Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nature ; 517(7533): 205-8, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25337874

RESUMEN

The gastrointestinal tracts of mammals are colonized by hundreds of microbial species that contribute to health, including colonization resistance against intestinal pathogens. Many antibiotics destroy intestinal microbial communities and increase susceptibility to intestinal pathogens. Among these, Clostridium difficile, a major cause of antibiotic-induced diarrhoea, greatly increases morbidity and mortality in hospitalized patients. Which intestinal bacteria provide resistance to C. difficile infection and their in vivo inhibitory mechanisms remain unclear. Here we correlate loss of specific bacterial taxa with development of infection, by treating mice with different antibiotics that result in distinct microbiota changes and lead to varied susceptibility to C. difficile. Mathematical modelling augmented by analyses of the microbiota of hospitalized patients identifies resistance-associated bacteria common to mice and humans. Using these platforms, we determine that Clostridium scindens, a bile acid 7α-dehydroxylating intestinal bacterium, is associated with resistance to C. difficile infection and, upon administration, enhances resistance to infection in a secondary bile acid dependent fashion. Using a workflow involving mouse models, clinical studies, metagenomic analyses, and mathematical modelling, we identify a probiotic candidate that corrects a clinically relevant microbiome deficiency. These findings have implications for the rational design of targeted antimicrobials as well as microbiome-based diagnostics and therapeutics for individuals at risk of C. difficile infection.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Clostridioides difficile/fisiología , Susceptibilidad a Enfermedades/microbiología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Microbiota/fisiología , Animales , Antibacterianos/farmacología , Evolución Biológica , Clostridioides difficile/efectos de los fármacos , Clostridium/metabolismo , Colitis/metabolismo , Colitis/microbiología , Colitis/prevención & control , Colitis/terapia , Heces/microbiología , Femenino , Humanos , Intestinos/efectos de los fármacos , Metagenoma/genética , Ratones , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Microbiota/genética , Simbiosis
2.
Blood ; 124(7): 1174-82, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-24939656

RESUMEN

Highly diverse bacterial populations inhabit the gastrointestinal tract and modulate host inflammation and promote immune tolerance. In allogeneic hematopoietic stem cell transplantation (allo-HSCT), the gastrointestinal mucosa is damaged, and colonizing bacteria are impacted, leading to an impaired intestinal microbiota with reduced diversity. We examined the impact of intestinal diversity on subsequent mortality outcomes following transplantation. Fecal specimens were collected from 80 recipients of allo-HSCT at the time of stem cell engraftment. Bacterial 16S rRNA gene sequences were characterized, and microbial diversity was estimated using the inverse Simpson index. Subjects were classified into high, intermediate, and low diversity groups and assessed for differences in outcomes. Mortality outcomes were significantly worse in patients with lower intestinal diversity; overall survival at 3 years was 36%, 60%, and 67% for low, intermediate, and high diversity groups, respectively (P = .019, log-rank test). Low diversity showed a strong effect on mortality after multivariate adjustment for other clinical predictors (transplant related mortality: adjusted hazard ratio, 5.25; P = .014). In conclusion, the diversity of the intestinal microbiota at engraftment is an independent predictor of mortality in allo-HSCT recipients. These results indicate that the intestinal microbiota may be an important factor in the success or failure in allo-HSCT.


Asunto(s)
Tracto Gastrointestinal/microbiología , Variación Genética , Trasplante de Células Madre Hematopoyéticas/métodos , Microbiota/genética , Adulto , Bacterias/clasificación , Bacterias/genética , Heces/microbiología , Femenino , Trasplante de Células Madre Hematopoyéticas/mortalidad , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Análisis Multivariante , Evaluación de Resultado en la Atención de Salud/métodos , Evaluación de Resultado en la Atención de Salud/estadística & datos numéricos , Filogenia , Pronóstico , Modelos de Riesgos Proporcionales , ARN Ribosómico 16S/genética , Tasa de Supervivencia , Trasplante Homólogo
3.
J Infect Dis ; 212(10): 1656-65, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25920320

RESUMEN

Antibiotic administration disrupts the intestinal microbiota, increasing susceptibility to pathogens such as Clostridium difficile. Metronidazole or oral vancomycin can cure C. difficile infection, and administration of these agents to prevent C. difficile infection in high-risk patients, although not sanctioned by Infectious Disease Society of America guidelines, has been considered. The relative impacts of metronidazole and vancomycin on the intestinal microbiota and colonization resistance are unknown. We investigated the effect of brief treatment with metronidazole and/or oral vancomycin on susceptibility to C. difficile, vancomycin-resistant Enterococcus, carbapenem-resistant Klebsiella pneumoniae, and Escherichia coli infection in mice. Although metronidazole resulted in transient loss of colonization resistance, oral vancomycin markedly disrupted the microbiota, leading to prolonged loss of colonization resistance to C. difficile infection and dense colonization by vancomycin-resistant Enterococcus, K. pneumoniae, and E. coli. Our results demonstrate that vancomycin, and to a lesser extent metronidazole, are associated with marked intestinal microbiota destruction and greater risk of colonization by nosocomial pathogens.


Asunto(s)
Antiinfecciosos/administración & dosificación , Infecciones Bacterianas/inmunología , Resistencia a la Enfermedad/efectos de los fármacos , Metronidazol/administración & dosificación , Vancomicina/administración & dosificación , Animales , Antiinfecciosos/efectos adversos , Infecciones Bacterianas/microbiología , Clostridioides difficile/aislamiento & purificación , Modelos Animales de Enfermedad , Escherichia coli/aislamiento & purificación , Femenino , Klebsiella pneumoniae/aislamiento & purificación , Metronidazol/efectos adversos , Ratones Endogámicos C57BL , Vancomicina/efectos adversos , Enterococos Resistentes a la Vancomicina/aislamiento & purificación
4.
Biol Blood Marrow Transplant ; 21(8): 1373-83, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25977230

RESUMEN

The relationship between intestinal microbiota composition and acute graft-versus-host disease (GVHD) after allogeneic blood/marrow transplantation (allo-BMT) is not well understood. Intestinal bacteria have long been thought to contribute to GVHD pathophysiology, but recent animal studies in nontransplant settings have found that anti-inflammatory effects are mediated by certain subpopulations of intestinal commensals. Hypothesizing that a more nuanced relationship may exist between the intestinal bacteria and GVHD, we evaluated the fecal bacterial composition of 64 patients 12 days after BMT. We found that increased bacterial diversity was associated with reduced GVHD-related mortality. Furthermore, harboring increased amounts of bacteria belonging to the genus Blautia was associated with reduced GVHD lethality in this cohort and was confirmed in another independent cohort of 51 patients from the same institution. Blautia abundance was also associated with improved overall survival. We evaluated the abundance of Blautia with respect to clinical factors and found that loss of Blautia was associated with treatment with antibiotics that inhibit anaerobic bacteria and receiving total parenteral nutrition for longer durations. We conclude that increased abundance of commensal bacteria belonging to the Blautia genus is associated with reduced lethal GVHD and improved overall survival.


Asunto(s)
Bacterias/metabolismo , Enfermedad Injerto contra Huésped/mortalidad , Intestinos/microbiología , Estudios de Cohortes , Femenino , Humanos , Mucosa Intestinal/metabolismo , Masculino , Factores de Riesgo , Análisis de Supervivencia
5.
J Am Heart Assoc ; : e033477, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39082397

RESUMEN

BACKGROUND: Self-care for adults with hypertension includes adherence to lifestyle behaviors and medication. For unpaid caregivers with hypertension, the burden of family caregiving may adversely impact self-care. We examined the association between caregiver strain and hypertension self-care among caregivers with hypertension. METHODS AND RESULTS: We included participants of the REGARDS (Reasons for Geographic and Racial Differences in Stroke) study who identified as caregivers and had hypertension. Caregiver strain, assessed by self-report, was categorized as "none/some" or "high." Hypertension self-care was assessed individually across 5 domains (Dietary Approaches to Stop Hypertension [DASH] diet, physical activity, alcohol use, cigarette smoking, and medication adherence) and a composite self-care score summing performance across them. The association between caregiver strain and hypertension self-care was examined with multivariable linear regression. Among the 2128 caregivers with hypertension, 18.1% reported high caregiver strain. Caregivers with high strain versus those with none/some were less adherent to the DASH diet (50.8% versus 38.9%, P<0.002), physically inactive (44.4% versus 36.2%, P<0.009), current smokers (19.7% versus 13.9%, P<0.004), and had lower overall self-care scores (6.6 [SD 1.7] versus 7.0 [SD 1.7], P<0.001). In an age-adjusted model, high caregiver strain was associated with worse hypertension self-care (ß=-0.37 [95% CI, -0.61 to -0.13]); this remained significant but was reduced in magnitude after adjustment for sociodemographics (ß=-0.35 [-0.59 to -0.11]), comorbidities (ß=-0.34 [-0.57 to -0.10]), caregiving intensity (ß=-0.34 [-0.59 to 0.10]), and psychological factors (ß=-0.26 [-0.51 to 0.00]). CONCLUSIONS: High caregiver strain was associated with worse hypertension self-care overall and across individual domains. Increased awareness of caregiver strain and its potential impact on hypertension self-care is warranted.

6.
Infect Immun ; 81(3): 965-73, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23319552

RESUMEN

Bacteria causing infections in hospitalized patients are increasingly antibiotic resistant. Classical infection control practices are only partially effective at preventing spread of antibiotic-resistant bacteria within hospitals. Because the density of intestinal colonization by the highly antibiotic-resistant bacterium vancomycin-resistant Enterococcus (VRE) can exceed 10(9) organisms per gram of feces, even optimally implemented hygiene protocols often fail. Decreasing the density of intestinal colonization, therefore, represents an important approach to limit VRE transmission. We demonstrate that reintroduction of a diverse intestinal microbiota to densely VRE-colonized mice eliminates VRE from the intestinal tract. While oxygen-tolerant members of the microbiota are ineffective at eliminating VRE, administration of obligate anaerobic commensal bacteria to mice results in a billionfold reduction in the density of intestinal VRE colonization. 16S rRNA gene sequence analysis of intestinal bacterial populations isolated from mice that cleared VRE following microbiota reconstitution revealed that recolonization with a microbiota that contains Barnesiella correlates with VRE elimination. Characterization of the fecal microbiota of patients undergoing allogeneic hematopoietic stem cell transplantation demonstrated that intestinal colonization with Barnesiella confers resistance to intestinal domination and bloodstream infection with VRE. Our studies indicate that obligate anaerobic bacteria belonging to the Barnesiella genus enable clearance of intestinal VRE colonization and may provide novel approaches to prevent the spread of highly antibiotic-resistant bacteria.


Asunto(s)
Bacteroidaceae/fisiología , Enterococcus faecium/efectos de los fármacos , Infecciones por Bacterias Grampositivas/terapia , Intestinos/microbiología , Resistencia a la Vancomicina , Animales , ADN Bacteriano , Femenino , Ratones , Ratones Endogámicos C57BL , ARN Bacteriano/genética , ARN Ribosómico 16S/genética
7.
Infect Immun ; 80(1): 62-73, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22006564

RESUMEN

Antibiotic-induced changes in the intestinal microbiota predispose mammalian hosts to infection with antibiotic-resistant pathogens. Clostridium difficile is a Gram-positive intestinal pathogen that causes colitis and diarrhea in patients following antibiotic treatment. Clindamycin predisposes patients to C. difficile colitis. Here, we have used Roche-454 16S rRNA gene pyrosequencing to longitudinally characterize the intestinal microbiota of mice following clindamycin treatment in the presence or absence of C. difficile infection. We show that a single dose of clindamycin markedly reduces the diversity of the intestinal microbiota for at least 28 days, with an enduring loss of ca. 90% of normal microbial taxa from the cecum. Loss of microbial complexity results in dramatic sequential expansion and contraction of a subset of bacterial taxa that are minor contributors to the microbial consortium prior to antibiotic treatment. Inoculation of clindamycin-treated mice with C. difficile (VPI 10463) spores results in rapid development of diarrhea and colitis, with a 4- to 5-day period of profound weight loss and an associated 40 to 50% mortality rate. Recovering mice resolve diarrhea and regain weight but remain highly infected with toxin-producing vegetative C. difficile bacteria and, in comparison to the acute stage of infection, have persistent, albeit ameliorated cecal and colonic inflammation. The microbiota of "recovered" mice remains highly restricted, and mice remain susceptible to C. difficile infection at least 10 days following clindamycin, suggesting that resolution of diarrhea and weight gain may result from the activation of mucosal immune defenses.


Asunto(s)
Antibacterianos/administración & dosificación , Bacterias/efectos de los fármacos , Clindamicina/administración & dosificación , Infecciones por Clostridium/inmunología , Colitis/inmunología , Susceptibilidad a Enfermedades , Tracto Gastrointestinal/metabolismo , Animales , Bacterias/clasificación , Bacterias/genética , Biodiversidad , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/mortalidad , Colitis/microbiología , Colitis/mortalidad , Diarrea/inmunología , Diarrea/microbiología , Diarrea/mortalidad , Femenino , Estudios Longitudinales , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ADN/métodos , Análisis de Supervivencia , Factores de Tiempo
8.
Clin Infect Dis ; 55(7): 905-14, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22718773

RESUMEN

BACKGROUND: Bacteremia is a frequent complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). It is unclear whether changes in the intestinal microbiota during allo-HSCT contribute to the development of bacteremia. We examined the microbiota of patients undergoing allo-HSCT, and correlated microbial shifts with the risk of bacteremia. METHODS: Fecal specimens were collected longitudinally from 94 patients undergoing allo-HSCT, from before transplant until 35 days after transplant. The intestinal microbiota was characterized by 454 pyrosequencing of the V1-V3 region of bacterial 16S ribosomal RNA genes. Microbial diversity was estimated by grouping sequences into operational taxonomic units and calculating the Shannon diversity index. Phylogenetic classification was obtained using the Ribosomal Database Project classifier. Associations of the microbiota with clinical predictors and outcomes were evaluated. RESULTS: During allo-HSCT, patients developed reduced diversity, with marked shifts in bacterial populations inhabiting the gut. Intestinal domination, defined as occupation of at least 30% of the microbiota by a single predominating bacterial taxon, occurred frequently. Commonly encountered dominating organisms included Enterococcus, Streptococcus, and various Proteobacteria. Enterococcal domination was increased 3-fold by metronidazole administration, whereas domination by Proteobacteria was reduced 10-fold by fluoroquinolone administration. As a predictor of outcomes, enterococcal domination increased the risk of Vancomycin-resistant Enterococcus bacteremia 9-fold, and proteobacterial domination increased the risk of gram-negative rod bacteremia 5-fold. CONCLUSIONS: During allo-HSCT, the diversity and stability of the intestinal flora are disrupted, resulting in domination by bacteria associated with subsequent bacteremia. Assessment of fecal microbiota identifies patients at highest risk for bloodstream infection during allo-HCST.


Asunto(s)
Bacteriemia/epidemiología , Bacteriemia/microbiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Adulto , Anciano , Biodiversidad , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Ribosómico/química , ADN Ribosómico/genética , Heces/microbiología , Femenino , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Filogenia , ARN Ribosómico 16S/genética , Factores de Riesgo , Análisis de Secuencia de ADN
9.
Nat Commun ; 7: 10391, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26837003

RESUMEN

The composition of the intestinal microbiota influences the development of inflammatory disorders. However, associating inflammatory diseases with specific microbial members of the microbiota is challenging, because clinically detectable inflammation and its treatment can alter the microbiota's composition. Immunologic checkpoint blockade with ipilimumab, a monoclonal antibody that blocks cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) signalling, is associated with new-onset, immune-mediated colitis. Here we conduct a prospective study of patients with metastatic melanoma undergoing ipilimumab treatment and correlate the pre-inflammation faecal microbiota and microbiome composition with subsequent colitis development. We demonstrate that increased representation of bacteria belonging to the Bacteroidetes phylum is correlated with resistance to the development of checkpoint-blockade-induced colitis. Furthermore, a paucity of genetic pathways involved in polyamine transport and B vitamin biosynthesis is associated with an increased risk of colitis. Identification of these biomarkers may enable interventions to reduce the risk of inflammatory complications following cancer immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Colitis/microbiología , Microbioma Gastrointestinal/genética , Melanoma/tratamiento farmacológico , ARN Ribosómico 16S/genética , Neoplasias Cutáneas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Bacteroidaceae , Bacteroidetes/genética , Bacteroidetes/aislamiento & purificación , Colitis/inducido químicamente , Femenino , Humanos , Ipilimumab , Masculino , Melanoma/secundario , Persona de Mediana Edad , Estudios Prospectivos , Factores de Riesgo , Análisis de Secuencia de ARN , Neoplasias Cutáneas/patología
10.
PLoS One ; 9(3): e90158, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24662889

RESUMEN

Clostridium difficile infection (CDI) is frequently diagnosed in recipients of allogeneic hematopoietic stem cell transplantation (allo-HSCT). We characterized early-transplant CDI and its associations, and analyzed serially-collected feces to determine intestinal carriage of toxigenic C. difficile. Fecal specimens were collected longitudinally from 94 patients during allo-HSCT hospitalization, from the start of pre-transplant conditioning until up to 35 days after stem cell infusion. Presence of C. difficile 16S rRNA and tcdB genes was determined. Clinical variables and specimen data were analyzed for association with development of CDI. Historical data from an additional 1144 allo-HSCT patients was also used. Fecal specimens from 37 patients (39%) were found to harbor C. difficile. Early-transplant CDI was diagnosed in 16 of 94 (17%) patients undergoing allo-HSCT; cases were generally mild and resembled non-CDI diarrhea associated with transplant conditioning. CDI was associated with preceding colonization with tcdB-positive C. difficile and conditioning regimen intensity. We found no associations between early-transplant CDI and graft-versus-host disease or CDI later in transplant. CDI occurs with high frequency during the early phase of allo-HSCT, where recipients are pre-colonized with toxigenic C. difficile. During this time, CDI incidence peaks during pre-transplant conditioning, and is correlated to intensity of the treatment. In this unique setting, high rates of CDI may be explained by prior colonization and chemotherapy; however, cases were generally mild and resembled non-infectious diarrhea due to conditioning, raising concerns of misdiagnosis. Further study of this unique population with more discriminating CDI diagnostic tests are warranted.


Asunto(s)
Clostridioides difficile/fisiología , Infecciones por Clostridium/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Adulto , Anciano , Estudios de Cohortes , Determinación de Punto Final , Heces/microbiología , Femenino , Humanos , Intestinos/microbiología , Masculino , Persona de Mediana Edad , Trasplante Homólogo/efectos adversos , Adulto Joven
11.
J Exp Med ; 209(8): 1445-56, 2012 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-22826298

RESUMEN

The intestinal microbiota contributes to the development of the immune system, and conversely, the immune system influences the composition of the microbiota. Toll-like receptors (TLRs) in the gut recognize bacterial ligands. Although TLR signaling represents a major arm of the innate immune system, the extent to which TLRs influence the composition of the intestinal microbiota remains unclear. We performed deep 16S ribosomal RNA sequencing to characterize the complex bacterial populations inhabiting the ileum and cecum of TLR- and MyD88-deficient mice. The microbiota of MyD88- and TLR-deficient mouse colonies differed markedly, with each colony harboring distinct and distinguishable bacterial populations in the small and large intestine. Comparison of MyD88-, TLR2-, TLR4-, TLR5-, and TLR9-deficient mice and their respective wild-type (WT) littermates demonstrated that the impact of TLR deficiency on the composition of the intestinal microbiota is minimal under homeostatic conditions and after recovery from antibiotic treatment. Thus, differences between TLR-deficient mouse colonies reflected long-term divergence of the microbiota after extended husbandry in isolation from each other. Long-term breeding of isolated mouse colonies results in changes of the intestinal microbiota that are communicated to offspring by maternal transmission, which account for marked compositional differences between WT and mutant mouse strains.


Asunto(s)
Ciego/inmunología , Íleon/inmunología , Inmunidad Innata/inmunología , Metagenoma/inmunología , Receptores Toll-Like/deficiencia , Receptores Toll-Like/inmunología , Animales , Antibacterianos/inmunología , Ciego/microbiología , Femenino , Íleon/microbiología , Inmunidad Innata/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptores Toll-Like/genética
12.
J Exp Med ; 209(5): 903-11, 2012 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-22547653

RESUMEN

Despite a growing understanding of the link between intestinal inflammation and resident gut microbes, longitudinal studies of human flora before initial onset of intestinal inflammation have not been reported. Here, we demonstrate in murine and human recipients of allogeneic bone marrow transplantation (BMT) that intestinal inflammation secondary to graft-versus-host disease (GVHD) is associated with major shifts in the composition of the intestinal microbiota. The microbiota, in turn, can modulate the severity of intestinal inflammation. In mouse models of GVHD, we observed loss of overall diversity and expansion of Lactobacillales and loss of Clostridiales. Eliminating Lactobacillales from the flora of mice before BMT aggravated GVHD, whereas reintroducing the predominant species of Lactobacillus mediated significant protection against GVHD. We then characterized gut flora of patients during onset of intestinal inflammation caused by GVHD and found patterns mirroring those in mice. We also identified increased microbial chaos early after allogeneic BMT as a potential risk factor for subsequent GVHD. Together, these data demonstrate regulation of flora by intestinal inflammation and suggest that flora manipulation may reduce intestinal inflammation and improve outcomes for allogeneic BMT recipients.


Asunto(s)
Biodiversidad , Trasplante de Médula Ósea/efectos adversos , Enterocolitis/microbiología , Enfermedad Injerto contra Huésped/complicaciones , Metagenoma/genética , Ampicilina , Animales , Secuencia de Bases , Sulfato de Dextran , Enterocolitis/etiología , Enterocolitis/patología , Heces/microbiología , Enfermedad Injerto contra Huésped/microbiología , Bacterias Grampositivas/aislamiento & purificación , Humanos , Ratones , Datos de Secuencia Molecular , Filogenia , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN , Especificidad de la Especie , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA