Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Mol Diagn ; 24(2): 158-166, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34775029

RESUMEN

Hospital-acquired infections pose significant costly global challenges to patient care. Rapid and sensitive methods to identify potential outbreaks are integral to infection control measures. Whole-genome sequencing (WGS)-based bacterial strain typing provides higher discriminatory power over standard nucleotide banding pattern-based methods such as repetitive sequence-based PCR (rep-PCR). However, integration of WGS into clinical epidemiology is limited by the lack of consensus in methodology and data analysis/interpretation. In this study, WGS was performed on genomic DNA extracted from 22 multidrug-resistant Pseudomonas aeruginosa (MDR-PA) isolates using next-generation sequencing. Resulting high-quality reads were analyzed for phylogenetic relatedness using a whole-genome multilocus sequence typing (wgMLST)-based software program and single-nucleotide variant phylogenomics (SNVPhyl). WGS-based results were compared with conventional MLST and archived rep-PCR results. Rep-PCR identified three independent clonal clusters of MDR-PA. Only one clonal cluster identified by rep-PCR, an endemic strain within the pediatric cystic fibrosis population at Texas Children's Hospital, was concordantly identified using wgMLST and SNVPhyl. Results were highly consistent between the three sequence-based analyses (conventional MLST, wgMLST, and SNVPhyl), and these results remained consistent with the addition of 74 MDR-PA genomes. These WGS-based methods provided greater resolution for strain discrimination than rep-PCR or standard MLST classification, and the ease of use of wgMLST software renders it clinically viable for analysis, interpretation, and reporting of WGS-based strain typing.


Asunto(s)
Pseudomonas aeruginosa , Secuencias Repetitivas de Ácidos Nucleicos , Técnicas de Tipificación Bacteriana/métodos , Niño , Humanos , Tipificación de Secuencias Multilocus/métodos , Filogenia , Reacción en Cadena de la Polimerasa/métodos , Pseudomonas aeruginosa/genética , Secuenciación Completa del Genoma/métodos
2.
mBio ; 12(2)2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33653893

RESUMEN

Multiple studies have implicated microbes in the development of inflammation, but the mechanisms remain unknown. Bacteria in the genus Fusobacterium have been identified in the intestinal mucosa of patients with digestive diseases; thus, we hypothesized that Fusobacterium nucleatum promotes intestinal inflammation. The addition of >50 kDa F. nucleatum conditioned media, which contain outer membrane vesicles (OMVs), to colonic epithelial cells stimulated secretion of the proinflammatory cytokines interleukin-8 (IL-8) and tumor necrosis factor (TNF). In addition, purified F. nucleatum OMVs, but not compounds <50 kDa, stimulated IL-8 and TNF production; which was decreased by pharmacological inhibition of Toll-like receptor 4 (TLR4). These effects were linked to downstream effectors p-ERK, p-CREB, and NF-κB. F. nucleatum >50-kDa compounds also stimulated TNF secretion, p-ERK, p-CREB, and NF-κB activation in human colonoid monolayers. In mice harboring a human microbiota, pretreatment with antibiotics and a single oral gavage of F. nucleatum resulted in inflammation. Compared to mice receiving vehicle control, mice treated with F. nucleatum showed disruption of the colonic architecture, with increased immune cell infiltration and depleted mucus layers. Analysis of mucosal gene expression revealed increased levels of proinflammatory cytokines (KC, TNF, IL-6, IFN-γ, and MCP-1) at day 3 and day 5 in F. nucleatum-treated mice compared to controls. These proinflammatory effects were absent in mice who received F. nucleatum without pretreatment with antibiotics, suggesting that an intact microbiome is protective against F. nucleatum-mediated immune responses. These data provide evidence that F. nucleatum promotes proinflammatory signaling cascades in the context of a depleted intestinal microbiome.IMPORTANCE Several studies have identified an increased abundance of Fusobacterium in the intestinal tracts of patients with colon cancer, liver cirrhosis, primary sclerosing cholangitis, gastroesophageal reflux disease, HIV infection, and alcoholism. However, the direct mechanism(s) of action of Fusobacterium on pathophysiological within the gastrointestinal tract is unclear. These studies have identified that F. nucleatum subsp. polymorphum releases outer membrane vesicles which activate TLR4 and NF-κB to stimulate proinflammatory signals in vitro Using mice harboring a human microbiome, we demonstrate that F. nucleatum can promote inflammation, an effect which required antibiotic-mediated alterations in the gut microbiome. Collectively, these results suggest a mechanism by which F. nucleatum may contribute to intestinal inflammation.


Asunto(s)
Membrana Externa Bacteriana/inmunología , Vesículas Extracelulares/inmunología , Fusobacterium nucleatum/inmunología , Fusobacterium nucleatum/metabolismo , Inflamación/microbiología , Animales , Células Cultivadas , Colon/citología , Medios de Cultivo/farmacología , Citocinas/análisis , Citocinas/inmunología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Femenino , Fusobacterium nucleatum/patogenicidad , Microbioma Gastrointestinal , Células HT29 , Humanos , Inflamación/inmunología , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/inmunología , Transducción de Señal , Receptor Toll-Like 4/inmunología
3.
Gut Microbes ; 12(1): 1788898, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-32804011

RESUMEN

Antibiotic resistance is one of the world's greatest public health challenges and adjunct probiotic therapies are strategies that could lessen this burden. Clostridioides difficile infection (CDI) is a prime example where adjunct probiotic therapies could decrease disease incidence through prevention. Human-derived Lactobacillus reuteri is a probiotic that produces the antimicrobial compound reuterin known to prevent C. difficile colonization of antibiotic-treated fecal microbial communities. However, the mechanism of inhibition is unclear. We show that reuterin inhibits C. difficile outgrowth from spores and vegetative cell growth, however, no effect on C. difficile germination or sporulation was observed. Consistent with published studies, we found that exposure to reuterin stimulated reactive oxygen species (ROS) in C. difficile, resulting in a concentration-dependent reduction in cell viability that was rescued by the antioxidant glutathione. Sublethal concentrations of reuterin enhanced the susceptibility of vegetative C. difficile to vancomycin and metronidazole treatment and reduced toxin synthesis by C. difficile. We also demonstrate that reuterin is protective against C. difficile toxin-mediated cellular damage in the human intestinal enteroid model. Overall, our results indicate that ROS are essential mediators of reuterin activity and show that reuterin production by L. reuteri is compatible as a therapeutic in a clinically relevant model.


Asunto(s)
Clostridioides difficile/efectos de los fármacos , Gliceraldehído/análogos & derivados , Propano/farmacología , Especies Reactivas de Oxígeno/metabolismo , Antibacterianos/farmacología , Clostridioides difficile/crecimiento & desarrollo , Clostridioides difficile/metabolismo , Clostridioides difficile/patogenicidad , Sinergismo Farmacológico , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Gliceraldehído/metabolismo , Gliceraldehído/farmacología , Humanos , Limosilactobacillus reuteri/metabolismo , Organoides/efectos de los fármacos , Organoides/microbiología , Estrés Oxidativo/efectos de los fármacos , Probióticos/metabolismo , Propano/metabolismo , Esporas Bacterianas/efectos de los fármacos , Esporas Bacterianas/crecimiento & desarrollo
4.
Am J Physiol Gastrointest Liver Physiol ; 318(5): G870-G888, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32223302

RESUMEN

Clostridioides difficile is an important nosocomial pathogen that produces toxins to cause life-threatening diarrhea and colitis. Toxins bind to epithelial receptors and promote the collapse of the actin cytoskeleton. C. difficile toxin activity is commonly studied in cancer-derived and immortalized cell lines. However, the biological relevance of these models is limited. Moreover, no model is available for examining C. difficile-induced enteritis, an understudied health problem. We hypothesized that human intestinal enteroids (HIEs) express toxin receptors and provide a new model to dissect C. difficile cytotoxicity in the small intestine. We generated biopsy-derived jejunal HIE and Vero cells, which stably express LifeAct-Ruby, a fluorescent label of F-actin, to monitor actin cytoskeleton rearrangement by live-cell microscopy. Imaging analysis revealed that toxins from pathogenic C. difficile strains elicited cell rounding in a strain-dependent manner, and HIEs were tenfold more sensitive to toxin A (TcdA) than toxin B (TcdB). By quantitative PCR, we paradoxically found that HIEs expressed greater quantities of toxin receptor mRNA and yet exhibited decreased sensitivity to toxins when compared with traditionally used cell lines. We reasoned that these differences may be explained by components, such as mucins, that are present in HIEs cultures, that are absent in immortalized cell lines. Addition of human-derived mucin 2 (MUC2) to Vero cells delayed cell rounding, indicating that mucus serves as a barrier to toxin-receptor binding. This work highlights that investigation of C. difficile infection in that HIEs can provide important insights into the intricate interactions between toxins and the human intestinal epithelium.NEW & NOTEWORTHY In this article, we developed a novel model of Clostridioides difficile-induced enteritis using jejunal-derived human intestinal enteroids (HIEs) transduced with fluorescently tagged F-actin. Using live-imaging, we identified that jejunal HIEs express high levels of TcdA and CDT receptors, are more sensitive to TcdA than TcdB, and secrete mucus, which delays toxin-epithelial interactions. This work also optimizes optically clear C. difficile-conditioned media suitable for live-cell imaging.


Asunto(s)
Clostridioides difficile/patogenicidad , Infecciones por Clostridium/microbiología , Enteritis/microbiología , Yeyuno/microbiología , ADP Ribosa Transferasas/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/microbiología , Citoesqueleto de Actina/ultraestructura , Animales , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Forma de la Célula , Chlorocebus aethiops , Clostridioides difficile/metabolismo , Infecciones por Clostridium/metabolismo , Infecciones por Clostridium/patología , Enteritis/metabolismo , Enteritis/patología , Enterotoxinas/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Yeyuno/metabolismo , Yeyuno/ultraestructura , Mucina 2/metabolismo , Organoides , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Factores de Tiempo , Células Vero , Virulencia
5.
Dig Dis Sci ; 65(3): 695-705, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32067143

RESUMEN

The human gastrointestinal (GI) tract contains communities of microbes (bacteria, fungi, viruses) that vary by anatomic location and impact human health. Microbial communities differ in composition based on age, diet, and location in the gastrointestinal tract. Differences in microbial composition have been associated with chronic disease states. In terms of function, microbial metabolites provide key signals that help maintain healthy human physiology. Alterations of the healthy gastrointestinal microbiome have been linked to the development of various disease states including inflammatory bowel disease, diabetes, and colorectal cancer. While the definition of a healthy GI microbiome cannot be precisely identified, features of a healthy gut microbiome include relatively greater biodiversity and relative abundances of specific phyla and genera. Microbes with desirable functional profiles for the human host have been identified, in addition to specific metabolic features of the microbiome. This article reviews the composition and function of the healthy human GI microbiome, including the relative abundances of different bacterial taxa and the specific metabolic pathways and classes of microbial metabolites contributing to human health and disease prevention.


Asunto(s)
Investigación Biomédica/tendencias , Microbioma Gastrointestinal/fisiología , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/fisiología , Estado de Salud , Investigación Biomédica/métodos , Humanos , Microbiota/fisiología
6.
Microbiol Resour Announc ; 8(43)2019 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-31649076

RESUMEN

Hybrid de novo assembly of Illumina/Nanopore sequence data produced complete circular sequences of the chromosome and a plasmid for the multidrug-resistant Pseudomonas aeruginosa Houston-1 strain. This provides a high-quality representative sequence for a lineage endemic to a pediatric cystic fibrosis care center at Texas Children's Hospital.

7.
BMC Microbiol ; 16(1): 242, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27756217

RESUMEN

BACKGROUND: Interleukin (IL)-8 is the key agent for initiating an inflammatory response to infection with Helicobacter pylori. Some strains of Lactobacillus spp. are known to colonize the stomach and suppress inflammation caused by H. pylori. In this study, we characterized two gastric-derived lactobacilli, Lactobacillus salivarius (LS) strains B37 and B60, capable of inhibiting H. pylori-induced IL-8 production by gastric epithelial cells. RESULTS: Conditioned media from LS-B37 and LS-B60 suppressed H. pylori-induced IL-8 production and mRNA expression from AGS cells without inhibiting H. pylori growth. These conditioned media suppressed the activation of NF-κB but did not suppress c-Jun activation. IL-8 inhibitory substances in conditioned media of LS-B37 and LS-B60 are heat-stable and larger than 100 kDa in size. The inhibitory activity of LS-B37 was abolished when the conditioned medium was treated with α-amylase but still remained when treated with either proteinase K, trypsin, lipase or lysozyme. The activity of LS-B60 was abolished when the conditioned medium was treated with either amylase or proteinase K but still remained when treated with lysozyme. Treatment with lipase and trypsin also significantly affected the inhibitory activity of LS-B60 although the conditioned medium retained IL-8 suppression statistically different from media control. CONCLUSIONS: These results suggest that L. salivarius strains B37 and B60 produce different immunomodulatory factors capable of suppressing H. pylori-induced IL-8 production from gastric epithelial cells. Our results suggest that the large, heat-stable immunomodulatory substance(s) present in the LCM of LS-B37 is a polysaccharide, while the one(s) of LS-B60 is either complex consisting of components of polysaccharide, lipid and protein or includes multiple components such as glycoprotein and lipoprotein.


Asunto(s)
Células Epiteliales/inmunología , Células Epiteliales/microbiología , Helicobacter pylori/patogenicidad , Interleucina-8/agonistas , Interleucina-8/metabolismo , Ligilactobacillus salivarius/inmunología , Ligilactobacillus salivarius/fisiología , Estómago/inmunología , Antiinflamatorios/uso terapéutico , Línea Celular , Medios de Cultivo Condicionados , Endopeptidasa K/farmacología , Células Epiteliales/metabolismo , Mucosa Gástrica/metabolismo , Regulación Bacteriana de la Expresión Génica , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Interleucina-8/genética , Lactobacillus/metabolismo , Ligilactobacillus salivarius/efectos de los fármacos , Lipasa/metabolismo , Muramidasa/metabolismo , FN-kappa B/metabolismo , Probióticos/uso terapéutico , ARN Mensajero/biosíntesis , Estómago/microbiología , Tripsina/metabolismo , alfa-Amilasas/farmacología
8.
Microbiologyopen ; 5(5): 802-818, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27353144

RESUMEN

Bacterial-derived compounds from the intestinal microbiome modulate host mucosal immunity. Identification and mechanistic studies of these compounds provide insights into host-microbial mutualism. Specific Lactobacillus reuteri strains suppress production of the proinflammatory cytokine, tumor necrosis factor (TNF), and are protective in a mouse model of colitis. Human-derived L. reuteri strain ATCC PTA 6475 suppresses intestinal inflammation and produces 5,10-methenyltetrahydrofolic acid polyglutamates. Insertional mutagenesis identified the bifunctional dihydrofolate synthase/folylpolyglutamate synthase type 2 (folC2) gene as essential for 5,10-methenyltetrahydrofolic acid polyglutamate biosynthesis, as well as for suppression of TNF production by activated human monocytes, and for the anti-inflammatory effect of L. reuteri 6475 in a trinitrobenzene sulfonic acid-induced mouse model of acute colitis. In contrast, folC encodes the enzyme responsible for folate polyglutamylation but does not impact TNF suppression by L. reuteri. Comparative transcriptomics between wild-type and mutant L. reuteri strains revealed additional genes involved in immunomodulation, including previously identified hdc genes involved in histidine to histamine conversion. The folC2 mutant yielded diminished hdc gene cluster expression and diminished histamine production, suggesting a link between folate and histadine/histamine metabolism. The identification of genes and gene networks regulating production of bacterial-derived immunoregulatory molecules may lead to improved anti-inflammatory strategies for digestive diseases.


Asunto(s)
Colitis/terapia , Limosilactobacillus reuteri/metabolismo , Complejos Multienzimáticos/metabolismo , Péptido Sintasas/metabolismo , Probióticos/uso terapéutico , Animales , Células Cultivadas , Colitis/inducido químicamente , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal/fisiología , Humanos , Inflamación/terapia , Ratones , Ratones Endogámicos BALB C , Mutagénesis Insercional , Tetrahidrofolatos/metabolismo , Ácido Trinitrobencenosulfónico , Factor de Necrosis Tumoral alfa/biosíntesis
9.
BMC Microbiol ; 14: 177, 2014 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-24989059

RESUMEN

BACKGROUND: Clostridium difficile is the main cause of hospital-acquired diarrhea and colitis known as C. difficile-associated disease (CDAD).With increased severity and failure of treatment in CDAD, new approaches for prevention and treatment, such as the use of probiotics, are needed. Since the pathogenesis of CDAD involves an inflammatory response with a massive influx of neutrophils recruited by interleukin (IL)-8, this study aimed to investigate the probiotic effects of Lactobacillus spp. on the suppression of IL-8 production in response to C. difficile infection. RESULTS: We screened Lactobacillus conditioned media from 34 infant fecal isolates for the ability to suppress C. difficile-induced IL-8 production from HT-29 cells. Factors produced by two vancomycin-resistant lactobacilli, L. rhamnosus L34 (LR-L34) and L.casei L39 (LC-L39), suppressed the secretion and transcription of IL-8 without inhibiting C. difficile viability or toxin production. Conditioned media from LR-L34 suppressed the activation of phospho-NF-κB with no effect on phospho-c-Jun. However, LC-L39 conditioned media suppressed the activation of both phospho-NF-κB and phospho-c-Jun. Conditioned media from LR-L34 and LC-L39 also decreased the production of C. difficile-induced GM-CSF in HT-29 cells. Immunomodulatory factors present in the conditioned media of both LR-L34 and LC-L39 are heat-stable up to 100°C and > 100 kDa in size. CONCLUSIONS: Our results suggest that L. rhamnosus L34 and L. casei L39 each produce factors capable of modulating inflammation stimulated by C. difficile. These vancomycin-resistant Lactobacillus strains are potential probiotics for treating or preventing CDAD.


Asunto(s)
Antibiosis , Clostridioides difficile/inmunología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Interleucina-8/metabolismo , Lacticaseibacillus casei/fisiología , Lacticaseibacillus rhamnosus/fisiología , Línea Celular , Humanos , Probióticos
10.
Genome Biol Evol ; 6(7): 1772-89, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24951561

RESUMEN

The vertebrate gut symbiont Lactobacillus reuteri has diversified into separate clades reflecting host origin. Strains show evidence of host adaptation, but how host-microbe coevolution influences microbial-derived effects on hosts is poorly understood. Emphasizing human-derived strains of L. reuteri, we combined comparative genomic analyses with functional assays to examine variations in host interaction among genetically distinct ecotypes. Within clade II or VI, the genomes of human-derived L. reuteri strains are highly conserved in gene content and at the nucleotide level. Nevertheless, they share only 70-90% of total gene content, indicating differences in functional capacity. Human-associated lineages are distinguished by genes related to bacteriophages, vitamin biosynthesis, antimicrobial production, and immunomodulation. Differential production of reuterin, histamine, and folate by 23 clade II and VI strains was demonstrated. These strains also differed with respect to their ability to modulate human cytokine production (tumor necrosis factor, monocyte chemoattractant protein-1, interleukin [IL]-1ß, IL-5, IL-7, IL-12, and IL-13) by myeloid cells. Microarray analysis of representative clade II and clade VI strains revealed global regulation of genes within the reuterin, vitamin B12, folate, and arginine catabolism gene clusters by the AraC family transcriptional regulator, PocR. Thus, human-derived L. reuteri clade II and VI strains are genetically distinct and their differences affect their functional repertoires and probiotic features. These findings highlight the biological impact of microbe:host coevolution and illustrate the functional significance of subspecies differences in the human microbiome. Consideration of host origin and functional differences at the subspecies level may have major impacts on probiotic strain selection and considerations of microbial ecology in mammalian species.


Asunto(s)
Evolución Molecular , Genómica , Limosilactobacillus reuteri/fisiología , Probióticos , Animales , Línea Celular , Humanos , Limosilactobacillus reuteri/genética , Análisis por Micromatrices , Filogenia
11.
Helicobacter ; 19(2): 144-55, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24387083

RESUMEN

BACKGROUND: Helicobacter pylori colonization of the gastric epithelium induces interleukin-8 (IL-8) production and inflammation leading to host cell damage. We searched for gastric-derived Lactobacillus with the ability to suppress H. pylori-induced inflammation. MATERIALS AND METHODS: Conditioned media from gastric-derived Lactobacillus spp. were tested for the ability to suppress H. pylori-induced IL-8 production in AGS gastric epithelial cells. IL-8 protein and mRNA levels were measured by ELISA and qPCR, respectively. The changes on host cell signaling pathway were analyzed by Western blotting and the anti-inflammatory effect was tested in a Sprague-Dawley rat model. RESULTS: Conditioned media from L. salivarius B101, L. rhamnosus B103, and L. plantarum XB7 suppressed IL-8 production and IL-8 mRNA expression in H. pylori-induced AGS cells without inhibiting H. pylori growth. Conditioned media from LS-B101, LR-B103, and LP-XB7 suppressed the activation of NF-κB in AGS cells, while strain LP-XB7 also suppressed c-Jun activation. The anti-inflammatory effect of LP-XB7 was further assessed in vivo using a H. pylori-infected Sprague-Dawley rat model. Strain LP-XB7 contributed to a delay in the detection and colonization of H. pylori in rat stomachs, attenuated gastric inflammation, and ameliorated gastric histopathology. Additionally, the administration of LP-XB7 correlated with the suppression of TNF-α and CINC-1 in sera, and suppression of CINC-1 in the gastric mucosa of H. pylori-infected rats. CONCLUSIONS: These results suggest that L. plantarum XB7 produces secreted factors capable of modulating inflammation during H. pylori infection, and this probiotic Lactobacillus strain shows promise as an adjunctive therapy for treating H. pylori-associated disease.


Asunto(s)
Antiinflamatorios/uso terapéutico , Infecciones por Helicobacter/terapia , Inflamación/inmunología , Interleucina-8/biosíntesis , Lactobacillus plantarum/inmunología , Probióticos/uso terapéutico , Adulto , Anciano , Animales , Medios de Cultivo Condicionados/farmacología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Femenino , Mucosa Gástrica/citología , Mucosa Gástrica/inmunología , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/inmunología , Humanos , Inmunomodulación , Inflamación/microbiología , Interleucina-8/genética , Proteínas Quinasas JNK Activadas por Mitógenos/biosíntesis , Masculino , Persona de Mediana Edad , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Estómago/microbiología , Estómago/patología , Factor de Transcripción ReIA/biosíntesis
12.
FEMS Microbiol Lett ; 293(1): 65-72, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19222575

RESUMEN

Human-derived lactobacilli were isolated from fecal samples of healthy volunteers. Forty-six isolates from different volunteers were selected and investigated for their immunomodulatory properties. Conditioned medium from each isolate was assessed for its effect on tumor necrosis factor (TNF) production in lipopolysaccharide-activated THP-1 monocytes. Of 46 Lactobacillus isolates, 12 significantly inhibited TNF production in varying magnitude. Lactobacillus strain TH58 displayed the most potent TNF-inhibitory activity (70% inhibition). In contrast, Lactobacillus strain TH14 exhibited immunostimulatory property by activating TNF production in THP-1 monocytes. Lactobacillus TH14 induced nuclear factor-kappaB (NF-kappaB) activation in the absence of lipopolysaccharide stimulation, whereas Lactobacillus TH58 had no effect on NF-kappaB signaling, irrespective of lipopolysaccharide stimulation. Strain TH58 was identified as Lactobacillus saerimneri and strain TH14 as Lactobacillus ruminis by sequence analysis of their 16S rRNA genes. This is the first report of a human isolate of L. saerimneri with TNF-inhibitory activity and L. ruminis, an indigenous species to humans, with TNF stimulatory activity. Our data suggest the potential use of these two strains as immunoprobiotic candidates.


Asunto(s)
Heces/microbiología , Regulación de la Expresión Génica , Lactobacillus/inmunología , Monocitos/inmunología , Probióticos , Factor de Necrosis Tumoral alfa/biosíntesis , Adolescente , Adulto , Línea Celular , Femenino , Humanos , Lactobacillus/clasificación , Lactobacillus/genética , Lactobacillus/aislamiento & purificación , Lipopolisacáridos/farmacología , Masculino , Monocitos/metabolismo , FN-kappa B/metabolismo , Especificidad de la Especie , Factor de Necrosis Tumoral alfa/genética , Adulto Joven
13.
FEMS Microbiol Lett ; 243(1): 1-8, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15667993

RESUMEN

Iron dependent regulatory proteins of the diphtheria toxin repressor family regulate transcription in a variety of bacterial species. These regulators have three domains. Domains 1 and 2 are required for DNA- and metal-binding while the role of the third domain is only partially defined. We compared full-length and carboxyl-terminally truncated variants of Corynebacterium diphtheriae DtxR and Mycobacterium tuberculosis IdeR for recognition by antibodies, DNA binding, and repressor activity. The third domain of DtxR contains immunodominant epitopes and is required for full repressor activity in an Escherichia coli reporter system, but it is not required for binding to DNA in vitro. In contrast, the third domain of IdeR is required both for full DNA binding activity in vitro and for repressor activity in vivo. DtxR and IdeR differ significantly in their requirements for domain 3 for DNA-binding and repressor activity.


Asunto(s)
Proteínas Bacterianas/genética , Corynebacterium diphtheriae/metabolismo , Proteínas de Unión al ADN/genética , Regulación Bacteriana de la Expresión Génica , Hierro/metabolismo , Mycobacterium tuberculosis/metabolismo , Proteínas Represoras/genética , Anticuerpos Antibacterianos/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Corynebacterium diphtheriae/genética , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Variación Genética , Mutación , Mycobacterium tuberculosis/genética , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA