Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Immunity ; 53(6): 1296-1314.e9, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33296687

RESUMEN

Temporal resolution of cellular features associated with a severe COVID-19 disease trajectory is needed for understanding skewed immune responses and defining predictors of outcome. Here, we performed a longitudinal multi-omics study using a two-center cohort of 14 patients. We analyzed the bulk transcriptome, bulk DNA methylome, and single-cell transcriptome (>358,000 cells, including BCR profiles) of peripheral blood samples harvested from up to 5 time points. Validation was performed in two independent cohorts of COVID-19 patients. Severe COVID-19 was characterized by an increase of proliferating, metabolically hyperactive plasmablasts. Coinciding with critical illness, we also identified an expansion of interferon-activated circulating megakaryocytes and increased erythropoiesis with features of hypoxic signaling. Megakaryocyte- and erythroid-cell-derived co-expression modules were predictive of fatal disease outcome. The study demonstrates broad cellular effects of SARS-CoV-2 infection beyond adaptive immune cells and provides an entry point toward developing biomarkers and targeted treatments of patients with COVID-19.


Asunto(s)
COVID-19/metabolismo , Células Eritroides/patología , Megacariocitos/fisiología , Células Plasmáticas/fisiología , SARS-CoV-2/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores , Circulación Sanguínea , COVID-19/inmunología , Células Cultivadas , Estudios de Cohortes , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Proteómica , Análisis de Secuencia de ARN , Índice de Severidad de la Enfermedad , Análisis de la Célula Individual
2.
Infection ; 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38700656

RESUMEN

PURPOSE: The influence of new SARS-CoV-2 variants on the post-COVID-19 condition (PCC) remains unanswered. Therefore, we examined the prevalence and predictors of PCC-related symptoms in patients infected with the SARS-CoV-2 variants delta or omicron. METHODS: We compared prevalences and risk factors of acute and PCC-related symptoms three months after primary infection (3MFU) between delta- and omicron-infected patients from the Cross-Sectoral Platform of the German National Pandemic Cohort Network. Health-related quality of life (HrQoL) was determined by the EQ-5D-5L index score and trend groups were calculated to describe changes of HrQoL between different time points. RESULTS: We considered 758 patients for our analysis (delta: n = 341; omicron: n = 417). Compared with omicron patients, delta patients had a similar prevalence of PCC at the 3MFU (p = 0.354), whereby fatigue occurred most frequently (n = 256, 34%). HrQoL was comparable between the groups with the lowest EQ-5D-5L index score (0.75, 95% CI 0.73-0.78) at disease onset. While most patients (69%, n = 348) never showed a declined HrQoL, it deteriorated substantially in 37 patients (7%) from the acute phase to the 3MFU of which 27 were infected with omicron. CONCLUSION: With quality-controlled data from a multicenter cohort, we showed that PCC is an equally common challenge for patients infected with the SARS-CoV-2 variants delta and omicron at least for the German population. Developing the EQ-5D-5L index score trend groups showed that over two thirds of patients did not experience any restrictions in their HrQoL due to or after the SARS-CoV-2 infection at the 3MFU. CLINICAL TRAIL REGISTRATION: The cohort is registered at ClinicalTrials.gov since February 24, 2021 (Identifier: NCT04768998).

3.
Infection ; 50(2): 423-436, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34625912

RESUMEN

PURPOSE: Reported antibiotic use in coronavirus disease 2019 (COVID-19) is far higher than the actual rate of reported bacterial co- and superinfection. A better understanding of antibiotic therapy in COVID-19 is necessary. METHODS: 6457 SARS-CoV-2-infected cases, documented from March 18, 2020, until February 16, 2021, in the LEOSS cohort were analyzed. As primary endpoint, the correlation between any antibiotic treatment and all-cause mortality/progression to the next more advanced phase of disease was calculated for adult patients in the complicated phase of disease and procalcitonin (PCT) ≤ 0.5 ng/ml. The analysis took the confounders gender, age, and comorbidities into account. RESULTS: Three thousand, six hundred twenty-seven cases matched all inclusion criteria for analyses. For the primary endpoint, antibiotic treatment was not correlated with lower all-cause mortality or progression to the next more advanced (critical) phase (n = 996) (both p > 0.05). For the secondary endpoints, patients in the uncomplicated phase (n = 1195), regardless of PCT level, had no lower all-cause mortality and did not progress less to the next more advanced (complicated) phase when treated with antibiotics (p > 0.05). Patients in the complicated phase with PCT > 0.5 ng/ml and antibiotic treatment (n = 286) had a significantly increased all-cause mortality (p = 0.029) but no significantly different probability of progression to the critical phase (p > 0.05). CONCLUSION: In this cohort, antibiotics in SARS-CoV-2-infected patients were not associated with positive effects on all-cause mortality or disease progression. Additional studies are needed. Advice of local antibiotic stewardship- (ABS-) teams and local educational campaigns should be sought to improve rational antibiotic use in COVID-19 patients.


Asunto(s)
Programas de Optimización del Uso de los Antimicrobianos , Tratamiento Farmacológico de COVID-19 , Adulto , Antibacterianos/uso terapéutico , Progresión de la Enfermedad , Humanos , SARS-CoV-2
4.
Ann Rheum Dis ; 80(10): 1306-1311, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33762264

RESUMEN

INTRODUCTION: In light of the SARS-CoV-2 pandemic, protecting vulnerable groups has become a high priority. Persons at risk of severe disease, for example, those receiving immunosuppressive therapies for chronic inflammatory cdiseases (CIDs), are prioritised for vaccination. However, data concerning generation of protective antibody titres in immunosuppressed patients are scarce. Additionally, mRNA vaccines represent a new vaccine technology leading to increased insecurity especially in patients with CID. OBJECTIVE: Here we present for the first time, data on the efficacy and safety of anti-SARS-CoV-2 mRNA vaccines in a cohort of immunosuppressed patients as compared with healthy controls. METHODS: 42 healthy controls and 26 patients with CID were included in this study (mean age 37.5 vs 50.5 years). Immunisations were performed according to national guidelines with mRNA vaccines. Antibody titres were assessed by ELISA before initial vaccination and 7 days after secondary vaccination. Disease activity and side effects were assessed prior to and 7 days after both vaccinations. RESULTS: Anti-SARS-CoV-2 antibodies as well as neutralising activity could be detected in all study participants. IgG titres were significantly lower in patients as compared with controls (2053 binding antibody units (BAU)/mL ±1218 vs 2685±1102). Side effects were comparable in both groups. No severe adverse effects were observed, and no patients experienced a disease flare. CONCLUSION: We show that SARS-CoV-2 mRNA vaccines lead to development of antibodies in immunosuppressed patients without considerable side effects or induction of disease flares. Despite the small size of this cohort, we were able to demonstrate the efficiency and safety of mRNA vaccines in our cohort.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/prevención & control , Huésped Inmunocomprometido/inmunología , Inmunogenicidad Vacunal/inmunología , Inflamación/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Estudios de Cohortes , Femenino , Humanos , Inmunosupresores/uso terapéutico , Inflamación/inmunología , Masculino , Persona de Mediana Edad , Enfermedades Reumáticas/tratamiento farmacológico , Enfermedades Reumáticas/inmunología , SARS-CoV-2 , Vacunas Sintéticas/inmunología , Vacunas de ARNm
5.
Infection ; 49(1): 63-73, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33001409

RESUMEN

PURPOSE: Knowledge regarding patients' clinical condition at severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) detection is sparse. Data in the international, multicenter Lean European Open Survey on SARS-CoV-2-Infected Patients (LEOSS) cohort study may enhance the understanding of COVID-19. METHODS: Sociodemographic and clinical characteristics of SARS-CoV-2-infected patients, enrolled in the LEOSS cohort study between March 16, 2020, and May 14, 2020, were analyzed. Associations between baseline characteristics and clinical stages at diagnosis (uncomplicated vs. complicated) were assessed using logistic regression models. RESULTS: We included 2155 patients, 59.7% (1,287/2,155) were male; the most common age category was 66-85 years (39.6%; 500/2,155). The primary COVID-19 diagnosis was made in 35.0% (755/2,155) during complicated clinical stages. A significant univariate association between age; sex; body mass index; smoking; diabetes; cardiovascular, pulmonary, neurological, and kidney diseases; ACE inhibitor therapy; statin intake and an increased risk for complicated clinical stages of COVID-19 at diagnosis was found. Multivariable analysis revealed that advanced age [46-65 years: adjusted odds ratio (aOR): 1.73, 95% CI 1.25-2.42, p = 0.001; 66-85 years: aOR 1.93, 95% CI 1.36-2.74, p < 0.001; > 85 years: aOR 2.38, 95% CI 1.49-3.81, p < 0.001 vs. individuals aged 26-45 years], male sex (aOR 1.23, 95% CI 1.01-1.50, p = 0.040), cardiovascular disease (aOR 1.37, 95% CI 1.09-1.72, p = 0.007), and diabetes (aOR 1.33, 95% CI 1.04-1.69, p = 0.023) were associated with complicated stages of COVID-19 at diagnosis. CONCLUSION: The LEOSS cohort identified age, cardiovascular disease, diabetes and male sex as risk factors for complicated disease stages at SARS-CoV-2 diagnosis, thus confirming previous data. Further data regarding outcomes of the natural course of COVID-19 and the influence of treatment are required.


Asunto(s)
COVID-19/epidemiología , Enfermedades Cardiovasculares/epidemiología , Diabetes Mellitus/epidemiología , Enfermedades Renales/epidemiología , Enfermedades Pulmonares/epidemiología , Pandemias , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Índice de Masa Corporal , COVID-19/diagnóstico , COVID-19/fisiopatología , COVID-19/virología , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/virología , Estudios de Cohortes , Comorbilidad , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/fisiopatología , Diabetes Mellitus/virología , Europa (Continente)/epidemiología , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Enfermedades Renales/diagnóstico , Enfermedades Renales/fisiopatología , Enfermedades Renales/virología , Modelos Logísticos , Enfermedades Pulmonares/diagnóstico , Enfermedades Pulmonares/fisiopatología , Enfermedades Pulmonares/virología , Masculino , Persona de Mediana Edad , SARS-CoV-2/patogenicidad , Índice de Severidad de la Enfermedad , Factores Sexuales
6.
J Antimicrob Chemother ; 74(10): 2916-2925, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31355848

RESUMEN

BACKGROUND: Chronic pulmonary infections by Pseudomonas aeruginosa require frequent intravenous antibiotic treatment in cystic fibrosis (CF) patients. Emergence of antimicrobial resistance is common in these patients, which to date has been investigated at long-term intervals only. OBJECTIVES: To investigate under close to real-time conditions the dynamics of the response by P. aeruginosa to a single course of antibiotic therapy and the potentially associated rapid spread of antimicrobial resistance, as well as the impact on the airway microbiome. METHODS: We investigated a cohort of adult CF patients that were treated with a single course of antimicrobial combination therapy. Using daily sampling during treatment, we quantified the expression of resistance by P. aeruginosa (median of six isolates per daily sample, 347 isolates in total), measured bacterial load by P. aeruginosa-specific quantitative PCR and characterized the airway microbiome with a 16S rRNA-based approach. WGS was performed to reconstruct intrapatient strain phylogenies. RESULTS: In two patients, we found rapid and large increases in resistance to meropenem and ceftazidime. Phylogenetic reconstruction of strain relationships revealed that resistance shifts are probably due to de novo evolution and/or the selection of resistant subpopulations. We observed high interindividual variation in the reduction of bacterial load, microbiome composition and antibiotic resistance. CONCLUSIONS: We show that CF-associated P. aeruginosa populations can quickly respond to antibiotic therapy and that responses are patient specific. Thus, resistance evolution can be a direct consequence of treatment, and drug efficacy can be lost much faster than usually assumed. The consideration of these patient-specific rapid resistance shifts can help to improve treatment of CF-associated infections, for example by deeper sampling of bacteria for diagnostics, repeated monitoring of pathogen susceptibility and switching between drugs.


Asunto(s)
Antibacterianos/farmacología , Pulmón/microbiología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/efectos de los fármacos , Resistencia betalactámica , beta-Lactamas/farmacología , Adulto , Antibacterianos/administración & dosificación , Carga Bacteriana , Análisis por Conglomerados , Estudios de Cohortes , Fibrosis Quística/complicaciones , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Ribosómico/química , ADN Ribosómico/genética , Femenino , Humanos , Masculino , Filogenia , Pseudomonas aeruginosa/aislamiento & purificación , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN , Adulto Joven , beta-Lactamas/administración & dosificación
8.
Artículo en Alemán | MEDLINE | ID: mdl-28470637

RESUMEN

Antimicrobial Stewardship (AMS) cannot be practised as a one-man show. A well-established AMS-team with formal authority and dedicated time given by the hospital management can manage its tasks also in exceptional situations as for example an outbreak due to a multi-drug-resistant pathogen. Know-how of clinical infectious diseases is mandatory for all members of the AMS-team. The AMS-team plays various roles in an outbreak situation with the rational use of last-resort antibiotics and optimization of the dosage by therapeutic drug monitoring being most important. Restrictive usage of antibiotics can decrease the antibiotic selection pressure and counteract with the development of new bacterial resistances. Usage of last-resort antibiotics in an outbreak situation leads to an exceptional increase of therapeutic costs with fewer patients at the same time. Interdisciplinary work of infection control, the AMS-team, the different clinical departments and the hospital management are important for the prevention and the management of outbreak situations due to multi-drug-resistant pathogens.


Asunto(s)
Antiinfecciosos/uso terapéutico , Programas de Optimización del Uso de los Antimicrobianos/organización & administración , Infección Hospitalaria/prevención & control , Farmacorresistencia Microbiana/efectos de los fármacos , Promoción de la Salud/organización & administración , Control de Infecciones/organización & administración , Modelos Organizacionales , Humanos
9.
BMC Microbiol ; 15: 158, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26242247

RESUMEN

BACKGROUND: Clostridium difficile can cause antibiotic-associated diarrhea and a possibility of outbreaks in hospital settings warrants molecular typing. A microarray was designed that included toxin genes (tcdA/B, cdtA/B), genes related to antimicrobial resistance, the slpA gene and additional variable genes. RESULTS: DNA of six reference strains and 234 clinical isolates from South-Western and Eastern Germany was subjected to linear amplification and labeling with dUTP-linked biotin. Amplicons were hybridized to microarrays providing information on the presence of target genes and on their alleles. Tested isolates were assigned to 37 distinct profiles that clustered mainly according to MLST-defined clades. Three additional profiles were predicted from published genome sequences, although they were not found experimentally. CONCLUSIONS: The microarray based assay allows rapid and high-throughput genotyping of clinical C. difficile isolates including toxin gene detection and strain assignment. Overall hybridization profiles correlated with MLST-derived clades.


Asunto(s)
Clostridioides difficile/clasificación , Clostridioides difficile/genética , Técnicas de Genotipaje/métodos , Análisis por Micromatrices/métodos , Tipificación Molecular/métodos , Análisis por Conglomerados , Genes Bacterianos , Genotipo , Alemania , Ensayos Analíticos de Alto Rendimiento , Técnicas de Amplificación de Ácido Nucleico , Hibridación de Ácido Nucleico , Coloración y Etiquetado/métodos , Factores de Tiempo
10.
Gut ; 62(11): 1591-601, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23236009

RESUMEN

OBJECTIVE: Antibiotic (AB) usage strongly affects microbial intestinal metabolism and thereby impacts human health. Understanding this process and the underlying mechanisms remains a major research goal. Accordingly, we conducted the first comparative omic investigation of gut microbial communities in faecal samples taken at multiple time points from an individual subjected to ß-lactam therapy. METHODS: The total (16S rDNA) and active (16S rRNA) microbiota, metagenome, metatranscriptome (mRNAs), metametabolome (high-performance liquid chromatography coupled to electrospray ionisation and quadrupole time-of-flight mass spectrometry) and metaproteome (ultra high performing liquid chromatography coupled to an Orbitrap MS(2) instrument [UPLC-LTQ Orbitrap-MS/MS]) of a patient undergoing AB therapy for 14 days were evaluated. RESULTS: Apparently oscillatory population dynamics were observed, with an early reduction in Gram-negative organisms (day 6) and an overall collapse in diversity and possible further colonisation by 'presumptive' naturally resistant bacteria (day 11), followed by the re-growth of Gram-positive species (day 14). During this process, the maximum imbalance in the active microbial fraction occurred later (day 14) than the greatest change in the total microbial fraction, which reached a minimum biodiversity and richness on day 11; additionally, major metabolic changes occurred at day 6. Gut bacteria respond to ABs early by activating systems to avoid the antimicrobial effects of the drugs, while 'presumptively' attenuating their overall energetic metabolic status and the capacity to transport and metabolise bile acid, cholesterol, hormones and vitamins; host-microbial interactions significantly improved after treatment cessation. CONCLUSIONS: This proof-of-concept study provides an extensive description of gut microbiota responses to follow-up ß-lactam therapy. The results demonstrate that ABs targeting specific pathogenic infections and diseases may alter gut microbial ecology and interactions with host metabolism at a much higher level than previously assumed.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Microbiota/efectos de los fármacos , beta-Lactamas/farmacología , Anciano , Bacterias/clasificación , Bacterias/aislamiento & purificación , Técnicas de Tipificación Bacteriana/métodos , Biodiversidad , ADN Bacteriano/análisis , Heces/microbiología , Tracto Gastrointestinal/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Metaboloma/efectos de los fármacos , ARN Bacteriano/análisis , ARN Ribosómico 16S/análisis
11.
J Infect Public Health ; 17(4): 642-649, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38458134

RESUMEN

BACKGROUND: Vulnerability to infectious diseases in refugees is dependent on country of origin, flight routes, and conditions. Information on specific medical needs of different groups of refugees is lacking. We assessed the prevalence of infectious diseases, immunity to vaccine-preventable diseases, and chronic medical conditions in children, adolescents, and adult refugees from Ukraine who arrived in Germany in 2022. METHODS: Using different media, we recruited Ukrainian refugees at 13 sites between 9-12/2022. An antigen test for acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection, serologies for a range of vaccine-preventable diseases, as well as interferon gamma release assays (IGRAs) for tuberculosis (TB), and SARS-CoV-2 were performed. We assessed personal and family history of chronic medical conditions, infectious diseases, vaccination status, and conditions during migration. RESULTS: Overall, 1793 refugees (1401 adults and 392 children/adolescents) were included. Most participants were females (n = 1307; 72·3%) and from Eastern or Southern Ukraine. TB IGRA was positive in 13% (n = 184) of the adults and in 2% (n = 7) of the children. Serology-based immunological response was insufficient in approximately 21% (360/1793) of the participants for measles, 32% (572/1793) for diphtheria, and 74% (1289/1793) for hepatitis B. CONCLUSIONS: We show evidence of low serological response to vaccine-preventable infections and increased LTBI prevalence in Ukrainian refugees. These findings should be integrated into guidelines for screening and treatment of infectious diseases in migrants and refugees in Germany and Europe. Furthermore, low immunity for vaccine-preventable diseases in Ukrainians independent of their refugee status, calls for tailor-made communication efforts.


Asunto(s)
Enfermedades Transmisibles , Pueblos de Europa Oriental , Refugiados , Tuberculosis , Enfermedades Prevenibles por Vacunación , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Enfermedades Transmisibles/epidemiología , Estudios Transversales , Alemania/epidemiología , Prevalencia , Tuberculosis/epidemiología , Tuberculosis/prevención & control , Universidades
12.
Z Evid Fortbild Qual Gesundhwes ; 176: 12-21, 2023 Feb.
Artículo en Alemán | MEDLINE | ID: mdl-36754716

RESUMEN

INTRODUCTION: This study describes the development and validation of structure indicators for clinical infectious disease (ID) care in German hospitals, which is important to adequately face the future challenges in ID medicine. METHODS: A team of experts developed the structure indicators in a three-stage, multicriteria decision-making process: (1) identification of potential structure indicators based on a literature review, (2) written assessment process, and (3) face-to-face discussion to reach consensus and final selection of appropriate structure indicators. A field study was conducted to assess the developed structure indicators. A score based on the structure indicators was determined for each hospital and validated via receiver operator characteristic (ROC) curves using externally validated ID expertise (German Society of ID (DGI) Centre). RESULTS: Based on a list of 45 potential structure indicators, 18 suitable indicators were developed for clinical ID care structures in German hospitals. Out of these, ten key indicators were defined for the general and coronavirus disease 2019- (COVID-19-) specific clinical ID care structures. In the field survey of clinical ID care provision for COVID-19 patients in 40 German hospitals, the participating facilities achieved 0 to 9 points (median 4) in the determined score. The area under the ROC curve was 0.893 (95% CI: 0.797, 0.988; p < 0.001). DISCUSSION/CONCLUSION: The structure indicators developed within the framework of a transparent and established development process can be used in the future to both capture the current state and future developments of ID care quality in Germany and enable comparisons.


Asunto(s)
COVID-19 , Enfermedades Transmisibles , Humanos , Alemania , Pandemias , Hospitales
13.
Med Klin Intensivmed Notfmed ; 116(1): 81-92, 2021 Feb.
Artículo en Alemán | MEDLINE | ID: mdl-33108477

RESUMEN

Antibiotic resistance is a part of bacterial evolution and therefore unavoidable. Scarcity of novel treatment options requires prudent use of available antibiotics in order to decelerate the spread of resistance. This is the aim of antibiotic stewardship (ABS) programmes. The implementation of strategies that optimize antibiotic prescription and therapy necessitates the deployment of personnel as well as of structural resources. Necessary requirements for staff and strategies based on their evidence are described in the updated German S3 ABS Guideline. In the future, patients with infectious diseases will benefit from accelerated microbiological diagnostics as early adequate treatment not only reduces antibiotic consumption but also improves patient outcome. In addition, training of infectious disease specialists will substantially contribute to enhanced quality of care of patients with infectious disease.


Asunto(s)
Programas de Optimización del Uso de los Antimicrobianos , Antibacterianos/uso terapéutico , Humanos
14.
Evol Med Public Health ; 2016(1): 182-94, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27193199

RESUMEN

BACKGROUND AND OBJECTIVES: Cystic fibrosis patients suffer from chronic lung infections that require long-term antibiotic therapy. Pseudomonas readily evolve resistance, rendering antibiotics ineffective. In vitro experiments suggest that resistant bacteria may be treated by exploiting their collateral sensitivity to other antibiotics. Here, we investigate correlations of sensitivity and resistance profiles of Pseudomonas aeruginosa that naturally adapted to antibiotics in the cystic fibrosis lung. METHODOLOGY: Resistance profiles for 13 antibiotics were obtained using broth dilution, E-test and VITEK mass spectroscopy. Genetic variants were determined from whole-genome sequences and interrelationships among isolates were analyzed using 13 MLST loci. RESULT: Our study focused on 45 isolates from 13 patients under documented treatment with antibiotics. Forty percent of these were clinically resistant and 15% multi-drug resistant. Colistin resistance was found once, despite continuous colistin treatment and even though colistin resistance can readily evolve experimentally in the laboratory. Patients typically harbored multiple genetically and phenotypically distinct clones. However, genetically similar clones often had dissimilar resistance profiles. Isolates showed mutations in genes encoding cell wall synthesis, alginate production, efflux pumps and antibiotic modifying enzymes. Cross-resistance was commonly observed within antibiotic classes and between aminoglycosides and ß-lactam antibiotics. No evidence was found for consistent phenotypic resistance to one antibiotic and sensitivity to another within one genotype. CONCLUSIONS AND IMPLICATIONS: Evidence supporting potential collateral sensitivity in clinical P. aeruginosa isolates remains equivocal. However, cross-resistance within antibiotic classes is common. Colistin therapy is promising since resistance to it was rare despite its intensive use in the studied patients.

15.
Z Evid Fortbild Qual Gesundhwes ; 109(7): 535-41, 2015.
Artículo en Alemán | MEDLINE | ID: mdl-26593770

RESUMEN

INTRODUCTION: One of the core strategies to optimize antiinfective therapy is to review antibiotic prescriptions. Therefore, Antibiotic Stewardship (ABS) team members either attend ward rounds or perform a chart review to provide feedback to and discuss with the attending physician. Acceptance and effectiveness of both options are discussed in this article. METHODS: Attending physicians were asked to complete a questionnaire evaluating ABS activities. The modality of the reviewing process and its effectiveness, as well as the feasibility of recommendations was assessed. As the degree of implementation of ABS recommendations decreased on a trauma ward, the reviewing process was changed from chart review to attending the daily ward rounds. In this setting, the duration of the reviewing process and the consumption of antiinfectives in recommended daily doses/100 patient days (RDD/100PT) were assessed, comparing the two intervention modalities. RESULTS: Attending physicians predominantly appreciated the modality and extent of ABS currently offered to them by the ABS team, rating it relevant and effective. Implementation of ABS recommendations was increased on the trauma ward by academic detailing during the daily ward round; the consumption of broad spectrum antibiotics was reduced. DISCUSSION: ABS team members with formal authority and dedicated time for antibiotic stewardship activities effectively optimize antiinfective therapies by reviewing antibiotic prescriptions. The interaction of ABS experts and attending physicians contributes fundamentally to the effectiveness and degree of implementation of ABS interventions.


Asunto(s)
Antibacterianos/uso terapéutico , Actitud del Personal de Salud , Infección Hospitalaria/tratamiento farmacológico , Farmacorresistencia Microbiana , Adhesión a Directriz/normas , Infectología/normas , Auditoría Médica , Mejoramiento de la Calidad/normas , Conducta Cooperativa , Utilización de Medicamentos/estadística & datos numéricos , Alemania , Humanos , Comunicación Interdisciplinaria , Derivación y Consulta/normas , Encuestas y Cuestionarios , Rondas de Enseñanza , Estudios de Tiempo y Movimiento , Centros Traumatológicos , Heridas y Lesiones/cirugía
16.
Gut Microbes ; 6(4): 243-54, 2015 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-26178862

RESUMEN

Gut microbiota play a key role in the host's health system. Broad antibiotic therapy is known to disrupt the microbial balance affecting pathogenic as well as host-associated microbes. The aim of the present study was to investigate the influence of antibiotic paromomycin on the luminal and mucosa-associated microbiota at the DNA (abundance) and RNA (potential activity) level as well as to identify possible differences. The influence of antibiotic treatment on intestinal microbiota was investigated in 5 healthy individuals (age range: 20-22 years). All participants received the antibiotic paromomycin for 3 d. Fecal samples as well as sigmoidal biopsies were collected before and immediately after cessation of antibiotic treatment as well as after a recovery phase of 42 d. Compartment- and treatment status-specific indicator operational taxonomic units (OTUs) as well as abundance- and activity-specific patterns were identified by 16S rRNA and 16S rRNA gene amplicon libraries and high-throughput pyrosequencing. Microbial composition of lumen and mucosa were significantly different at the DNA compared to the RNA level. Antibiotic treatment resulted in changes of the microbiota, affecting the luminal and mucosal bacteria in a similar way. Several OTUs were identified as compartment- and/or treatment status-specific. Abundance and activity patterns of some indicator OTUs differed considerably. The study shows fundamental changes in composition of gut microbiota under antibiotic therapy at both the potential activity and the abundance level at different treatment status. It may help to understand the complex processes of gut microbiota changes involved in resilience mechanisms and on development of antibiotic-associated clinical diseases.


Asunto(s)
Antibacterianos/administración & dosificación , Bacterias/clasificación , Bacterias/efectos de los fármacos , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Paromomicina/administración & dosificación , Adulto , Bacterias/genética , Biopsia , ADN Ribosómico/química , ADN Ribosómico/genética , Voluntarios Sanos , Humanos , Datos de Secuencia Molecular , Filogenia , ARN Ribosómico 16S/genética , Adulto Joven
17.
FEMS Microbiol Ecol ; 88(2): 272-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24490649

RESUMEN

Epibiotic biofilms have the potential to control major aspects of the biology and ecology of their hosts. Their composition and function may thus be essential for the health of the host. We tested the influence of salinity on the composition of epibacterial communities associated with the brown macroalga Fucus vesiculosus. Algal individuals were incubated at three salinities (5, 19, and 25) for 14 days and nonliving reference substrata (stones) were included in the experiment. Subsequently, the composition of their surface-associated bacterial communities was analyzed by 454 pyrosequencing of 16S rRNA gene sequences. Redundancy analysis revealed that the composition of epiphytic and epilithic communities significantly differed and were both affected by salinity. We found that 5% of 2494 epiphytic operational taxonomic units at 97% sequence similarity were responsible for the observed shifts. Epibacterial α-diversity was significantly lower at salinity 5 but did not differ between substrata. Our results indicate that salinity is an important factor in structuring alga-associated epibacterial communities with respect to composition and/or diversity. Whether direct or indirect mechanisms (via altered biotic interactions) may have been responsible for the observed shifts is discussed.


Asunto(s)
Bacterias/clasificación , Fucus/microbiología , Salinidad , Bacterias/genética , Biodiversidad
18.
PLoS One ; 9(2): e89417, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586762

RESUMEN

Clostridium difficile infections are an emerging health problem in the modern hospital environment. Severe alterations of the gut microbiome with loss of resistance to colonization against C. difficile are thought to be the major trigger, but there is no clear concept of how C. difficile infection evolves and which microbiological factors are involved. We sequenced 16S rRNA amplicons generated from DNA and RNA/cDNA of fecal samples from three groups of individuals by FLX technology: (i) healthy controls (no antibiotic therapy); (ii) individuals receiving antibiotic therapy (Ampicillin/Sulbactam, cephalosporins, and fluoroquinolones with subsequent development of C. difficile infection or (iii) individuals receiving antibiotic therapy without C. difficile infection. We compared the effects of the three different antibiotic classes on the intestinal microbiome and the effects of alterations of the gut microbiome on C. difficile infection at the DNA (total microbiota) and rRNA (potentially active) levels. A comparison of antibiotic classes showed significant differences at DNA level, but not at RNA level. Among individuals that developed or did not develop a C. difficile infection under antibiotics we found no significant differences. We identified single species that were up- or down regulated in individuals receiving antibiotics who developed the infection compared to non-infected individuals. We found no significant differences in the global composition of the transcriptionally active gut microbiome associated with C. difficile infections. We suggest that up- and down regulation of specific bacterial species may be involved in colonization resistance against C. difficile providing a potential therapeutic approach through specific manipulation of the intestinal microbiome.


Asunto(s)
Clostridioides difficile/efectos de los fármacos , Fluoroquinolonas/farmacología , Microbiota/efectos de los fármacos , beta-Lactamas/farmacología , Ampicilina/farmacología , Cefalosporinas/farmacología , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/tratamiento farmacológico , Diarrea/tratamiento farmacológico , Diarrea/microbiología , Fluoroquinolonas/uso terapéutico , Tracto Gastrointestinal/microbiología , Humanos , ARN Ribosómico 16S/genética , Sulbactam/farmacología
19.
Microbiologyopen ; 2(2): 338-49, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23568841

RESUMEN

The thallus surface of the brown macroalga Fucus vesiculosus is covered by a specific biofilm community. This biofilm supposedly plays an important role in the interaction between host and environment. So far, we know little about compositional or functional shifts of this epibiotic bacterial community under changing environmental conditions. In this study, the response of the microbiota to different temperatures with respect to cell density and community composition was analyzed by nonculture-based methods (denaturing gradient gel electrophoresis and 454 pyrosequencing of the 16S rRNA gene). Redundancy analysis showed that despite high variability among host individuals temperature accounted for 20% of the variation in the bacterial community composition, whereas cell density did not differ between groups. Across all samples, 4341 bacterial operational taxonomic units (OTUs) at a 97% similarity level were identified. Eight percent of OTUs were significantly correlated with low, medium, and high temperatures. Notably, the family Rhodobacteraceae increased in relative abundance from 20% to 50% with increasing temperature. OTU diversity (evenness and richness) was higher at 15 °C than at the lower and higher temperatures. Considering their known and presumed ecological functions for the host, change in the epibacterial community may entail shifts in the performance of the host alga.


Asunto(s)
Bacterias/aislamiento & purificación , Biopelículas , Fucus/microbiología , Algas Marinas/microbiología , Bacterias/clasificación , Bacterias/genética , Biodiversidad , ADN Bacteriano/genética , Electroforesis en Gel de Gradiente Desnaturalizante , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN , Temperatura
20.
PLoS One ; 8(11): e80201, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24282523

RESUMEN

The human intestinal microbiota performs many essential functions for the host. Antimicrobial agents, such as antibiotics (AB), are also known to disturb microbial community equilibrium, thereby having an impact on human physiology. While an increasing number of studies investigate the effects of AB usage on changes in human gut microbiota biodiversity, its functional effects are still poorly understood. We performed a follow-up study to explore the effect of ABs with different modes of action on human gut microbiota composition and function. Four individuals were treated with different antibiotics and samples were taken before, during and after the AB course for all of them. Changes in the total and in the active (growing) microbiota as well as the functional changes were addressed by 16S rRNA gene and metagenomic 454-based pyrosequencing approaches. We have found that the class of antibiotic, particularly its antimicrobial effect and mode of action, played an important role in modulating the gut microbiota composition and function. Furthermore, analysis of the resistome suggested that oscillatory dynamics are not only due to antibiotic-target resistance, but also to fluctuations in the surviving bacterial community. Our results indicated that the effect of AB on the human gut microbiota relates to the interaction of several factors, principally the properties of the antimicrobial agent, and the structure, functions and resistance genes of the microbial community.


Asunto(s)
Antibacterianos/farmacología , Microbiota/efectos de los fármacos , Biodiversidad , Farmacorresistencia Bacteriana/efectos de los fármacos , Estudios de Seguimiento , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Humanos , Metagenoma , ARN Ribosómico 16S/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA