Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 20(6): e1012252, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38833496

RESUMEN

Microbial pathogenicity often depends on the route of infection. For instance, P. aeruginosa or S. marcescens cause acute systemic infections when low numbers of bacteria are injected into D. melanogaster flies whereas flies succumb much slower to the continuous ingestion of these pathogens, even though both manage to escape from the gut compartment and reach the hemocoel. Here, we have developed a latent P. aeruginosa infection model by feeding flies on the bacteria for a short period. The bacteria stably colonize internal tissues yet hardly cause any damage since latently-infected flies live almost as long as noninfected control flies. The apparently dormant bacteria display particular characteristics in terms of bacterial colony morphology, composition of the outer cell wall, and motility. The virulence of these bacteria can however be reactivated upon wounding the host. We show that melanization but not the cellular or the systemic humoral response is the predominant host defense that establishes latency and may coerce the bacteria to a dormant state. In addition, the lasting activation of the melanization responses in latently-infected flies provides a degree of protection to the host against a secondary fungal infection. Latent infection by an ingested pathogen protects against a variety of homologous or heterologous systemic secondary infectious challenges, a situation previously described for the endosymbiotic Wolbachia bacteria, a guard against viral infections.


Asunto(s)
Drosophila melanogaster , Inmunidad Innata , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Animales , Drosophila melanogaster/microbiología , Drosophila melanogaster/inmunología , Pseudomonas aeruginosa/patogenicidad , Pseudomonas aeruginosa/inmunología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Virulencia , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno/inmunología
2.
iScience ; 26(9): 107490, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37636057

RESUMEN

Upon exposure to a bacterial pore-forming toxin, enterocytes rapidly purge their apical cytoplasm into the gut lumen, resulting in a thin intestinal epithelium. The enterocytes regain their original shape and thickness within 16 h after the ingestion of the bacteria. Here, we show that the regrowth of Drosophila enterocytes entails an inversion of metabolic fluxes from the organism back toward the intestine. We identify a proton-assisted transporter, Arcus, that is required for the reverse absorption of amino acids and the timely recovery of the intestinal epithelium. Arcus is required for a peak of amino acids appearing in the hemolymph shortly after infection. The regrowth of enterocytes involves the insulin signaling pathway and Myc. The purge decreases Myc mRNA levels, which subsequently remain at low levels in the arcus mutant. Interestingly, the action of arcus and Myc in the intestinal epithelium is not cell-autonomous, suggesting amino acid fluxes within the intestinal epithelium.

3.
Res Microbiol ; 174(6): 104089, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37348743

RESUMEN

Bacillus thuringiensis israelensis is largely regarded as the most selective, safe and ecofriendly biopesticide used for the control of insect vectors of human diseases. Bti enthomopathogenicity relies on the Cry and Cyt δ-endotoxins, produced as crystalline inclusions during sporulation. Insecticidal selectivity of Bti is mainly ascribed to the binding of the Cry toxins to receptors in the gut of target insects. However, the contribution of epithelial defenses in limiting Bti side effects in non-target species remains largely unexplored. Here, taking advantage of the genetically tractable Drosophila melanogaster model and its amenability for deciphering highly conserved innate immune defenses, we unravel a central role of the NF-κB factor Relish in the protection against the effects of ingested Bti spores in a non-susceptible host. Intriguingly, our data indicate that the Bti-induced Relish response is independent of its canonical activation downstream of peptidoglycan sensing and does not involve its longstanding role in the regulation of antimicrobial peptides encoding genes. In contrast, our data highlight a novel enterocyte specific function of Relish that is essential for preventing general septicemia following Bti oral infections strictly when producing δ-endotoxins. Altogether, our data provide novel insights into Bti-hosts interactions of prominent interest for the optimization and sustainability of insects' biocontrol strategies.


Asunto(s)
Bacillus thuringiensis , Endotoxinas , Animales , Humanos , Endotoxinas/genética , Endotoxinas/metabolismo , Endotoxinas/farmacología , Bacillus thuringiensis/genética , FN-kappa B/metabolismo , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Toxinas de Bacillus thuringiensis/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/farmacología
4.
Proc Natl Acad Sci U S A ; 120(12): e2205140120, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36917667

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Receptores Toll-Like/metabolismo , Transducción de Señal , Péptidos/metabolismo , Hongos/metabolismo , Bacterias Grampositivas/metabolismo
5.
EMBO Rep ; 24(1): e56036, 2023 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-36322050

RESUMEN

Host defense against infections encompasses both resistance, which targets microorganisms for neutralization or elimination, and resilience/disease tolerance, which allows the host to withstand/tolerate pathogens and repair damages. In Drosophila, the Toll signaling pathway is thought to mediate resistance against fungal infections by regulating the secretion of antimicrobial peptides, potentially including Bomanins. We find that Aspergillus fumigatus kills Drosophila Toll pathway mutants without invasion because its dissemination is blocked by melanization, suggesting a role for Toll in host defense distinct from resistance. We report that mutants affecting the Toll pathway or the 55C Bomanin locus are susceptible to the injection of two Aspergillus mycotoxins, restrictocin and verruculogen. The vulnerability of 55C deletion mutants to these mycotoxins is rescued by the overexpression of Bomanins specific to each challenge. Mechanistically, flies in which BomS6 is expressed in the nervous system exhibit an enhanced recovery from the tremors induced by injected verruculogen and display improved survival. Thus, innate immunity also protects the host against the action of microbial toxins through secreted peptides and thereby increases its resilience to infection.


Asunto(s)
Proteínas de Drosophila , Micotoxinas , Animales , Drosophila/genética , Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/metabolismo , Micotoxinas/metabolismo , Aspergillus/genética , Aspergillus/metabolismo , Inmunidad Innata
6.
Sci Rep ; 12(1): 3068, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35197500

RESUMEN

Serratia marcescens is an opportunistic bacterium that infects a wide range of hosts including humans. It is a potent pathogen in a septic injury model of Drosophila melanogaster since a few bacteria directly injected in the body cavity kill the insect within a day. In contrast, flies do not succumb to ingested bacteria for days even though some bacteria cross the intestinal barrier into the hemolymph within hours. The mechanisms by which S. marcescens attacks enterocytes and damages the intestinal epithelium remain uncharacterized. To better understand intestinal infections, we performed a genetic screen for loss of virulence of ingested S. marcescens and identified FliR, a structural component of the flagellum, as a virulence factor. Next, we compared the virulence of two flagellum mutants fliR and flhD in two distinct S. marcescens strains. Both genes are required for S. marcescens to escape the gut lumen into the hemocoel, indicating that the flagellum plays an important role for the passage of bacteria through the intestinal barrier. Unexpectedly, fliR but not flhD is involved in S. marcescens-mediated damages of the intestinal epithelium that ultimately contribute to the demise of the host. Our results therefore suggest a flagellum-independent role for fliR in bacterial virulence.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Drosophila melanogaster/microbiología , Flagelos/genética , Flagelos/fisiología , Gastroenteritis/microbiología , Mucosa Intestinal/microbiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Infecciones por Serratia , Serratia marcescens/genética , Serratia marcescens/patogenicidad , Animales , Modelos Animales de Enfermedad , Mucosa Intestinal/patología , Mutación , Virulencia/genética
7.
Immunogenetics ; 74(1): 35-62, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35092465

RESUMEN

Insects occupy a central position in the biosphere. They are able to resist infections even though they lack an adaptive immune system. Drosophila melanogaster has been used as a potent genetic model to understand innate immunity both in invertebrates and vertebrates. Its immune system includes both humoral and cellular arms. Here, we review how the distinct immune responses are triggered upon sensing infections, with an emphasis on the mechanisms that lead to systemic humoral immune responses. As in plants, the components of the cell wall of microorganisms are detected by dedicated receptors. There is also an induction of the systemic immune response upon sensing the proteolytic activities of microbial virulence factors. The antiviral response mostly relies on sensing double-stranded RNAs generated during the viral infection cycle. This event subsequently triggers either the viral short interfering RNA pathway or a cGAS-like/STING/NF-κB signaling pathway.


Asunto(s)
Drosophila melanogaster , Modelos Genéticos , Animales , Drosophila melanogaster/genética , Inmunidad Innata/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal/genética
8.
Elife ; 92020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32602836

RESUMEN

Single-cell RNA sequencing has revealed distinct subpopulations of hemocytes in fruit fly larvae.


Asunto(s)
Drosophila , Hemocitos , Animales , Larva , Análisis de Secuencia de ARN , Encuestas y Cuestionarios
9.
Front Immunol ; 11: 478958, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33746938

RESUMEN

The humoral immune response to bacterial or fungal infections in Drosophila relies largely on a transcriptional response mediated by the Toll and Immune deficiency NF-κB pathways. Antimicrobial peptides are potent effectors of these pathways and allow the organism to attack invading pathogens. Dorsal-related Immune Factor (DIF), a transcription factor regulated by the Toll pathway, is required in the host defense against fungal and some Gram-positive bacterial infections. The Mediator complex is involved in the initiation of transcription of most RNA polymerase B (PolB)-dependent genes by forming a functional bridge between transcription factors bound to enhancer regions and the gene promoter region and then recruiting the PolB pre-initiation complex. Mediator is formed by several modules that each comprises several subunits. The Med17 subunit of the head module of Mediator has been shown to be required for the expression of Drosomycin, which encodes a potent antifungal peptide, by binding to DIF. Thus, Mediator is expected to mediate the host defense against pathogens controlled by the Toll pathway-dependent innate immune response. Here, we first focus on the Med31 subunit of the middle module of Mediator and find that it is required in host defense against Aspergillus fumigatus, Enterococcus faecalis, and injected but not topically-applied Metarhizium robertsii. Thus, host defense against M. robertsii requires Dif but not necessarily Med31 in the two distinct infection models. The induction of some Toll-pathway-dependent genes is decreased after a challenge of Med31 RNAi-silenced flies with either A. fumigatus or E. faecalis, while these flies exhibit normal phagocytosis and melanization. We have further tested most Mediator subunits using RNAi by monitoring their survival after challenges to several other microbial infections known to be fought off through DIF. We report that the host defense against specific pathogens involves a distinct set of Mediator subunits with only one subunit for C. glabrata or Erwinia carotovora carotovora, at least one for M. robertsii or a somewhat extended repertoire for A. fumigatus (at least eight subunits) and E. faecalis (eight subunits), with two subunits, Med6 and Med11 being required only against A. fumigatus. Med31 but not Med17 is required in fighting off injected M. robertsii conidia. Thus, the involvement of Mediator in Drosophila innate immunity is more complex than expected.


Asunto(s)
Aspergillus fumigatus/inmunología , Proteínas de Drosophila/inmunología , Enterococcus faecalis/inmunología , Complejo Mediador/inmunología , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Complejo Mediador/genética , Metarhizium/inmunología
10.
EMBO Rep ; 19(5)2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29523648

RESUMEN

When Drosophila melanogaster feeds on Pseudomonas aeruginosa, some bacteria cross the intestinal barrier and eventually proliferate in the hemocoel. This process is limited by hemocytes through phagocytosis. P. aeruginosa requires the quorum-sensing regulator RhlR to elude the cellular immune response of the fly. RhlI synthesizes the autoinducer signal that activates RhlR. Here, we show that rhlI mutants are unexpectedly more virulent than rhlR mutants, both in fly and in nematode intestinal infection models, suggesting that RhlR has RhlI-independent functions. We also report that RhlR protects P. aeruginosa from opsonization mediated by the Drosophila thioester-containing protein 4 (Tep4). RhlR mutant bacteria show higher levels of Tep4-mediated opsonization, as compared to rhlI mutants, which prevents lethal bacteremia in the Drosophila hemocoel. In contrast, in a septic model of infection, in which bacteria are introduced directly into the hemocoel, Tep4 mutant flies are more resistant to wild-type P. aeruginosa, but not to the rhlR mutant. Thus, depending on the infection route, the Tep4 opsonin can either be protective or detrimental to host defense.


Asunto(s)
Proteínas Bacterianas/genética , ARN Helicasas DEAD-box/genética , Ligasas/genética , Fagocitosis , Pseudomonas aeruginosa/genética , Percepción de Quorum/genética , Factores de Transcripción/genética , Animales , Caenorhabditis elegans/microbiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/inmunología , Drosophila melanogaster/inmunología , Drosophila melanogaster/microbiología , Regulación Bacteriana de la Expresión Génica , Intestinos/inmunología , Intestinos/microbiología , Pseudomonas aeruginosa/patogenicidad , Receptores de Reconocimiento de Patrones/inmunología , Virulencia
11.
Cell Host Microbe ; 20(6): 716-730, 2016 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-27889464

RESUMEN

Besides digesting nutrients, the gut protects the host against invasion by pathogens. Enterocytes may be subjected to damage by both microbial and host defensive responses, causing their death. Here, we report a rapid epithelial response that alleviates infection stress and protects the enterocytes from the action of microbial virulence factors. Intestinal epithelia exposed to hemolysin, a pore-forming toxin secreted by Serratia marcescens, undergo an evolutionarily conserved process of thinning followed by the recovery of their initial thickness within a few hours. In response to hemolysin attack, Drosophila melanogaster enterocytes extrude most of their apical cytoplasm, including damaged organelles such as mitochondria, yet do not lyse. We identify two secreted peptides, the expression of which requires CyclinJ, that mediate the recovery phase in which enterocytes regain their original shape and volume. Epithelial thinning and recovery constitute a fast and efficient response to intestinal infections, with pore-forming toxins acting as alarm signals.


Asunto(s)
Toxinas Bacterianas/toxicidad , Sistema Digestivo/efectos de los fármacos , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Animales , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Citoplasma/efectos de los fármacos , Sistema Digestivo/inmunología , Sistema Digestivo/microbiología , Sistema Digestivo/patología , Modelos Animales de Enfermedad , Drosophila melanogaster , Enterocitos/patología , Proteínas Hemolisinas/metabolismo , Proteínas Hemolisinas/toxicidad , Enfermedades Intestinales/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Microvellosidades/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Infecciones por Serratia , Serratia marcescens/metabolismo , Serratia marcescens/patogenicidad , Sobrevida , Varroidae , Factores de Virulencia
12.
Immunobiology ; 218(4): 635-44, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23083631

RESUMEN

Drosophila melanogaster is a useful model to investigate mucosal immunity. The immune response to intestinal infections is mediated partly by the Immune deficiency (IMD) pathway, which only gets activated by a type of peptidoglycan lacking in several medically important Gram-positive bacterial species such as Staphylococcus. Thus, the intestinal host defense against such bacterial strains remains poorly known. Here, we have used Staphylococcus xylosus to develop a model of intestinal infections by Gram-positive bacteria. S. xylosus behaves as an opportunistic pathogen in a septic injury model, being able to kill only flies immunodeficient either for the Toll pathway or the cellular response. When ingested, it is controlled by IMD-independent host intestinal defenses, yet flies eventually die. Having excluded an overreaction of the immune response and the action of toxins, we find that flies actually succumb to starvation, likely as a result of a competition for sucrose between the bacteria and the flies. Fat stores of wild-type flies are severely reduced within a day, a period when sucrose is not yet exhausted in the feeding solution. Interestingly, the Toll pathway mutant MyD88 is more resistant to the ingestion of S. xylosus and to starvation than wild-type flies. MyD88 flies do not rapidly deplete their fat stores when starved, in contrast to wild-type flies. Thus, we have uncovered a novel function of MyD88 in the regulation of metabolism that appears to be independent of its known roles in immunity and development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Antígenos de Diferenciación/inmunología , Proteínas de Drosophila/inmunología , Inmunidad Innata , Inmunidad Mucosa , Enfermedades Intestinales/inmunología , Receptores Inmunológicos/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus/inmunología , Inanición/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Enfermedades Intestinales/genética , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/microbiología , Enfermedades Intestinales/patología , Mutación , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/patología , Inanición/genética , Inanición/metabolismo , Inanición/microbiología , Inanición/patología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo
13.
Fly (Austin) ; 6(3): 193-204, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22836791

RESUMEN

Drosophila melanogaster is a robust model to investigate many biological problems. It is however prone to some infections, which may endanger fly stocks if left unchecked for. One such infection is caused by an obligate fungal intracellular parasite, Tubulinosema ratisbonensis, which can be found in laboratory stocks. Here, we identify and briefly characterize a T. ratisbonensis strain that was infesting our Drosophila cultures and that required intensive measures to contain and eradicate the infection. We describe the phenotypes of infested stocks. We also report PCR-based techniques that allow the detection of infested stocks with a high sensitivity. We have developed a high-throughput qPCR assay that allows the efficient parallel screening of a large number of potentially-infested stocks. We also have investigated several prophylactic measures to prevent the further contamination of stocks, namely UV-exposure, ethanol treatment, bleaching, and desiccation. Bleaching was found to kill all spores. Other treatments were less effective but were found to be sufficient to prevent further contamination of noninfested stocks. Two treatments were efficacious in curing infested stocks (1) bleaching of eggs and subsequent raising of the larvae in clean vials; (2) fumagillin treatment. These cures only work on stocks that have not become too weak to withstand the procedures.


Asunto(s)
Apansporoblastina/genética , Drosophila melanogaster/microbiología , Animales , Apansporoblastina/fisiología , Secuencia de Bases , Cartilla de ADN , ADN de Hongos/química , ADN Ribosómico/química , Desinfección/métodos , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena de la Polimerasa/métodos , Alineación de Secuencia
14.
Science ; 325(5938): 340-3, 2009 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-19520911

RESUMEN

Innate immunity represents the first line of defense in animals. We report a genome-wide in vivo Drosophila RNA interference screen to uncover genes involved in susceptibility or resistance to intestinal infection with the bacterium Serratia marcescens. We first employed whole-organism gene suppression, followed by tissue-specific silencing in gut epithelium or hemocytes to identify several hundred genes involved in intestinal antibacterial immunity. Among the pathways identified, we showed that the JAK-STAT signaling pathway controls host defense in the gut by regulating stem cell proliferation and thus epithelial cell homeostasis. Therefore, we revealed multiple genes involved in antibacterial defense and the regulation of innate immunity.


Asunto(s)
Drosophila melanogaster/genética , Drosophila melanogaster/microbiología , Genoma de los Insectos , Inmunidad Innata/genética , Interferencia de ARN , Infecciones por Serratia/inmunología , Serratia marcescens/inmunología , Animales , Animales Modificados Genéticamente , Proliferación Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/inmunología , Células Epiteliales/citología , Células Epiteliales/fisiología , Hemocitos/inmunología , Hemocitos/metabolismo , Hemocitos/microbiología , Homeostasis , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Quinasas Janus/genética , Quinasas Janus/metabolismo , Modelos Animales , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Infecciones por Serratia/genética , Infecciones por Serratia/microbiología , Serratia marcescens/fisiología , Transducción de Señal , Células Madre/citología , Células Madre/fisiología
15.
PLoS Pathog ; 3(11): e173, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18039029

RESUMEN

Serratia marcescens is an entomopathogenic bacterium that opportunistically infects a wide range of hosts, including humans. In a model of septic injury, if directly introduced into the body cavity of Drosophila, this pathogen is insensitive to the host's systemic immune response and kills flies in a day. We find that S. marcescens resistance to the Drosophila immune deficiency (imd)-mediated humoral response requires the bacterial lipopolysaccharide O-antigen. If ingested by Drosophila, bacteria cross the gut and penetrate the body cavity. During this passage, the bacteria can be observed within the cells of the intestinal epithelium. In such an oral infection model, the flies succumb to infection only after 6 days. We demonstrate that two complementary host defense mechanisms act together against such food-borne infection: an antimicrobial response in the intestine that is regulated by the imd pathway and phagocytosis by hemocytes of bacteria that have escaped into the hemolymph. Interestingly, bacteria present in the hemolymph elicit a systemic immune response only when phagocytosis is blocked. Our observations support a model wherein peptidoglycan fragments released during bacterial growth activate the imd pathway and do not back a proposed role for phagocytosis in the immune activation of the fat body. Thanks to the genetic tools available in both host and pathogen, the molecular dissection of the interactions between S. marcescens and Drosophila will provide a useful paradigm for deciphering intestinal pathogenesis.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila/microbiología , Interacciones Huésped-Patógeno/fisiología , Intestinos/microbiología , Infecciones por Serratia/fisiopatología , Serratia marcescens/patogenicidad , Animales , Drosophila/inmunología , Hemolinfa/microbiología , Inmunohistoquímica , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Serratia/inmunología , Serratia marcescens/inmunología
16.
Genetics ; 175(2): 709-24, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17179093

RESUMEN

Few studies have investigated whether or not there is an interdependence between osmoregulation and vesicular trafficking. We previously showed that in Caenorhabditis elegans che-14 mutations affect osmoregulation, cuticle secretion, and sensory organ development. We report the identification of seven lethal mutations displaying che-14-like phenotypes, which define four new genes, rdy-1-rdy-4 (rod-like larval lethality and dye-filling defective). rdy-1, rdy-2, and rdy-4 mutations affect excretory canal function and cuticle formation. Moreover, rdy-1 and rdy-2 mutations reduce the amount of matrix material normally secreted by sheath cells in the amphid channel. In contrast, rdy-3 mutants have short cystic excretory canals, suggesting that it acts in a different process. rdy-1 encodes the vacuolar H+-ATPase a-subunit VHA-5, whereas rdy-2 encodes a new tetraspan protein. We suggest that RDY-1/VHA-5 acts upstream of RDY-2 and CHE-14 in some tissues, since it is required for their delivery to the epidermal, but not the amphid sheath, apical plasma membrane. Hence, the RDY-1/VHA-5 trafficking function appears essential in some cells and its proton pump function essential in others. Finally, we show that RDY-1/VHA-5 distribution changes prior to molting in parallel with that of actin microfilaments and propose a model for molting whereby actin provides a spatial cue for secretion.


Asunto(s)
Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Epidermis/metabolismo , Genes de Helminto , Equilibrio Hidroelectrolítico/genética , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Caenorhabditis elegans/citología , Caenorhabditis elegans/ultraestructura , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Células Epidérmicas , Epidermis/ultraestructura , Pruebas Genéticas , Proteínas de la Membrana/química , Datos de Secuencia Molecular , Muda , Mosaicismo , Mutagénesis , Mutación/genética , Fenotipo , ATPasas de Translocación de Protón Vacuolares/metabolismo
17.
J Cell Biol ; 173(6): 949-61, 2006 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-16785323

RESUMEN

Polarized intracellular trafficking in epithelia is critical in development, immunity, and physiology to deliver morphogens, defensins, or ion pumps to the appropriate membrane domain. The mechanisms that control apical trafficking remain poorly defined. Using Caenorhabditis elegans, we characterize a novel apical secretion pathway involving multivesicularbodies and the release of exosomes at the apical plasma membrane. By means of two different genetic approaches, we show that the membrane-bound V0 sector of the vacuolar H+-ATPase (V-ATPase) acts in this pathway, independent of its contribution to the V-ATPase proton pump activity. Specifically, we identified mutations in the V0 "a" subunit VHA-5 that affect either the V0-specific function or the V0+V1 function of the V-ATPase. These mutations allowed us to establish that the V0 sector mediates secretion of Hedgehog-related proteins. Our data raise the possibility that the V0 sector mediates exosome and morphogen release in mammals.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/enzimología , Vesículas Secretoras/fisiología , Transactivadores/metabolismo , ATPasas de Translocación de Protón Vacuolares/fisiología , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas Hedgehog , Modelos Biológicos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fenotipo , Estructura Terciaria de Proteína , Transporte de Proteínas/fisiología , Vesículas Secretoras/ultraestructura , Alineación de Secuencia , ATPasas de Translocación de Protón Vacuolares/química , ATPasas de Translocación de Protón Vacuolares/genética
18.
Dev Dyn ; 235(6): 1469-81, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16502424

RESUMEN

The Caenorhabditis elegans genome encodes ten proteins that share similarity with Hedgehog through the C-terminal Hint/Hog domain. While most genes are members of larger gene families, qua-1 is a single copy gene. Here we show that orthologs of qua-1 exist in many nematodes, including Brugia malayi, which shared a common ancestor with C. elegans about 300 million years ago. The QUA-1 proteins contain an N-terminal domain, the Qua domain, that is highly conserved, but whose molecular function is not known. We have studied the expression pattern of qua-1 in C. elegans using a qua-1::GFP transcriptional fusion. qua-1 is mainly expressed in hyp1 to hyp11 hypodermal cells, but not in seam cells. It is also expressed in intestinal and rectal cells, sensilla support cells, and the P cell lineage in L1. The expression of qua-1::GFP undergoes cyclical changes during development in phase with the molting cycle. It accumulates prior to molting and disappears between molts. Disruption of the qua-1 gene function through an internal deletion that causes a frame shift with premature stop in the middle of the gene results in strong lethality. The animals arrest in the early larval stages due to defects in molting. Electron microscopy reveals double cuticles due to defective ecdysis, but no obvious defects are seen in the hypodermis. Qua domain-only::GFP and full-length QUA-1::GFP fusion constructs are secreted and associated with the overlying cuticle, but only QUA-1::GFP rescues the mutant phenotype. Our results suggest that both the Hint/Hog domain and Qua domain are critically required for the function of QUA-1.


Asunto(s)
Relojes Biológicos/fisiología , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/genética , Proteínas Hedgehog/genética , Muda/genética , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/biosíntesis , Proteínas de Caenorhabditis elegans/fisiología , Femenino , Mutación del Sistema de Lectura , Proteínas Hedgehog/biosíntesis , Proteínas Hedgehog/fisiología , Datos de Secuencia Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA