interruptus, a key regulator of the flamenco locus in Drosophila melanogaster Here, we have identified a single unidirectional piRNA cluster in the A. aegypti genome that is the major source of EVE transcription fueling the generation of antisense small RNAs in mosquitoes. We propose that this region is a flamenco-like locus in A. aegypti due to its relatedness to the major unidirectional piRNA cluster in Drosophila melanogaster.


Asunto(s)
Aedes/genética , Genoma de los Insectos/genética , ARN Interferente Pequeño/genética , Retroelementos/genética , Animales , Sitios de Unión/genética , Cadherinas/genética , Culicidae/genética , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas de Homeodominio/genética , Regiones Promotoras Genéticas , Factores de Transcripción/genética
10.
Immunity ; 36(4): 555-7, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22520850

RESUMEN

The mammalian MyD88 signaling molecule participates in Toll receptor signaling within the cytoplasm. In this issue of Immunity, Marek and Kagan (2012) report that Drosophila (d)MyD88 acts instead at the plasma membrane to sort the signaling adaptor Tube.

11.
Curr Issues Mol Biol ; 34: 31-60, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31167955

RESUMEN

Insects, the most diverse group of animals, can be infected by an extraordinary diversity of viruses. Among them, arthropod-borne viruses can be transmitted to humans, while bee and silkworm viruses cause important economic losses. Like all invertebrates, insects rely solely on innate immunity to counter viral infections. Protein-based mechanisms, involving restriction factors and evolutionarily conserved signaling pathways regulating transcription factors of the NF-kB and STAT families, participate in the control of viral infections in insects. In addition, RNA-based responses play a major role in the silencing of viral RNAs. We review here our current state of knowledge on insect antiviral defense mechanisms, which include conserved as well as adaptive, insect-specific strategies. Identification of the innate immunity receptors that sense viral infection in insects remains a major challenge for the field.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de Insectos , Insectos/metabolismo , Insectos/virología , Animales , Biomarcadores , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Virus de Insectos/inmunología , Insectos/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal , Relación Estructura-Actividad
12.
Nat Immunol ; 9(12): 1425-32, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18953338

RESUMEN

Drosophila, like other invertebrates and plants, relies mainly on RNA interference for its defense against viruses. In flies, viral infection also triggers the expression of many genes. One of the genes induced, Vago, encodes a 18-kilodalton cysteine-rich polypeptide. Here we provide genetic evidence that the Vago gene product controlled viral load in the fat body after infection with drosophila C virus. Induction of Vago was dependent on the helicase Dicer-2. Dicer-2 belongs to the same DExD/H-box helicase family as do the RIG-I-like receptors, which sense viral infection and mediate interferon induction in mammals. We propose that this family represents an evolutionary conserved set of sensors that detect viral nucleic acids and direct antiviral responses.


Asunto(s)
Proteínas de Drosophila/inmunología , Drosophila/inmunología , Drosophila/virología , ARN Helicasas/inmunología , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Electroforesis en Gel de Poliacrilamida , Cuerpo Adiposo/inmunología , Cuerpo Adiposo/virología , Regulación de la Expresión Génica/inmunología , Humanos , Filogenia , ARN Helicasas/genética , ARN Helicasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasa III , Homología de Secuencia de Aminoácido , Transcripción Genética , Virosis/inmunología
13.
Nucleic Acids Res ; 45(15): 8993-9004, 2017 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-28911115

RESUMEN

Cricket paralysis virus (CrPV) is a dicistrovirus. Its positive-sense single-stranded RNA genome contains two internal ribosomal entry sites (IRESs). The 5' untranslated region (5'UTR) IRES5'UTR mediates translation of non-structural proteins encoded by ORF1 whereas the well-known intergenic region (IGR) IRESIGR is required for translation of structural proteins from open reading frame 2 in the late phase of infection. Concerted action of both IRES is essential for host translation shut-off and viral translation. IRESIGR has been extensively studied, in contrast the IRES5'UTR remains largely unexplored. Here, we define the minimal IRES element required for efficient translation initiation in drosophila S2 cell-free extracts. We show that IRES5'UTR promotes direct recruitment of the ribosome on the cognate viral AUG start codon without any scanning step, using a Hepatitis-C virus-related translation initiation mechanism. Mass spectrometry analysis revealed that IRES5'UTR recruits eukaryotic initiation factor 3, confirming that it belongs to type III class of IRES elements. Using Selective 2'-hydroxyl acylation analyzed by primer extension and DMS probing, we established a secondary structure model of 5'UTR and of the minimal IRES5'UTR. The IRES5'UTR contains a pseudoknot structure that is essential for proper folding and ribosome recruitment. Overall, our results pave the way for studies addressing the synergy and interplay between the two IRES from CrPV.


Asunto(s)
Regiones no Traducidas 5' , Dicistroviridae/genética , Sitios Internos de Entrada al Ribosoma , Biosíntesis de Proteínas , ARN Viral/química , Proteínas Virales/química , Animales , Secuencia de Bases , Línea Celular , Sistema Libre de Células/metabolismo , Dicistroviridae/crecimiento & desarrollo , Dicistroviridae/metabolismo , Drosophila melanogaster/virología , Gryllidae/virología , Interacciones Huésped-Patógeno , Conformación de Ácido Nucleico , Sistemas de Lectura Abierta , ARN Viral/genética , ARN Viral/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
14.
Proc Natl Acad Sci U S A ; 113(3): 698-703, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26739560

RESUMEN

Viruses are obligatory intracellular parasites that suffer strong evolutionary pressure from the host immune system. Rapidly evolving viral genomes can adapt to this pressure by acquiring genes that counteract host defense mechanisms. For example, many vertebrate DNA viruses have hijacked cellular genes encoding cytokines or cytokine receptors to disrupt host cell communication. Insect viruses express suppressors of RNA interference or apoptosis, highlighting the importance of these cell intrinsic antiviral mechanisms in invertebrates. Here, we report the identification and characterization of a family of proteins encoded by insect DNA viruses that are homologous to a 12-kDa circulating protein encoded by the virus-induced Drosophila gene diedel (die). We show that die mutant flies have shortened lifespan and succumb more rapidly than controls when infected with Sindbis virus. This reduced viability is associated with deregulated activation of the immune deficiency (IMD) pathway of host defense and can be rescued by mutations in the genes encoding the homolog of IKKγ or IMD itself. Our results reveal an endogenous pathway that is exploited by insect viruses to modulate NF-κB signaling and promote fly survival during the antiviral response.


Asunto(s)
Citocinas/química , Citocinas/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/inmunología , Drosophila melanogaster/metabolismo , Inmunidad , Homología de Secuencia de Aminoácido , Transducción de Señal , Infecciones por Alphavirus/genética , Secuencia de Aminoácidos , Animales , Citocinas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/virología , Inmunidad/genética , Datos de Secuencia Molecular , Mutación/genética , Virus Sindbis , Análisis de Supervivencia , Regulación hacia Arriba/genética
15.
An Acad Bras Cienc ; 91 Suppl 3: e20190122, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31166476

RESUMEN

Insects are the most diverse group of animals. They can be infected by an extraordinary diversity of viruses. Among them, arthropod-borne viruses (arboviruses) can be transmitted to humans. High-throughput sequencing of small RNAs from insects provides insight into their virome, which may help understand the dynamics of vector borne infectious diseases. Furthermore, investigating the mechanisms that restrict viral infections in insects points to genetic innovations that may inspire novel antiviral strategies.


Asunto(s)
Biodiversidad , Genoma Viral/genética , Insectos Vectores/virología , Virus de Insectos/clasificación , Virus ARN/clasificación , Animales , Insectos Vectores/clasificación , Virus de Insectos/genética , Virus ARN/genética
16.
Cell Mol Life Sci ; 74(11): 2039-2054, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28102430

RESUMEN

The fruit fly Drosophila melanogaster has been a valuable model to investigate the genetic mechanisms of innate immunity. Initially focused on the resistance to bacteria and fungi, these studies have been extended to include antiviral immunity over the last decade. Like all living organisms, insects are continually exposed to viruses and have developed efficient defense mechanisms. We review here our current understanding on antiviral host defense in fruit flies. A major antiviral defense in Drosophila is RNA interference, in particular the small interfering (si) RNA pathway. In addition, complex inducible responses and restriction factors contribute to the control of infections. Some of the genes involved in these pathways have been conserved through evolution, highlighting loci that may account for susceptibility to viral infections in humans. Other genes are not conserved and represent species-specific innovations.


Asunto(s)
Antivirales/inmunología , Drosophila melanogaster/inmunología , Inmunidad Innata , Animales , Drosophila melanogaster/virología , Humanos , Ácidos Nucleicos/inmunología , Interferencia de ARN , Virosis/inmunología , Virosis/virología
17.
J Virol ; 90(11): 5415-5426, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27009948

RESUMEN

UNLABELLED: Antiviral immunity in the model organism Drosophila melanogaster involves the broadly active intrinsic mechanism of RNA interference (RNAi) and virus-specific inducible responses. Here, using a panel of six viruses, we investigated the role of hemocytes and autophagy in the control of viral infections. Injection of latex beads to saturate phagocytosis, or genetic depletion of hemocytes, resulted in decreased survival and increased viral titers following infection with Cricket paralysis virus (CrPV), Flock House virus (FHV), and vesicular stomatitis virus (VSV) but had no impact on Drosophila C virus (DCV), Sindbis virus (SINV), and Invertebrate iridescent virus 6 (IIV6) infection. In the cases of CrPV and FHV, apoptosis was induced in infected cells, which were phagocytosed by hemocytes. In contrast, VSV did not trigger any significant apoptosis but we confirmed that the autophagy gene Atg7 was required for full virus resistance, suggesting that hemocytes use autophagy to recognize the virus. However, this recognition does not depend on the Toll-7 receptor. Autophagy had no impact on DCV, CrPV, SINV, or IIV6 infection and was required for replication of the sixth virus, FHV. Even in the case of VSV, the increases in titers were modest in Atg7 mutant flies, suggesting that autophagy does not play a major role in antiviral immunity in Drosophila Altogether, our results indicate that, while autophagy plays a minor role, phagocytosis contributes to virus-specific immune responses in insects. IMPORTANCE: Phagocytosis and autophagy are two cellular processes that involve lysosomal degradation and participate in Drosophila immunity. Using a panel of RNA and DNA viruses, we have addressed the contribution of phagocytosis and autophagy in the control of viral infections in this model organism. We show that, while autophagy plays a minor role, phagocytosis contributes to virus-specific immune responses in Drosophila This work brings to the front a novel facet of antiviral host defense in insects, which may have relevance in the control of virus transmission by vector insects or in the resistance of beneficial insects to viral pathogens.


Asunto(s)
Autofagia , Virus ADN/inmunología , Drosophila/inmunología , Drosophila/virología , Hemocitos/inmunología , Fagocitosis , Virus ARN/inmunología , Animales , Apoptosis , Proteína 7 Relacionada con la Autofagia/genética , Proteína 7 Relacionada con la Autofagia/metabolismo , Línea Celular , Drosophila/citología , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Interferencia de ARN , Virus Sindbis/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Replicación Viral
18.
Nucleic Acids Res ; 43(13): 6191-206, 2015 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-26040701

RESUMEN

Virus surveillance in vector insects is potentially of great benefit to public health. Large-scale sequencing of small and long RNAs has previously been used to detect viruses, but without any formal comparison of different strategies. Furthermore, the identification of viral sequences largely depends on similarity searches against reference databases. Here, we developed a sequence-independent strategy based on virus-derived small RNAs produced by the host response, such as the RNA interference pathway. In insects, we compared sequences of small and long RNAs, demonstrating that viral sequences are enriched in the small RNA fraction. We also noted that the small RNA size profile is a unique signature for each virus and can be used to identify novel viral sequences without known relatives in reference databases. Using this strategy, we characterized six novel viruses in the viromes of laboratory fruit flies and wild populations of two insect vectors: mosquitoes and sandflies. We also show that the small RNA profile could be used to infer viral tropism for ovaries among other aspects of virus biology. Additionally, our results suggest that virus detection utilizing small RNAs can also be applied to vertebrates, although not as efficiently as to plants and insects.


Asunto(s)
ARN Pequeño no Traducido/química , ARN Viral/química , Virus/aislamiento & purificación , Animales , Mapeo Contig , Femenino , Insectos/genética , Ovario/virología , Plantas/virología , Análisis de Secuencia de ARN , Vertebrados/virología , Tropismo Viral , Virus/genética
19.
J Biol Chem ; 289(30): 20470-6, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24947515

RESUMEN

The host defense of the model organism Drosophila is under the control of two major signaling cascades controlling transcription factors of the NF-B family, the Toll and the immune deficiency (IMD) pathways. The latter shares extensive similarities with the mammalian TNF-R pathway and was initially discovered for its role in anti-Gram-negative bacterial reactions. A previous interactome study from this laboratory reported that an unexpectedly large number of proteins are binding to the canonical components of the IMD pathway. Here, we focus on DNA methyltransferase-associated protein 1 (DMAP1), which this study identified as an interactant of Relish, a Drosophila transcription factor reminiscent of the mammalian p105 NF-B protein. We show that silencing of DMAP1 expression both in S2 cells and in flies results in a significant reduction of Escherichia coli-induced expression of antimicrobial peptides. Epistatic analysis indicates that DMAP1 acts in parallel or downstream of Relish. Co-immunoprecipitation experiments further reveal that, in addition to Relish, DMAP1 also interacts with Akirin and the Brahma-associated protein 55 kDa (BAP55). Taken together, these results reveal that DMAP1 is a novel nuclear modulator of the IMD pathway, possibly acting at the level of chromatin remodeling.


Asunto(s)
Proteínas de Drosophila/inmunología , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Inmunidad Innata/fisiología , FN-kappa B/inmunología , Proteínas Represoras/inmunología , Factores de Transcripción/inmunología , Animales , Línea Celular , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/inmunología , Proteínas de Drosophila/genética , Drosophila melanogaster , Epistasis Genética/genética , Epistasis Genética/inmunología , Infecciones por Escherichia coli/genética , FN-kappa B/genética , Proteínas Nucleares , Proteínas Represoras/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Factores de Transcripción/genética
20.
J Virol ; 88(24): 14057-69, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25253354

RESUMEN

UNLABELLED: Drosophila C virus (DCV) is a positive-sense RNA virus belonging to the Dicistroviridae family. This natural pathogen of the model organism Drosophila melanogaster is commonly used to investigate antiviral host defense in flies, which involves both RNA interference and inducible responses. Although lethality is used routinely as a readout for the efficiency of the antiviral immune response in these studies, virus-induced pathologies in flies still are poorly understood. Here, we characterize the pathogenesis associated with systemic DCV infection. Comparison of the transcriptome of flies infected with DCV or two other positive-sense RNA viruses, Flock House virus and Sindbis virus, reveals that DCV infection, unlike those of the other two viruses, represses the expression of a large number of genes. Several of these genes are expressed specifically in the midgut and also are repressed by starvation. We show that systemic DCV infection triggers a nutritional stress in Drosophila which results from intestinal obstruction with the accumulation of peritrophic matrix at the entry of the midgut and the accumulation of the food ingested in the crop, a blind muscular food storage organ. The related virus cricket paralysis virus (CrPV), which efficiently grows in Drosophila, does not trigger this pathology. We show that DCV, but not CrPV, infects the smooth muscles surrounding the crop, causing extensive cytopathology and strongly reducing the rate of contractions. We conclude that the pathogenesis associated with systemic DCV infection results from the tropism of the virus for an important organ within the foregut of dipteran insects, the crop. IMPORTANCE: DCV is one of the few identified natural viral pathogens affecting the model organism Drosophila melanogaster. As such, it is an important virus for the deciphering of host-virus interactions in insects. We characterize here the pathogenesis associated with DCV infection in flies and show that it results from the tropism of the virus for an essential but poorly characterized organ in the digestive tract, the crop. Our results may have relevance for other members of the Dicistroviridae, some of which are pathogenic to beneficial or pest insect species.


Asunto(s)
Dicistroviridae/crecimiento & desarrollo , Drosophila melanogaster/virología , Obstrucción Intestinal/virología , Animales , Dicistroviridae/fisiología , Femenino , Tracto Gastrointestinal/patología , Tracto Gastrointestinal/fisiopatología , Tracto Gastrointestinal/virología , Perfilación de la Expresión Génica , Músculo Liso/virología , Nodaviridae/crecimiento & desarrollo , Virus Sindbis/crecimiento & desarrollo , Tropismo Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA