Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 1341, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38351056

RESUMEN

The survival of animals depends, among other things, on their ability to identify threats in their surrounding environment. Senses such as olfaction, vision and taste play an essential role in sampling their living environment, including microorganisms, some of which are potentially pathogenic. This study focuses on the mechanisms of detection of bacteria by the Drosophila gustatory system. We demonstrate that the peptidoglycan (PGN) that forms the cell wall of bacteria triggers an immediate feeding aversive response when detected by the gustatory system of adult flies. Although we identify ppk23+ and Gr66a+ gustatory neurons as necessary to transduce fly response to PGN, we demonstrate that they play very different roles in the process. Time-controlled functional inactivation and in vivo calcium imaging demonstrate that while ppk23+ neurons are required in the adult flies to directly transduce PGN signal, Gr66a+ neurons must be functional in larvae to allow future adults to become PGN sensitive. Furthermore, the ability of adult flies to respond to bacterial PGN is lost when they hatch from larvae reared under axenic conditions. Recolonization of germ-free larvae, but not adults, with a single bacterial species, Lactobacillus brevis, is sufficient to restore the ability of adults to respond to PGN. Our data demonstrate that the genetic and environmental characteristics of the larvae are essential to make the future adults competent to respond to certain sensory stimuli such as PGN.


Asunto(s)
Proteínas de Drosophila , Microbiota , Animales , Drosophila , Percepción del Gusto/fisiología , Drosophila melanogaster/genética , Proteínas de Drosophila/genética , Larva/fisiología , Gusto/fisiología
2.
Elife ; 82019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31661076

RESUMEN

When facing microbes, animals engage in behaviors that lower the impact of the infection. We previously demonstrated that internal sensing of bacterial peptidoglycan reduces Drosophila female oviposition via NF-κB pathway activation in some neurons (Kurz et al., 2017). Although we showed that the neuromodulator octopamine is implicated, the identity of the involved neurons, as well as the physiological mechanism blocking egg-laying, remained unknown. In this study, we identified few ventral nerve cord and brain octopaminergic neurons expressing an NF-κB pathway component. We functionally demonstrated that NF-κB pathway activation in the brain, but not in the ventral nerve cord octopaminergic neurons, triggers an egg-laying drop in response to infection. Furthermore, we demonstrated via calcium imaging that the activity of these neurons can be directly modulated by peptidoglycan and that these cells do not control other octopamine-dependent behaviors such as female receptivity. This study shows that by sensing peptidoglycan and hence activating NF-κB cascade, a couple of brain neurons modulate a specific octopamine-dependent behavior to adapt female physiology status to their infectious state.


Asunto(s)
Encéfalo/citología , Drosophila/fisiología , FN-kappa B/metabolismo , Neuronas/efectos de los fármacos , Oviposición , Peptidoglicano/metabolismo , Animales , Drosophila/microbiología , Femenino , Octopamina/metabolismo
3.
Cell Host Microbe ; 23(2): 215-228.e4, 2018 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-29398649

RESUMEN

Gut-associated bacteria produce metabolites that both have a local influence on the intestinal tract and act at a distance on remote organs. In Drosophila, bacteria-derived peptidoglycan (PGN) displays such a dual role. PGN triggers local antimicrobial peptide production by enterocytes; it also activates systemic immune responses in fat-body cells and modulates fly behavior by acting on neurons. How these responses to a single microbiota-derived compound are simultaneously coordinated is not understood. We show here that the PGRP-LB locus generates both cytosolic and secreted PGN-cleaving enzymes. Through genetic analysis, we demonstrate that the cytosolic PGRP-LB isoforms cell-autonomously control the intensity of NF-κB activation in enterocytes, whereas the secreted isoform prevents massive and detrimental gut-derived PGN dissemination throughout the organism. This study explains how Drosophila are able to uncouple the modulation of local versus systemic responses to a single gut-bacteria-derived product by using isoform-specific enzymes.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Drosophila melanogaster/enzimología , Enterocitos/inmunología , Peptidoglicano/metabolismo , Animales , Animales Modificados Genéticamente/genética , Péptidos Catiónicos Antimicrobianos/inmunología , Drosophila melanogaster/genética , Drosophila melanogaster/inmunología , Activación Enzimática/genética , Cuerpo Adiposo/metabolismo , Microbioma Gastrointestinal/inmunología , Inmunidad Innata/inmunología , FN-kappa B/metabolismo , Pectobacterium carotovorum/inmunología , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología
4.
Elife ; 62017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28264763

RESUMEN

As infectious diseases pose a threat to host integrity, eukaryotes have evolved mechanisms to eliminate pathogens. In addition to develop strategies reducing infection, animals can engage in behaviors that lower the impact of the infection. The molecular mechanisms by which microbes impact host behavior are not well understood. We demonstrate that bacterial infection of Drosophila females reduces oviposition and that peptidoglycan, the component that activates Drosophila antibacterial response, is also the elicitor of this behavioral change. We show that peptidoglycan regulates egg-laying rate by activating NF-κB signaling pathway in octopaminergic neurons and that, a dedicated peptidoglycan degrading enzyme acts in these neurons to buffer this behavioral response. This study shows that a unique ligand and signaling cascade are used in immune cells to mount an immune response and in neurons to control fly behavior following infection. This may represent a case of behavioral immunity.


Asunto(s)
Drosophila/fisiología , Neuronas/metabolismo , Oviposición , Peptidoglicano/metabolismo , Receptores de Amina Biogénica/metabolismo , Animales , Drosophila/inmunología , FN-kappa B/metabolismo , Peptidoglicano/inmunología , Transducción de Señal
5.
J Insect Physiol ; 99: 25-32, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28232220

RESUMEN

When exposed to microorganisms, animals use several protective strategies. On one hand, as elegantly exemplified in Drosophila melanogaster, the innate immune system recognizes microbial compounds and triggers an antimicrobial response. On the other hand, behaviors preventing an extensive contact with the microbes and thus reducing the risk of infection have been described. However, these reactions ranging from microbes aversion to intestinal transit increase or food intake decrease have been rarely defined at the molecular level. In this study, we set up an experimental system that allowed us to rapidly identify and quantify food intake decreases in Drosophila larvae exposed to media contaminated with bacteria. Specifically, we report a robust dose-dependent food intake decrease following exposure to the bacteria Erwinia carotovora carotovora strain Ecc15. We demonstrate that this response does not require Imd innate immune pathway, but rather the olfactory neuronal circuitry, the Trpa1 receptor and the evf virulence factor. Finally, we show that Ecc15 induce the same behavior in the invasive pest insect Drosophila suzukii.


Asunto(s)
Drosophila/microbiología , Drosophila/fisiología , Pectobacterium carotovorum/patogenicidad , Animales , Proteínas Bacterianas/genética , Drosophila/inmunología , Proteínas de Drosophila/genética , Ingestión de Alimentos , Inmunidad Innata , Canales Iónicos , Larva/genética , Larva/microbiología , Larva/fisiología , Percepción Olfatoria , Pectobacterium carotovorum/genética , Canal Catiónico TRPA1/genética , Factores de Virulencia/genética
6.
BMC Biol ; 14: 35, 2016 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-27129311

RESUMEN

BACKGROUND: Caenorhabditis elegans has emerged over the last decade as a useful model for the study of innate immunity. Its infection with the pathogenic fungus Drechmeria coniospora leads to the rapid up-regulation in the epidermis of genes encoding antimicrobial peptides. The molecular basis of antimicrobial peptide gene regulation has been previously characterized through forward genetic screens. Reverse genetics, based on RNAi, provide a complementary approach to dissect the worm's immune defenses. RESULTS: We report here the full results of a quantitative whole-genome RNAi screen in C. elegans for genes involved in regulating antimicrobial peptide gene expression. The results will be a valuable resource for those contemplating similar RNAi-based screens and also reveal the limitations of such an approach. We present several strategies, including a comprehensive class clustering method, to overcome these limitations and which allowed us to characterize the different steps of the interaction between C. elegans and the fungus D. coniospora, leading to a complete description of the MAPK pathway central to innate immunity in C. elegans. The results further revealed a cross-tissue signaling, triggered by mitochondrial dysfunction in the intestine, that suppresses antimicrobial peptide gene expression in the nematode epidermis. CONCLUSIONS: Overall, our results provide an unprecedented system's level insight into the regulation of C. elegans innate immunity. They represent a significant contribution to our understanding of host defenses and will lead to a better comprehension of the function and evolution of animal innate immunity.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/inmunología , Caenorhabditis elegans/genética , Caenorhabditis elegans/inmunología , Inmunidad Innata/genética , Animales , Caenorhabditis elegans/microbiología , Clonación Molecular , Epidermis/inmunología , Estudios de Asociación Genética , Genoma de los Helmintos , Interacciones Huésped-Patógeno , Hypocreales , Mitocondrias/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Interferencia de ARN , Transducción de Señal , Regulación hacia Arriba
7.
Nat Immunol ; 15(9): 833-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25086774

RESUMEN

Immune defenses are triggered by microbe-associated molecular patterns or as a result of damage to host cells. The elicitors of immune responses in the nematode Caenorhabditis elegans are unclear. Using a genome-wide RNA-mediated interference (RNAi) screen, we identified the G protein-coupled receptor (GPCR) DCAR-1 as being required for the response to fungal infection and wounding. DCAR-1 acted in the epidermis to regulate the expression of antimicrobial peptides via a conserved p38 mitogen-activated protein kinase pathway. Through targeted metabolomics analysis we identified the tyrosine derivative 4-hydroxyphenyllactic acid (HPLA) as an endogenous ligand. Our findings reveal DCAR-1 and its cognate ligand HPLA to be triggers of the epidermal innate immune response in C. elegans and highlight the ancient role of GPCRs in host defense.


Asunto(s)
Proteínas de Caenorhabditis elegans/inmunología , Caenorhabditis elegans/inmunología , Epidermis/inmunología , Inmunidad Innata/inmunología , Micosis/inmunología , Fenilpropionatos/inmunología , Receptores Acoplados a Proteínas G/inmunología , Heridas y Lesiones/inmunología , Animales , Ligandos , Sistema de Señalización de MAP Quinasas/inmunología , Interferencia de ARN
8.
Dev Comp Immunol ; 42(2): 132-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24012871

RESUMEN

Host defense mechanisms are multi-layered and involve constitutive as well as inducible components. The dissection of these complex processes can be greatly facilitated using a reporter gene strategy with a transparent animal. In this study, we use Caenorhabditis elegans as a model host and introduce a new pathogen-inducible fluorescent reporter involving the promoter of f57f4.4, a gene encoding a putative component of the glycocalyx. We show that this reporter construct does not respond to heavy metal or hypertonic environments, but is specifically and locally induced in the intestine upon Photorhabus luminescens and Pseudomonas aeruginosa infections. We further demonstrate that its upregulation requires live pathogens as well as elements of the nematode p38 MAP kinase and TGF-beta pathways. In addition to introducing a new tool for the study of the interactions between C. elegans and a pathogen, our results suggest a role for the glycocalyx in gut immunity.


Asunto(s)
Proteínas de Caenorhabditis elegans/inmunología , Caenorhabditis elegans/inmunología , Infecciones por Enterobacteriaceae/inmunología , Glicocálix/inmunología , Glicoproteínas/inmunología , Infecciones por Pseudomonas/inmunología , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiología , Proteínas de Caenorhabditis elegans/genética , Genes Reporteros , Glicocálix/genética , Glicoproteínas/genética , Proteínas Fluorescentes Verdes/genética , Intestinos/inmunología , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Proteínas de la Membrana , Photorhabdus/inmunología , Regiones Promotoras Genéticas , Pseudomonas aeruginosa/inmunología , Interferencia de ARN , ARN Interferente Pequeño , Factor de Crecimiento Transformador beta/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
9.
Curr Drug Targets ; 12(7): 967-77, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21366520

RESUMEN

One approach to identify new drugs with antimicrobial activities is to screen large libraries of molecules directly for their capacity to block the growth of bacterial or fungal monocultures. A more relevant way to assess both a product's efficacy and its potential cytotoxicity is undoubtedly to use an in vivo infection system. Testing banks containing thousands of natural or chemically synthesized molecules with rodents is generally neither desirable nor feasible. Therefore, invertebrate model organisms could represent a valuable alternative. In this review, we present the worm C. elegans as a suitable host model for the evaluation and characterization of drug effects in a pathogenesis context. This simple organism has been of great value in many fields of biology and is currently intensely used in studies of host-pathogen interactions. Infection of C. elegans induces a number of defense mechanisms, some of which are similar to those seen in mammalian innate immunity. Further, it has been demonstrated that several microbial virulence mechanisms required for full pathogenicity in mammals are also necessary for infection in nematodes. Based on these facts, a number of innovative antimicrobial drug screens have been carried out successfully and the development of new tools to monitor the interaction between worm and microbes in vivo opens promising perspectives.


Asunto(s)
Caenorhabditis elegans/microbiología , Diseño de Fármacos , Modelos Biológicos , Animales , Antiinfecciosos/farmacología , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Interacciones Huésped-Patógeno , Humanos
10.
Cell Host Microbe ; 5(4): 341-52, 2009 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-19380113

RESUMEN

Like other multicellular organisms, the model nematode C. elegans responds to infection by inducing the expression of defense genes. Among the genes upregulated in response to a natural fungal pathogen is nlp-29, encoding an antimicrobial peptide. In a screen for mutants that fail to express nlp-29 following fungal infection, we isolated alleles of tpa-1, homologous to the mammalian protein kinase C (PKC) delta. Through epistasis analyses, we demonstrate that C. elegans PKC acts through the p38 MAPK pathway to regulate nlp-29. This involves G protein signaling and specific C-type phospholipases acting upstream of PKCdelta. Unexpectedly and unlike in mammals, tpa-1 does not act via D-type protein kinases, but another C. elegans PKC gene, pkc-3, functions nonredundantly with tpa-1 to control nlp-29 expression. Finally, the tribbles-like kinase nipi-3 acts upstream of PKCdelta in this antifungal immune signaling cascade. These findings greatly expand our understanding of the pathways involved in C. elegans innate immunity.


Asunto(s)
Proteínas de Caenorhabditis elegans/inmunología , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/inmunología , Hongos/inmunología , Inmunidad Innata , Proteínas Tirosina Quinasas/inmunología , Proteínas Tirosina Quinasas/metabolismo , Animales , Proteínas de Unión al GTP/metabolismo , Regulación Fúngica de la Expresión Génica , Modelos Biológicos , Proteína Quinasa C/metabolismo , Transducción de Señal , Fosfolipasas de Tipo C/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
11.
PLoS Pathog ; 4(7): e1000105, 2008 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-18636113

RESUMEN

Encounters with pathogens provoke changes in gene transcription that are an integral part of host innate immune responses. In recent years, studies with invertebrate model organisms have given insights into the origin, function, and evolution of innate immunity. Here, we use genome-wide transcriptome analysis to characterize the consequence of natural fungal infection in Caenorhabditis elegans. We identify several families of genes encoding putative antimicrobial peptides (AMPs) and proteins that are transcriptionally up-regulated upon infection. Many are located in small genomic clusters. We focus on the nlp-29 cluster of six AMP genes and show that it enhances pathogen resistance in vivo. The same cluster has a different structure in two other Caenorhabditis species. A phylogenetic analysis indicates that the evolutionary diversification of this cluster, especially in cases of intra-genomic gene duplications, is driven by natural selection. We further show that upon osmotic stress, two genes of the nlp-29 cluster are strongly induced. In contrast to fungus-induced nlp expression, this response is independent of the p38 MAP kinase cascade. At the same time, both involve the epidermal GATA factor ELT-3. Our results suggest that selective pressure from pathogens influences intra-genomic diversification of AMPs and reveal an unexpected complexity in AMP regulation as part of the invertebrate innate immune response.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/inmunología , Interacciones Huésped-Patógeno , Inmunidad Innata/fisiología , Hongos Mitospóricos/efectos de los fármacos , Animales , Caenorhabditis elegans/microbiología , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Expresión Génica , Genoma , Análisis de Secuencia por Matrices de Oligonucleótidos , Organismos Modificados Genéticamente , Filogenia , Selección Genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Biochem Biophys Res Commun ; 363(2): 438-43, 2007 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-17888400

RESUMEN

Animals and plants respond to bacterial infections and environmental stresses by inducing overlapping repertoires of defense genes. How the signals associated with infection and abiotic stresses are differentially integrated within a whole organism remains to be fully addressed. We show that the transcription of a Caenorhabditis elegans ABC transporter, pgp-5 is induced by both bacterial infection and heavy metal stress, but the magnitude and tissue distribution of its expression differs, depending on the type of stressor. PGP-5 contributes to resistance to bacterial infection and heavy metals. Using pgp-5 transcription as a read-out, we show that signals from both biotic and abiotic stresses are integrated by TIR-1, a TIR domain adaptor protein orthologous to human SARM, and a p38 MAP kinase signaling cassette. We further demonstrate that not all the TIR-1 isoforms are necessary for nematode resistance to infection, suggesting a molecular basis for the differential response to abiotic and biotic stress.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/inmunología , Proteínas de Caenorhabditis elegans/inmunología , Caenorhabditis elegans/inmunología , Proteínas del Citoesqueleto/inmunología , Inmunidad Innata/inmunología , Proteínas Protozoarias/inmunología , Infecciones por Pseudomonas/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Animales , Caenorhabditis elegans/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Metales Pesados/farmacología , Receptores Acoplados a Proteínas G
13.
Curr Opin Microbiol ; 10(1): 10-6, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17178462

RESUMEN

Diverse aspects of host-pathogen interactions have been studied using non-mammalian hosts such as Dictyostelium discoideum, Caenorhabditis elegans, Drosophila melanogaster and Danio rerio for more than 20 years. Over the past two years, the use of these model hosts to dissect bacterial virulence mechanisms has been expanded to include the important human pathogens Vibrio cholerae and Yersinia pestis. Innovative approaches using these alternative hosts have also been developed, enabling the isolation of new antimicrobials through screening large libraries of compounds in a C. elegans Enterococcus faecalis infection model. Host proteins required by Mycobacterium and Listeria during their invasion and intracellular growth have been uncovered using high-throughput dsRNA screens in a Drosophila cell culture system, and immune evasion mechanisms deployed by Pseudomonas aeruginosa during its infection of flies have been identified. Together, these reports further illustrate the potential and relevance of these non-mammalian hosts for modelling many facets of bacterial infection in mammals.


Asunto(s)
Bacterias/patogenicidad , Infecciones Bacterianas/microbiología , Caenorhabditis elegans/microbiología , Dictyostelium/microbiología , Drosophila melanogaster/microbiología , Modelos Animales , Pez Cebra/microbiología , Animales , Antibacterianos/farmacología , Regulación Bacteriana de la Expresión Génica , Virulencia/fisiología
14.
Aging Cell ; 3(4): 185-93, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15268752

RESUMEN

The free-living soil nematode Caenorhabditis elegans is a versatile model for the study of the genetic regulation of aging and of host-pathogen interactions. Many genes affecting multiple processes, such as neuroendocrine signalling, nutritional sensing and mitochondrial functions, have been shown to play important roles in determining the lifespan of C. elegans. The DAF-2-mediated insulin signalling pathway is the major pathway that regulates aging in this nematode and this role appears universal; neuroendrocrine signalling also affects aging in Drosophila and mice. Recent studies have shown that the innate immune function in C. elegans is modulated by signalling from the TGF-beta-like, the p38 MAPK and the DAF-2 insulin pathways. The requirement for the DAF-2 pathway in modulating aging and immunity suggests that these processes may be linked at the molecular level. It is well known that as humans age, immunosenescence occurs in which there is a general degradation of immune efficiency. However, the molecular mechanisms involved in this process remain unclear. In this review, we discuss the molecular mechanisms that modulate aging and immune response and attempt to suggest molecular links between these two processes.


Asunto(s)
Envejecimiento/fisiología , Caenorhabditis elegans/fisiología , Inmunidad Innata/fisiología , Envejecimiento/genética , Envejecimiento/inmunología , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/fisiología , Apoptosis/fisiología , Caenorhabditis elegans/genética , Caenorhabditis elegans/inmunología , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiología , Factores de Transcripción Forkhead , Regulación del Desarrollo de la Expresión Génica , Inmunidad Innata/inmunología , Larva/genética , Larva/inmunología , Larva/fisiología , Longevidad/genética , Longevidad/inmunología , Longevidad/fisiología , Modelos Biológicos , Muramidasa/genética , Muramidasa/fisiología , Neuropéptidos/fisiología , Receptor de Insulina/genética , Receptor de Insulina/fisiología , Receptores de Superficie Celular/fisiología , Saposinas/genética , Saposinas/fisiología , Transducción de Señal/inmunología , Transducción de Señal/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Factor de Crecimiento Transformador beta/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
15.
Immunol Rev ; 198: 36-58, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15199953

RESUMEN

Simple model organisms that are amenable to comprehensive experimental analysis can be used to elucidate the molecular genetic architecture of complex traits. They can thereby enhance our understanding of these traits in other organisms, including humans. Here, we describe the use of the nematode Caenorhabditis elegans as a tractable model system to study innate immunity. We detail our current understanding of the worm's immune system, which seems to be characterized by four main signaling cascades: a p38 mitogen-activated protein kinase, a transforming growth factor-beta-like, a programed cell death, and an insulin-like receptor pathway. Many details, especially regarding pathogen recognition and immune effectors, are only poorly characterized and clearly warrant further investigation. We additionally speculate on the evolution of the C. elegans immune system, taking into special consideration the relationship between immunity, stress responses and digestion, the diversification of the different parts of the immune system in response to multiple and/or coevolving pathogens, and the trade-off between immunity and host life history traits. Using C. elegans to address these different facets of host-pathogen interactions provides a fresh perspective on our understanding of the structure and complexity of innate immune systems in animals and plants.


Asunto(s)
Evolución Biológica , Caenorhabditis elegans/inmunología , Sistema Inmunológico , Secuencia de Aminoácidos , Animales , Bacterias/patogenicidad , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiología , Interacciones Huésped-Parásitos , Inmunidad Innata , Modelos Inmunológicos , Datos de Secuencia Molecular , Transducción de Señal
16.
Microbiology (Reading) ; 150(Pt 6): 1901-1910, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15184576

RESUMEN

The enzyme LuxS is responsible for the production of autoinducer-2 (AI-2), a molecule that has been implicated in quorum sensing in many bacterial species. This study investigated whether there is a luxS-dependent signalling system in the Gram-negative bacteria Serratia spp. Serratia marcescens is a broad-host-range pathogen and an important cause of nosocomial infections. Production of AI-2 activity was detected in S. marcescens ATCC 274 and Serratia ATCC 39006 and their luxS genes were sequenced. luxS mutants were constructed in these strains and were analysed to determine which phenotypes are regulated by luxS and therefore, potentially, by AI-2. The phenotypes of the luxS mutants included decreased carbapenem antibiotic production in Serratia ATCC 39006 and decreased prodigiosin and secreted haemolysin production in S. marcescens ATCC 274. The luxS mutant of S. marcescens ATCC 274 was also found to exhibit modestly reduced virulence in a Caenorhabditis elegans model. Finally, it was shown that the culture supernatant of a wild-type strain contains a signal, presumably AI-2, capable of complementing the prodigiosin defect of the luxS mutant of another strain, even when substantially diluted. It is concluded that luxS modulates virulence and antibiotic production in Serratia, in a strain-dependent manner, and that, for at least one phenotype, this regulation is via extracellular signalling.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Homoserina/análogos & derivados , Homoserina/metabolismo , Lactonas/metabolismo , Mutación , Serratia/metabolismo , Serratia/patogenicidad , Animales , Proteínas Bacterianas/genética , Caenorhabditis elegans/microbiología , Carbapenémicos/biosíntesis , Liasas de Carbono-Azufre , Datos de Secuencia Molecular , Prodigiosina/biosíntesis , Análisis de Secuencia de ADN , Serratia/genética , Serratia/crecimiento & desarrollo , Serratia marcescens/genética , Serratia marcescens/crecimiento & desarrollo , Serratia marcescens/metabolismo , Serratia marcescens/patogenicidad , Transducción de Señal , Especificidad de la Especie , Virulencia
17.
Nat Rev Genet ; 4(5): 380-90, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12728280

RESUMEN

Invaluable insights into how animals, humans included, defend themselves against infection have been provided by more than a decade of genetic studies that have used fruitflies. In the past few years, attention has also turned to another simple animal model, the nematode worm Caenorhabditis elegans. What exactly have we learned from the work in Drosophila? And will research with C. elegans teach us anything new about our response to pathogen attack?


Asunto(s)
Caenorhabditis elegans/inmunología , Modelos Genéticos , Animales , Drosophila melanogaster/inmunología , Inmunidad Innata/genética , Transducción de Señal
18.
EMBO J ; 22(7): 1451-60, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12660152

RESUMEN

The human opportunistic pathogen Serratia marcescens is a bacterium with a broad host range, and represents a growing problem for public health. Serratia marcescens kills Caenorhabditis elegans after colonizing the nematode's intestine. We used C.elegans to screen a bank of transposon-induced S.marcescens mutants and isolated 23 clones with an attenuated virulence. Nine of the selected bacterial clones also showed a reduced virulence in an insect model of infection. Of these, three exhibited a reduced cytotoxicity in vitro, and among them one was also markedly attenuated in its virulence in a murine lung infection model. For 21 of the 23 mutants, the transposon insertion site was identified. This revealed that among the genes necessary for full in vivo virulence are those that function in lipopolysaccharide (LPS) biosynthesis, iron uptake and hemolysin production. Using this system we also identified novel conserved virulence factors required for Pseudomonas aeruginosa pathogenicity. This study extends the utility of C.elegans as an in vivo model for the study of bacterial virulence and advances the molecular understanding of S.marcescens pathogenicity.


Asunto(s)
Caenorhabditis elegans/microbiología , Serratia marcescens/patogenicidad , Virulencia , Animales , Datos de Secuencia Molecular , Mutación , Serratia marcescens/genética
19.
Curr Biol ; 12(14): 1209-14, 2002 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-12176330

RESUMEN

The term innate immunity refers to a number of evolutionary ancient mechanisms that serve to defend animals and plants against infection. Genetically tractable model organisms, especially Drosophila, have contributed greatly to advances in our understanding of mammalian innate immunity. Essentially, nothing is known about immune responses in the nematode Caenorhabditis elegans. Using high-density cDNA microarrays, we show here that infection of C. elegans by the Gram-negative bacterium Serratia marcescens provokes a marked upregulation of the expression of many genes. Among the most robustly induced are genes encoding lectins and lysozymes, known to be involved in immune responses in other organisms. Certain infection-inducible genes are under the control of the DBL-1/TGFbeta pathway. We found that dbl-1 mutants exhibit increased susceptibility to infection. Conversely, overexpression of the lysozyme gene lys-1 augments the resistance of C. elegans to S. marcescens. These results constitute the first demonstration of inducible antibacterial defenses in C. elegans and open new avenues for the investigation of evolutionary conserved mechanisms of innate immunity.


Asunto(s)
Caenorhabditis elegans/inmunología , Serratia marcescens/fisiología , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/microbiología , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...