Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 7(1): 16222, 2017 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-29176735

RESUMO

In mammals, both sterile wounding and infection induce inflammation and activate the innate immune system, and the combination of both challenges may lead to severe health defects, revealing the importance of the balance between the intensity and resolution of the inflammatory response for the organism's fitness. Underlying mechanisms remain however elusive. Using Drosophila, we show that, upon infection with the entomopathogenic bacterium Pseudomonas entomophila (Pe), a sterile wounding induces a reduced resistance and increased host mortality. To identify the molecular mechanisms underlying the susceptibility of wounded flies to bacterial infection, we analyzed the very first steps of the process by comparing the transcriptome landscape of infected (simple hit flies, SH), wounded and infected (double hit flies, DH) and wounded (control) flies. We observed that overexpressed genes in DH flies compared to SH ones are significantly enriched in genes related to stress, including members of the JNK pathway. We demonstrated that the JNK pathway plays a central role in the DH phenotype by manipulating the Jra/dJun activity. Moreover, the CrebA/Creb3-like transcription factor (TF) and its targets were up-regulated in SH flies and we show that CrebA is required for mounting an appropriate immune response. Drosophila thus appears as a relevant model to investigate interactions between trauma and infection and allows to unravel key pathways involved.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Drosophila/metabolismo , MAP Quinase Quinase 4/metabolismo , Infecções por Pseudomonas/metabolismo , Ferimentos e Lesões/metabolismo , Animais , Drosophila melanogaster , Transdução de Sinais , Transcriptoma , Ferimentos e Lesões/microbiologia
2.
Methods Mol Biol ; 1196: 183-95, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25151164

RESUMO

The Hox family of transcription factors defines cell identity along the A/P axis of animal body plan by modulating expression of distinct sets of target genes in a tissue specific manner. Identifying such tissue specific target genes is indispensable if one wants to understand how Hox proteins mediate their context dependent function. Genome wide analysis of transcriptional activity in different tissues and contexts regarding Hox genes activity could help in reaching this goal. Such experiments rely on the possibility to selectively purify the cells of interest from developing embryos and to perform a transcriptomic analysis on such purified cell populations. By combining expression of a fluorescent protein and fluorescent activating cell sorting (FACS) technique, it is possible to obtain highly purified specific cell populations. In this chapter we describe the experimental procedure we have established in Drosophila-starting from a genetically marked small cell population (cardiomyocytes, 104 cells)-to dissociate the embryos in order to turn it into a suspension of individual cells, sort cells according to the expression of the introduced genetic marker and purify the total RNA content of the sorted cells. This can be used to analyze the transcriptome landscape of rare cell populations in wild type and mutant contexts. This technique has shown to be useful in the case of cardiac cells but is virtually applicable to any cell type and mutant backgrounds, provided that specific genetic markers are available.


Assuntos
Drosophila/genética , Embrião não Mamífero/metabolismo , RNA/isolamento & purificação , Animais , Drosophila/metabolismo , Citometria de Fluxo , Perfilação da Expressão Gênica , Especificidade de Órgãos/genética , RNA/genética , Transcriptoma
3.
J Insect Physiol ; 69: 126-35, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24907675

RESUMO

The intestinal physiology of Drosophila melanogaster can be monitored in an integrative, non-invasive manner by analysing graphical features of the excreta produced by flies fed on a dye-supplemented diet. This assay has been used by various labs to explore gut function and its regulation. To facilitate its use, we present here a free, stand-alone dedicated software tool for the analysis of fly excreta. The Ultimate Reader of Dung (T.U.R.D.) is designed to offer a flexible environment for a wide range of experimental designs, with special attention to automation and high-throughput processing. This software detects the distinctive changes in acid-base and water balance previously reported to occur in response to dietary challenges and mating. We have used T.U.R.D. to test the contribution of the bacterial environment of the flies to various intestinal parameters including the established diet- and mating-triggered responses. To this end, we have analysed the faecal patterns of flies reared in germ-free conditions, upon re-association with controlled microbiota and subjected to food-borne or systemic, non-lethal bacterial infections. We find that the tested faecal outputs are unchanged in all these conditions, suggesting that the impact of the bacterial environment on the intestinal features highlighted by faecal deposit analysis is minimal.


Assuntos
Defecação , Drosophila/microbiologia , Fezes , Vida Livre de Germes/fisiologia , Microbiota , Animais , Dieta , Drosophila/fisiologia , Feminino , Interações Hospedeiro-Patógeno , Masculino , Comportamento Sexual Animal , Software , Simbiose
4.
PLoS One ; 9(4): e94729, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24733183

RESUMO

Most metazoans engage in mutualistic interactions with their intestinal microbiota. Despite recent progress the molecular mechanisms through which microbiota exerts its beneficial influences on host physiology are still largely uncharacterized. Here we use axenic Drosophila melanogaster adults associated with a standardized microbiota composed of a defined set of commensal bacterial strains to study the impact of microbiota association on its host transcriptome. Our results demonstrate that Drosophila microbiota has a marked impact on the midgut transcriptome and promotes the expression of genes involved in host digestive functions and primary metabolism. We identify the IMD/Relish signaling pathway as a central regulator of this microbiota-mediated transcriptional response and we reveal a marked transcriptional trade-off between the midgut response to its beneficial microbiota and to bacterial pathogens. Taken together our results indicate that microbiota association potentiates host nutrition and host metabolic state, two key physiological parameters influencing host fitness. Our work paves the way to subsequent mechanistic studies to reveal how these microbiota-dependent transcriptional signatures translate into host physiological benefits.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/microbiologia , Regulação da Expressão Gênica , Microbiota , Fatores de Transcrição/metabolismo , Animais , Drosophila melanogaster/imunologia , Feminino , Sistema Imunitário , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Fenótipo , Transdução de Sinais , Transcriptoma
5.
Cell Host Microbe ; 13(1): 8-14, 2013 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-23332152

RESUMO

Given the complexity of the mammalian microbiota, there is a need for simple models to decipher the effector and regulatory mechanisms underlying host/microbiota mutualism. Approaches using Drosophila and its simple microbiota carry the potential to unravel the evolutionarily conserved mechanisms engaged in this association. Here, we review recent work carried out in this model, providing insights and exciting perspectives.


Assuntos
Drosophila/microbiologia , Drosophila/fisiologia , Metagenoma , Simbiose , Animais , Bactérias/isolamento & purificação , Fenômenos Fisiológicos Bacterianos , Intestinos/microbiologia
6.
Cell Metab ; 14(3): 403-14, 2011 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-21907145

RESUMO

There is growing evidence that intestinal bacteria are important beneficial partners of their metazoan hosts. Recent observations suggest a strong link between commensal bacteria, host energy metabolism, and metabolic diseases such as diabetes and obesity. As a consequence, the gut microbiota is now considered a "host" factor that influences energy uptake. However, the impact of intestinal bacteria on other systemic physiological parameters still remains unclear. Here, we demonstrate that Drosophila microbiota promotes larval growth upon nutrient scarcity. We reveal that Lactobacillus plantarum, a commensal bacterium of the Drosophila intestine, is sufficient on its own to recapitulate the natural microbiota growth-promoting effect. L. plantarum exerts its benefit by acting genetically upstream of the TOR-dependent host nutrient sensing system controlling hormonal growth signaling. Our results indicate that the intestinal microbiota should also be envisaged as a factor that influences the systemic growth of its host.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Mucosa Intestinal/metabolismo , Lactobacillus plantarum/fisiologia , Larva/crescimento & desenvolvimento , Transdução de Sinais/fisiologia , Simbiose/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Carga Bacteriana , Drosophila melanogaster/metabolismo , Metabolismo Energético/fisiologia , Feminino , Privação de Alimentos , Regulação da Expressão Gênica no Desenvolvimento , Intestinos/microbiologia , Larva/metabolismo , Serina-Treonina Quinases TOR/genética
7.
J Innate Immun ; 1(4): 322-34, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20375589

RESUMO

Drosophila phagocytes participate in development and immune responses through their abilities to perform phagocytosis and/or secrete extra-cellular matrix components, antimicrobial peptides, clotting factors and signalling molecules. However, our knowledge of their functional impact on development and host resistance to infection is limited. To address this, we have used a genetic cell ablation strategy to generate Drosophila individuals lacking functional phagocytes. Our results highlight the essential contribution of phagocytes to embryonic development including central nervous system morphogenesis. Phagocytes also ensure optimal viability during post-embryonic development through immune functions. The use of phagocyte-depleted flies reveals the contribution of phagocytes in the resistance of Drosophila adults upon systemic infections with specific bacteria. Phagocytes were not involved in the expression of antimicrobial peptides by the fat body indicating a clear separation between cellular and humoral immune responses at this stage. Finally, we confirm that phagocytosis is a critical effector mechanism of the cellular arm by demonstrating that phagocytosis contributes to resistance to infection with Staphylococcus aureus in adults. Our results highlight the power of this cell ablation strategy to reveal the contribution of phagocytes to specific biological processes. We now provide a blueprint of phagocyte importance during both development and innate immune responses in Drosophila.


Assuntos
Drosophila melanogaster/imunologia , Drosophila melanogaster/microbiologia , Fagócitos/imunologia , Fagocitose/imunologia , Animais , Sistema Nervoso Central/embriologia , Drosophila melanogaster/embriologia , Feminino , Fagocitose/genética , Fagossomos/genética , Fagossomos/imunologia , Staphylococcus aureus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...