Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 580(7805): E20, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32350466

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Cell ; 143(5): 694-702, 2010 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21111231

RESUMEN

Posttranslational modifications are increasingly recognized as key strategies used by bacterial and viral pathogens to modulate host factors critical for infection. A number of recent studies illustrate how pathogens use these posttranslational modifications to target central signaling pathways in the host cell, such as the NF-kB and MAP kinase pathways, which are essential for pathogens' replication, propagation, and evasion from host immune responses. These discoveries open new avenues for investigating the fundamental mechanisms of pathogen infection and the development of new therapeutics.


Asunto(s)
Bacterias/inmunología , Procesamiento Proteico-Postraduccional , Virus/inmunología , Bacterias/metabolismo , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Transducción de Señal , Virosis/inmunología , Virosis/virología , Virus/metabolismo
3.
J Virol ; 93(5)2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30541852

RESUMEN

Approximately 10% of the mouse genome is composed of endogenous retroviruses belonging to different families. In contrast to the situation in the human genome, several of these families correspond to recent, still-infectious elements capable of encoding complete viral particles. The mouse GLN endogenous retrovirus is one of these active families. We previously identified one fully functional provirus from the sequenced genome of the C57BL/6 mouse strain. The GLN envelope protein gives the infectious viral particles an ecotropic host range, and we had demonstrated that the receptor was neither CAT1 nor SMIT1, the two previously identified receptors for mouse ecotropic retroviral envelope proteins. In this study, we have identified SLC19A1, the reduced folate carrier, as the cellular protein used as a receptor by the GLN retrovirus. The ecotropic tropism exhibited by this envelope is due to the presence or absence of an N-linked glycosylation site in the first extracellular loop as well as the specific amino acid sequence of the extracellular domains of the receptor. Like all the other retroviral envelope proteins from the gammaretrovirus genus whose receptors have been identified, the GLN envelope protein uses a member of the solute carrier superfamily as a receptor.IMPORTANCE Endogenous retroviruses are genomic traces of past infections present in all vertebrates. Most of these elements degenerate over time and become nonfunctional, but the mouse genome still contains several families with full infection abilities. The GLN retrovirus is one of them, and its members encode particles that are able to infect only mouse cells. Here, we identified the cellular protein used as a receptor by GLN for cell entry. It is SLC19A1, the reduced folate carrier. We show that GLN infection is limited to mouse cells due to both a mutation in the mouse gene preventing the glycosylation of SLC19A1 and also other residues conserved within the rat but not in the hamster and human proteins. Like all other gammaretroviruses whose receptors have been identified, GLN uses a member of the solute carrier superfamily for cell entry, highlighting the role of these proteins for retroviral infection in mammals.


Asunto(s)
Gammaretrovirus/metabolismo , Productos del Gen env/genética , Receptores Virales/genética , Proteína Portadora de Folato Reducido/genética , Proteínas del Envoltorio Viral/genética , Acoplamiento Viral , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Gammaretrovirus/genética , Genoma/genética , Glicosilación , Células HEK293 , Especificidad del Huésped , Humanos , Ratones , Ratones Endogámicos C57BL , Ratas , Proteína Portadora de Folato Reducido/metabolismo , Infecciones por Retroviridae/virología
4.
BMC Microbiol ; 20(1): 340, 2020 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-33176677

RESUMEN

BACKGROUND: The use of animal models with depleted intestinal microbiota has recently increased thanks to the huge interest in the potential role of these micro-organisms in human health. In particular, depletion of gut bacteria using antibiotics has recently become popular as it represents a low cost and easy alternative to germ-free animals. Various regimens of antibiotics are used in the literature, which differ in composition, dose, length of treatment and mode of administration. In order to help investigators in choosing the most appropriate protocol for their studies, we compared here three modes of antibiotic delivery to deplete gut bacteria in C57Bl/6 mice. We delivered one of the most frequently used combination of antibiotics (a mix of ampicillin, neomycin, metronidazole and vancomycin) either ad libitum in drinking water or by oral gavage once or twice per day. RESULTS: We quantified the global bacterial density, as well as the abundance of specific bacterial and fungal taxa, in mouse feces in response to antibiotics exposure. We observed that oral gavage once a day with antibiotics is not a reliable method as it occasionally triggers hyperproliferation of bacteria belonging to the Escherichia/Shigella taxon and leads, as a consequence, to a moderate decrease in fecal bacterial density. Antibiotics delivery by oral gavage twice a day or in drinking water induces in contrast a robust and consistent depletion of mouse fecal bacteria, as soon as 4 days of treatment, and is associated with an increase in fecal moisture content. Extending exposure to antibiotics beyond 7 days does not improve total bacteria depletion efficiency and promotes fungal overgrowth. We show in addition that all tested protocols impact neither gut microbiota recolonization efficiency, 1 or 2 weeks after the stop of antibiotics, nor mice body composition after 1 week of treatment. CONCLUSIONS: Our study provides key experimental data and highlights important parameters to consider before selecting an appropriate protocol for antibiotic-mediated depletion of gut bacteria, in order to optimize the accuracy and the reproducibility of results and to facilitate comparison between studies.


Asunto(s)
Antibacterianos/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Administración Oral , Animales , Antibacterianos/farmacología , Bacterias/clasificación , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/crecimiento & desarrollo , Composición Corporal , Heces/microbiología , Hongos/clasificación , Hongos/efectos de los fármacos , Hongos/genética , Hongos/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL
5.
Mol Cell Proteomics ; 17(8): 1627-1636, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29752379

RESUMEN

Bacterial pathogens use various strategies to interfere with host cell functions. Among these strategies, bacteria modulate host gene transcription, thereby modifying the set of proteins synthetized by the infected cell. Bacteria can also target pre-existing host proteins and modulate their post-translational modifications or trigger their degradation. Analysis of protein levels variations in host cells during infection allows to integrate both transcriptional and post-transcriptional regulations induced by pathogens. Here, we focused on host proteome alterations induced by the toxin Listeriolysin O (LLO), secreted by the bacterial pathogen Listeria monocytogenes. We showed that a short-term treatment with LLO remodels the host cell proteome by specifically decreasing the abundance of 149 proteins. The same decrease in host protein levels was observed in different epithelial cell lines but not in macrophages. We show in particular that this proteome remodeling affects several ubiquitin and ubiquitin-like ligases and that LLO leads to major changes in the host ubiquitylome. Strikingly, this toxin-induced proteome remodeling involves only post-transcriptional regulations, as no modification in the transcription levels of the corresponding genes was observed. In addition, we could show that Perfringolysin O, another bacterial pore-forming toxin similar to LLO, also induces host proteome changes. Taken together, our data reveal that different bacterial pore-forming toxins induce important host proteome remodeling, that may impair epithelial cell functions.


Asunto(s)
Toxinas Bacterianas/toxicidad , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Proteínas de Choque Térmico/toxicidad , Proteínas Hemolisinas/toxicidad , Interacciones Huésped-Patógeno , Proteoma/metabolismo , Animales , Regulación hacia Abajo/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células HeLa , Células Hep G2 , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Células RAW 264.7 , Ubiquitinación/efectos de los fármacos
6.
Cell Microbiol ; 19(4)2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27739224

RESUMEN

Bacterial pathogens can interfere during infection with host cell organelles, such as mitochondria, the endoplasmic reticulum-Golgi system or nuclei. As important cellular functions are often compartmentalized in these organelles, their targeting allows pathogens to manipulate key host functions during infection. Here, we identify lysosomes as a new class of organelles targeted by the pathogenic bacterium Listeria monocytogenes. We demonstrate that extracellular Listeria, via secretion of the pore-forming toxin listeriolysin O, alters lysosomal integrity in epithelial cells but not in macrophages. Listeriolysin O induces lysosomal membrane permeabilization and release of lysosomal content, such as cathepsins proteases, which remain transiently active in the host cytosol. We furthermore show that other bacterial pore-forming toxins, such as perfringolysin O and pneumolysin, also induce lysosomes alteration. Together, our data unveil a novel activity of bacterial cholesterol-dependent cytolysins.


Asunto(s)
Células Epiteliales/microbiología , Proteínas de Choque Térmico/fisiología , Proteínas Hemolisinas/fisiología , Listeria monocytogenes/fisiología , Listeriosis/microbiología , Lisosomas/fisiología , Animales , Toxinas Bacterianas , Células CACO-2 , Permeabilidad de la Membrana Celular , Células HeLa , Células Hep G2 , Humanos , Listeriosis/patología , Lisosomas/microbiología , Ratones , Proteolisis , Células RAW 264.7
7.
Proc Natl Acad Sci U S A ; 111(34): 12432-7, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25114211

RESUMEN

SUMOylation is an essential ubiquitin-like modification involved in important biological processes in eukaryotic cells. Identification of small ubiquitin-related modifier (SUMO)-conjugated residues in proteins is critical for understanding the role of SUMOylation but remains experimentally challenging. We have set up a powerful and high-throughput method combining quantitative proteomics and peptide immunocapture to map SUMOylation sites and have analyzed changes in SUMOylation in response to stimuli. With this technique we identified 295 SUMO1 and 167 SUMO2 sites on endogenous substrates of human cells. We further used this strategy to characterize changes in SUMOylation induced by listeriolysin O, a bacterial toxin that impairs the host cell SUMOylation machinery, and identified several classes of host proteins specifically deSUMOylated in response to this toxin. Our approach constitutes an unprecedented tool, broadly applicable to various SUMO-regulated cellular processes in health and disease.


Asunto(s)
Proteína SUMO-1/química , Proteína SUMO-1/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/química , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación , Secuencia de Aminoácidos , Toxinas Bacterianas/toxicidad , Sitios de Unión , Células HeLa , Proteínas de Choque Térmico/toxicidad , Proteínas Hemolisinas/toxicidad , Humanos , Datos de Secuencia Molecular , Mapeo Peptídico/métodos , Proteómica/métodos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína SUMO-1/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Sumoilación/efectos de los fármacos
8.
Nature ; 464(7292): 1192-5, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20414307

RESUMEN

During infection, pathogenic bacteria manipulate the host cell in various ways to allow their own replication, propagation and escape from host immune responses. Post-translational modifications are unique mechanisms that allow cells to rapidly, locally and specifically modify activity or interactions of key proteins. Some of these modifications, including phosphorylation and ubiquitylation, can be induced by pathogens. However, the effects of pathogenic bacteria on SUMOylation, an essential post-translational modification in eukaryotic cells, remain largely unknown. Here we show that infection with Listeria monocytogenes leads to a decrease in the levels of cellular SUMO-conjugated proteins. This event is triggered by the bacterial virulence factor listeriolysin O (LLO), which induces a proteasome-independent degradation of Ubc9, an essential enzyme of the SUMOylation machinery, and a proteasome-dependent degradation of some SUMOylated proteins. The effect of LLO on Ubc9 is dependent on the pore-forming capacity of the toxin and is shared by other bacterial pore-forming toxins like perfringolysin O (PFO) and pneumolysin (PLY). Ubc9 degradation was also observed in vivo in infected mice. Furthermore, we show that SUMO overexpression impairs bacterial infection. Together, our results reveal that Listeria, and probably other pathogens, dampen the host response by decreasing the SUMOylation level of proteins critical for infection.


Asunto(s)
Listeria monocytogenes/patogenicidad , Listeriosis/metabolismo , Listeriosis/microbiología , Procesamiento Proteico-Postraduccional , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Animales , Toxinas Bacterianas/metabolismo , Línea Celular , Células HeLa , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Humanos , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Ratones , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Factores de Virulencia/metabolismo
9.
Biol Sex Differ ; 15(1): 6, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38217033

RESUMEN

RATIONALE: Patients with anorexia nervosa (AN) often present sleep disorders and circadian hormonal dysregulation. The role of the microbiota-gut-brain axis in the regulation of feeding behavior has emerged during the last decades but its relationships with the circadian rhythm remains poorly documented. Thus, we aimed to characterize the circadian clock genes expression in peripheral and central tissues in the activity-based anorexia mouse model (ABA), as well as the dynamics of the gut-microbiota composition. METHODS: From day 1 to day 17, male and female C57Bl/6 mice were submitted or not to the ABA protocol (ABA and control (CT) groups), which combines a progressive limited access to food and a free access to a running wheel. At day 17, fasted CT and ABA mice were euthanized after either resting (EoR) or activity (EoA) phase (n = 10-12 per group). Circadian clock genes expression was assessed by RT-qPCR on peripheral (liver, colon and ileum) and central (hypothalamic suprachiasmatic nucleus or SCN) tissues. Cecal bacterial taxa abundances were evaluated by qPCR. Data were compared by two-way ANOVA followed by post-tests. RESULTS: ABA mice exhibited a lower food intake, a body weight loss and an increase of diurnal physical activity that differ according with the sex. Interestingly, in the SCN, only ABA female mice exhibited altered circadian clock genes expression (Bmal1, Per1, Per2, Cry1, Cry2). In the intestinal tract, modification of clock genes expression was also more marked in females compared to males. For instance, in the ileum, female mice showed alteration of Bmal1, Clock, Per1, Per2, Cry1, Cry2 and Rev-erbα mRNA levels, while only Per2 and Cry1 mRNAs were affected by ABA model in males. By contrast, in the liver, clock genes expression was more markedly affected in males compared to females in response to ABA. Finally, circadian variations of gut-bacteria abundances were observed in both male and female mice and sex-dependent alteration were observed in response to the ABA model. CONCLUSIONS: This study shows that alteration of circadian clock genes expression at both peripheral and central levels occurs in response to the ABA model. In addition, our data underline that circadian variations of the gut-microbiota composition are sex-dependent.


Anorexia nervosa is an eating disorder with a female predominance. However, the underlying pathophysiological mechanisms are still incompletely understood. Patients with anorexia nervosa often show alterations in circadian rhythm, including sleep disorders and modifications in hormone circadian rhythm. The circadian rhythm is controlled in the central nervous system, particularly in the suprachiasmatic nucleus, but clocks have also been described in peripheral tissues. To better understand the putative role of circadian rhythm in the pathophysiology of anorexia nervosa, we have conducted an experimental study in a rodent model of anorexia nervosa called "activity-based anorexia" on both males and females. Interestingly, we observed that the expression of genes involved in the circadian rhythm is affected by the activity-based anorexia model in both the suprachiasmatic nucleus and peripheral tissues, such as the small intestine and liver. In addition, gut­microbiota also shows circadian variation. Interestingly, the anorexia-induced alterations of circadian variations (clock genes expression and gut­microbiota composition) are sex- and tissue-dependent. For instance, female mice exhibited more marked alterations in the ileum, whereas, in males, modifications were more pronounced in the liver. This study highlights sex-dependent alterations of circadian clock genes expression and of gut­microbiota in response to the anorexia rodent model. Further experiments should be performed to investigate the contribution of these mechanisms in the etiology of anorexia nervosa and the higher prevalence in females.


Asunto(s)
Factores de Transcripción ARNTL , Microbiota , Animales , Femenino , Masculino , Ratones , Anorexia , Factores de Transcripción ARNTL/genética , Ritmo Circadiano/genética , Expresión Génica , ARN Mensajero/metabolismo , Proteínas CLOCK
10.
PLoS Pathog ; 7(10): e1002309, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22028653

RESUMEN

The IAPE (Intracisternal A-type Particles elements with an Envelope) family of murine endogenous retroelements is present at more than 200 copies in the mouse genome. We had previously identified a single copy that proved to be fully functional, i.e. which can generate viral particles budding out of the cell and infectious on a series of cells, including human cells. We also showed that IAPE are the progenitors of the highly reiterated IAP elements. The latter are now strictly intracellular retrotransposons, due to the loss of the envelope gene and re-localisation of the associated particles in the course of evolution. In the present study we searched for the cellular receptor of the IAPE elements, by using a lentiviral human cDNA library and a pseudotype assay on transduced cells. We identified Ephrin A4, a GPI-anchored molecule involved in several developmental processes, as a receptor for the IAPE pseudotypes. We also found that the other 4 members of the Ephrin A family -but not those of the closely related Ephrin B family- were also able to mediate IAPE cell entry, thus significantly increasing the amount of possible cell types susceptible to IAPE infection. We show that these include mouse germline cells, as illustrated by immunohistochemistry experiments, consistent with IAPE genomic amplification by successive re-infection. We propose that the uncovered properties of the identified receptors played a role in the accumulation of IAPE elements in the mouse genome, and in the survival of a functional copy.


Asunto(s)
Retrovirus Endógenos/patogenicidad , Efrinas/metabolismo , Interacciones Huésped-Patógeno/fisiología , Infecciones por Retroviridae/virología , Animales , Chlorocebus aethiops , Retrovirus Endógenos/genética , Efrina-A4/genética , Efrina-A4/metabolismo , Efrinas/genética , Femenino , Regulación Viral de la Expresión Génica , Biblioteca de Genes , Genes de Partícula A Intracisternal/genética , Genes Virales , Células HEK293 , Humanos , Ratones , Ovario/metabolismo , Infecciones por Retroviridae/metabolismo , Células Vero , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA