Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 4290, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37463962

RESUMEN

Endo-lysosomes transport along microtubules and clustering in the perinuclear area are two necessary steps for microbes to activate specialized phagocyte functions. We report that RUN and FYVE domain-containing protein 3 (RUFY3) exists as two alternative isoforms distinguishable by the presence of a C-terminal FYVE domain and by their affinity for phosphatidylinositol 3-phosphate on endosomal membranes. The FYVE domain-bearing isoform (iRUFY3) is preferentially expressed in primary immune cells and up-regulated upon activation by microbes and Interferons. iRUFY3 is necessary for ARL8b + /LAMP1+ endo-lysosomes positioning in the pericentriolar organelles cloud of LPS-activated macrophages. We show that iRUFY3 controls macrophages migration, MHC II presentation and responses to Interferon-γ, while being important for intracellular Salmonella replication. Specific inactivation of rufy3 in phagocytes leads to aggravated pathologies in mouse upon LPS injection or bacterial pneumonia. This study highlights the role of iRUFY3 in controlling endo-lysosomal dynamics, which contributes to phagocyte activation and immune response regulation.


Asunto(s)
Presentación de Antígeno , Lipopolisacáridos , Animales , Ratones , Endosomas/metabolismo , Lipopolisacáridos/metabolismo , Lisosomas/metabolismo , Fagocitos
2.
Microb Cell ; 9(2): 24-41, 2022 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-35127930

RESUMEN

Salmonellae are bacteria that cause moderate to severe infections in humans, depending on the strain and the immune status of the infected host. These pathogens have the particularity of residing in the cells of the infected host. They are usually found in a vacuolar compartment that the bacteria shape with the help of effector proteins. Following invasion of a eukaryotic cell, the bacterial vacuole undergoes maturation characterized by changes in localization, composition and morphology. In particular, membrane tubules stretching over the microtubule cytoskeleton are formed from the bacterial vacuole. Although these tubules do not occur in all infected cells, they are functionally important and promote intracellular replication. This review focuses on the role and significance of membrane compartment remodeling observed in infected cells and the bacterial and host cell pathways involved.

3.
J Cell Sci ; 135(1)2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34878110

RESUMEN

When intracellular, pathogenic Salmonella reside in a membrane compartment composed of interconnected vacuoles and tubules, the formation of which depends on the translocation of bacterial effectors into the host cell. Cytoskeletons and their molecular motors are prime targets for these effectors. In this study, we show that the microtubule molecular motor KIF1Bß (a splice variant of KIF1B), a member of the kinesin-3 family, is a key element for the establishment of the Salmonella replication niche as its absence is detrimental to the stability of bacterial vacuoles and the formation of associated tubules. Kinesin-3 interacts with the Salmonella effector SifA but also with SKIP (also known as PLEKHM2), a host protein complexed to SifA. The interaction with SifA is essential for the recruitment of kinesin-3 on Salmonella vacuoles whereas that with SKIP is incidental. In the non-infectious context, however, the interaction with SKIP is essential for the recruitment and activity of kinesin-3 only on a fraction of the lysosomes. Finally, our results show that, in infected cells, the presence of SifA establishes a kinesin-1 and kinesin-3 recruitment pathway that is analogous to and functions independently of that mediated by the Arl8a and Arl8b GTPases. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Proteínas Bacterianas , Cinesinas , Factores de Ribosilacion-ADP , Proteínas Bacterianas/metabolismo , Glicoproteínas/metabolismo , Células HeLa , Humanos , Cinesinas/genética , Salmonella/metabolismo , Vacuolas/metabolismo
4.
Sci Rep ; 11(1): 4342, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33619301

RESUMEN

Induction of host cell autophagy by starvation was shown to enhance lysosomal delivery to mycobacterial phagosomes, resulting in the restriction of Mycobacterium tuberculosis reference strain H37Rv. Our previous study showed that strains belonging to M. tuberculosis Beijing genotype resisted starvation-induced autophagic elimination but the factors involved remained unclear. Here, we conducted RNA-Seq of macrophages infected with the autophagy-resistant Beijing strain (BJN) compared to macrophages infected with H37Rv upon autophagy induction by starvation. Results identified several genes uniquely upregulated in BJN-infected macrophages but not in H37Rv-infected cells, including those encoding Kxd1 and Plekhm2, which function in lysosome positioning towards the cell periphery. Unlike H37Rv, BJN suppressed enhanced lysosome positioning towards the perinuclear region and lysosomal delivery to its phagosome upon autophagy induction by starvation, while depletion of Kxd1 and Plekhm2 reverted such effects, resulting in restriction of BJN intracellular survival upon autophagy induction by starvation. Taken together, these data indicated that Kxd1 and Plekhm2 are important for the BJN strain to suppress lysosome positioning towards the perinuclear region and lysosomal delivery into its phagosome during autophagy induction by starvation to evade starvation-induced autophagic restriction.


Asunto(s)
Autofagia , Interacciones Huésped-Patógeno , Lisosomas/metabolismo , Lisosomas/microbiología , Mycobacterium tuberculosis/fisiología , Tuberculosis/metabolismo , Tuberculosis/microbiología , Autofagia/genética , Proteínas Portadoras/genética , Biología Computacional/métodos , Perfilación de la Expresión Génica , Ontología de Genes , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Anotación de Secuencia Molecular , Transcriptoma , Tuberculosis/genética , Tuberculosis/inmunología
5.
Methods Mol Biol ; 2182: 117-126, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32894491

RESUMEN

Salmonella enterica is a Gram-negative intracellular pathogen that causes a range of life-threatening diseases in humans and animals worldwide. In a systemic infection, the ability of Salmonella to survive/replicate in macrophages, particularly in the liver and spleen, is crucial for virulence. Transformed macrophage cell lines and primary macrophages prepared from mouse bone marrow are commonly used models for the study of Salmonella infection. However, these models raise technical or ethical issues that highlight the need for alternative methods. This chapter describes a technique for immortalizing early hematopoietic progenitor cells derived from wild-type or transgenic mice and using them to produce macrophages. It validates, through a specific example, the interest of this cellular approach for the study of Salmonella infection.


Asunto(s)
Células Precursoras de Granulocitos/microbiología , Proteínas de Homeodominio/metabolismo , Macrófagos/microbiología , Infecciones por Salmonella/microbiología , Animales , Línea Celular Transformada/metabolismo , Línea Celular Transformada/microbiología , Línea Celular Transformada/patología , Línea Celular Tumoral , Células Precursoras de Granulocitos/metabolismo , Hígado/metabolismo , Hígado/microbiología , Hígado/patología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/patología , Salmonella enterica/patogenicidad , Bazo/metabolismo , Bazo/microbiología , Bazo/patología , Virulencia/genética
6.
Cell Microbiol ; 22(12): e13260, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32902857

RESUMEN

Tetraspanins, a wide family composed of 33 transmembrane proteins, are associated with different types of proteins through which they arbitrate important cellular processes such as fusion, adhesion, invasion, tissue differentiation and immunological responses. Tetraspanins share a comparable structural design, which consists of four hydrophobic transmembrane domains with cytoplasmic and extracellular loops. They cooperate with different proteins, including other tetraspanins, receptors or signalling proteins to compose functional complexes at the cell surface, designated tetraspanin-enriched microdomains (TEM). Increasing evidences establish that tetraspanins are exploited by numerous intracellular pathogens as a doorway for entering and replicating within human cells. Although previous surveys focused mainly on viruses and parasites, it is now becoming clear that bacteria interact with tetraspanins, using TEM as a "gateway" to infection. In this review, we examine the biological functions of tetraspanins that are relevant to bacterial infective procedures and consider the available data that reveal how different bacteria benefit from host cell tetraspanins in infection and in the pathogenesis of diseases. We will also emphasise the stimulating potentials of targeting tetraspanins for preventing bacterial infectious diseases, using specific neutralising antibodies or anti-adhesion peptide-based therapies. Such innovative therapeutic opportunities may deliver alternatives for fighting difficult-to-manage and drug-resistant bacterial pathogens.


Asunto(s)
Infecciones Bacterianas/microbiología , Proteínas de la Membrana/metabolismo , Tetraspaninas/química , Tetraspaninas/metabolismo , Animales , Membrana Celular/metabolismo , Humanos , Ratones
7.
J Cell Sci ; 133(9)2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32409568

RESUMEN

Salmonella enterica is an intracellular bacterial pathogen. The formation of its replication niche, which is composed of a vacuole associated with a network of membrane tubules, depends on the secretion of a set of bacterial effector proteins whose activities deeply modify the functions of the eukaryotic host cell. By recruiting and regulating the activity of the kinesin-1 molecular motor, Salmonella effectors PipB2 and SifA play an essential role in the formation of the bacterial compartments. In particular, they allow the formation of tubules from the vacuole and their extension along the microtubule cytoskeleton, and thus promote membrane exchanges and nutrient supply. We have developed in vitro and in cellulo assays to better understand the specific role played by these two effectors in the recruitment and regulation of kinesin-1. Our results reveal a specific interaction between the two effectors and indicate that, contrary to what studies on infected cells suggested, interaction with PipB2 is sufficient to relieve the autoinhibition of kinesin-1. Finally, they suggest the involvement of other Salmonella effectors in the control of the activity of this molecular motor.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Salmonella enterica , Proteínas Bacterianas , Células HeLa , Humanos , Cinesinas/genética , Salmonella , Vacuolas
8.
Front Microbiol ; 11: 614, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32328049

RESUMEN

Salmonella enterica serotype Typhimurium is a bacterium that causes gastroenteritis and diarrhea in humans. The genome of S. Typhimurium codes for diverse virulence factors, among which are the toxin-antitoxin (TA) systems. SehAB is a type II TA, where SehA is the toxin and SehB is the antitoxin. It was previously reported that the absence of the SehB antitoxin affects the growth of S. Typhimurium. In addition, the SehB antitoxin can interact directly with the SehA toxin neutralizing its toxic effect as well as repressing its own expression. We identified conserved residues on SehB homologous proteins. Point mutations were introduced at both N- and C-terminal of SehB antitoxin to analyze the effect of these changes on its transcription repressor function, on its ability to form homodimers and on the virulence of S. Typhimurium. All changes in amino acid residues at both the N- and C-terminal affected the repressor function of SehB antitoxin and they were required for DNA-binding activity. Mutations in the amino acid residues at the N-terminal showed a lower capacity for homodimer formation of the SehB protein. However, none of the SehB point mutants were affected in the interaction with the SehA toxin. In terms of virulence, the eight single-amino acid mutations were attenuated for virulence in the mouse model. In agreement with our results, the eight amino acid residues of SehB antitoxin were required for its repressor activity, affecting both homodimerization and DNA-binding activity, supporting the notion that both activities of SehB antitoxin are required to confer virulence to Salmonella enterica.

9.
Cell Microbiol ; 22(4): e13164, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31953913

RESUMEN

The strategies by which intracellular pathogenic bacteria manipulate innate immunity to establish chronicity are poorly understood. Here, we show that Brucella abortus outer membrane protein Omp25 specifically binds the immune cell receptor SLAMF1 in vitro. The Omp25-dependent engagement of SLAMF1 by B. abortus limits NF-κB translocation in dendritic cells (DCs) with no impact on Brucella intracellular trafficking and replication. This in turn decreases pro-inflammatory cytokine secretion and impairs DC activation. The Omp25-SLAMF1 axis also dampens the immune response without affecting bacterial replication in vivo during the acute phase of Brucella infection in a mouse model. In contrast, at the chronic stage of infection, the Omp25/SLAMF1 engagement is essential for Brucella persistence. Interaction of a specific bacterial protein with an immune cell receptor expressed on the DC surface at the acute stage of infection is thus a powerful mechanism to support microbe settling in its replicative niche and progression to chronicity.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Brucella abortus/inmunología , Células Dendríticas/microbiología , Interacciones Huésped-Patógeno/inmunología , Inflamación , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Brucella abortus/genética , Brucella abortus/patogenicidad , Células Dendríticas/inmunología , Femenino , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Unión Proteica , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética
10.
Clin Infect Dis ; 70(11): 2405-2412, 2020 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-31309973

RESUMEN

BACKGROUND: Etiological diagnosis is a key to therapeutic adaptation and improved prognosis, particularly for infections such as endocarditis. In blood culture-negative endocarditis (BCNE), 22% of cases remain undiagnosed despite an updated comprehensive syndromic approach. This prompted us to develop a new diagnostic approach. METHODS: Eleven valves from 10 BCNE patients were analyzed using a method that combines human RNA bait-depletion with phi29 DNA polymerase-based multiple displacement amplification and shotgun DNA sequencing. An additional case in which a microbe was serendipitously visualized by immunofluorescence was analyzed using the same method, but after laser capture microdissection. RESULTS: Background DNA prevented any diagnosis in cases analyzed without microdissection because the majority of sequences were contaminants. Moraxella sequences were dramatically enriched in the stained microdissected region of the additional case. A consensus genome sequence of 2.4 Mbp covering more than 94% of the Moraxella osloensis KSH reference genome was reconstructed with 234X average coverage. Several antibiotic-resistance genes were observed. Etiological diagnosis was confirmed using Western blot and specific polymerase chain reaction with sequencing on a different valve sample. CONCLUSIONS: Microdissection could be a key to the metagenomic diagnosis of infectious diseases when a microbe is visualized but remains unidentified despite an updated optimal approach. Moraxella osloensis should be tested in blood culture-negative endocarditis.


Asunto(s)
Endocarditis Bacteriana , Endocarditis , Cultivo de Sangre , Endocarditis/diagnóstico , Endocarditis Bacteriana/diagnóstico , Humanos , Metagenómica , Moraxella
11.
Cell Microbiol ; 20(12): e12951, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30212607

RESUMEN

Cells infected with Salmonella are characterised by the appearance of membrane tubular structures that stretch from the bacterial vacuole. The formation of these tubules requires the translocation of Salmonella effector proteins within the infected cell. Different types of Salmonella-induced tubules with varying host protein compositions have been identified. This variability probably reflects the ability of these tubules to interact with different host compartments. Membrane tubules decorated with effector proteins but essentially devoid of host proteins and named LAMP1-negative (LNT) were observed. LNTs wrap around LAMP1-positive vesicles and may promote recruitment of lysosomal glycoproteins to bacterial vacuole and the formation of a replication niche. We conducted a biochemical and functional characterisation of LNTs. We show that the effector proteins SseF and SseG are necessary for their formation. The absence of these tubules is associated with decreased recruitment of LAMP1 to SCVs, decreased intracellular replication of Salmonella, and decreased virulence in mice. We found that the process leading to the recruitment of lysosomal glycoproteins to tubules involves the C-terminal domain of the effector protein SifA and the GTPase Arl8b. Overall, these data suggest that Salmonella-induced tubules promote the establishment of the replication niche by promoting recruitment of host proteins to the bacterial vacuole.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Bacterianas/metabolismo , Glicoproteínas/metabolismo , Interacciones Huésped-Patógeno/fisiología , Salmonella typhimurium/patogenicidad , Factores de Virulencia/metabolismo , Factores de Ribosilacion-ADP/genética , Animales , Proteínas Bacterianas/genética , Glicoproteínas/genética , Células HeLa , Humanos , Proteínas de Membrana de los Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Microtúbulos/metabolismo , Dominios Proteicos , Células RAW 264.7 , Infecciones por Salmonella/microbiología , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo , Vacuolas , Factores de Virulencia/genética
12.
PLoS Genet ; 13(1): e1006556, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28085879

RESUMEN

Bacterial pathogens often deliver effectors into host cells using type 3 secretion systems (T3SS), the extremity of which forms a translocon that perforates the host plasma membrane. The T3SS encoded by Salmonella pathogenicity island 1 (SPI-1) is genetically associated with an acyl carrier protein, IacP, whose role has remained enigmatic. In this study, using tandem affinity purification, we identify a direct protein-protein interaction between IacP and the translocon protein SipB. We show, by mass spectrometry and radiolabelling, that SipB is acylated, which provides evidence for a modification of the translocon that has not been described before. A unique and conserved cysteine residue of SipB is identified as crucial for this modification. Although acylation of SipB was not essential to virulence, we show that this posttranslational modification promoted SipB insertion into host-cell membranes and pore-forming activity linked to the SPI-1 T3SS. Cooccurrence of acyl carrier and translocon proteins in several γ- and ß-proteobacteria suggests that acylation of the translocon is conserved in these other pathogenic bacteria. These results also indicate that acyl carrier proteins, known for their involvement in metabolic pathways, have also evolved as cofactors of new bacterial protein lipidation pathways.


Asunto(s)
Proteína Transportadora de Acilo/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Acetilación , Proteína Transportadora de Acilo/genética , Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo , Procesamiento Proteico-Postraduccional , Salmonella typhimurium/genética , Salmonella typhimurium/metabolismo
13.
Cell Microbiol ; 19(4)2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27704705

RESUMEN

Iron-sulfur (Fe-S)-containing proteins contribute to various biological processes, including redox reactions or regulation of gene expression. Living organisms have evolved by developing distinct biosynthetic pathways to assemble these clusters, including iron sulfur cluster (ISC) and sulfur mobilization (SUF). Salmonella enterica serovar Typhimurium is an intracellular pathogen responsible for a wide range of infections, from gastroenteritis to severe systemic diseases. Salmonella possesses all known prokaryotic systems to assemble Fe-S clusters, including ISC and SUF. Because iron starvation and oxidative stress are detrimental for Fe-S enzyme biogenesis and because such environments are often met by Salmonella during its intracellular life, we investigated the role of the ISC and SUF machineries during the course of the infection. The iscU mutant, which is predicted to have no ISC system functioning, was found to be defective for epithelial cell invasion and for mice infection, whereas the sufBC mutant, which is predicted to have no SUF system functioning, did not present any defect. Moreover, the iscU mutant was highly impaired in the expression of Salmonella pathogenicity island 1 (Spi1) type III secretion system that is essential for the first stage of Salmonella infection. The Fe-S cluster sensor IscR, a transcriptional regulator matured by the ISC machinery, was shown to bind the promoter of hilD, which encodes the master regulator of Spi1. IscR was also demonstrated to repress hilD and subsequently Spi1 gene expression, consistent with the observation that an IscR mutant is hyper-invasive in epithelial cells. Collectively, our findings indicate that the ISC machinery plays a central role in Salmonella virulence through the ability of IscR to down-regulate Spi1 gene expression. At a broader level, this model illustrates an adaptive mechanism used by bacterial pathogens to modulate their infectivity according to iron and oxygen availability.


Asunto(s)
Proteínas Bacterianas/fisiología , Proteínas Hierro-Azufre/fisiología , Salmonella enterica/genética , Factores de Transcripción/fisiología , Sistemas de Secreción Tipo III/genética , Animales , Secuencia de Bases , Sitios de Unión , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Regulación hacia Abajo , Expresión Génica , Regulación Bacteriana de la Expresión Génica , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Unión Proteica , Células RAW 264.7 , Salmonella enterica/metabolismo , Sistemas de Secreción Tipo III/metabolismo
15.
Virulence ; 8(6): 975-992, 2017 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-27936347

RESUMEN

Salmonella enterica serovar Typhimurium (S. Typhimurium) is a Gram-negative pathogen that causes various host-specific diseases. During their life cycle, Salmonellae survive frequent exposures to a variety of environmental stresses, e.g. carbon-source starvation. The virulence of this pathogen relies on its ability to establish a replicative niche, named Salmonella-containing vacuole, inside host cells. However, the microenvironment of the SCV and the bacterial metabolic pathways required during infection are largely undefined. In this work we developed different biological probes whose expression is modulated by the environment and the physiological state of the bacterium. We constructed transcriptional reporters by fusing promoter regions to the gfpmut3a gene to monitor the expression profile of genes involved in glucose utilization and lipid catabolism. The induction of these probes by a specific metabolic change was first tested in vitro, and then during different conditions of infection in macrophages. We were able to determine that Entner-Doudoroff is the main metabolic pathway utilized by Salmonella during infection in mouse macrophages. Furthermore, we found sub-populations of bacteria expressing genes involved in pathways for the utilization of different sources of carbon. These populations are modified in presence of different metabolizable substrates, suggesting the coexistence of Salmonella with diverse metabolic states during the infection.


Asunto(s)
Adaptación Fisiológica , Citoplasma/microbiología , Salmonella typhimurium/fisiología , Vacuolas/microbiología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Citometría de Flujo , Regulación Bacteriana de la Expresión Génica , Macrófagos/microbiología , Redes y Vías Metabólicas , Ratones , Regiones Promotoras Genéticas , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidad , Virulencia
16.
Virulence ; 7(6): 669-78, 2016 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-27046257

RESUMEN

Salmonella-infected cells are characterized by the presence of intra-cellular membranous tubules that emerge from bacterial vacuoles and extend along microtubules. The formation of Salmonella-induced tubules depends on the Salmonella pathogenicity island 2-encoded type III secretion system (T3SS-2) that translocates bacterial effector proteins inside host cells. Effector proteins have enzymatic activities or allow for hijacking of cellular functions. The role of Salmonella-induced tubules in virulence remains unclear but their absence is correlated with virulence defects. This study describes the presence of inter-cellular tubules that arise between daughter cells during cytokinesis. Inter-cellular tubules connect bacterial vacuoles originally present in the parent cell and that have been apportioned between daughters. Their formation requires a functional T3SS-2 and effector proteins. Our data establish a correlation between the formation of inter-cellular tubules and the asymmetric distribution of bacterial vacuoles in daughters. Thus, by manipulating the distribution of bacteria in cytokinetic cells, Salmonella T3SS-2 effector proteins may increase bacterial spreading and the systemic character of the infection.


Asunto(s)
Proteínas Bacterianas/fisiología , Ciclo Celular , Citocinesis , Salmonella typhimurium/citología , Salmonella typhimurium/fisiología , Sistemas de Secreción Tipo III/metabolismo , Islas Genómicas , Células HeLa , Humanos , Microscopía Fluorescente , Salmonella typhimurium/ultraestructura , Sistemas de Secreción Tipo III/genética , Vacuolas , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
17.
Sci Rep ; 5: 12979, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26268777

RESUMEN

The virulence of Salmonella relies on the expression of effector proteins that the bacterium injects inside infected cells. Salmonella enters eukaryotic cells and resides in a vacuolar compartment on which a number of effector proteins such as SifA are found. SifA plays an essential role in Salmonella virulence. It is made of two distinct domains. The N-terminal domain of SifA interacts with the host protein SKIP. This interaction regulates vacuolar membrane dynamics. The C-terminal has a fold similar to other bacterial effector domains having a guanine nucleotide exchange factor activity. Although SifA interacts with RhoA, it does not stimulate the dissociation of GDP and the activation of this GTPase. Hence it remains unknown whether the C-terminal domain contributes to the function of SifA in virulence. We used a model of SKIP knockout mice to show that this protein mediates the host susceptibility to salmonellosis and to establish that SifA also contributes to Salmonella virulence independently of its interaction with SKIP. We establish that the C-terminal domain of SifA mediates this SKIP-independent contribution. Moreover, we show that the two domains of SifA are functionally linked and participate to the same signalling cascade that supports Salmonella virulence.


Asunto(s)
Proteínas Bacterianas/genética , Glicoproteínas/genética , Monoéster Fosfórico Hidrolasas/genética , Salmonella/metabolismo , Animales , Proteínas Bacterianas/química , GTP Fosfohidrolasas/química , GTP Fosfohidrolasas/metabolismo , Regulación Bacteriana de la Expresión Génica , Glicoproteínas/química , Guanosina Difosfato/metabolismo , Células HeLa , Humanos , Ratones , Ratones Noqueados , Microtúbulos/química , Microtúbulos/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Pliegue de Proteína , Estructura Terciaria de Proteína , Salmonella/química , Salmonella/patogenicidad , Proteínas de Unión al GTP rho/química , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
18.
Methods Mol Biol ; 1225: 81-92, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25253249

RESUMEN

Epitope tags are short peptide sequences that are particularly useful for the characterization of proteins against which no antibody has been developed. Influenza hemagglutinin (HA) tag is one of the most widely used epitope tags as several valuable monoclonal and polyclonal antibodies that can be used in various techniques are commercially available. Therefore, adding a HA tag to a protein of interest is quite helpful to get rapid and cost less information regarding its localization, its expression or its biological function. In this chapter, we describe a process, derived from the Datsenko and Wanner procedure, which allows the introduction of an internal 2HA tag sequence into a chromosomal gene of the bacterial pathogen Salmonella.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Cromosomas Bacterianos/genética , Genes Bacterianos , Marcaje Isotópico/métodos , Salmonella/genética , Secuencia de Aminoácidos , Epítopos/química , Células HeLa , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Recombinación Homóloga , Humanos , Mutación , Fragmentos de Péptidos/química , Plásmidos/genética , Reacción en Cadena de la Polimerasa , Reproducibilidad de los Resultados , Transformación Genética
19.
PLoS One ; 8(12): e82508, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24367519

RESUMEN

CD4(+) T cells display a variety of helper functions necessary for an efficient adaptive immune response against bacterial invaders. This work reports the in vivo identification and characterization of murine cytotoxic CD4(+) T cells (CD4(+) CTL) during Brucella abortus infection. These CD4(+) CTLs express granzyme B and exhibit immunophenotypic features consistent with fully differentiated T cells. They express CD25, CD44, CD62L ,CD43 molecules at their surface and produce IFN-γ. Moreover, these cells express neither the co-stimulatory molecule CD27 nor the memory T cell marker CD127. We show here that CD4(+) CTLs are capable of cytolytic action against Brucella-infected antigen presenting cells (APC) but not against Mycobacterium-infected APC. Cytotoxic CD4(+) T cell population appears at early stages of the infection concomitantly with high levels of IFN-γ and granzyme B expression. CD4(+) CTLs represent a so far uncharacterized immune cell sub-type triggered by early immune responses upon Brucella abortus infection.


Asunto(s)
Brucella abortus/inmunología , Brucella abortus/patogenicidad , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T Citotóxicos/metabolismo , Animales , Brucelosis/inmunología , Brucelosis/metabolismo , Femenino , Citometría de Flujo , Receptores de Hialuranos/metabolismo , Interferón gamma/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Selectina L/metabolismo , Leucosialina/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Confocal
20.
Curr Opin Microbiol ; 16(1): 38-44, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23295139

RESUMEN

Bacteria of the genus Salmonella express nanosyringe-like organelles called type three secretion systems (T3SSs). These systems promote the secretion of bacterial compounds and their translocation into host cells. Pathogenic Salmonella use two distinct T3SSs, with specialized functions, having the purpose to modify the biology of the host organism and to ensure a successful infection. The bacterial proteins translocated through the first T3SS (T3SS-1) facilitate the entry of Salmonella into host cells, whereas T3SS-2 is an important factor for shaping the intracellular replication niche. In addition both T3SSs have a strong impact on the host inflammation. For a long time the two T3SSs were thought to act separately. However, there is increasing evidence that their regulation depends not only on separate but also shared regulatory mechanisms and that their time of action during infection overlaps. Here, we review the current understanding of the structure and of the regulation of expression and activity of both T3SSs. The output image is multifaceted, as recent studies show that subpopulations of Salmonella present diverging patterns of expression and activity of T3SSs during important steps of infection. These diversities may advance the chances of Salmonella to outpace competitors and to well establish itself in its niche in the host.


Asunto(s)
Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos , Salmonella/metabolismo , Salmonella/patogenicidad , Factores de Virulencia/metabolismo , Animales , Regulación Bacteriana de la Expresión Génica , Humanos , Transporte de Proteínas , Salmonella/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA