Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
2.
Cell ; 159(5): 1086-1095, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25416947

RESUMO

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.


Assuntos
Dicistroviridae/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/virologia , Proteínas de Ligação ao GTP/metabolismo , Hepatócitos/virologia , Vírus de Insetos/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Linhagem Celular Tumoral , Drosophila melanogaster/metabolismo , Hepacivirus/metabolismo , Hepatócitos/metabolismo , Humanos , Modelos Moleculares , Fatores de Iniciação de Peptídeos/metabolismo , Biossíntese de Proteínas , Receptores de Quinase C Ativada , Sequências Reguladoras de Ácido Ribonucleico , Replicação Viral
3.
Immunity ; 49(2): 225-234.e4, 2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30119996

RESUMO

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.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/imunologia , Drosophila melanogaster/virologia , Quinase I-kappa B/metabolismo , Proteínas de Membrana/metabolismo , Fatores de Iniciação de Peptídeos/metabolismo , Infecções por Picornaviridae/imunologia , Animais , Linhagem Celular , Dicistroviridae/imunologia , Proteínas de Drosophila/genética , Quinase I-kappa B/genética , Proteínas de Membrana/genética , Fatores de Iniciação de Peptídeos/genética , Interferência de RNA , Fatores de Transcrição/metabolismo
4.
Proc Natl Acad Sci U S A ; 120(30): e2303462120, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37459549

RESUMO

Injection of OCs into adult male flies induces a strong transcriptomic response in the host flies featuring in particular genes encoding bona fide G coupled proteins, among which the gene for methuselah like 1 is prominent. The injection is followed after a 3-d lag period, by the proliferation of the oncogenic cells. We hypothesized that through the product of mthl1 the host might control, at least in part, this proliferation as a defense reaction. Through a combination of genetic manipulations of the mthl1 gene (loss of function and overexpression of mthl1), we document that indeed this gene has an antiproliferative effect. Parallel injections of primary embryonic Drosophila cells or of various microbes do not exhibit this effect. We further show that mthl1 controls the expression of a large number of genes coding for chemoreceptors and genes implicated in regulation of development. Of great potential interest is our observation that the expression of the mouse gene coding for the adhesion G-protein-coupled receptor E1 (Adgre1, also known as F4/80), a potential mammalian homologue of mthl1, is significantly induced by B16-F10 melanoma cell inoculation 3 d postinjection in both the bone marrow and spleen (nests of immature and mature myeloid-derived immune cells), respectively. This observation is compatible with a role of this GPCR in the early response to injected tumor cells in mice.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Masculino , Camundongos , Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Perfilação da Expressão Gênica , Mamíferos/genética , Células Mieloides/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
5.
Proc Natl Acad Sci U S A ; 120(12): e2205140120, 2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36917667

RESUMO

The Drosophila systemic immune response against many Gram-positive bacteria and fungi is mediated by the Toll pathway. How Toll-regulated effectors actually fulfill this role remains poorly understood as the known Toll-regulated antimicrobial peptide (AMP) genes are active only against filamentous fungi and not against Gram-positive bacteria or yeasts. Besides AMPs, two families of peptides secreted in response to infectious stimuli that activate the Toll pathway have been identified, namely Bomanins and peptides derived from a polyprotein precursor known as Baramicin A (BaraA). Unexpectedly, the deletion of a cluster of 10 Bomanins phenocopies the Toll mutant phenotype of susceptibility to infections. Here, we demonstrate that BaraA is required specifically in the host defense against Enterococcus faecalis and against the entomopathogenic fungus Metarhizium robertsii, albeit the fungal burden is not altered in BaraA mutants. BaraA protects the fly from the action of distinct toxins secreted by these Gram-positive and fungal pathogens, respectively, Enterocin V and Destruxin A. The injection of Destruxin A leads to the rapid paralysis of flies, whether wild type (WT) or mutant. However, a larger fraction of wild-type than BaraA flies recovers from paralysis within 5 to 10 h. BaraAs' function in protecting the host from the deleterious action of Destruxin is required in glial cells, highlighting a resilience role for the Toll pathway in the nervous system against microbial virulence factors. Thus, in complement to the current paradigm, innate immunity can cope effectively with the effects of toxins secreted by pathogens through the secretion of dedicated peptides, independently of xenobiotics detoxification pathways.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Receptores Toll-Like/metabolismo , Transdução de Sinais , Peptídeos/metabolismo , Fungos/metabolismo , Bactérias Gram-Positivas/metabolismo
7.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33737397

RESUMO

Oncogenic RasV12 cells [A. Simcox et al., PLoS Genet 4, e1000142 (2008)] injected into adult males proliferated massively after a lag period of several days, and led to the demise of the flies after 2 to 3 wk. The injection induced an early massive transcriptomic response that, unexpectedly, included more than 100 genes encoding chemoreceptors of various families. The kinetics of induction and the identities of the induced genes differed markedly from the responses generated by injections of microbes. Subsequently, hundreds of genes were up-regulated, attesting to intense catabolic activities in the flies, active tracheogenesis, and cuticulogenesis, as well as stress and inflammation-type responses. At 11 d after the injections, GFP-positive oncogenic cells isolated from the host flies exhibited a markedly different transcriptomic profile from that of the host and distinct from that at the time of their injection, including in particular up-regulated expression of genes typical for cells engaged in the classical antimicrobial response of Drosophila.


Assuntos
Perfilação da Expressão Gênica , Imunidade , Neoplasias/genética , Neoplasias/imunologia , Transcriptoma , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Modelos Animais de Doenças , Resistência à Doença , Drosophila , Genes Reporter , Humanos , Imunidade Inata
8.
Nat Immunol ; 9(12): 1425-32, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18953338

RESUMO

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.


Assuntos
Proteínas de Drosophila/imunologia , Drosophila/imunologia , Drosophila/virologia , RNA Helicases/imunologia , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Eletroforese em Gel de Poliacrilamida , Corpo Adiposo/imunologia , Corpo Adiposo/virologia , Regulação da Expressão Gênica/imunologia , Humanos , Filogenia , RNA Helicases/genética , RNA Helicases/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonuclease III , Homologia de Sequência de Aminoácidos , Transcrição Gênica , Viroses/imunologia
9.
Nat Immunol ; 9(1): 97-104, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18066067

RESUMO

During a genome-wide screen with RNA-mediated interference, we isolated CG8580 as a gene involved in the innate immune response of Drosophila melanogaster. CG8580, which we called Akirin, encoded a protein that acted in parallel with the NF-kappaB transcription factor downstream of the Imd pathway and was required for defense against Gram-negative bacteria. Akirin is highly conserved, and the human genome contains two homologs, one of which was able to rescue the loss-of-function phenotype in drosophila cells. Akirins were strictly localized to the nucleus. Knockout of both Akirin homologs in mice showed that one had an essential function downstream of the Toll-like receptor, tumor necrosis factor and interleukin (IL)-1beta signaling pathways leading to the production of IL-6. Thus, Akirin is a conserved nuclear factor required for innate immune responses.


Assuntos
Proteínas de Drosophila/biossíntese , Drosophila melanogaster/metabolismo , NF-kappa B/biossíntese , Proteínas Nucleares/fisiologia , Proteínas/fisiologia , Animais , Linhagem Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Humanos , Imunidade Inata , Interleucina-1beta/imunologia , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/genética , Proteínas/genética , Transdução de Sinais , Receptores Toll-Like/imunologia , Fator de Necrose Tumoral alfa/imunologia
10.
Proc Natl Acad Sci U S A ; 113(3): 698-703, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26739560

RESUMO

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.


Assuntos
Citocinas/química , Citocinas/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/imunologia , Drosophila melanogaster/metabolismo , Imunidade , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Infecções por Alphavirus/genética , Sequência de Aminoácidos , Animais , Citocinas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/virologia , Imunidade/genética , Dados de Sequência Molecular , Mutação/genética , Sindbis virus , Análise de Sobrevida , Regulação para Cima/genética
11.
EMBO J ; 33(20): 2332-48, 2014 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-25107474

RESUMO

Transcription of inflammatory genes in innate immune cells is coordinately regulated by transcription factors, including NF-κB, and chromatin modifiers. However, it remains unclear how microbial sensing initiates chromatin remodeling. Here, we show that Akirin2, an evolutionarily conserved nuclear protein, bridges NF-κB and the chromatin remodeling SWI/SNF complex by interacting with BRG1-Associated Factor 60 (BAF60) proteins as well as IκB-ζ, which forms a complex with the NF-κB p50 subunit. These interactions are essential for Toll-like receptor-, RIG-I-, and Listeria-mediated expression of proinflammatory genes including Il6 and Il12b in macrophages. Consistently, effective clearance of Listeria infection required Akirin2. Furthermore, Akirin2 and IκB-ζ recruitment to the Il6 promoter depend upon the presence of IκB-ζ and Akirin2, respectively, for regulation of chromatin remodeling. BAF60 proteins were also essential for the induction of Il6 in response to LPS stimulation. Collectively, the IκB-ζ-Akirin2-BAF60 complex physically links the NF-κB and SWI/SNF complexes in innate immune cell activation. By recruiting SWI/SNF chromatin remodellers to IκB-ζ, transcriptional coactivator for NF-κB, the conserved nuclear protein Akirin2 stimulates pro-inflammatory gene promoters in mouse macrophages during innate immune responses to viral or bacterial infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Regulação da Expressão Gênica , Imunidade Inata , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Núcleo Celular/metabolismo , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/genética , Citocinas/metabolismo , Feminino , Humanos , Listeria monocytogenes/fisiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Repressoras/genética , Deleção de Sequência , Ativação Transcricional
12.
J Virol ; 90(11): 5415-5426, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27009948

RESUMO

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.


Assuntos
Autofagia , Vírus de DNA/imunologia , Drosophila/imunologia , Drosophila/virologia , Hemócitos/imunologia , Fagocitose , Vírus de RNA/imunologia , Animais , Apoptose , Proteína 7 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/metabolismo , Linhagem Celular , Drosophila/citologia , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Interferência de RNA , Sindbis virus/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Replicação Viral
13.
Proc Natl Acad Sci U S A ; 110(8): 2957-62, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23378635

RESUMO

Chronic inflammation of the intestine is detrimental to mammals. Similarly, constant activation of the immune response in the gut by the endogenous flora is suspected to be harmful to Drosophila. Therefore, the innate immune response in the gut of Drosophila melanogaster is tightly balanced to simultaneously prevent infections by pathogenic microorganisms and tolerate the endogenous flora. Here we describe the role of the big bang (bbg) gene, encoding multiple membrane-associated PDZ (PSD-95, Discs-large, ZO-1) domain-containing protein isoforms, in the modulation of the gut immune response. We show that in the adult Drosophila midgut, BBG is present at the level of the septate junctions, on the apical side of the enterocytes. In the absence of BBG, these junctions become loose, enabling the intestinal flora to trigger a constitutive activation of the anterior midgut immune response. This chronic epithelial inflammation leads to a reduced lifespan of bbg mutant flies. Clearing the commensal flora by antibiotics prevents the abnormal activation of the gut immune response and restores a normal lifespan. We now provide genetic evidence that Drosophila septate junctions are part of the gut immune barrier, a function that is evolutionarily conserved in mammals. Collectively, our data suggest that septate junctions are required to maintain the subtle balance between immune tolerance and immune response in the Drosophila gut, which represents a powerful model to study inflammatory bowel diseases.


Assuntos
Proteínas de Drosophila/genética , Drosophila/imunologia , Tolerância Imunológica/genética , Proteínas de Membrana/genética , Animais , Longevidade
14.
Proc Natl Acad Sci U S A ; 110(26): 10717-22, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23749869

RESUMO

The Drosophila defense against pathogens largely relies on the activation of two signaling pathways: immune deficiency (IMD) and Toll. The IMD pathway is triggered mainly by Gram-negative bacteria, whereas the Toll pathway responds predominantly to Gram-positive bacteria and fungi. The activation of these pathways leads to the rapid induction of numerous NF-κB-induced immune response genes, including antimicrobial peptide genes. The IMD pathway shows significant similarities with the TNF receptor pathway. Recent evidence indicates that the IMD pathway is also activated in response to various noninfectious stimuli (i.e., inflammatory-like reactions). To gain a better understanding of the molecular machinery underlying the pleiotropic functions of this pathway, we first performed a comprehensive proteomics analysis to identify the proteins interacting with the 11 canonical members of the pathway initially identified by genetic studies. We identified 369 interacting proteins (corresponding to 291 genes) in heat-killed Escherichia coli-stimulated Drosophila S2 cells, 92% of which have human orthologs. A comparative analysis of gene ontology from fly or human gene annotation databases points to four significant common categories: (i) the NuA4, nucleosome acetyltransferase of H4, histone acetyltransferase complex, (ii) the switching defective/sucrose nonfermenting-type chromatin remodeling complex, (iii) transcription coactivator activity, and (iv) translation factor activity. Here we demonstrate that sumoylation of the IκB kinase homolog immune response-deficient 5 plays an important role in the induction of antimicrobial peptide genes through a highly conserved sumoylation consensus site during bacterial challenge. Taken together, the proteomics data presented here provide a unique avenue for a comparative functional analysis of proteins involved in innate immune reactions in flies and mammals.


Assuntos
Proteínas de Drosophila/imunologia , Drosophila/imunologia , Drosophila/microbiologia , Transdução de Sinais/imunologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Montagem e Desmontagem da Cromatina/genética , Montagem e Desmontagem da Cromatina/imunologia , Drosophila/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Escherichia coli/imunologia , Genes de Insetos , Histona Acetiltransferases/genética , Histona Acetiltransferases/imunologia , Histona Acetiltransferases/metabolismo , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mapas de Interação de Proteínas , Homologia de Sequência de Aminoácidos
15.
J Biol Chem ; 289(30): 20470-6, 2014 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-24947515

RESUMO

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.


Assuntos
Proteínas de Drosophila/imunologia , Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Imunidade Inata/fisiologia , NF-kappa B/imunologia , Proteínas Repressoras/imunologia , Fatores de Transcrição/imunologia , Animais , Linhagem Celular , Montagem e Desmontagem da Cromatina/genética , Montagem e Desmontagem da Cromatina/imunologia , Proteínas de Drosophila/genética , Drosophila melanogaster , Epistasia Genética/genética , Epistasia Genética/imunologia , Infecções por Escherichia coli/genética , NF-kappa B/genética , Proteínas Nucleares , Proteínas Repressoras/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fatores de Transcrição/genética
16.
J Virol ; 88(24): 14057-69, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25253354

RESUMO

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.


Assuntos
Dicistroviridae/crescimento & desenvolvimento , Drosophila melanogaster/virologia , Obstrução Intestinal/virologia , Animais , Dicistroviridae/fisiologia , Feminino , Trato Gastrointestinal/patologia , Trato Gastrointestinal/fisiopatologia , Trato Gastrointestinal/virologia , Perfilação da Expressão Gênica , Músculo Liso/virologia , Nodaviridae/crescimento & desenvolvimento , Sindbis virus/crescimento & desenvolvimento , Tropismo Viral
17.
J Immunol ; 190(2): 650-8, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23255357

RESUMO

The fruit fly Drosophila melanogaster is a good model to unravel the molecular mechanisms of innate immunity and has led to some important discoveries about the sensing and signaling of microbial infections. The response of Drosophila to virus infections remains poorly characterized and appears to involve two facets. On the one hand, RNA interference involves the recognition and processing of dsRNA into small interfering RNAs by the host RNase Dicer-2 (Dcr-2), whereas, on the other hand, an inducible response controlled by the evolutionarily conserved JAK-STAT pathway contributes to the antiviral host defense. To clarify the contribution of the small interfering RNA and JAK-STAT pathways to the control of viral infections, we have compared the resistance of flies wild-type and mutant for Dcr-2 or the JAK kinase Hopscotch to infections by seven RNA or DNA viruses belonging to different families. Our results reveal a unique susceptibility of hop mutant flies to infection by Drosophila C virus and cricket paralysis virus, two members of the Dicistroviridae family, which contrasts with the susceptibility of Dcr-2 mutant flies to many viruses, including the DNA virus invertebrate iridescent virus 6. Genome-wide microarray analysis confirmed that different sets of genes were induced following infection by Drosophila C virus or by two unrelated RNA viruses, Flock House virus and Sindbis virus. Overall, our data reveal that RNA interference is an efficient antiviral mechanism, operating against a large range of viruses, including a DNA virus. By contrast, the antiviral contribution of the JAK-STAT pathway appears to be virus specific.


Assuntos
Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Interferência de RNA/imunologia , Alphavirus/imunologia , Infecções por Alphavirus/genética , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/prevenção & controle , Animais , Animais Geneticamente Modificados , Infecções por Vírus de DNA/genética , Infecções por Vírus de DNA/imunologia , Infecções por Vírus de DNA/prevenção & controle , Proteínas de Drosophila/genética , Proteínas de Drosophila/imunologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/virologia , Regulação da Expressão Gênica , Janus Quinases/metabolismo , Masculino , Nodaviridae/imunologia , RNA Helicases/genética , RNA Helicases/imunologia , Infecções por Vírus de RNA/genética , Infecções por Vírus de RNA/imunologia , Infecções por Vírus de RNA/prevenção & controle , Ribonuclease III/genética , Ribonuclease III/imunologia , Fatores de Transcrição/metabolismo
18.
J Immunol ; 188(11): 5210-20, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22611248

RESUMO

The cytokine-induced activation cascade of NF-kappaB in mammals and the activation of the morphogen dorsal in Drosophila embryos show striking structural and functional similarities (Toll/IL-1, Cactus/I-kappaB, and dorsal/NF-kappaB). Here we demonstrate that these parallels extend to the immune response of Drosophila. In particular, the intracellular components of the dorsoventral signaling pathway (except for dorsal) and the extracellular Toll ligand, spätzle regulatory gene cassette, control expression of the antifungal peptide gene drosomycin in adults. We also show that mutations in the Toll signaling pathway dramatically reduce survival after fungal infection. Antibacterial genes are induced either by a distinct pathway involving the immune deficiency gene (imd) or by combined activation of both imd and dorsoventral pathways.


Assuntos
Antifúngicos/metabolismo , Proteínas de Ligação a DNA/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/imunologia , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Micoses/imunologia , Micoses/prevenção & controle , Fosfoproteínas/fisiologia , Receptores Toll-Like/fisiologia , Animais , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , História do Século XX , Família Multigênica/genética , Micoses/metabolismo
19.
Proc Natl Acad Sci U S A ; 108(29): 12024-9, 2011 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-21719711

RESUMO

The effects of the cellular environment on innate immunity remain poorly characterized. Here, we show that in Drosophila ATP-sensitive potassium channels (K(ATP)) mediate resistance to a cardiotropic RNA virus, Flock House virus (FHV). FHV viral load in the heart rapidly increases in K(ATP) mutant flies, leading to increased viremia and accelerated death. The effect of K(ATP) channels is dependent on the RNA interference genes Dcr-2, AGO2, and r2d2, indicating that an activity associated with this potassium channel participates in this antiviral pathway in Drosophila. Flies treated with the K(ATP) agonist drug pinacidil are protected against FHV infection, thus demonstrating the importance of this regulation of innate immunity by the cellular environment in the heart. In mice, the Coxsackievirus B3 replicates to higher titers in the hearts of mayday mutant animals, which are deficient in the Kir6.1 subunit of K(ATP) channels, than in controls. Together, our data suggest that K(ATP) channel deregulation can have a critical impact on innate antiviral immunity in the heart.


Assuntos
Drosophila/imunologia , Drosophila/virologia , Coração/virologia , Imunidade Inata/imunologia , Canais KATP/metabolismo , Nodaviridae/imunologia , Animais , Células HeLa , Humanos , Immunoblotting , Canais KATP/agonistas , Canais KATP/genética , Camundongos , Camundongos Endogâmicos C57BL , Nodaviridae/efeitos dos fármacos , Pinacidil/farmacologia , Interferência de RNA/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tolbutamida , Carga Viral/imunologia , Viremia
20.
Cell Rep ; 43(2): 113677, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38236774

RESUMO

Toll signaling is well known for its pivotal role in the host response against the invasion of external pathogens. Here, we investigate the potential involvement of Toll signaling in the intersection between the host and oncogenic cells. We show that loss of myeloid differentiation factor 88 (Myd88) leads to drastic fly death after the injection of RasV12-GFP oncogenic cells. Transcriptomic analyses show that challenging flies with oncogenic cells or bacteria leads to distinct inductions of Myd88-dependent genes. We note that downregulation of Myd88 in the tracheal system accounts for fly mortality, and ectopic tracheal complementation of Myd88 rescues the survival defect in Myd88 loss-of-function mutants following RasV12-GFP injection. Further, molecular and genetic evidence indicate that Toll signaling modulates fly resistance to RasV12-GFP cells through mediating airway function in a rolled-dependent manner. Collectively, our data indicate a critical role of Toll signaling in tracheal homeostasis and host survival after the injection of oncogenic cells.


Assuntos
Fator 88 de Diferenciação Mieloide , Traqueia , Transdução de Sinais , Regulação para Baixo , Homeostase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA