Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 382
Filtrar
1.
Sci Immunol ; 9(97): eadm7908, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38996009

RESUMO

Infections and neurodegenerative diseases induce neuroinflammation, but affected individuals often show nonneural symptoms including muscle pain and muscle fatigue. The molecular pathways by which neuroinflammation causes pathologies outside the central nervous system (CNS) are poorly understood. We developed multiple models to investigate the impact of CNS stressors on motor function and found that Escherichia coli infections and SARS-CoV-2 protein expression caused reactive oxygen species (ROS) to accumulate in the brain. ROS induced expression of the cytokine Unpaired 3 (Upd3) in Drosophila and its ortholog, IL-6, in mice. CNS-derived Upd3/IL-6 activated the JAK-STAT pathway in skeletal muscle, which caused muscle mitochondrial dysfunction and impaired motor function. We observed similar phenotypes after expressing toxic amyloid-ß (Aß42) in the CNS. Infection and chronic disease therefore activate a systemic brain-muscle signaling axis in which CNS-derived cytokines bypass the connectome and directly regulate muscle physiology, highlighting IL-6 as a therapeutic target to treat disease-associated muscle dysfunction.


Assuntos
Encéfalo , COVID-19 , Músculo Esquelético , Transdução de Sinais , Animais , Encéfalo/imunologia , Encéfalo/metabolismo , Transdução de Sinais/imunologia , Camundongos , Músculo Esquelético/imunologia , Músculo Esquelético/metabolismo , COVID-19/imunologia , Doença Crônica , Interleucina-6/metabolismo , Interleucina-6/imunologia , Infecções por Escherichia coli/imunologia , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/imunologia , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/imunologia , Proteínas de Drosophila/genética , SARS-CoV-2/imunologia , Drosophila melanogaster/imunologia , Peptídeos beta-Amiloides/metabolismo , Humanos , Camundongos Endogâmicos C57BL
2.
PLoS Pathog ; 20(6): e1012308, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38857285

RESUMO

Invertebrates lack the immune machinery underlying vertebrate-like acquired immunity. However, in many insects past infection by the same pathogen can 'prime' the immune response, resulting in improved survival upon reinfection. Here, we investigated the mechanistic basis and epidemiological consequences of innate immune priming in the fruit fly Drosophila melanogaster when infected with the gram-negative bacterial pathogen Providencia rettgeri. We find that priming in response to P. rettgeri infection is a long-lasting and sexually dimorphic response. We further explore the epidemiological consequences of immune priming and find it has the potential to curtail pathogen transmission by reducing pathogen shedding and spread. The enhanced survival of individuals previously exposed to a non-lethal bacterial inoculum coincided with a transient decrease in bacterial loads, and we provide strong evidence that the effect of priming requires the IMD-responsive antimicrobial-peptide Diptericin-B in the fat body. Further, we show that while Diptericin B is the main effector of bacterial clearance, it is not sufficient for immune priming, which requires regulation of IMD by peptidoglycan recognition proteins. This work underscores the plasticity and complexity of invertebrate responses to infection, providing novel experimental evidence for the effects of innate immune priming on population-level epidemiological outcomes.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Imunidade Inata , Providencia , Animais , Drosophila melanogaster/microbiologia , Drosophila melanogaster/imunologia , Providencia/imunologia , Proteínas de Drosophila/imunologia , Feminino , Masculino , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/transmissão , Peptídeos Antimicrobianos
3.
Cell Mol Life Sci ; 81(1): 230, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38780625

RESUMO

Insect host defense comprises two complementary dimensions, microbial killing-mediated resistance and microbial toxin neutralization-mediated resilience, both jointly providing protection against pathogen infections. Insect defensins are a class of effectors of innate immunity primarily responsible for resistance to Gram-positive bacteria. Here, we report a newly originated gene from an ancestral defensin via genetic deletion following gene duplication in Drosophila virilis, which confers an enhanced resilience to Gram-positive bacterial infection. This gene encodes an 18-mer arginine-rich peptide (termed DvirARP) with differences from its parent gene in its pattern of expression, structure and function. DvirARP specifically expresses in D. virilis female adults with a constitutive manner. It adopts a novel fold with a 310 helix and a two CXC motif-containing loop stabilized by two disulfide bridges. DvirARP exhibits no activity on the majority of microorganisms tested and only a weak activity against two Gram-positive bacteria. DvirARP knockout flies are viable and have no obvious defect in reproductivity but they are more susceptible to the DvirARP-resistant Staphylococcus aureus infection than the wild type files, which can be attributable to its ability in neutralization of the S. aureus secreted toxins. Phylogenetic distribution analysis reveals that DvirARP is restrictedly present in the Drosophila subgenus, but independent deletion variations also occur in defensins from the Sophophora subgenus, in support of the evolvability of this class of immune effectors. Our work illustrates for the first time how a duplicate resistance-mediated gene evolves an ability to increase the resilience of a subset of Drosophila species against bacterial infection.


Assuntos
Defensinas , Proteínas de Drosophila , Drosophila , Drosophila/classificação , Drosophila/genética , Drosophila/imunologia , Drosophila/microbiologia , Defensinas/química , Defensinas/genética , Defensinas/imunologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/imunologia , Animais , Deleção de Genes , Duplicação Gênica , Feminino , Dobramento de Proteína , Motivos de Aminoácidos , Toxinas Bacterianas/metabolismo , Staphylococcus aureus/fisiologia
4.
J Immunol ; 208(8): 1978-1988, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35379744

RESUMO

The Drosophila Toll signaling pathway mainly responds to Gram-positive (G+) bacteria or fungal infection, which is highly conserved with mammalian TLR signaling pathway. Although many positive and negative regulators involved in the immune response of the Toll pathway have been identified in Drosophila, the roles of long noncoding RNAs (lncRNAs) in Drosophila Toll immune responses are poorly understood to date. In this study, our results demonstrate that lncRNA-CR33942 is mainly expressed in the nucleus and upregulated after Micrococcus luteus infection. Especially, lncRNA-CR33942 not only modulates differential expressions of multiple antimicrobial peptide genes but also affects the Drosophila survival rate during response to G+ bacterial infection based on the transiently overexpressing and the knockdown lncRNA-CR33942 assays in vivo. Mechanically, lncRNA-CR33942 interacts with the NF-κB transcription factors Dorsal-related immunity factor/Dorsal to promote the transcriptions of antimicrobial peptides drosomycin and metchnikowin, thus enhancing Drosophila Toll immune responses. Taken together, this study identifies lncRNA-CR33942 as a positive regulator of Drosophila innate immune response to G+ bacterial infection to facilitate Toll signaling via interacting with Dorsal-related immunity factor/Dorsal. It would be helpful to reveal the roles of lncRNAs in Toll immune response in Drosophila and provide insights into animal innate immunity.


Assuntos
Peptídeos Antimicrobianos , Proteínas de Drosophila , Drosophila , RNA Longo não Codificante , Animais , Peptídeos Antimicrobianos/genética , Peptídeos Antimicrobianos/imunologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Drosophila/genética , Drosophila/imunologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/imunologia , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Imunidade Inata/genética , Imunidade Inata/imunologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/imunologia , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo
5.
Cell Rep ; 38(4): 110286, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35081354

RESUMO

Selective autophagy is a catabolic route that turns over specific cellular material for degradation by lysosomes, and whose role in the regulation of innate immunity is largely unexplored. Here, we show that the apical kinase of the Drosophila immune deficiency (IMD) pathway Tak1, as well as its co-activator Tab2, are both selective autophagy substrates that interact with the autophagy protein Atg8a. We also present a role for the Atg8a-interacting protein Sh3px1 in the downregulation of the IMD pathway, by facilitating targeting of the Tak1/Tab2 complex to the autophagy platform through its interaction with Tab2. Our findings show the Tak1/Tab2/Sh3px1 interactions with Atg8a mediate the removal of the Tak1/Tab2 signaling complex by selective autophagy. This in turn prevents constitutive activation of the IMD pathway in Drosophila. This study provides mechanistic insight on the regulation of innate immune responses by selective autophagy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Autofagia/imunologia , Proteínas de Drosophila/imunologia , Imunidade Inata/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , MAP Quinase Quinase Quinases/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Transdução de Sinais/imunologia
6.
Elife ; 112022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35076390

RESUMO

Expansion of the available repertoire of reagents for visualization and manipulation of proteins will help understand their function. Short epitope tags linked to proteins of interest and recognized by existing binders such as nanobodies facilitate protein studies by obviating the need to isolate new antibodies directed against them. Nanobodies have several advantages over conventional antibodies, as they can be expressed and used as tools for visualization and manipulation of proteins in vivo. Here, we characterize two short (<15aa) NanoTag epitopes, 127D01 and VHH05, and their corresponding high-affinity nanobodies. We demonstrate their use in Drosophila for in vivo protein detection and re-localization, direct and indirect immunofluorescence, immunoblotting, and immunoprecipitation. We further show that CRISPR-mediated gene targeting provides a straightforward approach to tagging endogenous proteins with the NanoTags. Single copies of the NanoTags, regardless of their location, suffice for detection. This versatile and validated toolbox of tags and nanobodies will serve as a resource for a wide array of applications, including functional studies in Drosophila and beyond.


Assuntos
Proteínas de Drosophila/imunologia , Drosophila melanogaster/imunologia , Epitopos/imunologia , Anticorpos de Domínio Único/metabolismo , Animais
7.
PLoS Genet ; 17(8): e1009718, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34370736

RESUMO

Insects depend on the innate immune response for defense against a wide array of pathogens. Central to Drosophila immunity are antimicrobial peptides (AMPs), released into circulation when pathogens trigger either of the two widely studied signal pathways, Toll or IMD. The Toll pathway responds to infection by Gram-positive bacteria and fungi while the IMD pathway is activated by Gram-negative bacteria. During activation of the IMD pathway, the NF-κB-like transcription factor Relish is phosphorylated and then cleaved, which is crucial for IMD-dependent AMP gene induction. Here we show that loss-of-function mutants of the unconventional histone variant H2Av upregulate IMD-dependent AMP gene induction in germ-free Drosophila larvae and adults. After careful dissection of the IMD pathway, we found that Relish has an epistatic relationship with H2Av. In the H2Av mutant larvae, SUMOylation is down-regulated, suggesting a possible role of SUMOylation in the immune phenotype. Eventually we demonstrated that Relish is mostly SUMOylated on amino acid K823. Loss of the potential SUMOylation site leads to significant auto-activation of Relish in vivo. Further work indicated that H2Av regulates Relish SUMOylation after physically interacting with Su(var)2-10, the E3 component of the SUMOylation pathway. Biochemical analysis suggested that SUMOylation of Relish prevents its cleavage and activation. Our findings suggest a new mechanism by which H2Av can negatively regulate, and thus prevent spontaneous activation of IMD-dependent AMP production, through facilitating SUMOylation of the NF-κB like transcription factor Relish.


Assuntos
Proteínas de Drosophila/metabolismo , Histonas/metabolismo , Imunidade Inata/genética , Fatores de Transcrição/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/imunologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Histonas/fisiologia , NF-kappa B/genética , Transdução de Sinais/genética , Sumoilação/genética , Receptores Toll-Like , Fatores de Transcrição/genética
8.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34341118

RESUMO

Male and female animals exhibit differences in infection outcomes. One possible source of sexually dimorphic immunity is the sex-specific costs of immune activity or pathology, but little is known about the independent effects of immune- versus microbe-induced pathology and whether these may differ for the sexes. Here, by measuring metabolic and physiological outputs in Drosophila melanogaster with wild-type and mutant immune responses, we test whether the sexes are differentially impacted by these various sources of pathology and identify a critical regulator of this difference. We find that the sexes exhibit differential immune activity but similar bacteria-derived metabolic pathology. We show that female-specific immune-inducible expression of PGRP-LB, a negative regulator of the immune deficiency (IMD) pathway, enables females to reduce immune activity in response to reductions in bacterial numbers. In the absence of PGRP-LB, females are more resistant to infection, confirming the functional importance of this regulation and suggesting that female-biased immune restriction comes at a cost.


Assuntos
Proteínas de Transporte/imunologia , Drosophila melanogaster/imunologia , Drosophila melanogaster/microbiologia , Animais , Proteínas de Transporte/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/imunologia , Drosophila melanogaster/genética , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/veterinária , Feminino , Regulação da Expressão Gênica , Imunidade Inata/fisiologia , Masculino , Mutação , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Fatores Sexuais , Transdução de Sinais/fisiologia , Triglicerídeos/metabolismo
9.
Viruses ; 13(8)2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34452330

RESUMO

Eukaryotic nucleic acid methyltransferase (MTase) proteins are essential mediators of epigenetic and epitranscriptomic regulation. DNMT2 belongs to a large, conserved family of DNA MTases found in many organisms, including holometabolous insects such as fruit flies and mosquitoes, where it is the lone MTase. Interestingly, despite its nomenclature, DNMT2 is not a DNA MTase, but instead targets and methylates RNA species. A growing body of literature suggests that DNMT2 mediates the host immune response against a wide range of pathogens, including RNA viruses. Curiously, although DNMT2 is antiviral in Drosophila, its expression promotes virus replication in mosquito species. We, therefore, sought to understand the divergent regulation, function, and evolution of these orthologs. We describe the role of the Drosophila-specific host protein IPOD in regulating the expression and function of fruit fly DNMT2. Heterologous expression of these orthologs suggests that DNMT2's role as an antiviral is host-dependent, indicating a requirement for additional host-specific factors. Finally, we identify and describe potential evidence of positive selection at different times throughout DNMT2 evolution within dipteran insects. We identify specific codons within each ortholog that are under positive selection and find that they are restricted to four distinct protein domains, which likely influence substrate binding, target recognition, and adaptation of unique intermolecular interactions. Collectively, our findings highlight the evolution of DNMT2 in Dipteran insects and point to structural, regulatory, and functional differences between mosquito and fruit fly homologs.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Dípteros/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/enzimologia , Drosophila melanogaster/microbiologia , Interações Hospedeiro-Patógeno , Wolbachia/fisiologia , Adaptação Biológica , Aedes/enzimologia , Aedes/genética , Aedes/imunologia , Aedes/microbiologia , Sequência de Aminoácidos , Animais , DNA (Citosina-5-)-Metiltransferases/química , DNA (Citosina-5-)-Metiltransferases/imunologia , Dípteros/classificação , Dípteros/enzimologia , Dípteros/imunologia , Proteínas de Drosophila/química , Proteínas de Drosophila/imunologia , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Evolução Molecular , Filogenia , Conformação Proteica , Alinhamento de Sequência , Wolbachia/genética
10.
Dev Comp Immunol ; 124: 104183, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34174242

RESUMO

The Toll signaling pathway is highly conserved from insects to mammals. Drosophila is a model species that is commonly used to study innate immunity. Although many studies have assessed protein-coding genes that regulate the Toll pathway, it is unclear whether long noncoding RNAs (lncRNAs) play regulatory roles in the Toll pathway. Here, we evaluated the expression of the lncRNA CR46018 in Drosophila. Our results showed that this lncRNA was significantly overexpressed after infection of Drosophila with Micrococcus luteus. A CR46018-overexpressing Drosophila strain was then constructed; we expected that CR46018 overexpression would enhance the expression of various antimicrobial peptides downstream of the Toll pathway, regardless of infection with M. luteus. RNA-seq analysis of CR46018-overexpressing Drosophila after infection with M. luteus showed that upregulated genes were mainly enriched in Toll and Imd signaling pathways. Moreover, bioinformatics predictions and RNA-immunoprecipitation experiments showed that CR46018 interacted with the transcription factors Dif and Dorsal to enhance the Toll pathway. During gram-positive bacterial infection, flies overexpressing CR46018 showed favorable survival compared with flies in the control group. Overall, our current work not only reveals a new immune regulatory factor, lncRNA-CR46018, and explores its potential regulatory model, but also provides a new perspective for the effect of immune disorders on the survival of Drosophila melanogaster.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/imunologia , Proteínas Nucleares/genética , Fosfoproteínas/genética , RNA Longo não Codificante/genética , Receptores Toll-Like/imunologia , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/imunologia , Drosophila melanogaster/genética , Drosophila melanogaster/microbiologia , Infecções por Bactérias Gram-Positivas/genética , Infecções por Bactérias Gram-Positivas/imunologia , Infecções por Bactérias Gram-Positivas/microbiologia , Imunidade Inata , Micrococcus luteus/fisiologia , RNA Longo não Codificante/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptores Toll-Like/genética
11.
Dev Comp Immunol ; 117: 103985, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33358662

RESUMO

The fruitfly Drosophila melanogaster is a valuable model to unravel mechanisms of innate immunity, in particular in the context of viral infections. RNA interference, and more specifically the small interfering RNA pathway, is a major component of antiviral immunity in drosophila. In addition, the contribution of inducible transcriptional responses to the control of viruses in drosophila and other invertebrates is increasingly recognized. In particular, the recent discovery of a STING-IKKß-Relish signalling cassette in drosophila has confirmed that NF-κB transcription factors play an important role in the control of viral infections, in addition to bacterial and fungal infections. Here, we review recent developments in the field, which begin to shed light on the mechanisms involved in sensing of viral infections and in signalling leading to production of antiviral effectors.


Assuntos
Proteínas de Drosophila/imunologia , Drosophila melanogaster/imunologia , Imunidade Inata/imunologia , Interferência de RNA/imunologia , Transdução de Sinais/imunologia , Vírus/imunologia , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/virologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Quinase I-kappa B/genética , Quinase I-kappa B/imunologia , Quinase I-kappa B/metabolismo , Imunidade Inata/genética , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , RNA Viral/genética , RNA Viral/imunologia , RNA Viral/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo , Vírus/genética
12.
Elife ; 92020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33372660

RESUMO

Studies in different animal model systems have revealed the impact of odors on immune cells; however, any understanding on why and how odors control cellular immunity remained unclear. We find that Drosophila employ an olfactory-immune cross-talk to tune a specific cell type, the lamellocytes, from hematopoietic-progenitor cells. We show that neuronally released GABA derived upon olfactory stimulation is utilized by blood-progenitor cells as a metabolite and through its catabolism, these cells stabilize Sima/HIFα protein. Sima capacitates blood-progenitor cells with the ability to initiate lamellocyte differentiation. This systemic axis becomes relevant for larvae dwelling in wasp-infested environments where chances of infection are high. By co-opting the olfactory route, the preconditioned animals elevate their systemic GABA levels leading to the upregulation of blood-progenitor cell Sima expression. This elevates their immune-potential and primes them to respond rapidly when infected with parasitic wasps. The present work highlights the importance of the olfaction in immunity and shows how odor detection during animal development is utilized to establish a long-range axis in the control of blood-progenitor competency and immune-priming.


Assuntos
Fenômenos Bioquímicos/imunologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Células-Tronco Hematopoéticas/citologia , Hemócitos/citologia , Animais , Drosophila/imunologia , Drosophila/metabolismo , Proteínas de Drosophila/imunologia , Drosophila melanogaster/imunologia , Hematopoese/imunologia , Larva/metabolismo , Vespas/imunologia
13.
PLoS Genet ; 16(6): e1008861, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32525870

RESUMO

In metazoan germlines, the piRNA pathway acts as a genomic immune system, employing small RNA-mediated silencing to defend host DNA from the harmful effects of transposable elements (TEs). Expression of genomic TEs is proposed to initiate self regulation by increasing the production of repressive piRNAs, thereby "adapting" piRNA-mediated control to the most active TE families. Surprisingly, however, piRNA pathway proteins, which execute piRNA biogenesis and enforce silencing of targeted sequences, evolve rapidly and adaptively in animals. If TE silencing is ensured through piRNA biogenesis, what necessitates changes in piRNA pathway proteins? Here we used interspecific complementation to test for functional differences between Drosophila melanogaster and D. simulans alleles of three adaptively evolving piRNA pathway proteins: Armitage, Aubergine and Spindle-E. In contrast to piRNA-mediated transcriptional regulators examined in previous studies, these three proteins have cytoplasmic functions in piRNA maturation and post-transcriptional silencing. Across all three proteins we observed interspecific divergence in the regulation of only a handful of TE families, which were more robustly silenced by the heterospecific piRNA pathway protein. This unexpected result suggests that unlike transcriptional regulators, positive selection has not acted on cytoplasmic piRNA effector proteins to enhance their function in TE repression. Rather, TEs may evolve to "escape" silencing by host proteins. We further discovered that D. simulans alleles of aub and armi exhibit enhanced off-target effects on host transcripts in a D. melanogaster background, as well as modest reductions in the efficiency of piRNA biogenesis, suggesting that promiscuous binding of D. simulans Aub and Armi proteins to host transcripts reduces their participation in piRNA production. Avoidance of genomic auto-immunity may therefore be a critical target of selection. Our observations suggest that piRNA effector proteins are subject to an evolutionary trade-off between defending the host genome from the harmful effect of TEs while also minimizing collateral damage to host genes.


Assuntos
Autoimunidade/genética , Elementos de DNA Transponíveis/imunologia , Drosophila simulans/genética , Evolução Molecular , Genoma de Inseto/imunologia , RNA Interferente Pequeno/biossíntese , Alelos , Animais , Animais Geneticamente Modificados , Citoplasma/genética , Citoplasma/metabolismo , Elementos de DNA Transponíveis/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/imunologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Drosophila melanogaster/metabolismo , Drosophila simulans/metabolismo , Feminino , Regulação da Expressão Gênica/imunologia , Genoma de Inseto/genética , Masculino , Mutação , Interferência de RNA/imunologia
14.
Sheng Wu Gong Cheng Xue Bao ; 36(6): 1198-1208, 2020 Jun 25.
Artigo em Chinês | MEDLINE | ID: mdl-32597069

RESUMO

In this research, we studied the formation of Drosophila melanogaster FADD (Fas-associated death domain-containing protein) amyloid fiber and its influence on signal transduction in IMD (Immune deficiency) signaling pathway to better understand the regulation mechanism of Drosophila innate immune signaling pathway, which will provide reference for the immune regulation in other species. First, we purified dFADD protein expressed in Escherichia coli and performed Sulfur flavin T binding and transmission electron microscopy to identify the dFADD amyloid fibers formed in vitro. Then we investigated the formation of dFADD polymers in S2 cells using SDD-AGE and confocal microscope. We also constructed dFADD mutants to find out which domain is essential to fiber formation and its effect on IMD signal transduction. Our results revealed that dFADD could be polymerized to form amyloid fiber polymers in vitro and inside the cells. Formation of fibers relies on DED (Death-effector domain) domain of dFADD, since DED domain-deleted mutant existed as a monomer. Dual luciferase reporter assay showed that intact DED domain was required for the induction of downstream antimicrobial peptides, indicating that fiber formation was the key to IMD signal transduction. Our study revealed the role of dFADD in mediating the cascade between IMD and Dredd in the IMD signaling pathway by forming amyloid fibers, suggesting an evolutionarily conserved regulatory mechanism of innate immune signaling pathway.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Imunidade Inata , Transdução de Sinais , Animais , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/imunologia , Drosophila melanogaster/imunologia , Proteína de Domínio de Morte Associada a Fas/biossíntese , Proteína de Domínio de Morte Associada a Fas/imunologia , Imunidade Inata/imunologia
16.
PLoS Pathog ; 16(4): e1008458, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32339205

RESUMO

The Immune Deficiency (IMD) pathway in Drosophila melanogaster is activated upon microbial challenge with Gram-negative bacteria to trigger the innate immune response. In order to decipher this nuclear factor κB (NF-κB) signaling pathway, we undertook an in vitro RNAi screen targeting E3 ubiquitin ligases specifically and identified the HECT-type E3 ubiquitin ligase Hyperplastic discs (Hyd) as a new actor in the IMD pathway. Hyd mediated Lys63 (K63)-linked polyubiquitination of the NF-κB cofactor Akirin was required for efficient binding of Akirin to the NF-κB transcription factor Relish. We showed that this Hyd-dependent interaction was required for the transcription of immunity-related genes that are activated by both Relish and Akirin but was dispensable for the transcription of genes that depend solely on Relish. Therefore Hyd is key in NF-κB transcriptional selectivity downstream of the IMD pathway. Drosophila depleted of Akirin or Hyd failed to express the full set of genes encoding immune-induced anti-microbial peptides and succumbed to immune challenges. We showed further that UBR5, the mammalian homolog of Hyd, was also required downstream of the NF-κB pathway for the activation of Interleukin 6 (IL6) transcription by LPS or IL-1ß in cultured human cells. Our findings link the action of an E3 ubiquitin ligase to the activation of immune effector genes, deepening our understanding of the involvement of ubiquitination in inflammation and identifying a potential target for the control of inflammatory diseases.


Assuntos
Proteínas de Drosophila/imunologia , Drosophila melanogaster/imunologia , Proteínas Nucleares/imunologia , Fatores de Transcrição/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Drosophila , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/microbiologia , Bactérias Gram-Negativas/fisiologia , Células HeLa , Humanos , Imunidade Inata , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
17.
Int J Mol Sci ; 21(7)2020 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-32244587

RESUMO

Peptidoglycan recognition proteins (PGRPs) play an important role in the defense against invading microbes via the recognition of the immunogenic substance peptidoglycan (PGN). Bees possess fewer PGRPs than Drosophila melanogaster and Anopheles gambiae but retain two important immune pathways, the Toll pathway and the Imd pathway, which can be triggered by the recognition of Dap-type PGN by PGRP-LCx with the assistance of PGRP-LCa in Drosophila. There are three isoforms of PGRP-LC including PGRP-LCx, PGRP-LCa and PGRP-LCy in Drosophila. Our previous study showed that a single PGRP-LC exists in bumblebees. In this present study, we prove that the bumblebee Bombus lantschouensis PGRP-LC (Bl-PGRP-LC) can respond to an infection with Gram-negative bacterium Escherichia coli through binding to the Dap-type PGNs directly, and that E. coli infection induces the quick and strong upregulation of PGRP-LC, abaecin and defensin. Moreover, the Bl-PGRP-LC exhibits a very strong affinity for the Dap-type PGN, much stronger than the affinity exhibited by the PGRP-LC from the more eusocial honeybee Apis mellifera (Am-PGRP-LC). In addition, mutagenesis experiments showed that the residue His390 is the anchor residue for the binding to the Dap-type PGN and forms a hydrogen bond with MurNAc rather than meso-Dap, which interacts with the anchor residue Arg413 of PGRP-LCx in Drosophila. Therefore, bumblebee PGRP-LC possesses exclusive characteristics for the immune response among insect PGRPs.


Assuntos
Abelhas/imunologia , Proteínas de Transporte/imunologia , Escherichia coli/imunologia , Peptidoglicano/imunologia , Sequência de Aminoácidos , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/imunologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Abelhas/química , Abelhas/microbiologia , Proteínas de Transporte/classificação , Proteínas de Transporte/genética , Defensinas/genética , Defensinas/imunologia , Defensinas/metabolismo , Proteínas de Drosophila/imunologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Drosophila melanogaster/metabolismo , Escherichia coli/fisiologia , Bactérias Gram-Negativas/classificação , Bactérias Gram-Negativas/imunologia , Bactérias Gram-Negativas/fisiologia , Bactérias Gram-Positivas/classificação , Bactérias Gram-Positivas/imunologia , Bactérias Gram-Positivas/fisiologia , Proteínas de Insetos/genética , Proteínas de Insetos/imunologia , Proteínas de Insetos/metabolismo , Peptidoglicano/química , Peptidoglicano/metabolismo , Ligação Proteica , Conformação Proteica , Isoformas de Proteínas/classificação , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Homologia de Sequência de Aminoácidos
18.
Infect Genet Evol ; 82: 104308, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32240802

RESUMO

Aspergilloses are opportunistic infections in animals and humans caused by several Aspergillus species, including Aspergillus flavus. Although the immune system of Drosophila melanogaster is extensively studied, little is known about the fly's specific responses to infection by A. flavus. We compared gene expression levels during induced infections in D. melanogaster by a virulent A. flavus isolate and a less virulent isolate, as well as from uninfected flies as a control. We found that 1081 of the 14,554 gene regions detected were significantly differentially expressed among treatments. Some of these up- and down- regulated genes were previously shown to be involved in defense responses against pathogens. Some are known to be involved in vitelline membrane formation in flies. Other up- and down-regulated genes are of unknown function. Understanding expression of these genes during the process of infection in flies should improve our knowledge of innate immunity in invertebrates, and by extension, in vertebrates as well.


Assuntos
Aspergillus flavus/patogenicidade , Proteínas de Drosophila/imunologia , Drosophila melanogaster/imunologia , Drosophila melanogaster/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/genética , Imunidade Inata/genética
19.
J Immunol ; 204(8): 2143-2155, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32198143

RESUMO

Negative regulation of innate immunity is essential to avoid autoinflammation. In Drosophila melanogaster, NF-κB signaling-mediated immune responses are negatively regulated at multiple levels. Using a Drosophila RNA interference in vitro screen, we identified a set of genes inhibiting immune activation. Four of these genes encode members of the chromatin remodeling Osa-containing Brahma (BAP) complex. Silencing additional two genes of the BAP complex was shown to have the same phenotype, confirming its role in immune regulation in vitro. In vivo, the knockdown of osa and brahma was shown to enhance the expression of the Toll pathway-mediated antimicrobial peptides when the flies were challenged with Gram-positive bacteria Micrococcus luteus In this setting, osa knockdown had a particularly strong effect on immune effectors that are predominantly activated by the Imd pathway. Accordingly, Drosophila NF-κB Relish expression was increased by osa silencing. These transcriptional changes were associated with enhanced survival from M. luteus + E. faecalis infection. Besides regulating the expression of immune effector genes, osa RNA interference decreased the expression of a large group of genes involved in metabolism, particularly proteolysis. Of note, the expression of the recently characterized, immune-inducible gene Induced by Infection (IBIN) was diminished in osa knockdown flies. Although IBIN has been shown to modulate metabolism upon infection, the expression of selected Osa-regulated metabolism genes was not rescued by overexpressing IBIN. We conclude that the BAP complex regulates expression of genes involved in metabolism at least partially independent or downstream of IBIN Moreover, Osa affects the NF-κB-mediated immune response by regulating Drosophila NF-κB factor Relish expression.


Assuntos
Proteínas de Ciclo Celular/imunologia , Proteínas de Ligação a DNA/imunologia , Proteínas de Drosophila/imunologia , Drosophila melanogaster/genética , Drosophila melanogaster/imunologia , Imunidade Inata/imunologia , Transativadores/imunologia , Fatores de Transcrição/imunologia , Animais , Proteínas de Ciclo Celular/genética , Células Cultivadas , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , NF-kappa B/imunologia , Transativadores/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
20.
Proc Natl Acad Sci U S A ; 117(13): 7317-7325, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32188787

RESUMO

Iron sequestration is a recognized innate immune mechanism against invading pathogens mediated by iron-binding proteins called transferrins. Despite many studies on antimicrobial activity of transferrins in vitro, their specific in vivo functions are poorly understood. Here we use Drosophila melanogaster as an in vivo model to investigate the role of transferrins in host defense. We find that systemic infections with a variety of pathogens trigger a hypoferremic response in flies, namely, iron withdrawal from the hemolymph and accumulation in the fat body. Notably, this hypoferremia to infection requires Drosophila nuclear factor κB (NF-κB) immune pathways, Toll and Imd, revealing that these pathways also mediate nutritional immunity in flies. Next, we show that the iron transporter Tsf1 is induced by infections downstream of the Toll and Imd pathways and is necessary for iron relocation from the hemolymph to the fat body. Consistent with elevated iron levels in the hemolymph, Tsf1 mutants exhibited increased susceptibility to Pseudomonas bacteria and Mucorales fungi, which could be rescued by chemical chelation of iron. Furthermore, using siderophore-deficient Pseudomonas aeruginosa, we discover that the siderophore pyoverdine is necessary for pathogenesis in wild-type flies, but it becomes dispensable in Tsf1 mutants due to excessive iron present in the hemolymph of these flies. As such, our study reveals that, similar to mammals, Drosophila uses iron limitation as an immune defense mechanism mediated by conserved iron-transporting proteins transferrins. Our in vivo work, together with accumulating in vitro studies, supports the immune role of insect transferrins against infections via an iron withholding strategy.


Assuntos
Proteínas de Drosophila/metabolismo , Ferro/metabolismo , Transferrina/metabolismo , Animais , Proteínas de Drosophila/imunologia , Drosophila melanogaster , Hemolinfa/imunologia , Hemolinfa/metabolismo , Imunidade Inata , Ferro/imunologia , NF-kappa B/metabolismo , Pseudomonas aeruginosa/metabolismo , Sideróforos/metabolismo , Transferrina/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA