Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Hepatol ; 75(6): 1452-1464, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34453966

RESUMEN

Hepatic encephalopathy (HE) is a complication of cirrhosis characterised by neuropsychiatric and motor dysfunction. Microbiota-host interactions play an important role in HE pathogenesis. Therapies targeting microbial community composition and function have been explored for the treatment of HE. Prebiotics, probiotics and faecal microbiota transplant (FMT) have been used with the aim of increasing the abundance of potentially beneficial taxa, while antibiotics have been used to decrease the abundance of potentially harmful taxa. Other microbiome therapeutics, including postbiotics and absorbents, have been used to target microbial products. Microbiome-targeted therapies for HE have had some success, notably lactulose and rifaximin, with probiotics and FMT also showing promise. However, there remain several challenges to the effective application of microbiome therapeutics in HE, including the resilience of the microbiome to sustainable change and unpredictable clinical outcomes from microbiota alterations. Future work in this space should focus on rigorous trial design, microbiome therapy selection, and a personalised approach to HE.


Asunto(s)
Encefalopatía Hepática/tratamiento farmacológico , Microbiota/efectos de los fármacos , Trasplante de Microbiota Fecal/métodos , Trasplante de Microbiota Fecal/estadística & datos numéricos , Interacciones Microbiota-Huesped/efectos de los fármacos , Humanos , Prebióticos/administración & dosificación , Probióticos/uso terapéutico
2.
mBio ; 12(2)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33785619

RESUMEN

Clostridioides difficile, a Gram-positive, spore-forming bacterium, is the primary cause of infectious nosocomial diarrhea. Antibiotics are a major risk factor for C. difficile infection (CDI), as they disrupt the gut microbial community, enabling increased germination of spores and growth of vegetative C. difficile To date, the only single-species bacterial preparation that has demonstrated efficacy in reducing recurrent CDI in humans is nontoxigenic C. difficile Using multiple infection models, we determined that precolonization with a less virulent strain is sufficient to protect from challenge with a lethal strain of C. difficile, surprisingly even in the absence of adaptive immunity. Additionally, we showed that protection is dependent on high levels of colonization by the less virulent strain and that it is mediated by exclusion of the invading strain. Our results suggest that reduction of amino acids, specifically glycine following colonization by the first strain of C. difficile, is sufficient to decrease germination of the second strain, thereby limiting colonization by the lethal strain.IMPORTANCE Antibiotic-associated colitis is often caused by infection with the bacterium Clostridioides difficile In this study, we found that reduction of the amino acid glycine by precolonization with a less virulent strain of C. difficile is sufficient to decrease germination of a second strain. This finding demonstrates that the axis of competition for nutrients can include multiple life stages. This work is important, as it is the first to identify a possible mechanism through which precolonization with C. difficile, a current clinical therapy, provides protection from reinfection. Furthermore, our work suggests that targeting nutrients utilized by all life stages could be an improved strategy for bacterial therapeutics that aim to restore colonization resistance in the gut.


Asunto(s)
Antibiosis , Terapia Biológica , Clostridioides difficile/fisiología , Infecciones por Clostridium/prevención & control , Animales , Clostridioides difficile/clasificación , Clostridioides difficile/crecimiento & desarrollo , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/microbiología , Femenino , Glicina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microbiota , Esporas Bacterianas/clasificación , Esporas Bacterianas/genética , Esporas Bacterianas/crecimiento & desarrollo , Esporas Bacterianas/fisiología , Virulencia
3.
mSphere ; 5(1)2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31915217

RESUMEN

Dietary fiber provides a variety of microbiota-mediated benefits ranging from anti-inflammatory metabolites to pathogen colonization resistance. A healthy gut microbiota protects against Clostridioides difficile colonization. Manipulation of these microbes through diet may increase colonization resistance to improve clinical outcomes. The primary objective of this study was to identify how the dietary fiber xanthan gum affects the microbiota and C. difficile colonization. We added 5% xanthan gum to the diet of C57BL/6 mice and examined its effect on the microbiota through 16S rRNA gene amplicon sequencing and short-chain fatty acid analysis. Following either cefoperazone or an antibiotic cocktail administration, we challenged mice with C. difficile and measured colonization by monitoring the CFU. Xanthan gum administration is associated with increases in fiber-degrading taxa and short-chain fatty acid concentrations. However, by maintaining both the diversity and absolute abundance of the microbiota during antibiotic treatment, the protective effects of xanthan gum administration on the microbiota were more prominent than the enrichment of these fiber-degrading taxa. As a result, mice that were on the xanthan gum diet experienced limited to no C. difficile colonization. Xanthan gum administration alters mouse susceptibility to C. difficile colonization by maintaining the microbiota during antibiotic treatment. While antibiotic-xanthan gum interactions are not well understood, xanthan gum has previously been used to bind drugs and alter their pharmacokinetics. Thus, xanthan gum may alter the activity of the oral antibiotics used to make the microbiota susceptible. Future research should further characterize how this and other common dietary fibers interact with drugs.IMPORTANCE A healthy gut bacterial community benefits the host by breaking down dietary nutrients and protecting against pathogens. Clostridioides difficile capitalizes on the absence of this community to cause diarrhea and inflammation. Thus, a major clinical goal is to find ways to increase resistance to C. difficile colonization by either supplementing with bacteria that promote resistance or a diet to enrich for those already present in the gut. In this study, we describe an interaction between xanthan gum, a human dietary additive, and the microbiota resulting in an altered gut environment that is protective against C. difficile colonization.


Asunto(s)
Antibacterianos/uso terapéutico , Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/prevención & control , Fibras de la Dieta/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Polisacáridos Bacterianos/administración & dosificación , Animales , Cefoperazona/uso terapéutico , Infecciones por Clostridium/microbiología , Suplementos Dietéticos , Susceptibilidad a Enfermedades , Heces/microbiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Organismos Libres de Patógenos Específicos
4.
J Nutr ; 149(7): 1170-1179, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31051496

RESUMEN

BACKGROUND: The intestinal microbiome is an important determinant of inflammatory balance in the colon that may affect response to dietary agents. OBJECTIVE: This is a secondary analysis of a clinical trial, the Fish Oil Study, to determine whether interindividual differences in colonic bacteria are associated with variability in the reduction of colonic prostaglandin E2 (PGE2) concentrations after personalized supplementation with ω-3 (n-3) fatty acids. METHODS: Forty-seven healthy adults (17 men, 30 women, ages 26-75 y) provided biopsy samples of colonic mucosa and luminal stool brushings before and after personalized ω-3 fatty acid supplementation that was based on blood fatty acid responses. Samples were analyzed using 16S ribosomal RNA sequencing. The data analyses focused on changes in bacterial community diversity. Linear regression was used to evaluate factors that predict a reduction in colonic PGE2. RESULTS: At baseline, increased bacterial diversity, as measured by the Shannon and Inverse Simpson indexes in both biopsy and luminal brushing samples, was positively correlated with dietary fiber intakes and negatively correlated with fat intakes. Dietary supplementation with ω-3 fatty acids increased the Yue and Clayton community dis-similarity index between the microbiome in luminal brushings and colon biopsy samples post-supplementation (P = 0.015). In addition, there was a small group of individuals with relatively high Prevotella abundance who were resistant to the anti-inflammatory effects of ω-3 fatty acid supplementation. In linear regression analyses, increases in diversity of the bacteria in the luminal brushing samples, but not in the biopsy samples, were significant predictors of lower colonic PGE2 concentrations post-supplementation in models that included baseline PGE2, baseline body mass index, and changes in colonic eicosapentaenoic acid-to-arachidonic acid ratios. The changes in bacterial diversity contributed to 6-8% of the interindividual variance in change in colonic PGE2 (P = 0.001). CONCLUSIONS: Dietary supplementation with ω-3 fatty acids had little effect on intestinal bacteria in healthy humans; however, an increase in diversity in the luminal brushings significantly predicted reductions in colonic PGE2. This trial was registered at www.clinicaltrials.gov as NCT01860352.


Asunto(s)
Bacterias/clasificación , Colon/microbiología , Suplementos Dietéticos , Dinoprostona/metabolismo , Ácidos Grasos Omega-3/administración & dosificación , Adulto , Anciano , Colon/metabolismo , Femenino , Microbioma Gastrointestinal , Humanos , Masculino , Persona de Mediana Edad
5.
mSphere ; 4(2)2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30894434

RESUMEN

Between October 2016 and June 2017, a C57BL/6J mouse colony that was undergoing a pre- and perinatal methyl donor supplementation diet intervention to study the impact of parental nutrition on offspring susceptibility to disease was found to suffer from an epizootic of unexpected deaths. Necropsy revealed the presence of severe colitis, and further investigation linked these outbreak deaths to a Clostridium difficile strain of ribotype 027 that we term 16N203. C. difficile infection (CDI) is associated with antibiotic use in humans. Current murine models of CDI rely on antibiotic pretreatment to establish clinical phenotypes. In this report, the C. difficile outbreak occurs in F1 mice linked to alterations in the parental diet. The diagnosis of CDI in the affected mice was confirmed by cecal/colonic histopathology, the presence of C. difficile bacteria in fecal/colonic culture, and detection of C. difficile toxins. F1 mice from parents fed the methyl supplementation diet also had significantly reduced survival (P < 0.0001) compared with F1 mice from parents fed the control diet. When we tested the 16N203 outbreak strain in an established mouse model of antibiotic-induced CDI, we confirmed that this strain is pathogenic. Our serendipitous observations from this spontaneous outbreak of C. difficile in association with a pre- and perinatal methyl donor diet suggest the important role that diet may play in host defense and CDI risk factors.IMPORTANCEClostridium difficile infection (CDI) has become the leading cause of infectious diarrhea in hospitals worldwide, owing its preeminence to the emergence of hyperendemic strains, such as ribotype 027 (RT027). A major CDI risk factor is antibiotic exposure, which alters gut microbiota, resulting in the loss of colonization resistance. Current murine models of CDI also depend on pretreatment of animals with antibiotics to establish disease. The outbreak that we report here is unique in that the CDI occurred in mice with no antibiotic exposure and is associated with a pre- and perinatal methyl supplementation donor diet intervention study. Our investigation subsequently reveals that the outbreak strain that we term 16N203 is an RT027 strain, and this isolated strain is also pathogenic in an established murine model of CDI (with antibiotics). Our report of this spontaneous outbreak offers additional insight into the importance of environmental factors, such as diet, and CDI susceptibility.


Asunto(s)
Infecciones por Clostridium/etiología , Dieta/efectos adversos , Suplementos Dietéticos/efectos adversos , Brotes de Enfermedades , Animales , Betaína/metabolismo , Colina/metabolismo , Clostridioides difficile/aislamiento & purificación , Clostridioides difficile/patogenicidad , Susceptibilidad a Enfermedades/etiología , Femenino , Masculino , Metionina/metabolismo , Ratones , Ratones Endogámicos C57BL , Nutrición Parenteral/métodos , Ribotipificación , Factores de Riesgo
6.
Infect Dis Clin North Am ; 29(1): 109-22, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25677705

RESUMEN

This article discusses the use of fecal microbiota transplantation (FMT) for the treatment of recurrent Clostridium difficile infection (CDI). The disruption of the normal gut microbiota is central to the pathogenesis of CDI, and disruption persists in recurrent disease. The use of FMT for recurrent CDI is characterized by a high response rate and short term safety is excellent, although the long-term effects of FMT are as yet unknown.


Asunto(s)
Terapia Biológica/métodos , Clostridioides difficile , Infecciones por Clostridium/terapia , Heces/microbiología , Enfermedades Intestinales/microbiología , Infecciones por Clostridium/microbiología , Manejo de la Enfermedad , Tracto Gastrointestinal/microbiología , Humanos , Enfermedades Intestinales/terapia , Microbiota
7.
mBio ; 5(3): e00893-14, 2014 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-24939885

RESUMEN

UNLABELLED: Clostridium difficile infection is one of the most common health care-associated infections, and up to 40% of patients suffer from recurrence of disease following standard antibiotic therapy. Recently, fecal microbiota transplantation (FMT) has been successfully used to treat recurrent C. difficile infection. It is hypothesized that FMT aids in recovery of a microbiota capable of colonization resistance to C. difficile. However, it is not fully understood how this occurs. Here we investigated changes in the fecal microbiota structure following FMT in patients with recurrent C. difficile infection, and imputed a hypothetical functional profile based on the 16S rRNA profile using a predictive metagenomic tool. Increased relative abundance of Bacteroidetes and decreased abundance of Proteobacteria were observed following FMT. The fecal microbiota of recipients following transplantation was more diverse and more similar to the donor profile than the microbiota prior to transplantation. Additionally, we observed differences in the imputed metagenomic profile. In particular, amino acid transport systems were overrepresented in samples collected prior to transplantation. These results suggest that functional changes accompany microbial structural changes following this therapy. Further identification of the specific community members and functions that promote colonization resistance may aid in the development of improved treatment methods for C. difficile infection. IMPORTANCE: Within the last decade, Clostridium difficile infection has surpassed other bacterial infections to become the leading cause of nosocomial infections. Antibiotic use, which disrupts the gut microbiota and its capability in providing colonization resistance against C. difficile, is a known risk factor in C. difficile infection. In particular, recurrent C. difficile remains difficult to treat with standard antibiotic therapy. Fecal microbiota transplantation (FMT) has provided a successful treatment method for some patients with recurrent C. difficile infection, but its mechanism and long-term effects remain unknown. Our results provide insight into the structural and potential metabolic changes that occur following FMT, which may aid in the development of new treatment methods for C. difficile infection.


Asunto(s)
Bacterias/aislamiento & purificación , Terapia Biológica , Clostridioides difficile/fisiología , Infecciones por Clostridium/terapia , Heces/microbiología , Microbiota , Adulto , Anciano , Anciano de 80 o más Años , Bacterias/clasificación , Bacterias/genética , Infecciones por Clostridium/microbiología , Femenino , Tracto Gastrointestinal/microbiología , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Filogenia
8.
Gastroenterology ; 146(6): 1547-53, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24503131

RESUMEN

Antibiotic-associated infection with the bacterial pathogen Clostridium difficile is a major cause of morbidity and increased health care costs. C difficile infection follows disruption of the indigenous gut microbiota by antibiotics. Antibiotics create an environment within the intestine that promotes C difficile spore germination, vegetative growth, and toxin production, leading to epithelial damage and colitis. Studies of patients with C difficile infection and animal models have shown that the indigenous microbiota can inhibit expansion and persistence of C difficile. Although the specific mechanisms of these processes are not known, they are likely to interfere with key aspects of the pathogen's physiology, including spore germination and competitive growth. Increasing our understanding of how the intestinal microbiota manage C difficile could lead to better means of controlling this important nosocomial pathogen.


Asunto(s)
Clostridioides difficile/crecimiento & desarrollo , Enterocolitis Seudomembranosa/microbiología , Intestinos/microbiología , Microbiota , Animales , Antibacterianos/efectos adversos , Ácidos y Sales Biliares/metabolismo , Terapia Biológica/métodos , Clostridioides difficile/metabolismo , Clostridioides difficile/patogenicidad , Modelos Animales de Enfermedad , Disbiosis , Enterocolitis Seudomembranosa/metabolismo , Enterocolitis Seudomembranosa/prevención & control , Heces/microbiología , Interacciones Huésped-Patógeno , Humanos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Microbiota/efectos de los fármacos , Probióticos/uso terapéutico
10.
Curr Opin Gastroenterol ; 29(6): 628-32, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24100717

RESUMEN

PURPOSE OF REVIEW: The use of faecal microbiota transplantation (FMT) as treatment for recurrent Clostridium difficile infection (CDI) has increased rapidly over the past few years. In this review, we highlight clinical studies of FMT for treatment of recurrent CDI and discuss the safety, standardization and future of this treatment option. The major risk factor for CDI is prior antibiotic use, which results in an altered state of the gut microbiota characterized by decreased microbial diversity. This altered gut microbiota increases the patient's susceptibility to CDI. In patients with recurrent CDI, the microbiota remains in a state with decreased diversity, and FMT from a healthy individual restores the gut microbiota and subsequently colonization resistance against the pathogen. RECENT FINDINGS: Recent studies have shown the success rate for FMT as treatment for recurrent CDI being greater than 90%. Standardized, frozen preparations of faeces can be used, which increases the availability of faeces for FMT and decreases the cost of screening individual donors. In addition, there have been recent advances in identifying a defined microbial community isolated from faeces that can restore colonization resistance against C. difficile. SUMMARY: The use of FMT is a successful treatment for recurrent CDI when primary treatment options have failed. However, more work needs to define potential long-term consequences of this treatment and understand how specific members of the gut microbiota can restore colonization resistance against C. difficile.


Asunto(s)
Terapia Biológica/métodos , Enterocolitis Seudomembranosa/terapia , Heces/microbiología , Microbiota , Terapia Biológica/efectos adversos , Terapia Biológica/normas , Enterocolitis Seudomembranosa/microbiología , Humanos , Intestinos/microbiología , Ensayos Clínicos Controlados Aleatorios como Asunto , Recurrencia
11.
Teach Learn Med ; 22(1): 3-7, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20391276

RESUMEN

BACKGROUND AND PURPOSE: The relationship between osteopathic (COMLEX) and allopathic (USMLE) exam scores in not well described. We sought to describe the relationship between COMLEX and USMLE scores for osteopathic medical students who reported scores for both exams during application to internal medicine residency. METHODS: Cross sectional study of 588 matched USMLE/COMLEX Step 1 scores and 241 matched Step 2 scores. Means, standard deviations, and pairwise correlations for matched scores were calculated. RESULTS: USMLE and COMLEX paired scores resulted in a Pearson's correlation of 0.85 for Step 1 scores and 0.79 for Step 2 scores. COMLEX means were 560 and 561 for Level 1 and 2 and USMLE means were 209 and 215 for Step 1 and 2. CONCLUSIONS: USMLE and COMLEX scores are strongly related for internal medicine residency applicants who took both exams. Our sample performed approximately 0.8 standard deviation higher that the national COMLEX means on both Level 1 and 2 but at the national mean on USMLE Step 1 and 2. The variation in scoring methodology and potential differences between the study sample and individual applicant characteristics suggesteject caution should be taken in using a given COMLEX score as a predictor of performance on the USMLE.


Asunto(s)
Evaluación Educacional/métodos , Medicina Osteopática/educación , Estudiantes de Medicina , Acreditación , Estudios Transversales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA