Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 183(3): 650-665.e15, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33031742

RESUMEN

Endocannabinoids are host-derived lipid hormones that fundamentally impact gastrointestinal (GI) biology. The use of cannabis and other exocannabinoids as anecdotal treatments for various GI disorders inspired the search for mechanisms by which these compounds mediate their effects, which led to the discovery of the mammalian endocannabinoid system. Dysregulated endocannabinoid signaling was linked to inflammation and the gut microbiota. However, the effects of endocannabinoids on host susceptibility to infection has not been explored. Here, we show that mice with elevated levels of the endocannabinoid 2-arachidonoyl glycerol (2-AG) are protected from enteric infection by Enterobacteriaceae pathogens. 2-AG directly modulates pathogen function by inhibiting virulence programs essential for successful infection. Furthermore, 2-AG antagonizes the bacterial receptor QseC, a histidine kinase encoded within the core Enterobacteriaceae genome that promotes the activation of pathogen-associated type three secretion systems. Taken together, our findings establish that endocannabinoids are directly sensed by bacteria and can modulate bacterial function.


Asunto(s)
Endocannabinoides/metabolismo , Enterobacteriaceae/patogenicidad , Animales , Ácidos Araquidónicos/química , Ácidos Araquidónicos/metabolismo , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Bacterianos/metabolismo , Citrobacter rodentium/patogenicidad , Colon/microbiología , Colon/patología , Endocannabinoides/química , Infecciones por Enterobacteriaceae/microbiología , Femenino , Microbioma Gastrointestinal , Glicéridos/química , Glicéridos/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Monoacilglicerol Lipasas/metabolismo , Salmonella/patogenicidad , Virulencia
2.
Nature ; 553(7687): 208-211, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29323293

RESUMEN

Inflammatory diseases of the gastrointestinal tract are frequently associated with dysbiosis, characterized by changes in gut microbial communities that include an expansion of facultative anaerobic bacteria of the Enterobacteriaceae family (phylum Proteobacteria). Here we show that a dysbiotic expansion of Enterobacteriaceae during gut inflammation could be prevented by tungstate treatment, which selectively inhibited molybdenum-cofactor-dependent microbial respiratory pathways that are operational only during episodes of inflammation. By contrast, we found that tungstate treatment caused minimal changes in the microbiota composition under homeostatic conditions. Notably, tungstate-mediated microbiota editing reduced the severity of intestinal inflammation in mouse models of colitis. We conclude that precision editing of the microbiota composition by tungstate treatment ameliorates the adverse effects of dysbiosis in the inflamed gut.


Asunto(s)
Colitis/tratamiento farmacológico , Colitis/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Intestinos/microbiología , Anaerobiosis/efectos de los fármacos , Animales , Respiración de la Célula/efectos de los fármacos , Disbiosis/tratamiento farmacológico , Disbiosis/microbiología , Enterobacteriaceae/efectos de los fármacos , Enterobacteriaceae/crecimiento & desarrollo , Enterobacteriaceae/metabolismo , Femenino , Inflamación/tratamiento farmacológico , Inflamación/microbiología , Inflamación/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Intestinos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Molibdeno/metabolismo , Compuestos de Tungsteno/farmacología , Compuestos de Tungsteno/uso terapéutico
3.
Proc Natl Acad Sci U S A ; 117(22): 12387-12393, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32409599

RESUMEN

Microbiota, host and dietary metabolites/signals compose the rich gut chemical environment, which profoundly impacts virulence of enteric pathogens. Enterohemorrhagic Escherichia coli (EHEC) engages a syringe-like machinery named type-III secretion system (T3SS) to inject effectors within host cells that lead to intestinal colonization and disease. We previously conducted a high-throughput screen to identify metabolic pathways that affect T3SS expression. Here we show that in the presence of arginine, the arginine sensor ArgR, identified through this screen, directly activates expression of the genes encoding the T3SS. Exogenously added arginine induces EHEC virulence gene expression in vitro. Congruently, a mutant deficient in arginine transport (ΔartP) had decreased virulence gene expression. ArgR also augments murine disease caused by Citrobacter rodentium, which is a murine pathogen extensively employed as a surrogate animal model for EHEC. The source of arginine sensed by C. rodentium is not dietary. At the peak of C. rodentium infection, increased arginine concentration in the colon correlated with down-regulation of the host SLC7A2 transporter. This increase in the concentration of colonic arginine promotes virulence gene expression in C. rodentium Arginine is an important modulator of the host immune response to pathogens. Here we add that arginine also directly impacts bacterial virulence. These findings suggest that a delicate balance between host and pathogen responses to arginine occur during disease progression.


Asunto(s)
Citrobacter rodentium/metabolismo , Infecciones por Enterobacteriaceae/microbiología , Escherichia coli Enterohemorrágica/metabolismo , Infecciones por Escherichia coli/microbiología , Regulación Bacteriana de la Expresión Génica , Animales , Arginina/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Citrobacter rodentium/genética , Citrobacter rodentium/patogenicidad , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/patogenicidad , Humanos , Ratones , Ratones Endogámicos C3H , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
4.
Sensors (Basel) ; 23(16)2023 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-37631733

RESUMEN

Microneedle puncture is a standard minimally invasive treatment and surgical method, which is widely used in extracting blood, tissues, and their secretions for pathological examination, needle-puncture-directed drug therapy, local anaesthesia, microwave ablation needle therapy, radiotherapy, and other procedures. The use of robots for microneedle puncture has become a worldwide research hotspot, and medical imaging navigation technology plays an essential role in preoperative robotic puncture path planning, intraoperative assisted puncture, and surgical efficacy detection. This paper introduces medical imaging technology and minimally invasive puncture robots, reviews the current status of research on the application of medical imaging navigation technology in minimally invasive puncture robots, and points out its future development trends and challenges.


Asunto(s)
Ablación por Radiofrecuencia , Robótica , Punciones , Agujas , Tecnología
5.
Sensors (Basel) ; 22(11)2022 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-35684751

RESUMEN

Laser welding is an indispensable link in most types of industrial production. The realization of welding automation by industrial robots can greatly improve production efficiency. In the research and development of the welding seam tracking system, information on the position of the weld joint needs to be obtained accurately. For laser welding images with strong and complex interference, a weld tracking module was designed to capture real-time images of the weld, and a total of 737, 1920 × 1200 pixel weld images were captured using the device, of which 637 were used to create the dataset, and the other 100 were used as images to test the segmentation success rate. Based on the pixel-level segmentation capability of the semantic segmentation network, this study used an encoder-decoder architecture to design a lightweight network structure and introduced a channel attention mechanism. Compared to ERF-Net, SegNet, and DFA-Net, the network model in this paper has a fast segmentation speed and higher segmentation accuracy, with a success rate of 96% and remarkable segmentation results.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Soldadura , Automatización , Procesamiento de Imagen Asistido por Computador/métodos , Rayos Láser , Semántica
6.
PLoS Pathog ; 15(4): e1007745, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31009517

RESUMEN

The mechanisms by which the gut luminal environment is disturbed by the immune system to foster pathogenic bacterial growth and survival remain incompletely understood. Here, we show that STAT2 dependent type I IFN signaling contributes to the inflammatory environment by disrupting hypoxia enabling the pathogenic S. Typhimurium to outgrow the microbiota. Stat2-/- mice infected with S. Typhimurium exhibited impaired type I IFN induced transcriptional responses in cecal tissue and reduced bacterial burden in the intestinal lumen compared to infected wild-type mice. Although inflammatory pathology was similar between wild-type and Stat2-/- mice, we observed decreased hypoxia in the gut tissue of Stat2-/- mice. Neutrophil numbers were similar in wild-type and Stat2-/- mice, yet Stat2-/- mice showed reduced levels of myeloperoxidase activity. In vitro, the neutrophils from Stat2-/- mice produced lower levels of superoxide anion upon stimulation with the bacterial ligand N-formylmethionyl-leucyl-phenylalanine (fMLP) in the presence of IFNα compared to neutrophils from wild-type mice, indicating that the neutrophils were less functional in Stat2-/- mice. Cytochrome bd-II oxidase-mediated respiration enhances S. Typhimurium fitness in wild-type mice, while in Stat2-/- deficiency, this respiratory pathway did not provide a fitness advantage. Furthermore, luminal expansion of S. Typhimurium in wild-type mice was blunted in Stat2-/- mice. Compared to wild-type mice which exhibited a significant perturbation in Bacteroidetes abundance, Stat2-/- mice exhibited significantly less perturbation and higher levels of Bacteroidetes upon S. Typhimurium infection. Our results highlight STAT2 dependent type I IFN mediated inflammation in the gut as a novel mechanism promoting luminal expansion of S. Typhimurium.


Asunto(s)
Disbiosis/inmunología , Gastroenteritis/inmunología , Inflamación/inmunología , Interferón Tipo I/inmunología , Factor de Transcripción STAT2/fisiología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Animales , Células Cultivadas , Disbiosis/metabolismo , Disbiosis/patología , Femenino , Gastroenteritis/metabolismo , Gastroenteritis/microbiología , Gastroenteritis/patología , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Interferón Tipo I/metabolismo , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Neutrófilos/patología , Factor de Transcripción STAT1/fisiología , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/patología
7.
Phytother Res ; 35(2): 1010-1022, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32914480

RESUMEN

Rosmarinic acid (RA) is a water-soluble phenolic compound extracted from Boraginaceae and Lamiaceae. This study was designed to investigate the role and mechanism of action of RA in improving nonalcoholic fatty liver disease (NAFLD). Male SD rats maintained on a high fat diet and L02 cells stimulated with oleic acid were treated with RA. Our results showed that RA significantly reduced total cholesterol, triglycerides, low-density lipoprotein cholesterol, alanine aminotransferase, aspartate aminotransferase, and malondialdehyde levels and increased high-density lipoprotein cholesterol, superoxide dismutase and adenosine triphosphate levels both in vivo and in vitro. Hematoxylin and eosin staining and oil red O staining showed that RA had a good lipid-lowering effect and substantial protective effects on liver injury. Transmission electron microscopy and JC-1 fluorescence results showed that RA could improve mitochondrial damage in hepatocytes. Additionally, flow cytometry results indicated that RA inhibited ROS generation and apoptosis in L02 cells. The impaired hepatocytes were restored by using RA in NAFLD models characterized by down-regulating YAP1 and TAZ, meanwhile up-regulating PPARγ and PGC-1α. When YAP1 was over-expressed, RA reduced the expression of YAP1; however, the action of RA was significantly blocked by silencing YAP1. The experimental results indicated that RA markedly alleviated NAFLD by repairing mitochondrial damage and regulating the YAP1/TAZ-PPARγ/PGC-1α signaling pathway.


Asunto(s)
Cinamatos/uso terapéutico , Depsidos/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , PPAR gamma/efectos de los fármacos , Animales , Cinamatos/farmacología , Depsidos/farmacología , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Transfección , Ácido Rosmarínico
8.
Infect Immun ; 87(4)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30617205

RESUMEN

During Salmonella enterica serovar Typhimurium infection, host inflammation alters the metabolic environment of the gut lumen to favor the outgrowth of the pathogen at the expense of the microbiota. Inflammation-driven changes in host cell metabolism lead to the release of l-lactate and molecular oxygen from the tissue into the gut lumen. Salmonella utilizes lactate as an electron donor in conjunction with oxygen as the terminal electron acceptor to support gut colonization. Here, we investigated transcriptional regulation of the respiratory l-lactate dehydrogenase LldD in vitro and in mouse models of Salmonella infection. The two-component system ArcAB repressed transcription of l-lactate utilization genes under anaerobic conditions in vitro The ArcAB-mediated repression of lldD transcription was relieved under microaerobic conditions. Transcription of lldD was induced by l-lactate but not d-lactate. A mutant lacking the regulatory protein LldR failed to induce lldD transcription in response to l-lactate. Furthermore, the lldR mutant exhibited reduced transcription of l-lactate utilization genes and impaired fitness in murine models of infection. These data provide evidence that the host-derived metabolites oxygen and l-lactate serve as cues for Salmonella to regulate lactate oxidation metabolism on a transcriptional level.


Asunto(s)
Mucosa Intestinal/microbiología , Ácido Láctico/metabolismo , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Femenino , Regulación Bacteriana de la Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Infecciones por Salmonella/microbiología , Salmonella typhimurium/enzimología , Salmonella typhimurium/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
PLoS Pathog ; 13(1): e1006186, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28129393

RESUMEN

Legionella pneumophila, the etiological agent of Legionnaires' disease, replicates intracellularly in protozoan and human hosts. Successful colonization and replication of this pathogen in host cells requires the Dot/Icm type IVB secretion system, which translocates approximately 300 effector proteins into the host cell to modulate various cellular processes. In this study, we identified RavK as a Dot/Icm substrate that targets the host cytoskeleton and reduces actin filament abundance in mammalian cells upon ectopic expression. RavK harbors an H95EXXH99 motif associated with diverse metalloproteases, which is essential for the inhibition of yeast growth and for the induction of cell rounding in HEK293T cells. We demonstrate that the actin protein itself is the cellular target of RavK and that this effector cleaves actin at a site between residues Thr351 and Phe352. Importantly, RavK-mediated actin cleavage also occurs during L. pneumophila infection. Cleavage by RavK abolishes the ability of actin to form polymers. Furthermore, an F352A mutation renders actin resistant to RavK-mediated cleavage; expression of the mutant in mammalian cells suppresses the cell rounding phenotype caused by RavK, further establishing that actin is the physiological substrate of RavK. Thus, L. pneumophila exploits components of the host cytoskeleton by multiple effectors with distinct mechanisms, highlighting the importance of modulating cellular processes governed by the actin cytoskeleton in the intracellular life cycle of this pathogen.


Asunto(s)
Citoesqueleto de Actina/patología , Citoesqueleto/patología , Interacciones Huésped-Patógeno/fisiología , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/metabolismo , Sistemas de Secreción Tipo IV/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Immunoblotting , Inmunoprecipitación , Enfermedad de los Legionarios/patología , Espectrometría de Masas , Ratones
10.
Biometals ; 32(3): 369-384, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30788645

RESUMEN

Host-associated microbial communities provide critical functions for their hosts. Transition metals are essential for both the mammalian host and the majority of commensal bacteria. As such, access to transition metals is an important component of host-microbe interactions in the gastrointestinal tract. In mammals, transition metal ions are often sequestered by metal binding proteins to limit microbial access under homeostatic conditions. In response to invading pathogens, the mammalian host further decreases availability of these micronutrients by regulating their trafficking or releasing high-affinity metal chelating proteins, a process termed nutritional immunity. Bacterial pathogens have evolved several mechanisms to subvert nutritional immunity. Here, we provide an overview on how metal ion availability shapes host-microbe interactions in the gut with a particular focus on intestinal inflammatory diseases.


Asunto(s)
Interacciones Microbiota-Huesped , Intestinos/microbiología , Elementos de Transición/metabolismo , Animales , Humanos
11.
PLoS Pathog ; 11(3): e1004704, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25738962

RESUMEN

The intracellular bacterial pathogen Legionella pneumophila provokes strong host responses and has proven to be a valuable model for the discovery of novel immunosurveillance pathways. Our previous work revealed that an environmental isolate of L. pneumophila induces a noncanonical form of cell death, leading to restriction of bacterial replication in primary mouse macrophages. Here we show that such restriction also occurs in infections with wild type clinical isolates. Importantly, we found that a lysine to arginine mutation at residue 88 (K88R) in the ribosome protein RpsL that not only confers bacterial resistance to streptomycin, but more importantly, severely attenuated the induction of host cell death and enabled L. pneumophila to replicate in primary mouse macrophages. Although conferring similar resistance to streptomycin, a K43N mutation in RpsL does not allow productive intracellular bacterial replication. Further analysis indicated that RpsL is capable of effectively inducing macrophage death via a pathway involved in lysosomal membrane permeabilization; the K88R mutant elicits similar responses but is less potent. Moreover, cathepsin B, a lysosomal protease that causes cell death after being released into the cytosol upon the loss of membrane integrity, is required for efficient RpsL-induced macrophage death. Furthermore, despite the critical role of cathepsin B in delaying RpsL-induced cell death, macrophages lacking cathepsin B do not support productive intracellular replication of L. pneumophila harboring wild type RpsL. This suggests the involvement of other yet unidentified components in the restriction of bacterial replication. Our results identified RpsL as a regulator in the interactions between bacteria such as L. pneumophila and primary mouse macrophages by triggering unique cellular pathways that restrict intracellular bacterial replication.


Asunto(s)
Proteínas Bacterianas/inmunología , Enfermedad de los Legionarios/inmunología , Macrófagos/inmunología , Proteínas Ribosómicas/inmunología , Animales , Muerte Celular , Citosol/inmunología , Citosol/metabolismo , Immunoblotting , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Legionella pneumophila , Lisosomas/inmunología , Lisosomas/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
12.
PLoS Pathog ; 11(7): e1005020, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26134274

RESUMEN

Type VI secretion systems (T6SSs) are widespread multi-component machineries that translocate effectors into either eukaryotic or prokaryotic cells, for virulence or for interbacterial competition. Herein, we report that the T6SS-4 from Yersinia pseudotuberculosis displays an unexpected function in the transportation of Zn2+ to combat diverse stresses and host immunity. Environmental insults such as oxidative stress induce the expression of T6SS-4 via OxyR, the transcriptional factor that also regulates many oxidative response genes. Zinc transportation is achieved by T6SS-4-mediated translocation of a novel Zn2+-binding protein substrate YezP (YPK_3549), which has the capacity to rescue the sensitivity to oxidative stress exhibited by T6SS-4 mutants when added to extracellular milieu. Disruption of the classic zinc transporter ZnuABC together with T6SS-4 or yezP results in mutants that almost completely lost virulence against mice, further highlighting the importance of T6SS-4 in resistance to host immunity. These results assigned an unconventional role to T6SSs, which will lay the foundation for studying novel mechanisms of metal ion uptake by bacteria and the role of this process in their resistance to host immunity and survival in harmful environments.


Asunto(s)
Adaptación Fisiológica/inmunología , Sistemas de Secreción Tipo VI/metabolismo , Infecciones por Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/metabolismo , Yersinia pseudotuberculosis/patogenicidad , Animales , Western Blotting , Ensayo de Cambio de Movilidad Electroforética , Ratones , Reacción en Cadena de la Polimerasa , Reacción en Cadena en Tiempo Real de la Polimerasa , Estrés Fisiológico/inmunología , Sistemas de Secreción Tipo VI/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología , Zinc/metabolismo
13.
Proc Natl Acad Sci U S A ; 109(34): 13567-72, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22872863

RESUMEN

Legionella pneumophila is an opportunistic intracellular pathogen that causes sporadic and epidemic cases of Legionnaires' disease. Emerging data suggest that Legionella infection involves the subversion of host phosphoinositide (PI) metabolism. However, how this bacterium actively manipulates PI lipids to benefit its infection is still an enigma. Here, we report that the L. pneumophila virulence factor SidF is a phosphatidylinositol polyphosphate 3-phosphatase that specifically hydrolyzes the D3 phosphate of PI(3,4)P(2) and PI(3,4,5)P(3). This activity is necessary for anchoring of PI(4)P-binding effectors to bacterial phagosomes. Crystal structures of SidF and its complex with its substrate PI(3,4)P(2) reveal striking conformational rearrangement of residues at the catalytic site to form a cationic pocket that specifically accommodates the D4 phosphate group of the substrate. Thus, our findings unveil a unique Legionella PI phosphatase essential for the establishment of lipid identity of bacterial phagosomes.


Asunto(s)
Proteínas Bacterianas/química , Legionella pneumophila/enzimología , Monoéster Fosfórico Hidrolasas/química , Catálisis , Dominio Catalítico , Cristalografía por Rayos X/métodos , Hidrólisis , Lípidos/química , Conformación Molecular , Oxidación-Reducción , Fagocitosis , Fagosomas/metabolismo , Fosfatidilinositoles/química , Estructura Terciaria de Proteína , Especificidad por Sustrato
14.
Cell Microbiol ; 15(11): 1783-95, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23773455

RESUMEN

The intracellular pathogen Legionella pneumophila is able to strike a balance between the death and survival of the host cell during infection. Despite the presence of high level of active caspase 3, the executioner caspase of apoptotic cell death, infected permissive macrophages are markedly resistant to exogenous apoptotic stimuli. Several bacterial molecules capable of promoting the cell survival pathways have been identified, but proteins involved in the activation of caspase 3 remain unknown. To study the mechanism of L. pneumophila-mediated caspase 3 activation, we tested all known Dot/Icm substrates for their ability to activate caspase 3. Five effectors capable of causing caspase 3 activation upon transient expression were identified. Among these, by using its ability to activate caspase 3 by inducing the release of cytochrome c from the mitochondria, we demonstrated that VipD is a phospholipase A2, which hydrolyses phosphatidylethanolamine (PE) and phosphocholine (PC) on the mitochondrial membrane in a manner that appears to require host cofactor(s). The lipase activity leads to the production of free fatty acids and 2-lysophospholipids, which destabilize the mitochondrial membrane and may contribute to the release of cytochrome c and the subsequent caspase 3 activation. Furthermore, we found that whereas it is not detectably defectively in caspase 3 activation in permissive cells, amutant lacking all of these five genes is less potent in inducing apoptosis in dendritic cells. Our results reveal that activation of host cell death pathways by L. pneumophila is a result of the effects of multiple bacterial proteins with diverse biochemical functions.


Asunto(s)
Proteínas Bacterianas/metabolismo , Caspasa 3/biosíntesis , Interacciones Huésped-Patógeno , Legionella pneumophila/metabolismo , Factores de Virulencia/metabolismo , Animales , Apoptosis , Células Cultivadas , Células Dendríticas/microbiología , Humanos , Ratones
15.
Infect Immun ; 81(9): 3077-88, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23753633

RESUMEN

Legionella pneumophila, the etiological agent for Legionnaires' disease, is ubiquitous in the aqueous environment, where it replicates as an intracellular parasite of free-living protozoa. Our understanding of L. pneumophila pathogenicity is obtained mostly from study of derivatives of several clinical isolates, which employ almost identical virulent determinants to exploit host functions. To determine whether environmental L. pneumophila isolates interact similarly with the model host systems, we analyzed intracellular replication of several recently isolated such strains and found that these strains cannot productively grow in bone marrow-derived macrophages of A/J mice, which are permissive for all examined laboratory strains. By focusing on one strain called LPE509, we found that its deficiency in intracellular replication in primary A/J macrophages is not caused by the lack of important pathogenic determinants because this strain replicates proficiently in two protozoan hosts and the human macrophage U937 cell. We also found that in the early phase of infection, the trafficking of this strain in A/J macrophages is similar to that of JR32, a derivative of strain Philadelphia 1. Furthermore, infection of these cells by LPE509 caused extensive cell death in a process that requires the Dot/Icm type IV secretion system. Finally, we showed that the cell death is caused neither by the activation of the NAIP5/NLRC4 inflammasome nor by the recently described caspase 11-dependent pathway. Our results revealed that some environmental L. pneumophila strains are unable to overcome the defense conferred by primary macrophages from mice known to be permissive for laboratory L. pneumophila strains. These results also suggest the existence of a host immune surveillance mechanism differing from those currently known in responding to L. pneumophila infection.


Asunto(s)
Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/patología , Macrófagos/microbiología , Macrófagos/patología , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasas/genética , Caspasas/metabolismo , Muerte Celular/fisiología , Línea Celular Tumoral , Femenino , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Enfermedad de los Legionarios/genética , Enfermedad de los Legionarios/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo , Monocitos/microbiología , Monocitos/patología , Proteína Inhibidora de la Apoptosis Neuronal/genética , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Células U937
16.
Cell Chem Biol ; 30(5): 417-419, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-37207630

RESUMEN

In this issue of Cell Chemical Biology, Lettl et al.1 identify complex I as a suitable target for selective killing of Helicobacter pylori. The unique composition of complex I in H. pylori enables precision targeting of the carcinogenic pathogen while sparing representative species of the gut microbiota.


Asunto(s)
Microbioma Gastrointestinal , Infecciones por Helicobacter , Helicobacter pylori , Humanos , Infecciones por Helicobacter/tratamiento farmacológico
17.
Gut Microbes ; 15(1): 2221813, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37317012

RESUMEN

The Salmonella biofilm-associated amyloid protein, curli, is a dominant instigator of systemic inflammation and autoimmune responses following Salmonella infection. Systemic curli injections or infection of mice with Salmonella Typhimurium induce the major features of reactive arthritis, an autoimmune disorder associated with Salmonella infection in humans. In this study, we investigated the link between inflammation and microbiota in exacerbating autoimmunity. We studied C57BL/6 mice from two sources, Taconic Farms and Jackson Labs. Mice from Taconic Farms have been reported to have higher basal levels of the inflammatory cytokine IL - 17 than do mice from Jackson Labs due to the differences in their microbiota. When we systemically injected mice with purified curli, we observed a significant increase in diversity in the microbiota of Jackson Labs mice but not in that of the Taconic mice. In Jackson Labs, mice, the most striking effect was the expansion of Prevotellaceae. Furthermore, there were increases in the relative abundance of the family Akkermansiaceae and decreases in families Clostridiaceae and Muribaculaceae in Jackson Labs mice. Curli treatment led to significantly aggravated immune responses in the Taconic mice compared to Jackson Labs counterparts. Expression and production of IL - 1ß, a cytokine known to promote IL - 17 production, as well as expression of Tnfa increased in the gut mucosa of Taconic mice in the first 24 hours after curli injections, which correlated with significant increases in the number of neutrophils and macrophages in the mesenteric lymph nodes. A significant increase in the expression of Ccl3 in colon and cecum of Taconic mice injected with curli was detected. Taconic mice injected with curli also had elevated levels of inflammation in their knees. Overall, our data suggest that autoimmune responses to bacterial ligands, such as curli, are amplified in individuals with a microbiome that promote inflammation.


Asunto(s)
Artritis , Microbioma Gastrointestinal , Microbiota , Infecciones por Salmonella , Humanos , Animales , Ratones , Ratones Endogámicos C57BL , Inmunidad Mucosa , Proteínas Amiloidogénicas , Inflamación , Bacteroidetes
18.
mBio ; 14(1): e0244422, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36475774

RESUMEN

Chicks are ideal to follow the development of the intestinal microbiota and to understand how a pathogen perturbs this developing population. Taxonomic/metagenomic analyses captured the development of the chick microbiota in unperturbed chicks and in chicks infected with Salmonella enterica serotype Typhimurium (STm) during development. Taxonomic analysis suggests that colonization by the chicken microbiota takes place in several waves. The cecal microbiota stabilizes at day 12 posthatch with prominent Gammaproteobacteria and Clostridiales. Introduction of S. Typhimurium at day 4 posthatch disrupted the expected waves of intestinal colonization. Taxonomic and metagenomic shotgun sequencing analyses allowed us to identify species present in uninfected chicks. Untargeted metabolomics suggested different metabolic activities in infected chick microbiota. This analysis and gas chromatography-mass spectrometry on ingesta confirmed that lactic acid in cecal content coincides with the stable presence of enterococci in STm-infected chicks. Unique metabolites, including 2-isopropylmalic acid, an intermediate in the biosynthesis of leucine, were present only in the cecal content of STm-infected chicks. The metagenomic data suggested that the microbiota in STm-infected chicks contained a higher abundance of genes, from STm itself, involved in branched-chain amino acid synthesis. We generated an ilvC deletion mutant (STM3909) encoding ketol-acid-reductoisomerase, a gene required for the production of l-isoleucine and l-valine. ΔilvC mutants are disadvantaged for growth during competitive infection with the wild type. Providing the ilvC gene in trans restored the growth of the ΔilvC mutant. Our integrative approach identified biochemical pathways used by STm to establish a colonization niche in the chick intestine during development. IMPORTANCE Chicks are an ideal model to follow the development of the intestinal microbiota and to understand how a pathogen perturbs this developing population. Using taxonomic and metagenomic analyses, we captured the development of chick microbiota to 19 days posthatch in unperturbed chicks and in chicks infected with Salmonella enterica serotype Typhimurium (STm). We show that normal development of the microbiota takes place in waves and is altered in the presence of a pathogen. Metagenomics and metabolomics suggested that branched-chain amino acid biosynthesis is especially important for Salmonella growth in the infected chick intestine. Salmonella mutants unable to make l-isoleucine and l-valine colonize the chick intestine poorly. Restoration of the pathway for biosynthesis of these amino acids restored the colonizing ability of Salmonella. Integration of multiple analyses allowed us to correctly identify biochemical pathways used by Salmonella to establish a niche for colonization in the chick intestine during development.


Asunto(s)
Microbiota , Enfermedades de las Aves de Corral , Salmonelosis Animal , Animales , Pollos/microbiología , Isoleucina , Salmonella typhimurium/metabolismo , Ciego/microbiología , Aminoácidos de Cadena Ramificada/metabolismo , Valina/metabolismo , Salmonelosis Animal/microbiología , Enfermedades de las Aves de Corral/microbiología
19.
bioRxiv ; 2023 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-37425782

RESUMEN

During intestinal inflammation, host nutritional immunity starves microbes of essential micronutrients such as iron. Pathogens scavenge iron using siderophores, which is counteracted by the host using lipocalin-2, a protein that sequesters iron-laden siderophores, including enterobactin. Although the host and pathogens compete for iron in the presence of gut commensal bacteria, the roles of commensals in nutritional immunity involving iron remain unexplored. Here, we report that the gut commensal Bacteroides thetaiotaomicron acquires iron in the inflamed gut by utilizing siderophores produced by other bacteria including Salmonella, via a secreted siderophore-binding lipoprotein termed XusB. Notably, XusB-bound siderophores are less accessible to host sequestration by lipocalin-2 but can be "re-acquired" by Salmonella , allowing the pathogen to evade nutritional immunity. As the host and pathogen have been the focus of studies of nutritional immunity, this work adds commensal iron metabolism as a previously unrecognized mechanism modulating the interactions between pathogen and host nutritional immunity.

20.
Cell Host Microbe ; 31(10): 1639-1654.e10, 2023 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-37776864

RESUMEN

During intestinal inflammation, host nutritional immunity starves microbes of essential micronutrients, such as iron. Pathogens scavenge iron using siderophores, including enterobactin; however, this strategy is counteracted by host protein lipocalin-2, which sequesters iron-laden enterobactin. Although this iron competition occurs in the presence of gut bacteria, the roles of commensals in nutritional immunity involving iron remain unexplored. Here, we report that the gut commensal Bacteroides thetaiotaomicron acquires iron and sustains its resilience in the inflamed gut by utilizing siderophores produced by other bacteria, including Salmonella, via a secreted siderophore-binding lipoprotein XusB. Notably, XusB-bound enterobactin is less accessible to host sequestration by lipocalin-2 but can be "re-acquired" by Salmonella, allowing the pathogen to evade nutritional immunity. Because the host and pathogen have been the focus of studies of nutritional immunity, this work adds commensal iron metabolism as a previously unrecognized mechanism modulating the host-pathogen interactions and nutritional immunity.


Asunto(s)
Infecciones por Salmonella , Sideróforos , Humanos , Lipocalina 2/metabolismo , Sideróforos/metabolismo , Enterobactina/metabolismo , Bacterias/metabolismo , Hierro/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA