Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 16(7): 698-707, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26006013

RESUMEN

The epithelium is the main entry point for many viruses, but the processes that protect barrier surfaces against viral infections are incompletely understood. Here we identified interleukin 22 (IL-22) produced by innate lymphoid cell group 3 (ILC3) as an amplifier of signaling via interferon-λ (IFN-λ), a synergism needed to curtail the replication of rotavirus, the leading cause of childhood gastroenteritis. Cooperation between the receptor for IL-22 and the receptor for IFN-λ, both of which were 'preferentially' expressed by intestinal epithelial cells (IECs), was required for optimal activation of the transcription factor STAT1 and expression of interferon-stimulated genes (ISGs). These data suggested that epithelial cells are protected against viral replication by co-option of two evolutionarily related cytokine networks. These data may inform the design of novel immunotherapy for viral infections that are sensitive to interferons.


Asunto(s)
Citocinas/inmunología , Expresión Génica/inmunología , Interleucinas/inmunología , Infecciones por Rotavirus/inmunología , Animales , Células CACO-2 , Línea Celular , Chlorocebus aethiops , Citocinas/genética , Citocinas/farmacología , Perros , Sinergismo Farmacológico , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Expresión Génica/efectos de los fármacos , Células HT29 , Humanos , Immunoblotting , Interleucinas/genética , Interleucinas/farmacología , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Intestinos/virología , Células de Riñón Canino Madin Darby , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Receptores de Citocinas/genética , Receptores de Citocinas/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Rotavirus/genética , Infecciones por Rotavirus/virología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Células Vero , Interleucina-22
2.
Infection ; 52(2): 661-665, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38030862

RESUMEN

PURPOSE: Carbapenemase-producing Enterobacterales (CPE) pose a serious threat for healthcare facilities worldwide, yet the mode of transmission is often unclear. Recently, we recorded an increase of blaOXA-48-harboring isolates at our hospital associated with patients with previous medical treatment in the Ukraine. We used long-read whole genome sequencing (lrWGS) to characterize these isolates including their plasmids. METHODS: Samples were collected as part of clinical routine diagnostic or screening of multi-drug resistance bacteria (MDRB). Antimicrobial susceptibility testing was performed and all MDRB (n = 10) were sequenced by lrWGS for genotyping, identification of antimicrobial resistance (AMR) genes, and characterization of plasmids. RESULTS: While routine analysis of core genome multilocus sequence typing (cgMLST) did not show any genetic similarities between isolates, we found an unexpected high similarity in the plasmid diversity of different Enterobacterales in patients with previous medical treatment in the Ukraine. This included an IncL/M plasmid carrying blaOXA-48 and additional small non-AMR-coding plasmids. CONCLUSION: Our results show that lrWGS can be used in the routine setting to uncover similarities in plasmids and may give further information about potential epidemiologic associations. In the future, analysis of both AMR and non-AMR plasmids may provide an additional layer of information for molecular surveillance of CPE.


Asunto(s)
Escherichia coli , beta-Lactamasas , Humanos , beta-Lactamasas/genética , Plásmidos/genética , Escherichia coli/genética , Antibacterianos/farmacología , Tipificación de Secuencias Multilocus/métodos , Klebsiella pneumoniae/genética , Pruebas de Sensibilidad Microbiana
3.
BMC Biol ; 21(1): 269, 2023 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996810

RESUMEN

BACKGROUND: Microbiome analysis is becoming a standard component in many scientific studies, but also requires extensive quality control of the 16S rRNA gene sequencing data prior to analysis. In particular, when investigating low-biomass microbial environments such as human skin, contaminants distort the true microbiome sample composition and need to be removed bioinformatically. We introduce MicrobIEM, a novel tool to bioinformatically remove contaminants using negative controls. RESULTS: We benchmarked MicrobIEM against five established decontamination approaches in four 16S rRNA amplicon sequencing datasets: three serially diluted mock communities (108-103 cells, 0.4-80% contamination) with even or staggered taxon compositions and a skin microbiome dataset. Results depended strongly on user-selected algorithm parameters. Overall, sample-based algorithms separated mock and contaminant sequences best in the even mock, whereas control-based algorithms performed better in the two staggered mocks, particularly in low-biomass samples (≤ 106 cells). We show that a correct decontamination benchmarking requires realistic staggered mock communities and unbiased evaluation measures such as Youden's index. In the skin dataset, the Decontam prevalence filter and MicrobIEM's ratio filter effectively reduced common contaminants while keeping skin-associated genera. CONCLUSIONS: MicrobIEM's ratio filter for decontamination performs better or as good as established bioinformatic decontamination tools. In contrast to established tools, MicrobIEM additionally provides interactive plots and supports selecting appropriate filtering parameters via a user-friendly graphical user interface. Therefore, MicrobIEM is the first quality control tool for microbiome experts without coding experience.


Asunto(s)
Bacterias , Microbiota , Humanos , Bacterias/genética , Benchmarking , ARN Ribosómico 16S/genética , Descontaminación , Microbiota/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos
4.
J Clin Immunol ; 42(6): 1301-1309, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35655107

RESUMEN

Hyper-IgE syndromes (HIES) are a group of inborn errors of immunity (IEI) caused by monogenic defects such as in the gene STAT3 (STAT3-HIES). Patients suffering from HIES show an increased susceptibility to Staphylococcus aureus (S. aureus) including skin abscesses and pulmonary infections. To assess if the underlying immune defect of STAT3-HIES patients influences the resistance patterns, pathogenicity factors or strain types of S. aureus. We characterized eleven S. aureus strains isolated from STAT3-HIES patients (n = 4) by whole genome sequencing (WGS) to determine presence of resistance and virulence genes. Additionally, we used multi-locus sequence typing (MLST) and protein A (spa) typing to classify these isolates. Bacterial isolates collected from this cohort of STAT3-HIES patients were identified as common spa types in Germany. Only one of the isolates was classified as methicillin-resistant S. aureus (MRSA). For one STAT3 patient WGS illustrated that infection and colonization occurred with different S. aureus isolates rather than one particular clone. The identified S. aureus carriage profile on a molecular level suggests that S. aureus strain type in STAT3-HIES patients is determined by local epidemiology rather than the underlying immune defect highlighting the importance of microbiological assessment prior to antibiotic treatment.


Asunto(s)
Síndrome de Job , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Antibacterianos , Humanos , Síndrome de Job/diagnóstico , Síndrome de Job/genética , Tipificación de Secuencias Multilocus , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
5.
Curr Microbiol ; 80(1): 37, 2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-36526801

RESUMEN

Clostridioides difficile infection (CDI) often manifests as diarrhea, particularly in adults of older age or with underlying comorbidities. However, only severe cases are notifiable in Germany. Moreover, failure to collect a stool specimen from inpatients with diarrhea or incomplete testing may lead to underdiagnosis and underreporting of CDI. We assessed the frequency of diarrhea, stool specimen collection, and CDI testing to estimate CDI underdiagnosis and underreporting among hospitalized adults. In a ten-day point-prevalence study (2019-2021) of nine hospitals in a defined area (Muenster/Coesfeld, North Rhine-Westphalia, Germany), all diarrhea cases (≥ 3 loose stools in 24 h) among adult inpatients were captured via medical record screening and nurse interviews. Patient characteristics, symptom onset, putative origin, antibiotic consumption, and diagnostic stool sampling were collected in a case report form (CRF). Diagnostic results were retrieved from the respective hospital laboratories. Among 6998 patients screened, 476 (7%) diarrhea patients were identified, yielding a hospital-based incidence of 201 cases per 10,000 patient-days. Of the diarrheal patients, 186 (39%) had a stool sample collected, of which 160 (86%) were tested for CDI, meaning that the overall CDI testing rate among diarrhea patients was 34%. Toxigenic C. difficile was detected in 18 (11%) of the tested samples. The frequency of stool specimen collection and CDI testing among hospitalized diarrhea patients was suboptimal. Thus, CDI incidence in Germany is likely underestimated. To assess the complete burden of CDI in German hospitals, further investigations are needed.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium , Adulto , Humanos , Infecciones por Clostridium/diagnóstico , Infecciones por Clostridium/epidemiología , Diarrea/diagnóstico , Diarrea/epidemiología , Heces , Manejo de Especímenes
6.
Handb Exp Pharmacol ; 268: 53-65, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34228203

RESUMEN

Over the last few decades, allergic diseases have been steadily increasing worldwide, a phenomenon that is not yet completely understood. Recent evidence, however, suggests that alterations in the microbiome may be a contributing factor. The microbiome refers to all microorganisms in a habitat including bacteria, fungi, and viruses. Using modern sequencing technologies, we are now capable of detecting and analyzing the human microbiome in more detail than ever before. Epidemiological and experimental studies have indicated that a complex intestinal microbiome supports the development of the immune system during childhood, thus providing protection from allergic diseases, including food allergy. The microbiome becomes an important part of human physiology and forms dynamic relationships with our various barrier systems. For example, bacterial dysbiosis is a hallmark of atopic eczema and correlates with disease progression. Similarly, the lung and nasopharyngeal microbiome is altered in patients with asthma and allergic rhinitis. While these results are interesting, the underlying mechanisms are still unclear and need to be investigated with functional studies. This review gives a short overview of the terminology and methods used in microbiome research before highlighting results concerning the lung, skin, and intestinal microbiome in allergic diseases.


Asunto(s)
Dermatitis Atópica , Hipersensibilidad a los Alimentos , Microbiota , Rinitis Alérgica , Disbiosis , Humanos
7.
Clin Infect Dis ; 72(2): 265-270, 2021 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33501962

RESUMEN

BACKGROUND: Coronavirus disease 2019 (COVID-19) is a life-threatening respiratory condition caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and was initially detected in China in December 2019. Currently, in Germany >140 000 cases of COVID-19 are confirmed. Here we report a nosocomial outbreak of SARS-CoV-2 infections in the pediatric dialysis unit of the University Hospital Münster (UHM). METHODS: Single-step real-time reverse-transcription polymerase chain reaction (rRT-PCR) from nasopharyngeal swabs was used to diagnose the index patient and identify infected contacts. Epidemiological links were analyzed by patient interviews and medical record reviews. In addition, each contact was assessed for exposure to the index case and monitored for clinical symptoms. Cycle threshold (Ct) values of all positive test results were compared between symptomatic and asymptomatic cases. RESULTS: Forty-eight cases were involved in this nosocomial outbreak. Nine contact cases developed laboratory-confirmed COVID-19 infections. Two SARS-CoV-2-positive cases remained clinically asymptomatic. Eleven cases reported flulike symptoms without positive results. Ct values were significantly lower in cases presenting typical COVID-19 symptoms, suggesting high viral shedding (P = .007). CONCLUSIONS: Person-to-person transmission was at the heart of a hospital outbreak of SARS-CoV-2 between healthcare workers (HCWs) and patients in the pediatric dialysis unit at UHM. Semiquantitative rRT-PCR results suggest that individuals with high viral load pose a risk to spread SARS-CoV-2 in the hospital setting. Our epidemiological observation highlights the need to develop strategies to trace and monitor SARS-CoV-2-infected HCWs to prevent COVID-19 outbreaks in the hospital setting.


Asunto(s)
COVID-19 , Infección Hospitalaria , Niño , China/epidemiología , Infección Hospitalaria/epidemiología , Brotes de Enfermedades , Alemania , Humanos , Diálisis Renal , SARS-CoV-2
8.
Nature ; 494(7436): 261-5, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23334414

RESUMEN

At mucosal surfaces, the immune system should not initiate inflammatory immune responses to the plethora of antigens constantly present in the environment, but should remain poised to unleash a potent assault on intestinal pathogens. The transcriptional programs and regulatory factors required for immune cells to switch from homeostatic (often tissue-protective) function to potent antimicrobial immunity are poorly defined. Mucosal retinoic-acid-receptor-related orphan receptor-γt-positive (RORγt(+)) innate lymphoid cells (ILCs) are emerging as an important innate lymphocyte population required for immunity to intestinal infections. Various subsets of RORγt(+) ILCs have been described but the transcriptional programs controlling their specification and fate remain largely unknown. Here we provide evidence that the transcription factor T-bet determines the fate of a distinct lineage of CCR6(-)RORγt(+) ILCs. Postnatally emerging CCR6(-)RORγt(+) ILCs upregulated T-bet and this was controlled by cues from the commensal microbiota and interleukin-23 (IL-23). In contrast, CCR6(+)RORγt(+) ILCs, which arise earlier during ontogeny, did not express T-bet. T-bet instructed the expression of T-bet target genes such as interferon-γ (IFN-γ) and of the natural cytotoxicity receptor NKp46. Mice genetically lacking T-bet showed normal development of CCR6(-)RORγt(+) ILCs, but they could not differentiate into NKp46-expressing RORγt(+) ILCs (that is, IL-22-producing natural killer (NK-22) cells) and failed to produce IFN-γ. The production of IFN-γ by T-bet-expressing CCR6(-)RORγt(+) ILCs was essential for the release of mucus-forming glycoproteins required to protect the epithelial barrier during Salmonella enterica infection. Salmonella infection also causes severe enterocolitis that is at least partly driven by IFN-γ. Mice deficient for T-bet or depleted of ILCs developed only mild enterocolitis. Thus, graded expression of T-bet in CCR6(-)RORγt(+) ILCs facilitates the differentiation of IFN-γ-producing CCR6(-)RORγt(+) ILCs required to protect the epithelial barrier against Salmonella infections. Co-expression of T-bet and RORγt, which is also found in subsets of IL-17-producing T-helper (T(H)17) cells, may be an evolutionarily conserved transcriptional program that originally developed as part of the innate defence against infections but that also confers an increased risk of immune-mediated pathology.


Asunto(s)
Linaje de la Célula , Inmunidad Innata/inmunología , Linfocitos/citología , Linfocitos/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores CCR6/deficiencia , Proteínas de Dominio T Box/metabolismo , Animales , Antígenos Ly/genética , Diferenciación Celular , Células Cultivadas , Enterocolitis/inmunología , Enterocolitis/metabolismo , Enterocolitis/patología , Epitelio/inmunología , Epitelio/metabolismo , Epitelio/microbiología , Interferón gamma/biosíntesis , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-23/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Linfocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Moco/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Receptores CCR6/metabolismo , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/metabolismo , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad
11.
J Immunol ; 191(6): 3037-3048, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23960236

RESUMEN

MicroRNAs (MiRs) are small, noncoding RNAs that regulate gene expression posttranscriptionally. In this study, we show that MiR-210 is induced by Oct-2, a key transcriptional mediator of B cell activation. Germline deletion of MiR-210 results in the development of autoantibodies from 5 mo of age. Overexpression of MiR-210 in vivo resulted in cell autonomous expansion of the B1 lineage and impaired fitness of B2 cells. Mice overexpressing MiR-210 exhibited impaired class-switched Ab responses, a finding confirmed in wild-type B cells transfected with a MiR-210 mimic. In vitro studies demonstrated defects in cellular proliferation and cell cycle entry, which were consistent with the transcriptomic analysis demonstrating downregulation of genes involved in cellular proliferation and B cell activation. These findings indicate that Oct-2 induction of MiR-210 provides a novel inhibitory mechanism for the control of B cells and autoantibody production.


Asunto(s)
Autoanticuerpos/biosíntesis , Linfocitos B/metabolismo , Activación de Linfocitos/inmunología , MicroARNs/biosíntesis , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Animales , Autoanticuerpos/inmunología , Linfocitos B/inmunología , Separación Celular , Inmunoprecipitación de Cromatina , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/inmunología , Factor 2 de Transcripción de Unión a Octámeros/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Transcriptoma
12.
Front Cell Infect Microbiol ; 14: 1343858, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38469349

RESUMEN

Introduction: The emergence of extended-spectrum ß-lactamase (ESBL)-producing Enterobacteriaceae is an urgent and alarming One Health problem. This study aimed to investigate duplications of plasmid-encoded ESBL genes and their impact on antimicrobial resistance (AMR) phenotypes in clinical and screening isolates. Methods: Multi-drug-resistant bacteria from hospitalized patients were collected during routine clinical surveillance from January 2022 to June 2023, and their antimicrobial susceptibility patterns were determined. Genotypes were extracted from long-read whole-genome sequencing data. Furthermore, plasmids and other mobile genetic elements associated with ESBL genes were characterized, and the ESBL genes were correlated to ceftazidime minimal inhibitory concentration (MIC). Results: In total, we identified four cases of plasmid-encoded ESBL gene duplications that match four genetically similar plasmids during the 18-month surveillance period: five Escherichia coli and three Klebsiella pneumoniae isolates. As the ESBL genes were part of transposable elements, the surrounding sequence regions were duplicated as well. In-depth analysis revealed insertion sequence (IS)-mediated transposition mechanisms. Isolates with duplicated ESBL genes exhibited a higher MIC for ceftazidime in comparison to isolates with a single gene copy (3-256 vs. 1.5-32 mg/L, respectively). Conclusion: ESBL gene duplications led to an increased phenotypic resistance against ceftazidime. Our data suggest that ESBL gene duplications by an IS-mediated transposition are a relevant mechanism for how AMR develops in the clinical setting and is part of the microevolution of plasmids.


Asunto(s)
Antibacterianos , Ceftazidima , Humanos , Ceftazidima/farmacología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , beta-Lactamasas/genética , Duplicación de Gen , Escherichia coli , Plásmidos/genética , Enterobacteriaceae/genética , Klebsiella pneumoniae , Pruebas de Sensibilidad Microbiana
13.
Microbes Infect ; 26(4): 105306, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38316375

RESUMEN

Staphylococcus aureus is one of the major pathogens isolated from the airways of people with cystic fibrosis (pwCF). Recently, we described a mucoid S. aureus phenotype from respiratory specimens of pwCF, which constitutively overproduced biofilm that consisted of polysaccharide intercellular adhesin (PIA) due to a 5bp-deletion (5bp-del) in the intergenic region of the intercellular adhesin (ica) locus. Since we were not able to identify the 5bp-del in mucoid isolates of two pwCF with long-term S. aureus persistence and in a number of mucoid isolates of pwCF from a prospective multicenter study, these strains were (i) characterized phenotypically, (ii) investigated for biofilm formation, and (iii) molecular typed by spa-sequence typing. To screen for mutations responsible for mucoidy, the ica operon of all mucoid isolates was analyzed by Sanger sequencing. Whole genome sequencing was performed for selected isolates. For all mucoid isolates without the 5 bp-del, various mutations in icaR, which is the transcriptional repressor of the icaADBC operon. Mucoid and non-mucoid strains belonged to the same spa-type. Transformation of PIA-overproducing S. aureus with a vector expressing the intact icaR gene restored the non-mucoid phenotype. Altogether, we demonstrated a new mechanism for the emergence of mucoid S. aureus isolates of pwCF.


Asunto(s)
Biopelículas , Fibrosis Quística , Mutación , Infecciones Estafilocócicas , Staphylococcus aureus , Fibrosis Quística/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/aislamiento & purificación , Humanos , Biopelículas/crecimiento & desarrollo , Infecciones Estafilocócicas/microbiología , Operón/genética , Polisacáridos Bacterianos/genética , Polisacáridos Bacterianos/metabolismo , Proteínas Represoras/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Estudios Prospectivos , Secuenciación Completa del Genoma , Sistema Respiratorio/microbiología
14.
Artículo en Inglés | MEDLINE | ID: mdl-37681832

RESUMEN

Water systems in health care facilities can form reservoirs for Gram-negative bacteria. While planning a new neonatal intensive care unit (NICU), we performed a retrospective evaluation of potential risks from water-diverting systems on the existing NICU of our tertiary care University Hospital. During 2017 to 2023, we recorded nine nosocomial cluster events with bacterial pathogens in our NICU. Of these, three clusters of Gram-negative bacteria were potentially related to sink drains: A Klebsiella oxytoca, a Pseudomonas aeruginosa, and an Enterobacter hormaechei cluster were uncovered by clinical routine screening of patients and breastmilk samples. They were confirmed using whole-genome sequencing and a subsequent core genome multilocus sequence typing (cgMLST) algorithm. Our observations highlight that the implementation of sink drains in a NICU may have negative effects on patients' safety. Construction planning should concentrate on the avoidance of washbasins in patient rooms when redesigning sensitive areas such as NICUs.


Asunto(s)
Algoritmos , Unidades de Cuidado Intensivo Neonatal , Recién Nacido , Humanos , Estudios Retrospectivos , Instituciones de Salud , Leche Humana
15.
Microorganisms ; 11(7)2023 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-37512978

RESUMEN

Staphylococcus epidermidis (S. epidermidis) is part of the human skin flora but can also cause nosocomial infections, such as device-associated infections, especially in vulnerable patient groups. Here, we investigated clinical isolates of linezolid-resistant S. epidermidis (LRSE) collected from blood cultures at the University Hospital Münster (UHM) during the period 2020-2022. All detected isolates were subjected to whole genome sequencing (WGS) and the relatedness of the isolates was determined using core genome multilocus sequence typing (cgMLST). The 15 LRSE isolates detected were classified as multilocus sequence type (ST) 2 carrying the staphylococcal cassette chromosome mec (SCCmec) type III. All isolates showed high-level resistance for linezolid by gradient tests. However, no isolate carried the cfr gene that is often associated with linezolid resistance. Analysis of cgMLST data sets revealed a cluster of six closely related LRSE isolates, suggesting a transmission event on a hematological/oncological ward at our hospital. Among the included patients, the majority of patients affected by LRSE infections had underlying hematological malignancies. This confirms previous observations that this patient group is particularly vulnerable to LRSE infection. Our data emphasize that the surveillance of LRSE in the hospital setting is a necessary step to prevent the spread of multidrug-resistant S. epidermidis among vulnerable patient groups, such as patients with hematological malignancies, immunosuppression or patients in intensive care units.

16.
Microorganisms ; 10(3)2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-35336067

RESUMEN

Plasmid transfer is one important mechanism how antimicrobial resistance can spread between different species, contributing to the rise of multidrug resistant bacteria (MDRB) worldwide. Here were present whole genome sequencing (WGS) data of two MDRB isolates, an Escherichia coli and a Klebsiella quasipneumoniae, which were isolated from a single patient. Detailed analysis of long-read sequencing data identified an identical F2:A-:B- lncFII plasmid containing blaCTX-M-27 in both isolates, suggesting horizontal plasmid exchange between the two species. As the plasmid of the E. coli strain carried multiple copies of the resistance cassette, the genomic data correlated with the increased antimicrobial resistance (AMR) detected for this isolate. Our case report demonstrates how long-read sequencing data of MDRB can be used to investigate the role of plasmid mediate resistance in the healthcare setting and explain resistance phenotypes.

17.
Future Microbiol ; 17: 589-598, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35341325

RESUMEN

Aim: To estimate the burden of parenting and caregiving duties among clinical microbiologists in Germany and to identify workplace-related support systems and barriers to engaging in career-relevant activities. Methods: A cross-sectional web-based survey was conducted. Participants were asked to answer 37 questions, of which 24 specifically addressed parenting and caregiving duties. Results: Only few workplace-related support systems are currently available, and experiences of job-related disadvantages were frequently reported (27 of 47; 57.4%). Main barriers were a lack of flexible working hours and reliable childcare. Sociocultural norms and a lack of role models were perceived as detrimental. Conclusion: More support systems and a credible culture of family friendliness are needed to prevent jeopardizing the academic potential of young parents.


Asunto(s)
Responsabilidad Parental , Estudios Transversales , Humanos , Encuestas y Cuestionarios
18.
Microorganisms ; 10(4)2022 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-35456740

RESUMEN

Since March 2020, the COVID-19 pandemic forced hospitals worldwide to intensify their infection control measures to prevent health care-associated transmission of SARS-CoV-2. The correct use of personal protective equipment, especially the application of masks, was quickly identified as priority to reduce transmission with this pathogen. Here, we report a nosocomial cluster of methicillin-resistant Staphylococcus aureus (MRSA) that occurred during the COVID-19 pandemic in a gynecology/obstetrics department, despite these intensified contact precautions. Five MRSA originating from clinical samples after surgical intervention led to an outbreak investigation. Firstly, this included environmental sampling of the operation theatre (OT) and, secondly, a point prevalence screening of patients and health care workers (HCW). All detected MRSA were subjected to whole genome sequencing (WGS) and isolate relatedness was determined using core genome multilocus sequence typing (cgMLST). WGS revealed one MRSA cluster with genetically closely related five patient and two HCW isolates differing in a single cgMLST allele at maximum. The outbreak was terminated after implementation of infection control bundle strategies. Although contact precaution measures, which are also part of MRSA prevention bundle strategies, were intensified during the COVID-19 pandemic, this MRSA outbreak could take place. This illustrates the importance of adherence to classical infection prevention strategies.

19.
Microbiol Resour Announc ; 11(4): e0007222, 2022 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35258326

RESUMEN

Staphylococcus aureus is a widely distributed, opportunistic pathogen and has been linked to the human skin disease atopic dermatitis (AD). Here, we present 44 complete and 4 draft genome sequences of S. aureus strains isolated from the nose and skin of AD patients and healthy controls from a German study cohort.

20.
Antimicrob Resist Infect Control ; 9(1): 154, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32962759

RESUMEN

BACKGROUND: Currently, hospitals have been forced to divert substantial resources to cope with the ongoing coronavirus disease 2019 (COVID-19) pandemic. It is unclear if this situation will affect long-standing infection prevention practices and impact on healthcare associated infections. Here, we report a nosocomial cluster of vancomycin-resistant enterococci (VRE) that occurred on a COVID-19 dedicated intensive care unit (ICU) despite intensified contact precautions during the current pandemic. Whole genome sequence-based typing (WGS) was used to investigate genetic relatedness of VRE isolates collected from COVID-19 and non-COVID-19 patients during the outbreak and to compare them to environmental VRE samples. METHODS: Five VRE isolated from patients (three clinical and two screening samples) as well as 11 VRE and six vancomycin susceptible Enterococcus faecium (E. faecium) samples from environmental sites underwent WGS during the outbreak investigation. Isolate relatedness was determined using core genome multilocus sequence typing (cgMLST). RESULTS: WGS revealed two genotypic distinct VRE clusters with genetically closely related patient and environmental isolates. The cluster was terminated by enhanced infection control bundle strategies. CONCLUSIONS: Our results illustrate the importance of continued adherence to infection prevention and control measures during the COVID-19 pandemic to prevent VRE transmission and healthcare associated infections.


Asunto(s)
Coinfección/tratamiento farmacológico , Infección Hospitalaria/epidemiología , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/epidemiología , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Betacoronavirus , COVID-19 , Coinfección/microbiología , Infecciones por Coronavirus/patología , Infección Hospitalaria/tratamiento farmacológico , Infección Hospitalaria/microbiología , Brotes de Enfermedades , Genoma Bacteriano/genética , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Control de Infecciones , Unidades de Cuidados Intensivos , Tipificación de Secuencias Multilocus , Pandemias , Neumonía Viral/patología , Prevención Primaria , SARS-CoV-2 , Enterococos Resistentes a la Vancomicina/genética , Enterococos Resistentes a la Vancomicina/aislamiento & purificación , Secuenciación Completa del Genoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA