Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 631(8021): 635-644, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38961291

RESUMEN

Innate immune pattern recognition receptors, such as the Toll-like receptors (TLRs), are key mediators of the immune response to infection and central to our understanding of health and disease1. After microbial detection, these receptors activate inflammatory signal transduction pathways that involve IκB kinases, mitogen-activated protein kinases, ubiquitin ligases and other adaptor proteins. The mechanisms that connect the proteins in the TLR pathways are poorly defined. To delineate TLR pathway activities, we engineered macrophages to enable microscopy and proteomic analysis of the endogenous myddosome constituent MyD88. We found that myddosomes form transient contacts with activated TLRs and that TLR-free myddosomes are dynamic in size, number and composition over the course of 24 h. Analysis using super-resolution microscopy revealed that, within most myddosomes, MyD88 forms barrel-like structures that function as scaffolds for effector protein recruitment. Proteomic analysis demonstrated that myddosomes contain proteins that act at all stages and regulate all effector responses of the TLR pathways, and genetic analysis defined the epistatic relationship between these effector modules. Myddosome assembly was evident in cells infected with Listeria monocytogenes, but these bacteria evaded myddosome assembly and TLR signalling during cell-to-cell spread. On the basis of these findings, we propose that the entire TLR signalling pathway is executed from within the myddosome.


Asunto(s)
Macrófagos , Transducción de Señal , Receptores Toll-Like , Animales , Humanos , Ratones , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Listeriosis/microbiología , Listeriosis/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Factor 88 de Diferenciación Mieloide/metabolismo , Proteómica , Receptores Toll-Like/metabolismo , Microscopía , Inmunidad Innata
2.
J Immunol ; 206(2): 323-328, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33288542

RESUMEN

The NOX2 NADPH oxidase (NOX2) produces reactive oxygen species to kill phagosome-confined bacteria. However, we previously showed that Listeria monocytogenes is able to avoid the NOX2 activity in phagosomes and escape to the cytosol. Thus, despite the established role of NOX2 limiting L. monocytogenes infection in mice, the underlying mechanisms of this antibacterial activity remain unclear. In this article, we report that NOX2 controls systemic L. monocytogenes spread through modulation of the type I IFN response, which is known to be exploited by L. monocytogenes during infection. NOX2 deficiency results in increased expression of IFN-stimulated genes in response to type I IFN and leads to 1) promotion of cell-to-cell spread by L. monocytogenes, 2) defective leukocyte recruitment to infection foci, and 3) production of anti-inflammatory effectors IL-10 and thioredoxin 1. Our findings report a novel antimicrobial role for NOX2 through modulation of type I IFN responses to control bacterial dissemination.


Asunto(s)
Inflamación/inmunología , Interferón Tipo I/metabolismo , Leucocitos/inmunología , Listeria monocytogenes/fisiología , Listeriosis/inmunología , Macrófagos/metabolismo , NADPH Oxidasa 2/metabolismo , Animales , Movimiento Celular , Células Cultivadas , Interleucina-10/metabolismo , Listeriosis/transmisión , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 2/genética , Tiorredoxinas
3.
Cell ; 134(5): 743-56, 2008 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-18775308

RESUMEN

Inflammatory bowel disease (IBD) has been attributed to aberrant mucosal immunity to the intestinal microbiota. The transcription factor XBP1, a key component of the endoplasmic reticulum (ER) stress response, is required for development and maintenance of secretory cells and linked to JNK activation. We hypothesized that a stressful environmental milieu in a rapidly proliferating tissue might instigate a proinflammatory response. We report that Xbp1 deletion in intestinal epithelial cells (IECs) results in spontaneous enteritis and increased susceptibility to induced colitis secondary to both Paneth cell dysfunction and an epithelium that is overly reactive to inducers of IBD such as bacterial products (flagellin) and TNFalpha. An association of XBP1 variants with both forms of human IBD (Crohn's disease and ulcerative colitis) was identified and replicated (rs35873774; p value 1.6 x 10(-5)) with novel, private hypomorphic variants identified as susceptibility factors. Hence, intestinal inflammation can originate solely from XBP1 abnormalities in IECs, thus linking cell-specific ER stress to the induction of organ-specific inflammation.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Retículo Endoplásmico/inmunología , Enfermedades Inflamatorias del Intestino/genética , Factores de Transcripción/inmunología , Animales , Apoptosis , Colitis Ulcerosa/genética , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Predisposición Genética a la Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Listeria monocytogenes/inmunología , Ratones , Ratones Transgénicos , Células de Paneth/citología , Células de Paneth/metabolismo , Factores de Transcripción del Factor Regulador X , Proteína 1 de Unión a la X-Box
4.
J Strength Cond Res ; 37(8): 1634-1642, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-36723062

RESUMEN

ABSTRACT: Aben, HGJ, Hills, SP, Higgins, D, Cooke, CB, Davis, D, Jones, B, and Russell, M. The efficacy of a multimodal recovery strategy implemented after a high-intensity rugby league training session. J Strength Cond Res 37(8): 1634-1642, 2023-The efficacy of a multimodal recovery strategy implemented within 4 hours of rugby league (RL) training was investigated using repeated-measures, randomized, crossover methods in 10 professional academy RL players (age: 17 ± 1 years). Following standardized training (5,383 m covered, 350-m high-speed running, 28 repeated high-intensity efforts, 24 collisions), players completed a multimodal recovery (REC) strategy (i.e., ∼640 kcal meal + ∼1,285 kcal snacks or drinks, cold-water immersion, sleep hygiene recommendations) or control (i.e., ∼640 kcal meal: CONT) practices. Isometric mid-thigh pulls (IMTP), countermovement jumps (CMJ), and wellness questionnaires were completed before (-3 hours) and after (+24, +48 hours) training. The recovery strategy influenced IMTP peak force ( p = 0.026), but between-trial differences were undetectable. No other between-trial effects (all p > 0.05) were seen for IMTP, CMJ, or wellness variables. Training-induced reductions in CMJ peak power (-4 ± 6% vs baseline: 4,878 ± 642 W) at +24 hours ( p = 0.016) dissipated by +48 hours. Fatigue and lower-body soreness reduced by 16 ± 19% ( p = 0.01) and 32 ± 44% ( p = 0.024) at +48 hours versus +24 hours, respectively. Relative to CONT (i.e., posttraining nutrition), the effects of a single bout of recovery practices appeared limited when implemented after RL-specific training. Therefore, when training included limited collisions, balanced postexercise meals appeared equally effective relative to a multimodal recovery strategy. Transient changes in performance and wellness variables after training may have implications for practitioners. Consecutive training sessions, including a high frequency and intensity of eccentric muscle actions, should be carefully planned, especially near match-play.


Asunto(s)
Rendimiento Atlético , Fútbol Americano , Carrera , Adolescente , Humanos , Rendimiento Atlético/fisiología , Fatiga , Fútbol Americano/fisiología , Rugby
5.
Immunity ; 37(5): 930-46, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23123061

RESUMEN

Carcinoembryonic antigen cell adhesion molecule like I (CEACAM1) is expressed on activated T cells and signals through either a long (L) cytoplasmic tail containing immune receptor tyrosine based inhibitory motifs, which provide inhibitory function, or a short (S) cytoplasmic tail with an unknown role. Previous studies on peripheral T cells show that CEACAM1-L isoforms predominate with little to no detectable CEACAM1-S isoforms in mouse and human. We show here that this was not the case in tissue resident T cells of intestines and gut associated lymphoid tissues, which demonstrated predominant expression of CEACAM1-S isoforms relative to CEACAM1-L isoforms in human and mouse. This tissue resident predominance of CEACAM1-S expression was determined by the intestinal environment where it served a stimulatory function leading to the regulation of T cell subsets associated with the generation of secretory IgA immunity, the regulation of mucosal commensalism, and defense of the barrier against enteropathogens.


Asunto(s)
Antígeno Carcinoembrionario/inmunología , Inmunidad Mucosa/inmunología , Intestinos/inmunología , Linfocitos T/inmunología , Secuencias de Aminoácidos/genética , Secuencias de Aminoácidos/inmunología , Animales , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Citoplasma/genética , Citoplasma/inmunología , Citoplasma/metabolismo , Homeostasis , Inmunidad Mucosa/genética , Inmunoglobulina A/genética , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Mucosa Intestinal/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Activación de Linfocitos , Metagenoma/inmunología , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Isoformas de Proteínas , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Linfocitos T/metabolismo , Tirosina/genética , Tirosina/inmunología , Tirosina/metabolismo
6.
Nature ; 509(7499): 230-4, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24739967

RESUMEN

Efferocytosis, the process by which dying or dead cells are removed by phagocytosis, has an important role in development, tissue homeostasis and innate immunity. Efferocytosis is mediated, in part, by receptors that bind to exofacial phosphatidylserine (PS) on cells or cellular debris after loss of plasma membrane asymmetry. Here we show that a bacterial pathogen, Listeria monocytogenes, can exploit efferocytosis to promote cell-to-cell spread during infection. These bacteria can escape the phagosome in host cells by using the pore-forming toxin listeriolysin O (LLO) and two phospholipase C enzymes. Expression of the cell surface protein ActA allows L. monocytogenes to activate host actin regulatory factors and undergo actin-based motility in the cytosol, eventually leading to formation of actin-rich protrusions at the cell surface. Here we show that protrusion formation is associated with plasma membrane damage due to LLO's pore-forming activity. LLO also promotes the release of bacteria-containing protrusions from the host cell, generating membrane-derived vesicles with exofacial PS. The PS-binding receptor TIM-4 (encoded by the Timd4 gene) contributes to efficient cell-to-cell spread by L. monocytogenes in macrophages in vitro and growth of these bacteria is impaired in Timd4(-/-) mice. Thus, L. monocytogenes promotes its dissemination in a host by exploiting efferocytosis. Our results indicate that PS-targeted therapeutics may be useful in the fight against infections by L. monocytogenes and other bacteria that use similar strategies of cell-to-cell spread during infection.


Asunto(s)
Extensiones de la Superficie Celular/microbiología , Listeria monocytogenes/fisiología , Fagocitosis , Actinas/metabolismo , Animales , Toxinas Bacterianas/metabolismo , Membrana Celular/metabolismo , Membrana Celular/microbiología , Membrana Celular/patología , Extensiones de la Superficie Celular/metabolismo , Citoplasma/metabolismo , Citoplasma/microbiología , Femenino , Células HeLa , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Humanos , Listeria monocytogenes/patogenicidad , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/microbiología , Proteínas de la Membrana/metabolismo , Ratones , Fagosomas/metabolismo , Fagosomas/microbiología , Fosfatidilserinas/metabolismo , Fosfolipasas de Tipo C/metabolismo , Vacuolas/metabolismo , Vacuolas/microbiología
7.
Proc Natl Acad Sci U S A ; 114(24): 6334-6339, 2017 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28559314

RESUMEN

Listeria monocytogenes is a common food-borne pathogen that can disseminate from the intestine and infect multiple organs. Here, we used sequence tag-based analysis of microbial populations (STAMP) to investigate Lmonocytogenes population dynamics during infection. We created a genetically barcoded library of murinized Lmonocytogenes and then used deep sequencing to track the pathogen's dissemination routes and quantify its founding population (Nb) sizes in different organs. We found that the pathogen disseminates from the gastrointestinal tract to distal sites through multiple independent routes and that Nb sizes vary greatly among tissues, indicative of diverse host barriers to infection. Unexpectedly, comparative analyses of sequence tags revealed that fecally excreted organisms are largely derived from the very small number of L. monocytogenes cells that colonize the gallbladder. Immune depletion studies suggest that distinct innate immune cells restrict the pathogen's capacity to establish replicative niches in the spleen and liver. Finally, studies in germ-free mice suggest that the microbiota plays a critical role in the development of the splenic, but not the hepatic, barriers that prevent L. monocytogenes from seeding these organs. Collectively, these observations illustrate the potency of the STAMP approach to decipher the impact of host factors on population dynamics of pathogens during infection.


Asunto(s)
Listeria monocytogenes/patogenicidad , Listeriosis/inmunología , Animales , Carga Bacteriana , Código de Barras del ADN Taxonómico , Femenino , Vesícula Biliar/inmunología , Vesícula Biliar/microbiología , Microbioma Gastrointestinal , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Vida Libre de Gérmenes , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Listeria monocytogenes/genética , Listeria monocytogenes/inmunología , Listeriosis/microbiología , Hígado/inmunología , Hígado/microbiología , Ratones , Ratones Endogámicos BALB C , Bazo/inmunología , Bazo/microbiología
8.
J Immunol ; 198(7): 2772-2784, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28258198

RESUMEN

Vaccine strategies to enhance CD8+ CTL responses remain a current challenge because they should overcome the plasmatic and endosomal membranes for favoring exogenous Ag access to the cytosol of APCs. As a way to avoid this hurdle, sticholysin (St) II, a pore-forming protein from the Caribbean Sea anemone Stichodactyla helianthus, was encapsulated with OVA into liposomes (Lp/OVA/StII) to assess their efficacy to induce a CTL response. OVA-specific CD8+ T cells transferred to mice immunized with Lp/OVA/StII experienced a greater expansion than when the recipients were injected with the vesicles without St, mostly exhibiting a memory phenotype. Consequently, Lp/OVA/StII induced a more potent effector function, as shown by CTLs, in vivo assays. Furthermore, treatment of E.G7-OVA tumor-bearing mice with Lp/OVA/StII significantly reduced tumor growth being more noticeable in the preventive assay. The contribution of CD4+ and CD8+ T cells to CTL and antitumor activity, respectively, was elucidated. Interestingly, the irreversibly inactive variant of the StI mutant StI W111C, encapsulated with OVA into Lp, elicited a similar OVA-specific CTL response to that observed with Lp/OVA/StII or vesicles encapsulating recombinant StI or the reversibly inactive StI W111C dimer. These findings suggest the relative independence between StII pore-forming activity and its immunomodulatory properties. In addition, StII-induced in vitro maturation of dendritic cells might be supporting these properties. These results are the first evidence, to our knowledge, that StII, a pore-forming protein from a marine eukaryotic organism, encapsulated into Lp functions as an adjuvant to induce a robust specific CTL response.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra el Cáncer/inmunología , Venenos de Cnidarios/administración & dosificación , Neoplasias Experimentales/patología , Linfocitos T Citotóxicos/efectos de los fármacos , Animales , Venenos de Cnidarios/inmunología , Femenino , Citometría de Flujo , Liposomas/inmunología , Ratones , Ratones Endogámicos C57BL , Linfocitos T Citotóxicos/inmunología
9.
Infect Immun ; 86(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29263107

RESUMEN

Listeria monocytogenes is a Gram-positive intracellular pathogen that causes a severe invasive disease. Upon infecting a host cell, L. monocytogenes upregulates the transcription of numerous factors necessary for productive infection. VirR is the response regulator component of a two-component regulatory system in L. monocytogenes In this report, we have identified the putative ABC transporter encoded by genes lmo1746-lmo1747 as necessary for VirR function. We have designated lmo1746-lmo1747 virAB We constructed an in-frame deletion of virAB and determined that the ΔvirAB mutant exhibited reduced transcription of VirR-regulated genes. The ΔvirAB mutant also showed defects in in vitro plaque formation and in vivo virulence that were similar to those of a ΔvirR deletion mutant. Since VirR is important for innate resistance to antimicrobial agents, we determined the MICs of nisin and bacitracin for ΔvirAB bacteria. We found that VirAB expression was necessary for nisin resistance but was dispensable for resistance to bacitracin. This result suggested a VirAB-independent mechanism of VirR regulation in response to bacitracin. Lastly, we found that the ΔvirR and ΔvirAB mutants had no deficiency in growth in broth culture, intracellular replication, or production of the ActA surface protein, which facilitates actin-based motility and cell-to-cell spread. However, the ΔvirR and ΔvirAB mutants produced shorter actin tails during intracellular infection, which suggested that these mutants have a reduced ability to move and spread via actin-based motility. These findings have demonstrated that L. monocytogenes VirAB functions in a pathway with VirR to regulate the expression of genes necessary for virulence and resistance to antimicrobial agents.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Nisina/farmacología , Factores de Transcripción/genética , Transportadoras de Casetes de Unión a ATP/genética , Animales , Proteínas Bacterianas/genética , Farmacorresistencia Bacteriana , Femenino , Regulación Bacteriana de la Expresión Génica , Humanos , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/genética , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Regulón , Factores de Transcripción/metabolismo , Virulencia
10.
Cell Microbiol ; 19(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27582004

RESUMEN

Type I interferons (IFNs) play a critical role in antiviral immune responses, but can be deleterious to the host during some bacterial infections. Listeria monocytogenes (Lm) induces a type I IFN response by activating cytosolic antiviral surveillance pathways. This is beneficial to the bacteria as mice lacking the type I IFN receptor (IFNAR1-/- ) are resistant to systemic infection by Lm. The mechanisms by which type I IFNs promote Lm infection are unclear. Here, we show that IFNAR1 is required for dissemination of Lm within infection foci in livers of infected mice and for efficient cell-to-cell spread in vitro in macrophages. IFNAR1 promotes ActA polarization and actin-based motility in the cytosol of host cells. Our studies suggest type I IFNs directly impact the intracellular life cycle of Lm and provide new insight into the mechanisms used by bacterial pathogens to exploit the type I IFN response.


Asunto(s)
Interacciones Huésped-Patógeno , Interferón Tipo I/metabolismo , Listeria monocytogenes/crecimiento & desarrollo , Animales , Modelos Animales de Enfermedad , Listeriosis/microbiología , Listeriosis/patología , Hígado/microbiología , Hígado/patología , Macrófagos/microbiología , Ratones , Receptor de Interferón alfa y beta/metabolismo
11.
J Infect Dis ; 211(7): 1185-95, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25281757

RESUMEN

The Gram-positive bacterium Listeria monocytogenes is a facultative intracellular pathogen whose virulence depends on its ability to spread from cell to cell within an infected host. Although the actin-related protein 2/3 (Arp2/3) complex is necessary and sufficient for Listeria actin tail assembly, previous studies suggest that other actin polymerization factors, such as formins, may participate in protrusion formation. Here, we show that Arp2/3 localized to only a minor portion of the protrusion. Moreover, treatment of L. monocytogenes-infected HeLa cells with a formin FH2-domain inhibitor significantly reduced protrusion length. In addition, the Diaphanous-related formins 1-3 (mDia1-3) localized to protrusions, and knockdown of mDia1, mDia2, and mDia3 substantially decreased cell-to-cell spread of L. monocytogenes. Rho GTPases are known to be involved in formin activation. Our studies also show that knockdown of several Rho family members significantly influenced bacterial cell-to-cell spread. Collectively, these findings identify a Rho GTPase-formin network that is critically involved in the cell-to-cell spread of L. monocytogenes.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Extensiones de la Superficie Celular/metabolismo , Listeria monocytogenes/fisiología , Proteínas de Unión al GTP rho/metabolismo , Proteína 2 Relacionada con la Actina/genética , Proteína 2 Relacionada con la Actina/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/genética , Proteína 3 Relacionada con la Actina/genética , Proteína 3 Relacionada con la Actina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/genética , Extensiones de la Superficie Celular/efectos de los fármacos , Extensiones de la Superficie Celular/ultraestructura , Forminas , Técnicas de Silenciamiento del Gen , Genes Reporteros , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Listeria monocytogenes/patogenicidad , Modelos Biológicos , Estructura Terciaria de Proteína , Tionas/farmacología , Uracilo/análogos & derivados , Uracilo/farmacología , Proteínas de Unión al GTP rho/genética
12.
Nature ; 451(7176): 350-4, 2008 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-18202661

RESUMEN

Listeria monocytogenes is an intracellular bacterial pathogen that replicates rapidly in the cytosol of host cells during acute infection. Surprisingly, these bacteria were found to occupy vacuoles in liver granuloma macrophages during persistent infection of severe combined immunodeficient (SCID) mice. Here we show that L. monocytogenes can replicate in vacuoles within macrophages. In livers of SCID mice infected for 21 days, we observed bacteria in large LAMP1(+) compartments that we termed spacious Listeria-containing phagosomes (SLAPs). SLAPs were also observed in vitro, and were found to be non-acidic and non-degradative compartments that are generated in an autophagy-dependent manner. The replication rate of bacteria in SLAPs was found to be reduced compared to the rate of those in the cytosol. Listeriolysin O (LLO, encoded by hly), a pore-forming toxin essential for L. monocytogenes virulence, was necessary and sufficient for SLAP formation. A L. monocytogenes mutant with low LLO expression was impaired for phagosome escape but replicated slowly in SLAPs over a 72 h period. Therefore, our studies reveal a role for LLO in promoting L. monocytogenes replication in vacuoles and suggest a mechanism by which this pathogen can establish persistent infection in host macrophages.


Asunto(s)
Toxinas Bacterianas/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/patogenicidad , Macrófagos/citología , Macrófagos/microbiología , Vacuolas/microbiología , Animales , Autofagia , Toxinas Bacterianas/genética , Enfermedad Crónica , Proteínas de Choque Térmico/genética , Proteínas Hemolisinas/genética , Listeria monocytogenes/metabolismo , Listeriosis/microbiología , Listeriosis/patología , Hígado/microbiología , Proteínas de Membrana de los Lisosomas/metabolismo , Ratones , Ratones SCID , Fagosomas/metabolismo , Fagosomas/microbiología , Vacuolas/metabolismo , Virulencia
13.
J Bacteriol ; 195(15): 3331-40, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23687268

RESUMEN

Listeria monocytogenes is a Gram-positive, facultative intracellular pathogen capable of causing severe invasive disease with high mortality rates in humans. While previous studies have largely elucidated the bacterial and host cell mechanisms necessary for invasion, vacuolar escape, and subsequent cell-to-cell spread, the L. monocytogenes factors required for rapid replication within the restrictive environment of the host cell cytosol are poorly understood. In this report, we describe a differential fluorescence-based genetic screen utilizing fluorescence-activated cell sorting (FACS) and high-throughput microscopy to identify L. monocytogenes mutants defective in optimal intracellular replication. Bacteria harboring deletions within the identified gene menD or pepP were defective for growth in primary murine macrophages and plaque formation in monolayers of L2 fibroblasts, thus validating the ability of the screening method to identify intracellular replication-defective mutants. Genetic complementation of the menD and pepP deletion strains rescued the in vitro intracellular infection defects. Furthermore, the menD deletion strain displayed a general extracellular replication defect that could be complemented by growth under anaerobic conditions, while the intracellular growth defect of this strain could be complemented by the addition of exogenous menaquinone. As prior studies have indicated the importance of aerobic metabolism for L. monocytogenes infection, these findings provide further evidence for the importance of menaquinone and aerobic metabolism for L. monocytogenes pathogenesis. Lastly, both the menD and pepP deletion strains were attenuated during in vivo infection of mice. These findings demonstrate that the differential fluorescence-based screening approach provides a powerful tool for the identification of intracellular replication determinants in multiple bacterial systems.


Asunto(s)
Citosol/microbiología , Genética Microbiana/métodos , Listeria monocytogenes/patogenicidad , Biología Molecular/métodos , Factores de Virulencia/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/microbiología , Citometría de Flujo , Colorantes Fluorescentes/análisis , Eliminación de Gen , Prueba de Complementación Genética , Pruebas Genéticas , Ensayos Analíticos de Alto Rendimiento , Listeria monocytogenes/genética , Listeria monocytogenes/crecimiento & desarrollo , Listeriosis/microbiología , Listeriosis/patología , Macrófagos/microbiología , Ratones , Microscopía Fluorescente , Coloración y Etiquetado/métodos , Virulencia
14.
PLoS Pathog ; 7(8): e1002153, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21829361

RESUMEN

Facultative bacterial pathogens must adapt to multiple stimuli to persist in the environment or establish infection within a host. Temperature is often utilized as a signal to control expression of virulence genes necessary for infection or genes required for persistence in the environment. However, very little is known about the molecular mechanisms that allow bacteria to adapt and respond to temperature fluctuations. Listeria monocytogenes (Lm) is a food-borne, facultative intracellular pathogen that uses flagellar motility to survive in the extracellular environment and to enhance initial invasion of host cells during infection. Upon entering the host, Lm represses transcription of flagellar motility genes in response to mammalian physiological temperature (37°C) with a concomitant temperature-dependent up-regulation of virulence genes. We previously determined that down-regulation of flagellar motility is required for virulence and is governed by the reciprocal activities of the MogR transcriptional repressor and the bifunctional flagellar anti-repressor/glycosyltransferase, GmaR. In this study, we determined that GmaR is also a protein thermometer that controls temperature-dependent transcription of flagellar motility genes. Two-hybrid and gel mobility shift analyses indicated that the interaction between MogR and GmaR is temperature sensitive. Using circular dichroism and limited proteolysis, we determined that GmaR undergoes a temperature-dependent conformational change as temperature is elevated. Quantitative analysis of GmaR in Lm revealed that GmaR is degraded in the absence of MogR and at 37°C (when the MogR:GmaR complex is less stable). Since MogR represses transcription of all flagellar motility genes, including transcription of gmaR, changes in the stability of the MogR:GmaR anti-repression complex, due to conformational changes in GmaR, mediates repression or de-repression of flagellar motility genes in Lm. Thus, GmaR functions as a thermo-sensing anti-repressor that incorporates temperature signals into transcriptional control of flagellar motility. To our knowledge, this is the first example of a protein thermometer that functions as an anti-repressor to control a developmental process in bacteria.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Flagelos/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Listeria monocytogenes/metabolismo , Transducción de Señal/fisiología , Factores de Virulencia/biosíntesis , Proteínas Bacterianas/genética , Flagelos/genética , Calor , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Factores de Virulencia/genética
15.
Nature ; 434(7034): 767-72, 2005 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-15815631

RESUMEN

An estimated eight million people are infected each year with the pathogen Mycobacterium tuberculosis, and more than two million die annually. Yet only about 10% of those infected develop tuberculosis. Genetic variation within host populations is known to be significant in humans and animals, but the nature of genetic control of host resistance to tuberculosis remains poorly understood. Previously we mapped a new genetic locus on mouse chromosome 1, designated sst1 (for supersusceptibility to tuberculosis 1). Here we show that this locus mediates innate immunity in sst1 congenic mouse strains and identify a candidate gene, Intracellular pathogen resistance 1 (Ipr1), within the sst1 locus. The Ipr1 gene is upregulated in the sst1 resistant macrophages after activation and infection, but it is not expressed in the sst1 susceptible macrophages. Expression of the Ipr1 transgene in the sst1 susceptible macrophages limits the multiplication not only of M. tuberculosis but also of Listeria monocytogenes and switches a cell death pathway of the infected macrophages from necrosis to apoptosis. Our data indicate that the Ipr1 gene product might have a previously undocumented function in integrating signals generated by intracellular pathogens with mechanisms controlling innate immunity, cell death and pathogenesis.


Asunto(s)
Inmunidad Innata/genética , Inmunidad Innata/inmunología , Macrófagos/inmunología , Transactivadores/genética , Transactivadores/metabolismo , Tuberculosis/genética , Tuberculosis/inmunología , Animales , Animales Congénicos , Apoptosis , Trasplante de Médula Ósea , Progresión de la Enfermedad , Predisposición Genética a la Enfermedad/genética , Humanos , Interferón gamma/inmunología , Listeria monocytogenes/inmunología , Pulmón/microbiología , Pulmón/patología , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor , Datos de Secuencia Molecular , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Necrosis , Proteínas Nucleares/química , Proteínas Nucleares/genética , Polimorfismo Genético/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tasa de Supervivencia , Transactivadores/química , Transgenes/genética , Tuberculosis/microbiología , Tuberculosis/patología
16.
Structure ; 17(5): 769-77, 2009 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-19446532

RESUMEN

The MogR transcriptional repressor of the intracellular pathogen Listeria monocytogenes recognizes AT-rich binding sites in promoters of flagellar genes to downregulate flagellar gene expression during infection. We describe here the 1.8 A resolution crystal structure of MogR bound to the recognition sequence 5' ATTTTTTAAAAAAAT 3' present within the flaA promoter region. Our structure shows that MogR binds as a dimer. Each half-site is recognized in the major groove by a helix-turn-helix motif and in the minor groove by a loop from the symmetry-related molecule, resulting in a "crossover" binding mode. This oversampling through minor groove interactions is important for specificity. The MogR binding site has structural features of A-tract DNA and is bent by approximately 52 degrees away from the dimer. The structure explains how MogR achieves binding specificity in the AT-rich genome of L. monocytogenes and explains the evolutionary conservation of A-tract sequence elements within promoter regions of MogR-regulated flagellar genes.


Asunto(s)
Secuencia Rica en At , Proteínas Bacterianas/química , Proteínas Represoras/química , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Sitios de Unión , ADN/metabolismo , ADN Bacteriano/química , ADN Bacteriano/metabolismo , Flagelina/química , Flagelina/genética , Flagelina/metabolismo , Secuencias Hélice-Giro-Hélice , Listeria monocytogenes/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , Conformación Proteica , Proteínas Represoras/metabolismo
17.
Mol Immunol ; 131: 144-154, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33422341

RESUMEN

Sticholysins (Sts) I and II (StI and StII) are pore-forming proteins (PFPs), purified from the Caribbean Sea anemone Stichodactyla helianthus. StII encapsulated into liposomes induces a robust antigen-specific cytotoxic CD8+ T lymphocytes (CTL) response and in its free form the maturation of bone marrow-derived dendritic cells (BM-DCs). It is probable that the latter is partially supporting in part the immunomodulatory effect on the CTL response induced by StII-containing liposomes. In the present work, we demonstrate that the StII's ability of inducing maturation of BM-DCs is also shared by StI, an isoform of StII. Using heat-denatured Sts we observed a significant reduction in the up-regulation of maturation markers indicating that both PFP's ability to promote maturation of BM-DCs is dependent on their conformational characteristics. StII-mediated DC maturation was abrogated in BM-DCs from toll-like receptor (TLR) 4 and myeloid differentiation primary response gene 88 (MyD88)-knockout mice but not in cells from TLR2-knockout mice. Furthermore, the antigen-specific CTL response induced by StII-containing liposomes was reduced in TLR4-knockout mice. These results indicate that StII, and probably by extension StI, has the ability to induce maturation of DCs through a TLR4/MyD88-dependent pathway, and that this activation contributes to the CTL response generated by StII-containing liposomes.


Asunto(s)
Venenos de Cnidarios/metabolismo , Células Dendríticas/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Compuestos Orgánicos/metabolismo , Transducción de Señal/fisiología
18.
Nat Commun ; 12(1): 4999, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34404769

RESUMEN

The type I interferon (IFN) signaling pathway has important functions in resistance to viral infection, with the downstream induction of interferon stimulated genes (ISG) protecting the host from virus entry, replication and spread. Listeria monocytogenes (Lm), a facultative intracellular foodborne pathogen, can exploit the type I IFN response as part of their pathogenic strategy, but the molecular mechanisms involved remain unclear. Here we show that type I IFN suppresses the antibacterial activity of phagocytes to promote systemic Lm infection. Mechanistically, type I IFN suppresses phagosome maturation and proteolysis of Lm virulence factors ActA and LLO, thereby promoting phagosome escape and cell-to-cell spread; the antiviral protein, IFN-induced transmembrane protein 3 (IFITM3), is required for this type I IFN-mediated alteration. Ifitm3-/- mice are resistant to systemic infection by Lm, displaying decreased bacterial spread in tissues, and increased immune cell recruitment and pro-inflammatory cytokine signaling. Together, our findings show how an antiviral mechanism in phagocytes can be exploited by bacterial pathogens, and implicate IFITM3 as a potential antimicrobial therapeutic target.


Asunto(s)
Antibacterianos/farmacología , Listeria/efectos de los fármacos , Listeriosis/inmunología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Fagocitos/inmunología , Fagocitos/microbiología , Animales , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Interferón Tipo I/metabolismo , Listeria monocytogenes/inmunología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagosomas/inmunología , Células RAW 264.7 , Transcriptoma , Factores de Virulencia , Internalización del Virus/efectos de los fármacos
19.
Mol Microbiol ; 74(2): 421-35, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19796338

RESUMEN

Flagellar motility in Listeria monocytogenes (Lm) is restricted to temperatures below 37 degrees C due to the opposing activities of the MogR transcriptional repressor and the GmaR antirepressor. Previous studies have suggested that both the DegU response regulator and MogR regulate expression of GmaR. In this report, we further define the role of DegU for GmaR production and flagellar motility. We demonstrate that deletion of the receiver domain of DegU has no effect on flagellar motility in Lm. Using transcriptional reporter fusions, we determined that gmaR is cotranscribed within an operon initiating with fliN. Furthermore, the fliN-gmaR promoter (p(fliN-gmaR)) is transcriptionally activated by DegU and is also MogR-repressed. DNA affinity purification, gel mobility shift and footprinting analyses revealed that both DegU and MogR directly bind fliN-gmaR promoter region DNA and that the binding sites do not overlap. Quantitative analysis of gmaR transcripts in Delta mogR bacteria indicated that transcriptional activation of p(fliN-gmaR) by DegU is not inherently temperature-dependent. However, GmaR protein was not detectable at 37 degrees C in Delta mogR bacteria, indicating that a temperature-dependent, post-transcriptional mechanism limits GmaR production to temperatures below 37 degrees C. Our findings reveal that flagellar motility in Lm is governed by both temperature-dependent transcriptional and post-transcriptional regulation of the GmaR antirepressor.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/metabolismo , Flagelos/fisiología , Listeria monocytogenes/genética , Temperatura , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Sitios de Unión , Huella de ADN , ADN Bacteriano/genética , Proteínas de Unión al ADN/genética , Flagelos/genética , Flagelos/metabolismo , Regulación Bacteriana de la Expresión Génica , Listeria monocytogenes/metabolismo , Listeria monocytogenes/fisiología , Datos de Secuencia Molecular , Operón , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Procesamiento Proteico-Postraduccional , Alineación de Secuencia , Activación Transcripcional
20.
Cell Microbiol ; 11(9): 1382-98, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19500109

RESUMEN

Listeria monocytogenes is a bacterial pathogen that replicates within the cytosol of infected host cells. The ability to rapidly escape the phagocytic vacuole is essential for efficient intracellular replication. In the murine model of infection, the pore-forming cytolysin listeriolysin O (LLO) is absolutely required for vacuolar dissolution, as LLO-deficient (DeltaLLO) mutants remain trapped within vacuoles. In contrast, in many human cell types DeltaLLO L. monocytogenes are capable of vacuolar escape at moderate to high frequencies. To better characterize the mechanism of LLO-independent vacuolar escape in human cells, we conducted an RNA interference screen to identify vesicular trafficking factors that play a role in altering vacuolar escape efficiency of DeltaLLO L. monocytogenes. RNA interference knockdown of 18 vesicular trafficking factors resulted in increased LLO-independent vacuolar escape. Our results suggest that knockdown of one factor, RABEP1 (rabaptin-5), decreased the maturation of vacuoles containing DeltaLLO L. monocytogenes. Thus, we provide evidence that increased vacuolar escape of DeltaLLO L. monocytogenes in human cells correlates with slower vacuolar maturation. We also determined that increased LLO-independent dissolution of vacuoles during RABEP1 knockdown required the bacterial broad-range phospholipase C (PC-PLC). We hypothesize that slowing the kinetics of vacuolar maturation generates an environment conducive for vacuolar escape mediated by the bacterial phospholipases.


Asunto(s)
Toxinas Bacterianas/toxicidad , Proteínas de Choque Térmico/toxicidad , Proteínas Hemolisinas/toxicidad , Listeria monocytogenes/inmunología , Listeria monocytogenes/patogenicidad , Vacuolas/microbiología , Vacuolas/fisiología , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Interferencia de ARN , Proteínas de Transporte Vesicular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA