Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Front Microbiol ; 11: 1422, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32765433

RESUMEN

In developed countries, hematological malignancies (HM) account for 8 to 10% of cancers diagnosed annually and one-third of patients with HM (HMP) are expected to die from their disease. The former wide spectrum "magic bullet," imipenem, has been ousted by the emergence of carbapenem resistant (CR) pathogens. In endemic areas, infections with CR-bacteria occur in vulnerable patients, notably in HMP, who suffer from high mortality related to infectious complications. In this work, we reviewed epidemiologic and clinical factors associated with CR-infections in adult HMP and data on CR-related mortality and antibiotic treatments in this population. We found that resistance profile of strains involved in HMP infections, mainly bacteremia, reflect local epidemiology. Significant risk factors for infections with CR-bacteria include sex male, age around 50 years old, acute leukemia, selvage chemotherapy, neutropenia, and digestive colonization by CR-bacteria. Mortality rate is high in HMP infected with CR-Enterobacteriaceae, more particularly in case of acute myeloid leukemia and unresolved neutropenia, due to inappropriate empiric management and delayed administration of targeted antibiotics, such as tigecycline, colistin, or new associations of active drugs. Thus, we developed an algorithm for clinicians, assessing the incremental risk for CR-bacterial infection occurrence and mortality in febrile HMP, to guide decisions related to empirical therapeutic strategies.

2.
Nat Rev Rheumatol ; 16(5): 282-292, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32157196

RESUMEN

In the past three decades, extraordinary advances have been made in the understanding of the pathogenesis of, and treatment options for, inflammatory arthritides, including rheumatoid arthritis and spondyloarthritis. The use of methotrexate and subsequently biologic therapies (such as TNF inhibitors, among others) and oral small molecules have substantially improved clinical outcomes for many patients with inflammatory arthritis; for others, however, these agents do not substantially improve their symptoms. The emerging field of pharmacomicrobiomics, which investigates the effect of variations within the human gut microbiome on drugs, has already provided important insights into these therapeutics. Pharmacomicrobiomic studies have demonstrated that human gut microorganisms and their enzymatic products can affect the bioavailability, clinical efficacy and toxicity of a wide array of drugs through direct and indirect mechanisms. This discipline promises to facilitate the advent of microbiome-based precision medicine approaches in inflammatory arthritis, including strategies for predicting response to treatment and for modulating the microbiome to improve response to therapy or reduce drug toxicity.


Asunto(s)
Artritis Reumatoide/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Medicina de Precisión/métodos , Espondiloartritis/microbiología , Animales , Antirreumáticos/metabolismo , Antirreumáticos/farmacocinética , Antirreumáticos/uso terapéutico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Autoinmunidad/efectos de los fármacos , Disponibilidad Biológica , Microbioma Gastrointestinal/genética , Humanos , Metotrexato/metabolismo , Metotrexato/farmacocinética , Metotrexato/uso terapéutico , Ratones , Espondiloartritis/tratamiento farmacológico , Espondiloartritis/patología , Inhibidores del Factor de Necrosis Tumoral/metabolismo , Inhibidores del Factor de Necrosis Tumoral/farmacocinética , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico
3.
Arthritis Rheumatol ; 72(4): 645-657, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31729183

RESUMEN

OBJECTIVE: To characterize the ecological effects of biologic therapies on the gut bacterial and fungal microbiome in psoriatic arthritis (PsA)/spondyloarthritis (SpA) patients. METHODS: Fecal samples from PsA/SpA patients pre- and posttreatment with tumor necrosis factor inhibitors (TNFi; n = 15) or an anti-interleukin-17A monoclonal antibody inhibitor (IL-17i; n = 14) underwent sequencing (16S ribosomal RNA, internal transcribed spacer and shotgun metagenomics) and computational microbiome analysis. Fecal levels of fatty acid metabolites and cytokines/proteins implicated in PsA/SpA pathogenesis or intestinal inflammation were correlated with sequence data. Additionally, ileal biopsies obtained from SpA patients who developed clinically overt Crohn's disease (CD) after treatment with IL-17i (n = 5) were analyzed for expression of IL-23/Th17-related cytokines, IL-25/IL-17E-producing cells, and type 2 innate lymphoid cells (ILC2s). RESULTS: There were significant shifts in abundance of specific taxa after treatment with IL-17i compared to TNFi, particularly Clostridiales (P = 0.016) and Candida albicans (P = 0.041). These subclinical alterations correlated with changes in bacterial community co-occurrence, metabolic pathways, IL-23/Th17-related cytokines, and various fatty acids. Ileal biopsies showed that clinically overt CD was associated with expansion of IL-25/IL-17E-producing tuft cells and ILC2s (P < 0.05), compared to pre-IL-17i treatment levels. CONCLUSION: In a subgroup of SpA patients, the initiation of IL-17A blockade correlated with features of subclinical gut inflammation and intestinal dysbiosis of certain bacterial and fungal taxa, most notably C albicans. Further, IL-17i-related CD was associated with overexpression of IL-25/IL-17E-producing tuft cells and ILC2s. These results may help to explain the potential link between inhibition of a specific IL-17 pathway and the (sub)clinical gut inflammation observed in SpA.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Artritis Psoriásica/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Inflamación/metabolismo , Interleucina-17/inmunología , Espondiloartritis/tratamiento farmacológico , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Artritis Psoriásica/metabolismo , Artritis Psoriásica/microbiología , Femenino , Humanos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Intestinos/microbiología , Masculino , Persona de Mediana Edad , Espondiloartritis/metabolismo , Espondiloartritis/microbiología , Inhibidores del Factor de Necrosis Tumoral/farmacología
4.
Artículo en Inglés | MEDLINE | ID: mdl-31767720

RESUMEN

Multidrug-resistant Enterobacteriaceae (MRE) colonize the intestine asymptomatically from where they can breach into the bloodstream and cause life-threatening infections, especially in heavily colonized patients. Despite the clinical relevance of MRE colonization levels, we know little about how they vary in hospitalized patients and the clinical factors that determine those levels. Here, we conducted one of the largest studies of MRE fecal levels by tracking longitudinally 133 acute leukemia patients and monitoring their MRE levels over time through extensive culturing. MRE were defined as Enterobacteriaceae species that acquired nonsusceptibility to ≥1 agent in ≥3 antimicrobial categories. In addition, due to the selective media used, the MRE had to be resistant to third-generation cephalosporins. MRE were detected in 60% of the patients, but their fecal levels varied considerably among patients and within the same patient (>6 and 4 orders of magnitude, respectively). Multivariate analysis of clinical metadata revealed an impact of intravenous beta-lactams (i.e., meropenem and piperacillin-tazobactam), which significantly diminished the fecal MRE levels in hospitalized patients. Consistent with a direct action of beta-lactams, we found an effect only when the patient was colonized with strains sensitive to the administered beta-lactam (P < 0.001) but not with nonsusceptible strains. We report previously unobserved inter- and intraindividual heterogeneity in MRE fecal levels, suggesting that quantitative surveillance is more informative than qualitative surveillance of hospitalized patients. In addition, our study highlights the relevance of incorporating antibiotic treatment and susceptibility data of gut-colonizing pathogens for future clinical studies and in clinical decision-making.


Asunto(s)
Antibacterianos/efectos adversos , Farmacorresistencia Bacteriana Múltiple , Enterobacteriaceae/efectos de los fármacos , Heces/microbiología , beta-Lactamas/efectos adversos , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Cefalosporinas/farmacología , Medios de Cultivo , Hospitalización , Humanos , Inyecciones Intravenosas , Leucemia/complicaciones , Pruebas de Sensibilidad Microbiana , Estudios Prospectivos , beta-Lactamas/administración & dosificación , beta-Lactamas/farmacología
5.
Artículo en Inglés | MEDLINE | ID: mdl-31687131

RESUMEN

Background: The emergence of carbapenemase-producing (CP) Citrobacter freundii poses a significant threat to public health, especially in high-risk populations. In this study, whole genome sequencing was used to characterize the carbapenem resistance mechanism of three C. freundii clinical isolates recovered from fecal samples of patients with acute leukemia (AL) from Spain. Materials and methods: Twelve fecal samples, collected between 2013 and 2015 from 9 patients with AL, were screened for the presence of CP strains by selecting them on MacConkey agar supplemented with ertapenem (0.5 mg/L). Bacteria were identified by MALDI-TOF mass spectrometry and were phenotypically characterized. Whole genome sequencing of C. freundii isolates was performed using the MinION and MiSeq Illumina sequencers. Bioinformatic analysis was performed in order to identify the molecular support of carbapenem resistance and to study the genetic environment of carbapenem resistance encoding genes. Results: Three carbapenem-resistant C. freundii strains (imipenem MIC≥32 mg/L) corresponding to three different AL patients were isolated. Positive modified Carba NP test results suggested carbapenemase production. The genomes of each C. freundii tested were assembled into a single chromosomal contig and plasmids contig. In all the strains, the carbapenem resistance was due to the coproduction of OXA-48 and VIM-1 enzymes encoded by genes located on chromosome and on an IncHI2 plasmid, respectively. According to the MLST and the SNPs analysis, all strains belonged to the same clone ST169. Conclusion: We report in our study, the intestinal carrying of C. freundii clone ST169 coproducing OXA-48 and VIM-1 identified in leukemic patients.


Asunto(s)
Proteínas Bacterianas/genética , Citrobacter freundii/clasificación , Citrobacter freundii/genética , Infecciones por Enterobacteriaceae/epidemiología , Infecciones por Enterobacteriaceae/etiología , Genoma Bacteriano , Genómica , Leucemia/complicaciones , beta-Lactamasas/genética , Citrobacter freundii/efectos de los fármacos , Genómica/métodos , Humanos , Vigilancia en Salud Pública , España/epidemiología
6.
J Infect Chemother ; 25(8): 605-609, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31023570

RESUMEN

BACKGROUND: Bacterial infections in immunocompromised patients are associated with a high mortality and morbidity rate. In this high-risk group, the presence of multidrug-resistant (MDR) bacteria, particularly bacteria that harbor a transferable antibiotic resistance gene, complicates the management of bacterial infections. In this study, we investigated the presence of the transferable colistin resistance mcr genes in patients with leukemia in Spain. METHODS: 217 fecal samples collected in 2013-2015 from 56 patients with acute leukemia and colonized with MDR Enterobacteriaceae strains, were screened on September 2017 for the presence of the colistin resistance mcr genes (mcr-1 to -5) by multiplex PCR. mcr positive strains selected on LBJMR and MacConkey supplemented with colistin (2 µg/ml) media were phenotypically and molecularly characterized by antimicrobial susceptibility testing, minimum inhibitory concentration, multilocus sequence typing and plasmid characterization. RESULTS: Among 217 fecal samples, 5 samples collected from 3 patients were positive for the presence of the mcr-1 colistin-resistance gene. Four Escherichia coli strains were isolated and exhibited resistance to colistin with MIC = 4 µg/ml. Other genes conferring the resistance to ß-lactam antibiotics have also been identified in mcr-1 positive strains, including blaTEM-206 and blaTEM-98. Three different sequence types were identified, including ST1196, ST140 and ST10. Plasmid characterization allowed us to detect the mcr-1 colistin resistance gene on conjugative IncP plasmid type. CONCLUSION: To the best of our knowledge, we have identified the mcr-1 gene for the first time in leukemia patients in Spain. In light of these results, strict measures have been implemented to prevent its dissemination.


Asunto(s)
Colistina/farmacología , Farmacorresistencia Bacteriana/genética , Proteínas de Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Leucemia/microbiología , Plásmidos/genética , Antibacterianos/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Humanos , Pruebas de Sensibilidad Microbiana/métodos , Persona de Mediana Edad , España , beta-Lactamasas/genética
7.
Proteomes ; 7(1)2019 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-30626002

RESUMEN

The microbiome has a strong impact on human health and disease and is, therefore, increasingly studied in a clinical context. Metaproteomics is also attracting considerable attention, and such data can be efficiently generated today owing to improvements in mass spectrometry-based proteomics. As we will discuss in this study, there are still major challenges notably in data analysis that need to be overcome. Here, we analyzed 212 fecal samples from 56 hospitalized acute leukemia patients with multidrug-resistant Enterobactericeae (MRE) gut colonization using metagenomics and metaproteomics. This is one of the largest clinical metaproteomic studies to date, and the first metaproteomic study addressing the gut microbiome in MRE colonized acute leukemia patients. Based on this substantial data set, we discuss major current limitations in clinical metaproteomic data analysis to provide guidance to researchers in the field. Notably, the results show that public metagenome databases are incomplete and that sample-specific metagenomes improve results. Furthermore, biological variation is tremendous which challenges clinical study designs and argues that longitudinal measurements of individual patients are a valuable future addition to the analysis of patient cohorts.

8.
EMBO J ; 37(5)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29378774

RESUMEN

Intestinal homeostasis relies on a continuous dialogue between the commensal bacteria and the immune system. Natural killer T (NKT) cells, which recognize CD1d-restricted microbial lipids and self-lipids, contribute to the regulation of mucosal immunity, yet the mechanisms underlying their functions remain poorly understood. Here, we demonstrate that NKT cells respond to intestinal lipids and CD11c+ cells (including dendritic cells (DCs) and macrophages) are essential to mediate lipid presentation within the gut ultimately controlling intestinal NKT cell homeostasis and activation. Conversely, CD1d and NKT cells participate in the control of the intestinal bacteria composition and compartmentalization, in the regulation of the IgA repertoire and in the induction of regulatory T cells within the gut. These changes in intestinal homeostasis require CD1d expression on DC/macrophage populations as mice with conditional deletion of CD1d on CD11c+ cells exhibit dysbiosis and altered immune homeostasis. These results unveil the importance of CD11c+ cells in controlling lipid-dependent immunity in the intestinal compartment and reveal an NKT cell-DC crosstalk as a key mechanism for the regulation of gut homeostasis.


Asunto(s)
Mucosa Intestinal/inmunología , Lípidos de la Membrana/inmunología , Células T Asesinas Naturales/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD1d/biosíntesis , Antígenos CD1d/genética , Antígenos CD1d/inmunología , Antígeno CD11c/metabolismo , Células Dendríticas/inmunología , Disbiosis/genética , Microbioma Gastrointestinal/inmunología , Inmunoglobulina A/inmunología , Interleucina-4/inmunología , Mucosa Intestinal/microbiología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
Best Pract Res Clin Haematol ; 28(2-3): 155-61, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26590773

RESUMEN

While allogeneic hematopoietic stem cell transplantations have a curative potential, infections and graft-versus-host disease remain significant problems. The intestinal microbiota can influence responses to cancer chemotherapy and the role of the microbiota in affecting allogeneic hematopoietic stem cell transplantation outcomes is increasingly appreciated. The following paper discusses the most recent developments in this area.


Asunto(s)
Bacteriemia/tratamiento farmacológico , Microbioma Gastrointestinal , Enfermedad Injerto contra Huésped/prevención & control , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas/mortalidad , Infecciones Oportunistas/tratamiento farmacológico , Antibacterianos/administración & dosificación , Antibacterianos/efectos adversos , Antineoplásicos/uso terapéutico , Bacteriemia/etiología , Bacteriemia/microbiología , Bacteriemia/mortalidad , Ensayos Clínicos como Asunto , Enfermedad Injerto contra Huésped/microbiología , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/patología , Neoplasias Hematológicas/microbiología , Neoplasias Hematológicas/mortalidad , Neoplasias Hematológicas/patología , Humanos , Metronidazol/administración & dosificación , Metronidazol/efectos adversos , Infecciones Oportunistas/etiología , Infecciones Oportunistas/microbiología , Infecciones Oportunistas/mortalidad , Factores de Riesgo , Análisis de Supervivencia , Vancomicina/administración & dosificación , Vancomicina/efectos adversos , Enterococos Resistentes a la Vancomicina/crecimiento & desarrollo , Enterococos Resistentes a la Vancomicina/patogenicidad
10.
Arthritis Rheumatol ; 67(1): 128-39, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25319745

RESUMEN

OBJECTIVE: To characterize the diversity and taxonomic relative abundance of the gut microbiota in patients with never-treated, recent-onset psoriatic arthritis (PsA). METHODS: High-throughput 16S ribosomal RNA pyrosequencing was utilized to compare the community composition of gut microbiota in patients with PsA (n = 16), patients with psoriasis of the skin (n = 15), and healthy, matched control subjects (n = 17). Samples were further assessed for the presence and levels of fecal and serum secretory IgA (sIgA), proinflammatory proteins, and fatty acids. RESULTS: The gut microbiota observed in patients with PsA and patients with skin psoriasis was less diverse when compared to that in healthy controls. This could be attributed to the reduced presence of several taxa. Samples from both patient groups showed a relative decrease in abundance of Coprococcus species, while samples from PsA patients were also characterized by a significant reduction in Akkermansia, Ruminococcus, and Pseudobutyrivibrio. Supernatants of fecal samples from PsA patients revealed an increase in sIgA levels and decrease in RANKL levels. Analysis of fatty acids revealed low fecal quantities of hexanoate and heptanoate in both patients with PsA and patients with psoriasis. CONCLUSION: Patients with PsA and patients with skin psoriasis had a lower relative abundance of multiple intestinal bacteria. Although some genera were concomitantly decreased in both conditions, PsA samples had a lower abundance of reportedly beneficial taxa. This gut microbiota profile in PsA was similar to that previously described in patients with inflammatory bowel disease and was associated with changes in specific inflammatory proteins unique to this group, and distinct from that in patients with skin psoriasis and healthy controls. Thus, the role of the gut microbiome in the continuum of psoriasis-PsA pathogenesis and the associated immune response merits further study.


Asunto(s)
Artritis Psoriásica/microbiología , Disbiosis/microbiología , Tracto Gastrointestinal/microbiología , Enfermedades Inflamatorias del Intestino/microbiología , Microbiota , Adulto , Artritis Psoriásica/metabolismo , Estudios de Casos y Controles , Citocinas/metabolismo , Disbiosis/metabolismo , Ácidos Grasos/metabolismo , Heces/microbiología , Femenino , Tracto Gastrointestinal/metabolismo , Humanos , Inmunoglobulina A/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Masculino , Persona de Mediana Edad , Psoriasis/metabolismo , Psoriasis/microbiología , Ligando RANK/metabolismo
11.
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
13.
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
14.
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
15.
Arthritis Rheum ; 64(10): 3083-94, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22576262

RESUMEN

OBJECTIVE: To profile the abundance and diversity of subgingival oral microbiota in patients with never-treated, new-onset rheumatoid arthritis (RA). METHODS: Periodontal disease (PD) status, clinical activity, and sociodemographic factors were determined in patients with new-onset RA, patients with chronic RA, and healthy subjects. Multiplexed-454 pyrosequencing was used to compare the composition of subgingival microbiota and establish correlations between the presence/abundance of bacteria and disease phenotypes. Anti-Porphyromonas gingivalis antibody testing was performed to assess prior exposure to the bacterial pathogen P gingivalis. RESULTS: The more advanced forms of periodontitis were already present at disease onset in patients with new-onset RA. The subgingival microbiota observed in patients with new-onset RA was distinct from that found in healthy controls. In most cases, however, these microbial differences could be attributed to the severity of PD and were not inherent to RA. The presence and abundance of P gingivalis were also directly associated with the severity of PD and were not unique to RA. The presence of P gingivalis was not correlated with anti-citrullinated protein antibody (ACPA) titers. Overall exposure to P gingivalis was similar between patients with new-onset RA and controls, observed in 78% of patients and 83% of controls. The presence and abundance of Anaeroglobus geminatus correlated with the presence of ACPAs/rheumatoid factor. Prevotella and Leptotrichia species were the only characteristic taxa observed in patients with new-onset RA irrespective of PD status. CONCLUSION: Patients with new-onset RA exhibited a high prevalence of PD at disease onset, despite their young age and paucity of smoking history. The subgingival microbiota profile in patients with new-onset RA was similar to that in patients with chronic RA and healthy subjects whose PD was of comparable severity. Although colonization with P gingivalis correlated with the severity of PD, overall exposure to P gingivalis was similar among the groups. The role of A geminatus and Prevotella/Leptotrichia species in this process merits further study.


Asunto(s)
Artritis Reumatoide/microbiología , Metagenoma , Boca/microbiología , Periodontitis/microbiología , Adulto , Artritis Reumatoide/complicaciones , Artritis Reumatoide/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Boca/inmunología , Periodontitis/complicaciones , Periodontitis/inmunología , Porphyromonas gingivalis/inmunología , Índice de Severidad de la Enfermedad , Factores Socioeconómicos , Encuestas y Cuestionarios
16.
J Clin Invest ; 120(12): 4332-41, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21099116

RESUMEN

Bloodstream infection by highly antibiotic-resistant bacteria, such as vancomycin-resistant Enterococcus (VRE), is a growing clinical problem that increasingly defies medical intervention. Identifying patients at high risk for bacterial sepsis remains an important clinical challenge. Recent studies have shown that antibiotics can alter microbial diversity in the intestine. Here, we characterized these effects using 16s rDNA pyrosequencing and demonstrated that antibiotic treatment of mice enabled exogenously administered VRE to efficiently and nearly completely displace the normal microbiota of the small and large intestine. In the clinical setting, we found that intestinal domination by VRE preceded bloodstream infection in patients undergoing allogeneic hematopoietic stem cell transplantation. Our results demonstrate that antibiotics perturb the normal commensal microbiota and set the stage for intestinal domination by bacteria associated with hospital-acquired infections. Thus, high-throughput DNA sequencing of the intestinal microbiota could identify patients at high risk of developing bacterial sepsis.


Asunto(s)
Enterococcus/efectos de los fármacos , Intestinos/microbiología , Animales , Antibacterianos/efectos adversos , Bacteriemia/etiología , Bacteriemia/microbiología , Secuencia de Bases , Infección Hospitalaria/etiología , Infección Hospitalaria/microbiología , ADN Bacteriano/genética , ADN Ribosómico/genética , Enterococcus/genética , Enterococcus/aislamiento & purificación , Infecciones por Bacterias Grampositivas/etiología , Infecciones por Bacterias Grampositivas/microbiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Intestinos/efectos de los fármacos , Metagenoma/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Resistencia a la Vancomicina
17.
J Infect Dis ; 201(4): 534-43, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20064069

RESUMEN

Treatment of vancomycin-resistant Enterococcus (VRE) infections is limited by the paucity of effective antibiotics. Administration of broad-spectrum antibiotics promotes VRE colonization by down-regulating homeostatic innate immune defenses. Intestinal epithelial cells and Paneth cells express antimicrobial factors on direct or indirect stimulation of the Toll-like receptor (TLR)-myeloid differentiation factor 88-mediated pathway by microbe-derived molecules. Here, we demonstrate that the TLR5 agonist flagellin restores antibiotic-impaired innate immune defenses and restricts colonization with VRE. Flagellin stimulates the expression of RegIIIgamma, a secreted C-type lectin that kills gram-positive bacteria, including VRE. Systemic administration of flagellin induces RegIIIgamma expression in intestinal epithelial cells and Paneth cells along the entire length of the small intestine. Induction of RegIIIgamma requires TLR5 expression in hematopoietic cells and is dependent on interleukin 22 expression. Systemic administration of flagellin to antibiotic-treated mice dramatically reduces VRE colonization. By enhancing mucosal resistance to multidrug-resistant organisms, flagellin administration may provide a clinically useful approach to prevent infections in patients treated with broad-spectrum antibiotics.


Asunto(s)
Enterococcus/efectos de los fármacos , Flagelina/farmacología , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Receptor Toll-Like 5/agonistas , Resistencia a la Vancomicina/efectos de los fármacos , Animales , Antibacterianos/farmacología , Recuento de Células , Citocinas/biosíntesis , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Infecciones por Bacterias Grampositivas/genética , Infecciones por Bacterias Grampositivas/inmunología , Inmunidad Innata/efectos de los fármacos , Inmunohistoquímica , Interleucinas/inmunología , Interleucinas/metabolismo , Intestino Delgado/citología , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Pancreatitis , Proteínas/genética , Proteínas/inmunología , Proteínas/metabolismo , Receptor Toll-Like 5/inmunología , Receptor Toll-Like 5/metabolismo , Resistencia a la Vancomicina/inmunología , Interleucina-22
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA