Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(18): e2119396119, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35476524

RESUMEN

Combatting Clostridioides difficile infections, a dominant cause of hospital-associated infections with incidence and resulting deaths increasing worldwide, is complicated by the frequent emergence of new virulent strains. Here, we employ whole-genome sequencing, high-throughput phenotypic screenings, and genome-scale models of metabolism to evaluate the genetic diversity of 451 strains of C. difficile. Constructing the C. difficile pangenome based on this set revealed 9,924 distinct gene clusters, of which 2,899 (29%) are defined as core, 2,968 (30%) are defined as unique, and the remaining 4,057 (41%) are defined as accessory. We develop a strain typing method, sequence typing by accessory genome (STAG), that identifies 176 genetically distinct groups of strains and allows for explicit interrogation of accessory gene content. Thirty-five strains representative of the overall set were experimentally profiled on 95 different nutrient sources, revealing 26 distinct growth profiles and unique nutrient preferences; 451 strain-specific genome scale models of metabolism were constructed, allowing us to computationally probe phenotypic diversity in 28,864 unique conditions. The models create a mechanistic link between the observed phenotypes and strain-specific genetic differences and exhibit an ability to correctly predict growth in 76% of measured cases. The typing and model predictions are used to identify and contextualize discriminating genetic features and phenotypes that may contribute to the emergence of new problematic strains.


Asunto(s)
Clostridioides difficile , Infección Hospitalaria , Clostridioides , Clostridioides difficile/genética , Variación Genética , Humanos , Biología de Sistemas
2.
Am J Pathol ; 191(4): 704-719, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33516788

RESUMEN

The intestinal microbiota influences the development and function of the mucosal immune system. However, the exact mechanisms by which commensal microbes modulate immunity is not clear. We previously demonstrated that commensal Bacteroides ovatus ATCC 8384 reduces mucosal inflammation. Herein, we aimed to identify immunomodulatory pathways employed by B. ovatus. In germ-free mice, mono-association with B. ovatus shifted the CD11b+/CD11c+ and CD103+/CD11c+ dendritic cell populations. Because indole compounds are known to modulate dendritic cells, B. ovatus cell-free supernatant was screened for tryptophan metabolites by liquid chromatography-tandem mass spectrometry and larger quantities of indole-3-acetic acid were detected. Analysis of cecal and fecal samples from germ-free and B. ovatus mono-associated mice confirmed that B. ovatus could elevate indole-3-acetic acid concentrations in vivo. Indole metabolites have previously been shown to stimulate immune cells to secrete the reparative cytokine IL-22. Addition of B. ovatus cell-free supernatant to immature bone marrow-derived dendritic cells stimulated IL-22 secretion. The ability of IL-22 to drive repair in the intestinal epithelium was confirmed using a physiologically relevant human intestinal enteroid model. Finally, B. ovatus shifted the immune cell populations in trinitrobenzene sulfonic acid-treated mice and up-regulated colonic IL-22 expression, effects that correlated with decreased inflammation. Our data suggest that B. ovatus-produced indole-3-acetic acid promotes IL-22 production by immune cells, yielding beneficial effects on colitis.


Asunto(s)
Bacteroides/efectos de los fármacos , Colon/metabolismo , Inflamación/tratamiento farmacológico , Interleucinas/metabolismo , Ácido Trinitrobencenosulfónico/farmacología , Animales , Colitis/tratamiento farmacológico , Colitis/metabolismo , Colon/efectos de los fármacos , Citocinas/metabolismo , Sulfato de Dextran/metabolismo , Humanos , Inflamación/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Ratones , Interleucina-22
3.
J Pediatr Gastroenterol Nutr ; 74(2): 227-235, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34724447

RESUMEN

OBJECTIVES: Fecal microbiota transplantation (FMT) is arguably the most effective treatment for recurrent Clostridioides difficile infection (rCDI). Clinical reports on pediatric FMT have not systematically evaluated microbiome restoration in patients with co-morbidities. Here, we determined whether FMT recipient age and underlying co-morbidity influenced clinical outcomes and microbiome restoration when treated from shared fecal donor sources. METHODS: Eighteen rCDI patients participating in a single-center, open-label prospective cohort study received fecal preparation from a self-designated (single case) or two universal donors. Twelve age-matched healthy children and four pediatric ulcerative colitis (UC) cases from an independent serial FMT trial, but with a shared fecal donor were examined as controls for microbiome restoration using 16S rRNA gene sequencing of longitudinal fecal specimens. RESULTS: FMT was significantly more effective in rCDI recipients without underlying chronic co-morbidities where fecal microbiome composition in post-transplant responders was restored to levels of healthy children. Microbiome reconstitution was not associated with symptomatic resolution in some rCDI patients who had co-morbidities. Significant elevation in Bacteroidaceae, Bifidobacteriaceae, Lachnospiraceae, Ruminococcaceae, and Erysipelotrichaceae was consistently observed in pediatric rCDI responders, while Enterobacteriaceae decreased, correlating with augmented complex carbohydrate degradation capacity. CONCLUSION: Recipient background disease was a significant risk factor influencing FMT outcomes. Special attention should be taken when considering FMT for pediatric rCDI patients with underlying co-morbidities.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium , Niño , Infecciones por Clostridium/terapia , Trasplante de Microbiota Fecal , Heces , Humanos , Morbilidad , Estudios Prospectivos , ARN Ribosómico 16S/genética , Recurrencia , Resultado del Tratamiento
4.
BMC Microbiol ; 21(1): 154, 2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-34030655

RESUMEN

BACKGROUND: Bifidobacteria are commensal microbes of the mammalian gastrointestinal tract. In this study, we aimed to identify the intestinal colonization mechanisms and key metabolic pathways implemented by Bifidobacterium dentium. RESULTS: B. dentium displayed acid resistance, with high viability over a pH range from 4 to 7; findings that correlated to the expression of Na+/H+ antiporters within the B. dentium genome. B. dentium was found to adhere to human MUC2+ mucus and harbor mucin-binding proteins. Using microbial phenotyping microarrays and fully-defined media, we demonstrated that in the absence of glucose, B. dentium could metabolize a variety of nutrient sources. Many of these nutrient sources were plant-based, suggesting that B. dentium can consume dietary substances. In contrast to other bifidobacteria, B. dentium was largely unable to grow on compounds found in human mucus; a finding that was supported by its glycosyl hydrolase (GH) profile. Of the proteins identified in B. dentium by proteomic analysis, a large cohort of proteins were associated with diverse metabolic pathways, indicating metabolic plasticity which supports colonization of the dynamic gastrointestinal environment. CONCLUSIONS: Taken together, we conclude that B. dentium is well adapted for commensalism in the gastrointestinal tract.


Asunto(s)
Bifidobacterium/metabolismo , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Ácidos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bifidobacterium/genética , Bifidobacterium/crecimiento & desarrollo , Tracto Gastrointestinal/fisiología , Genoma Bacteriano , Glucosa/metabolismo , Humanos , Simbiosis
5.
Anaerobe ; 70: 102387, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34044101

RESUMEN

BACKGROUND: Fecal microbiota transplantation (FMT) aims to cure Clostridioides difficile infection (CDI) through reestablishing a healthy microbiome and restoring colonization resistance. Although often effective after one infusion, patients with continued microbiome disruptions may require multiple FMTs. In this N-of-1 study, we use a systems biology approach to evaluate CDI in a patient receiving chronic suppressive antibiotics with four failed FMTs over two years. METHODS: Seven stool samples were obtained between 2016-18 while the patient underwent five FMTs. Stool samples were cultured for C. difficile and underwent microbial characterization and functional gene analysis using shotgun metagenomics. C. difficile isolates were characterized through ribotyping, whole genome sequencing, metabolic pathway analysis, and minimum inhibitory concentration (MIC) determinations. RESULTS: Growing ten strains from each sample, the index and first four recurrent cultures were single strain ribotype F078-126, the fifth was a mixed culture of ribotypes F002 and F054, and the final culture was ribotype F002. One single nucleotide polymorphism (SNP) variant was identified in the RNA polymerase (RNAP) ß-subunit RpoB in the final isolated F078-126 strain when compared to previous F078-126 isolates. This SNV was associated with metabolic shifts but phenotypic differences in fidaxomicin MIC were not observed. Microbiome differences were observed over time during vancomycin therapy and after failed FMTs. CONCLUSION: This study highlights the importance of antimicrobial stewardship in patients receiving FMT. Continued antibiotics play a destructive role on a transplanted microbiome and applies selection pressure for resistance to the few antibiotics available to treat CDI.


Asunto(s)
Clostridioides difficile/fisiología , Infecciones por Clostridium/terapia , Trasplante de Microbiota Fecal , Antibacterianos/administración & dosificación , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/genética , Clostridioides difficile/aislamiento & purificación , Infecciones por Clostridium/tratamiento farmacológico , Infecciones por Clostridium/microbiología , Heces , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Polimorfismo de Nucleótido Simple , Insuficiencia del Tratamiento
6.
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
7.
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
8.
Anaerobe ; 54: 83-91, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30099125

RESUMEN

BACKGROUND: Morbidity and mortality associated with Clostridioides (formerly Clostridium) difficile infection (CDI) rises progressively with advanced age (≥65 years) due in part to perturbations of the gut microbiota and immune dysfunction. Epidemiological data of community-acquired CDI suggests increased susceptibility may begin earlier during middle-age (45-64 years) but the causation remains unknown. METHODS: Middle-aged (12-14 months) and young (2-4 months) adult mice were infected with C. difficile, and disease severity, gut microbiome and innate immune response were compared. Cytokine reconstitution studies were performed in infected middle-aged mice. RESULTS: Infection of middle-aged mice with C. difficile led to greater disease compared to young controls, which was associated with increases in C. difficile burden and toxin titers, and elevated bacterial translocation. With the exception of an expansion of C. difficile in middle-aged mice, microbiome analysis revealed no age-related differences. In contrast, middle-aged mice displayed a significant defect in neutrophil recruitment to the colon, with diminished levels of innate immune cytokines IL-6, IL-23 and IL-22. Importantly, recombinant IL-22 administration during CDI reduced morbidity and prevented death in middle-aged mice. CONCLUSION: Increased susceptibility to C. difficile occurs in middle-aged mice modeling the community-acquired CDI demographics and is driven by an impaired innate immune response.


Asunto(s)
Envejecimiento/inmunología , Clostridioides difficile/fisiología , Infecciones por Clostridium/inmunología , Interleucinas/inmunología , Neutrófilos/inmunología , Factores de Edad , Animales , Clostridioides difficile/inmunología , Infecciones por Clostridium/genética , Infecciones por Clostridium/microbiología , Femenino , Microbioma Gastrointestinal , Humanos , Inmunidad Innata , Interleucinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Interleucina-22
9.
Infect Immun ; 85(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28760934

RESUMEN

Integration of antibiotic and probiotic therapy has the potential to lessen the public health burden of antimicrobial-associated diseases. Clostridium difficile infection (CDI) represents an important example where the rational design of next-generation probiotics is being actively pursued to prevent disease recurrence. Because intrinsic resistance to clinically relevant antibiotics used to treat CDI (vancomycin, metronidazole, and fidaxomicin) is a desired trait in such probiotic species, we screened several bacteria and identified Lactobacillus reuteri to be a promising candidate for adjunct therapy. Human-derived L. reuteri bacteria convert glycerol to the broad-spectrum antimicrobial compound reuterin. When supplemented with glycerol, strains carrying the pocR gene locus were potent reuterin producers, with L. reuteri 17938 inhibiting C. difficile growth at a level on par with the level of growth inhibition by vancomycin. Targeted pocR mutations and complementation studies identified reuterin to be the precursor-induced antimicrobial agent. Pathophysiological relevance was demonstrated when the codelivery of L. reuteri with glycerol was effective against C. difficile colonization in complex human fecal microbial communities, whereas treatment with either glycerol or L. reuteri alone was ineffective. A global unbiased microbiome and metabolomics analysis independently confirmed that glycerol precursor delivery with L. reuteri elicited changes in the composition and function of the human microbial community that preferentially targets C. difficile outgrowth and toxicity, a finding consistent with glycerol fermentation and reuterin production. Antimicrobial resistance has thus been successfully exploited in the natural design of human microbiome evasion of C. difficile, and this method may provide a prototypic precursor-directed probiotic approach. Antibiotic resistance and substrate bioavailability may therefore represent critical new determinants of probiotic efficacy in clinical trials.


Asunto(s)
Antibacterianos/biosíntesis , Clostridioides difficile/crecimiento & desarrollo , Infecciones por Clostridium/prevención & control , Gliceraldehído/análogos & derivados , Glicerol/administración & dosificación , Limosilactobacillus reuteri/metabolismo , Probióticos , Propano/metabolismo , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Proteínas Bacterianas/genética , Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/inmunología , Infecciones por Clostridium/terapia , Descubrimiento de Drogas/métodos , Farmacorresistencia Bacteriana , Heces/microbiología , Fermentación , Microbioma Gastrointestinal , Gliceraldehído/metabolismo , Gliceraldehído/farmacología , Gliceraldehído/uso terapéutico , Glicerol/inmunología , Glicerol/metabolismo , Humanos , Metabolómica , Propano/farmacología , Propano/uso terapéutico , Vancomicina/farmacología
10.
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
11.
Anaerobe ; 41: 37-43, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27180006

RESUMEN

Alteration of the gut microbial community structure and function through antibiotic use increases susceptibility to colonization by Clostridium difficile and other enteric pathogens. However, the mechanisms that mediate colonization resistance remain elusive. As the leading definable cause of infectious diarrhea, toxigenic C. difficile represents a burden for patients and health care systems, underscoring the need for better diagnostics and treatment strategies. Next-generation sequence data has increased our understanding of how the gut microbiota is influenced by many factors including diet, disease, aging and drugs. However, a microbial-based biomarker differentiating C. difficile infection from antibiotic-associated diarrhea has not been identified. Metabolomics profiling, which is highly responsive to changes in physiological conditions, have shown promise in differentiating subtle disease phenotypes that exhibit a nearly identical microbiome community structure, suggesting metabolite-based biomarkers may be an ideal diagnostic for identifying patients with CDI. This review focuses on the current understanding of structural and functional changes to the gut microbiota during C. difficile infection obtained from studies assessing the microbiome and metabolome of samples from patients and murine models.


Asunto(s)
Clostridioides difficile/fisiología , Enterocolitis Seudomembranosa/microbiología , Microbioma Gastrointestinal/inmunología , Animales , Susceptibilidad a Enfermedades , Enterocolitis Seudomembranosa/inmunología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Interacciones Huésped-Patógeno , Humanos , Metaboloma
12.
Anaerobe ; 41: 51-57, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27180657

RESUMEN

With the end of the golden era of antibiotic discovery, the emergence of a new post-antibiotic age threatens to thrust global health and modern medicine back to the pre-antibiotic era. Antibiotic overuse has resulted in the natural evolution and selection of multi-drug resistant bacteria. One major public health threat, Clostridium difficile, is now the single leading cause of hospital-acquired bacterial infections and is by far the most deadly enteric pathogen for the U.S. POPULATION: Due to the high morbidity and mortality and increasing incidence that coincides with antibiotic use, non-traditional therapeutics are ideal alternatives to current treatment methods and also provide an avenue towards prevention. Despite the need for alternative therapies to antibiotics and the safety of most probiotics on the market, researchers are inundated with regulatory issues that hinder the translational science required to push these therapies forward. This review discusses the regulatory challenges of probiotic research, expert opinion regarding the application of probiotics to C. difficile infection and the efficacy of probiotics in preventing this disease.


Asunto(s)
Clostridioides difficile/fisiología , Enterocolitis Seudomembranosa/prevención & control , Probióticos/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Humanos
13.
Anaerobe ; 40: 54-7, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27180007

RESUMEN

Lifeway(®) kefir, a fermented milk product containing 12 probiotic organisms, is reported to show promise as an alternative to fecal microbiota transplantation for recurrent Clostridium difficile infection (CDI). We employed a murine CDI model to study the probiotic protective mechanisms and unexpectedly determined that kefir drastically increased disease severity. Our results emphasize the need for further independent clinical testing of kefir as alternative therapy in recurrent CDI.


Asunto(s)
Clostridioides difficile/efectos de los fármacos , Enterocolitis Seudomembranosa/patología , Kéfir/efectos adversos , Probióticos/efectos adversos , Animales , Clostridioides difficile/crecimiento & desarrollo , Clostridioides difficile/patogenicidad , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Enterocolitis Seudomembranosa/microbiología , Enterocolitis Seudomembranosa/mortalidad , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Índice de Severidad de la Enfermedad , Análisis de Supervivencia
14.
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
15.
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
16.
Trop Med Int Health ; 18(7): 879-86, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23600592

RESUMEN

OBJECTIVE: We aimed to improve paediatric inpatient surveillance at a busy referral hospital in Malawi with two new programmes: (i) the provision of vital sign equipment and implementation of an inpatient triage programme (ITAT) that includes a simplified paediatric severity-of-illness score, and (ii) task shifting ITAT to a new cadre of healthcare workers called 'vital sign assistants' (VSAs). METHODS: This study, conducted on the paediatric inpatient ward of a large referral hospital in Malawi, was divided into three phases, each lasting 4 weeks. In Phase A, we collected baseline data. In Phase B, we provided three new automated vital sign poles and implemented ITAT with current hospital staff. In Phase C, VSAs were introduced and performed ITAT. Our primary outcome measures were the number of vital sign assessments performed and clinician notifications to reassess patients with high ITAT scores. RESULTS: We enrolled 3994 patients who received 5155 vital sign assessments. Assessment frequency was equal between Phases A (0.67 assessments/patient) and B (0.61 assessments/patient), but increased 3.6-fold in Phase C (2.44 assessments/patient, P < 0.001). Clinician notifications increased from Phases A (84) and B (113) to Phase C (161, P = 0.002). Inpatient mortality fell from Phase A (9.3%) to Phases B (5.7) and C (6.9%). CONCLUSION: ITAT with VSAs improved vital sign assessments and nearly doubled clinician notifications of patients needing further assessment due to high ITAT scores, while equipment alone made no difference. Task shifting ITAT to VSAs may improve outcomes in paediatric hospitals in the developing world.


Asunto(s)
Técnicos Medios en Salud , Hospitalización , Hospitales , Índice de Severidad de la Enfermedad , Triaje , Signos Vitales , Niño Hospitalizado , Preescolar , Comunicación , Femenino , Personal de Salud , Mortalidad Hospitalaria , Humanos , Lactante , Malaui/epidemiología , Masculino , Mejoramiento de la Calidad , Derivación y Consulta , Triaje/métodos , Triaje/organización & administración , Triaje/normas , Carga de Trabajo
17.
Nat Protoc ; 18(2): 490-529, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36352124

RESUMEN

Interest in the communication between the gastrointestinal tract and central nervous system, known as the gut-brain axis, has prompted the development of quantitative analytical platforms to analyze microbe- and host-derived signals. This protocol enables investigations into connections between microbial colonization and intestinal and brain neurotransmitters and contains strategies for the comprehensive evaluation of metabolites in in vitro (organoids) and in vivo mouse model systems. Here we present an optimized workflow that includes procedures for preparing these gut-brain axis model systems: (stage 1) growth of microbes in defined media; (stage 2) microinjection of intestinal organoids; and (stage 3) generation of animal models including germ-free (no microbes), specific-pathogen-free (complete gut microbiota) and specific-pathogen-free re-conventionalized (germ-free mice associated with a complete gut microbiota from a specific-pathogen-free mouse), and Bifidobacterium dentium and Bacteroides ovatus mono-associated mice (germ-free mice colonized with a single gut microbe). We describe targeted liquid chromatography-tandem mass spectrometry-based metabolomics methods for analyzing microbially derived short-chain fatty acids and neurotransmitters from these samples. Unlike other protocols that commonly examine only stool samples, this protocol includes bacterial cultures, organoid cultures and in vivo samples, in addition to monitoring the metabolite content of stool samples. The incorporation of three experimental models (microbes, organoids and animals) enhances the impact of this protocol. The protocol requires 3 weeks of murine colonization with microbes and ~1-2 weeks for liquid chromatography-tandem mass spectrometry-based instrumental and quantitative analysis, and sample post-processing and normalization.


Asunto(s)
Eje Cerebro-Intestino , Espectrometría de Masas en Tándem , Animales , Ratones , Cromatografía Liquida , Vida Libre de Gérmenes , Metabolómica/métodos , Bacterias , Mamíferos , Organoides
18.
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
19.
iScience ; 25(5): 104158, 2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35494230

RESUMEN

Gut microbes can synthesize multiple neuro-active metabolites. We profiled neuro-active compounds produced by the gut commensal Bacteroides ovatus in vitro and in vivo by LC-MS/MS. We found that B. ovatus generates acetic acid, propionic acid, isobutyric acid, and isovaleric acid. In vitro, B. ovatus consumed tryptophan and glutamate and synthesized the neuro-active compounds glutamine and GABA. Consistent with our LC-MS/MS-based in vitro data, we observed elevated levels of acetic acid, propionic acid, isobutyric acid, and isovaleric acid in the intestines of B. ovatus mono-associated mice compared with germ-free controls. B. ovatus mono-association also increased the concentrations of intestinal GABA and decreased the concentrations of tryptophan and glutamine compared with germ-free controls. Computational network analysis revealed unique links between SCFAs, neuro-active compounds, and colonization status. These results highlight connections between microbial colonization and intestinal neurotransmitter concentrations, suggesting that B. ovatus selectively influences the presence of intestinal neurotransmitters.

20.
Microb Cell Fact ; 10: 55, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21777454

RESUMEN

BACKGROUND: Lactobacillus reuteri harbors the genes responsible for glycerol utilization and vitamin B12 synthesis within a genetic island phylogenetically related to gamma-Proteobacteria. Within this island, resides a gene (lreu_1750) that based on its genomic context has been suggested to encode the regulatory protein PocR and presumably control the expression of the neighboring loci. However, this functional assignment is not fully supported by sequence homology, and hitherto, completely lacks experimental confirmation. RESULTS: In this contribution, we have overexpressed and inactivated the gene encoding the putative PocR in L. reuteri. The comparison of these strains provided metabolic and transcriptional evidence that this regulatory protein controls the expression of the operons encoding glycerol utilization and vitamin B12 synthesis. CONCLUSIONS: We provide clear experimental evidence for assigning Lreu_1750 as PocR in Lactobacillus reuteri. Our genome-wide transcriptional analysis further identifies the loci contained in the PocR regulon. The findings reported here could be used to improve the production-yield of vitamin B12, 1,3-propanediol and reuterin, all industrially relevant compounds.


Asunto(s)
Proteínas Bacterianas/metabolismo , Glicerol/farmacología , Limosilactobacillus reuteri/metabolismo , Factores de Transcripción/metabolismo , Vitamina B 12/biosíntesis , Secuencia de Aminoácidos , Proteínas Bacterianas/clasificación , Perfilación de la Expresión Génica , Sitios Genéticos , Gliceraldehído/análogos & derivados , Gliceraldehído/metabolismo , Concentración de Iones de Hidrógeno , Datos de Secuencia Molecular , Filogenia , Propano/metabolismo , Glicoles de Propileno/metabolismo , Alineación de Secuencia , Factores de Transcripción/clasificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA