Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 18.514
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Biochem ; 86: 567-583, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28654325

RESUMEN

Multidrug resistance is a global threat as the clinically available potent antibiotic drugs are becoming exceedingly scarce. For example, increasing drug resistance among gram-positive bacteria is responsible for approximately one-third of nosocomial infections. As ribosomes are a major target for these drugs, they may serve as suitable objects for novel development of next-generation antibiotics. Three-dimensional structures of ribosomal particles from Staphylococcus aureus obtained by X-ray crystallography have shed light on fine details of drug binding sites and have revealed unique structural motifs specific for this pathogenic strain, which may be used for the design of novel degradable pathogen-specific, and hence, environmentally friendly drugs.


Asunto(s)
Antibacterianos/síntesis química , Proteínas Bacterianas/química , Diseño de Fármacos , Ribosomas/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Antibacterianos/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Infección Hospitalaria/tratamiento farmacológico , Infección Hospitalaria/microbiología , Cristalografía por Rayos X , Deinococcus/efectos de los fármacos , Deinococcus/genética , Deinococcus/metabolismo , Farmacorresistencia Bacteriana Múltiple , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Modelos Moleculares , Ribosomas/metabolismo , Ribosomas/ultraestructura , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Thermus thermophilus/efectos de los fármacos , Thermus thermophilus/genética , Thermus thermophilus/metabolismo
2.
Cell ; 169(5): 849-861.e13, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28502769

RESUMEN

We examined the evolutionary history of leading multidrug resistant hospital pathogens, the enterococci, to their origin hundreds of millions of years ago. Our goal was to understand why, among the vast diversity of gut flora, enterococci are so well adapted to the modern hospital environment. Molecular clock estimation, together with analysis of their environmental distribution, phenotypic diversity, and concordance with host fossil records, place the origins of the enterococci around the time of animal terrestrialization, 425-500 mya. Speciation appears to parallel the diversification of hosts, including the rapid emergence of new enterococcal species following the End Permian Extinction. Major drivers of speciation include changing carbohydrate availability in the host gut. Life on land would have selected for the precise traits that now allow pathogenic enterococci to survive desiccation, starvation, and disinfection in the modern hospital, foreordaining their emergence as leading hospital pathogens.


Asunto(s)
Evolución Biológica , Enterococcus/genética , Animales , Enfermedades Transmisibles Emergentes/microbiología , Infección Hospitalaria/microbiología , Farmacorresistencia Bacteriana , Enterococcus/clasificación , Enterococcus/citología , Enterococcus/efectos de los fármacos , Especiación Genética , Interacciones Huésped-Patógeno , Larva/microbiología , Mariposas Nocturnas/crecimiento & desarrollo , Mariposas Nocturnas/microbiología , Filogenia , ARN Ribosómico 16S/genética
3.
PLoS Genet ; 19(7): e1010646, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37498819

RESUMEN

The Gram-negative bacterial pathogen Acinetobacter baumannii is a major cause of hospital-acquired opportunistic infections. The increasing spread of pan-drug resistant strains makes A. baumannii top-ranking among the ESKAPE pathogens for which novel routes of treatment are urgently needed. Comparative genomics approaches have successfully identified genetic changes coinciding with the emergence of pathogenicity in Acinetobacter. Genes that are prevalent both in pathogenic and a-pathogenic Acinetobacter species were not considered ignoring that virulence factors may emerge by the modification of evolutionarily old and widespread proteins. Here, we increased the resolution of comparative genomics analyses to also include lineage-specific changes in protein feature architectures. Using type IVa pili (T4aP) as an example, we show that three pilus components, among them the pilus tip adhesin ComC, vary in their Pfam domain annotation within the genus Acinetobacter. In most pathogenic Acinetobacter isolates, ComC displays a von Willebrand Factor type A domain harboring a finger-like protrusion, and we provide experimental evidence that this finger conveys virulence-related functions in A. baumannii. All three genes are part of an evolutionary cassette, which has been replaced at least twice during A. baumannii diversification. The resulting strain-specific differences in T4aP layout suggests differences in the way how individual strains interact with their host. Our study underpins the hypothesis that A. baumannii uses T4aP for host infection as it was shown previously for other pathogens. It also indicates that many more functional complexes may exist whose precise functions have been adjusted by modifying individual components on the domain level.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Infección Hospitalaria , Humanos , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Filogenia , Infección Hospitalaria/microbiología , Infecciones por Acinetobacter/microbiología , Hospitales , Antibacterianos
4.
Clin Microbiol Rev ; 37(2): e0013523, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38421181

RESUMEN

SUMMARYClostridioides difficile infection (CDI) is one of the major issues in nosocomial infections. This bacterium is constantly evolving and poses complex challenges for clinicians, often encountered in real-life scenarios. In the face of CDI, we are increasingly equipped with new therapeutic strategies, such as monoclonal antibodies and live biotherapeutic products, which need to be thoroughly understood to fully harness their benefits. Moreover, interesting options are currently under study for the future, including bacteriophages, vaccines, and antibiotic inhibitors. Surveillance and prevention strategies continue to play a pivotal role in limiting the spread of the infection. In this review, we aim to provide the reader with a comprehensive overview of epidemiological aspects, predisposing factors, clinical manifestations, diagnostic tools, and current and future prophylactic and therapeutic options for C. difficile infection.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium , Humanos , Infecciones por Clostridium/epidemiología , Infecciones por Clostridium/prevención & control , Infecciones por Clostridium/terapia , Factores de Riesgo , Infección Hospitalaria/epidemiología , Infección Hospitalaria/prevención & control , Infección Hospitalaria/microbiología , Antibacterianos/uso terapéutico , Historia del Siglo XXI
5.
CA Cancer J Clin ; 68(5): 340-355, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29985544

RESUMEN

Therapies in oncology have evolved rapidly over the last years. At the same pace, supportive care for patients receiving cancer therapy has also evolved, allowing patients to safely receive the newest advances in treatment in both an inpatient and outpatient basis. The recognition of the role of infection control and prevention (ICP) in the outcomes of patients living with cancer has been such that it is now a requirement for hospitals and involves multidisciplinary groups. Some unique aspects of ICP for patients with cancer that have gained momentum over the past few decades include catheter-related infections, multidrug-resistant organisms, community-acquired viral infections, and the impact of the health care environment on the horizontal transmission of organisms. Furthermore, as the potential for infections to cross international borders has increased, alertness for outbreaks or new infections that occur outside the area have become constant. As the future approaches, ICP in immunocompromised hosts will continue to integrate emerging disciplines, such as antibiotic stewardship and the microbiome, and new techniques for environmental cleaning and for controlling the spread of infections, such as whole-genome sequencing. CA Cancer J Clin 2018;000:000-000. © 2018 American Cancer Society.


Asunto(s)
Instituciones Oncológicas/normas , Huésped Inmunocomprometido , Control de Infecciones/métodos , Control de Infecciones/normas , Neoplasias/inmunología , Atención Ambulatoria/normas , Antibacterianos/uso terapéutico , Antifúngicos/uso terapéutico , Programas de Optimización del Uso de los Antimicrobianos , Infecciones Relacionadas con Catéteres/prevención & control , Infección Hospitalaria/microbiología , Infección Hospitalaria/prevención & control , Dieta , Farmacorresistencia Bacteriana Múltiple , Inocuidad de los Alimentos , Desinfección de las Manos , Humanos , Aislamiento de Pacientes , Estados Unidos , Virosis/prevención & control
6.
Emerg Infect Dis ; 30(13): S41-S48, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38561639

RESUMEN

Serratia marcescens is an environmental gram-negative bacterium that causes invasive disease in rare cases. During 2020-2022, an outbreak of 21 invasive Serratia infections occurred in a prison in California, USA. Most (95%) patients had a history of recent injection drug use (IDU). We performed whole-genome sequencing and found isolates from 8 patients and 2 pieces of IDU equipment were closely related. We also identified social interactions among patients. We recovered S. marcescens from multiple environmental samples throughout the prison, including personal containers storing Cell Block 64 (CB64), a quaternary ammonium disinfectant solution. CB64 preparation and storage conditions were suboptimal for S. marcescens disinfection. The outbreak was likely caused by contaminated CB64 and propagated by shared IDU equipment and social connections. Ensuring appropriate preparation, storage, and availability of disinfectants and enacting interventions to counteract disease spread through IDU can reduce risks for invasive Serratia infections in California prisons.


Asunto(s)
Infección Hospitalaria , Desinfectantes , Prisioneros , Infecciones por Serratia , Humanos , Serratia marcescens/genética , Infecciones por Serratia/epidemiología , Prisiones , Infección Hospitalaria/microbiología , Brotes de Enfermedades , California/epidemiología
7.
Emerg Infect Dis ; 30(5): 908-915, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38666567

RESUMEN

Considering patient room shortages and prevalence of other communicable diseases, reassessing the isolation of patients with Clostridioides difficile infection (CDI) is imperative. We conducted a retrospective study to investigate the secondary CDI transmission rate in a hospital in South Korea, where patients with CDI were not isolated. Using data from a real-time locating system and electronic medical records, we investigated patients who had both direct and indirect contact with CDI index patients. The primary outcome was secondary CDI transmission, identified by whole-genome sequencing. Among 909 direct and 2,711 indirect contact cases, 2 instances of secondary transmission were observed (2 [0.05%] of 3,620 cases), 1 transmission via direct contact and 1 via environmental sources. A low level of direct contact (113 minutes) was required for secondary CDI transmission. Our findings support the adoption of exhaustive standard preventive measures, including environmental decontamination, rather than contact isolation of CDI patients in nonoutbreak settings.


Asunto(s)
Clostridioides difficile , Infecciones por Clostridium , Humanos , Infecciones por Clostridium/transmisión , Infecciones por Clostridium/epidemiología , Infecciones por Clostridium/microbiología , Clostridioides difficile/genética , Clostridioides difficile/aislamiento & purificación , República de Corea/epidemiología , Estudios Retrospectivos , Femenino , Masculino , Infección Hospitalaria/epidemiología , Infección Hospitalaria/transmisión , Infección Hospitalaria/microbiología , Factores de Tiempo , Anciano , Persona de Mediana Edad , Adulto , Trazado de Contacto
8.
Emerg Infect Dis ; 30(6): 1069-1076, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38781679

RESUMEN

Antimicrobial resistance in healthcare-associated bacterial pathogens and the infections they cause are major public health threats affecting nearly all healthcare facilities. Antimicrobial-resistant bacterial infections can occur when colonizing pathogenic bacteria that normally make up a small fraction of the human microbiota increase in number in response to clinical perturbations. Such infections are especially likely when pathogens are resistant to the collateral effects of antimicrobial agents that disrupt the human microbiome, resulting in loss of colonization resistance, a key host defense. Pathogen reduction is an emerging strategy to prevent transmission of, and infection with, antimicrobial-resistant healthcare-associated pathogens. We describe the basis for pathogen reduction as an overall prevention strategy, the evidence for its effectiveness, and the role of the human microbiome in colonization resistance that also reduces the risk for infection once colonized. In addition, we explore ideal attributes of current and future pathogen-reducing approaches.


Asunto(s)
Antibacterianos , Infección Hospitalaria , Farmacorresistencia Bacteriana , Humanos , Infección Hospitalaria/prevención & control , Infección Hospitalaria/microbiología , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Microbiota/efectos de los fármacos , Infecciones Bacterianas/prevención & control , Infecciones Bacterianas/microbiología , Control de Infecciones/métodos , Bacterias/efectos de los fármacos
9.
Antimicrob Agents Chemother ; 68(7): e0057424, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38899928

RESUMEN

FIM-1 metallo-ß-lactamase was previously detected in sporadic Pseudomonas aeruginosa clinical isolates. Here, we report on FIM-1-positive P. aeruginosa from two patients who had shared the same ward in a long-term acute care rehabilitation hospital. Whole-genome sequencing analysis revealed close relatedness of these isolates, which belonged to an ST235 sublineage (clade 8/14) different from those previously reported. Results highlighted the occurrence of clonal diversity among FIM-positive strains and the possibility of their cross-transmission in some healthcare settings.


Asunto(s)
Antibacterianos , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Secuenciación Completa del Genoma , beta-Lactamasas , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/efectos de los fármacos , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Humanos , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/tratamiento farmacológico , Antibacterianos/farmacología , Pruebas de Sensibilidad Microbiana , Hospitales de Rehabilitación , Infección Hospitalaria/microbiología , Masculino
10.
Antimicrob Agents Chemother ; 68(5): e0171623, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38506550

RESUMEN

Outbreaks caused by vancomycin-resistant enterococci that transcend jurisdictional boundaries are occurring worldwide. This study focused on a vancomycin-resistant enterococcus outbreak that occurred between 2018 and 2021 across two cities in Hiroshima, Japan. The study involved genetic and phylogenetic analyses using whole-genome sequencing of 103 isolates of vancomycin-resistant enterococci to identify the source and transmission routes of the outbreak. Phylogenetic analysis was performed using core genome multilocus sequence typing and core single-nucleotide polymorphisms; infection routes between hospitals were inferred using BadTrIP. The outbreak was caused by Enterococcus faecium sequence type (ST) 80 carrying the vanA plasmid, which was derived from strain A10290 isolated in India. Of the 103 isolates, 93 were E. faecium ST80 transmitted across hospitals. The circular vanA plasmid of the Hiroshima isolates was similar to the vanA plasmid of strain A10290 and transferred from E. faecium ST80 to other STs of E. faecium and other Enterococcus species by conjugation. The inferred transmission routes across hospitals suggest the existence of a central hospital serving as a hub, propagating vancomycin-resistant enterococci to multiple hospitals. Our study highlights the importance of early intervention at the key central hospital to prevent the spread of the infection to small medical facilities, such as nursing homes, with limited medical resources and a high number of vulnerable individuals.


Asunto(s)
Brotes de Enfermedades , Enterococcus faecium , Infecciones por Bacterias Grampositivas , Tipificación de Secuencias Multilocus , Filogenia , Plásmidos , Enterococos Resistentes a la Vancomicina , Secuenciación Completa del Genoma , Enterococcus faecium/genética , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/aislamiento & purificación , Japón/epidemiología , Humanos , Enterococos Resistentes a la Vancomicina/genética , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/aislamiento & purificación , Plásmidos/genética , Infecciones por Bacterias Grampositivas/transmisión , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/epidemiología , Infección Hospitalaria/microbiología , Infección Hospitalaria/transmisión , Infección Hospitalaria/epidemiología , Proteínas Bacterianas/genética , Antibacterianos/farmacología , Ligasas de Carbono-Oxígeno/genética , Pruebas de Sensibilidad Microbiana , Polimorfismo de Nucleótido Simple , Hospitales , Vancomicina/farmacología , Genoma Bacteriano/genética
11.
J Clin Microbiol ; 62(6): e0014924, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38690881

RESUMEN

We identified 23 cases of Mycobacterium immunogenum respiratory acquisition linked to a colonized plumbing system at a new hospital addition. We conducted a genomic and epidemiologic investigation to assess for clonal acquisition of M. immunogenum from hospital water sources and improve understanding of genetic distances between M. immunogenum isolates. We performed whole-genome sequencing on 28 M. immunogenum isolates obtained from August 2013 to July 2021 from patients and water sources on four intensive care and intermediate units at an academic hospital. Study hospital isolates were recovered from 23 patients who experienced de novo respiratory isolation of M. immunogenum and from biofilms obtained from five tap water outlets. We also analyzed 10 M. immunogenum genomes from previously sequenced clinical (n = 7) and environmental (n = 3) external control isolates. The 38-isolate cohort clustered into three clades with pairwise single-nucleotide polymorphism (SNP) distances ranging from 0 to 106,697 SNPs. We identified two clusters of study hospital isolates in Clade 1 and one cluster in Clade 2 for which clinical and environmental isolates differed by fewer than 10 SNPs and had less than 0.5% accessory genome variation. A less restrictive combined threshold of 40 SNPs and 5% accessory genes reliably captured additional isolates that met clinical criteria for hospital acquisition, but 12 (4%) of 310 epidemiologically unrelated isolate pairs also met this threshold. Core and accessory genome analyses confirmed respiratory acquisition of multiple clones of M. immunogenum from hospital water sources to patients. When combined with epidemiologic investigation, genomic thresholds accurately distinguished hospital acquisition.


Asunto(s)
Polimorfismo de Nucleótido Simple , Secuenciación Completa del Genoma , Humanos , Genoma Bacteriano , Hospitales , Agua Potable/microbiología , Mycobacterium/genética , Mycobacterium/clasificación , Mycobacterium/aislamiento & purificación , Masculino , Microbiología del Agua , Genómica , Femenino , Persona de Mediana Edad , Anciano , Infección Hospitalaria/microbiología , Infección Hospitalaria/epidemiología , Infecciones por Mycobacterium no Tuberculosas/epidemiología , Infecciones por Mycobacterium no Tuberculosas/microbiología , Adulto
12.
J Clin Microbiol ; 62(7): e0020324, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38934681

RESUMEN

In this study, we investigated the genomic changes in a major methicillin-resistant Staphylococcus aureus (MRSA) clone following a significant outbreak at a hospital. Whole-genome sequencing of MRSA isolates was utilized to explore the genomic evolution of post-outbreak MRSA strains. The epidemicity of the clone declined over time, coinciding with the introduction of multimodal infection control measures. A genome-wide association study (GWAS) identified multiple genes significantly associated with either high or low epidemic success, indicating alterations in mobilome, virulence, and defense mechanisms. Random Forest models pinpointed a gene related to fibrinogen binding as the most influential predictor of epidemicity. The decline of the MRSA clone may be attributed to various factors, including the implementation of new infection control measures, single nucleotide polymorphisms accumulation, and the genetic drift of a given clone. This research underscores the complex dynamics of MRSA clones, emphasizing the multifactorial nature of their evolution. The decline in epidemicity seems linked to alterations in the clone's genetic profile, with a probable shift towards decreased virulence and adaptation to long-term carriage. Understanding the genomic basis for the decline of epidemic clones is crucial to develop effective strategies for their surveillance and management, as well as to gain insights into the evolutionary dynamics of pathogen genomes.


Asunto(s)
Infección Hospitalaria , Brotes de Enfermedades , Evolución Molecular , Genoma Bacteriano , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Secuenciación Completa del Genoma , Humanos , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/epidemiología , Infección Hospitalaria/microbiología , Infección Hospitalaria/epidemiología , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Staphylococcus aureus Resistente a Meticilina/clasificación , Genoma Bacteriano/genética , Estudio de Asociación del Genoma Completo , Polimorfismo de Nucleótido Simple , Epidemiología Molecular
13.
J Antimicrob Chemother ; 79(7): 1529-1539, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38751093

RESUMEN

OBJECTIVES: Comprehensive data on the genomic epidemiology of hospital-associated Klebsiella pneumoniae in Ghana are scarce. This study investigated the genomic diversity, antimicrobial resistance patterns, and clonal relationships of 103 clinical K. pneumoniae isolates from five tertiary hospitals in Southern Ghana-predominantly from paediatric patients aged under 5 years (67/103; 65%), with the majority collected from urine (32/103; 31%) and blood (25/103; 24%) cultures. METHODS: We generated hybrid Nanopore-Illumina assemblies and employed Pathogenwatch for genotyping via Kaptive [capsular (K) locus and lipopolysaccharide (O) antigens] and Kleborate (antimicrobial resistance and hypervirulence) and determined clonal relationships using core-genome MLST (cgMLST). RESULTS: Of 44 distinct STs detected, ST133 was the most common, comprising 23% of isolates (n = 23/103). KL116 (28/103; 27%) and O1 (66/103; 64%) were the most prevalent K-locus and O-antigen types. Single-linkage clustering highlighted the global spread of MDR clones such as ST15, ST307, ST17, ST11, ST101 and ST48, with minimal allele differences (1-5) from publicly available genomes worldwide. Conversely, 17 isolates constituted novel clonal groups and lacked close relatives among publicly available genomes, displaying unique genetic diversity within our study population. A significant proportion of isolates (88/103; 85%) carried resistance genes for ≥3 antibiotic classes, with the blaCTX-M-15 gene present in 78% (n = 80/103). Carbapenem resistance, predominantly due to blaOXA-181 and blaNDM-1 genes, was found in 10% (n = 10/103) of the isolates. CONCLUSIONS: Our findings reveal a complex genomic landscape of K. pneumoniae in Southern Ghana, underscoring the critical need for ongoing genomic surveillance to manage the substantial burden of antimicrobial resistance.


Asunto(s)
Antibacterianos , Variación Genética , Infecciones por Klebsiella , Klebsiella pneumoniae , Tipificación de Secuencias Multilocus , Centros de Atención Terciaria , Humanos , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/aislamiento & purificación , Centros de Atención Terciaria/estadística & datos numéricos , Ghana/epidemiología , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/epidemiología , Antibacterianos/farmacología , Preescolar , Lactante , Pruebas de Sensibilidad Microbiana , Genotipo , Femenino , Masculino , Niño , Farmacorresistencia Bacteriana Múltiple/genética , Infección Hospitalaria/microbiología , Infección Hospitalaria/epidemiología , Genoma Bacteriano , Farmacorresistencia Bacteriana/genética , Adulto , Epidemiología Molecular
14.
Int J Med Microbiol ; 314: 151605, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38290401

RESUMEN

The threat of multidrug-resistant organisms (MDROs) and antimicrobial resistance (AMR) are real and increasing every day. They affect not only healthcare systems but also communities, causing economic and public health concerns. Governments must take action to tackle AMR and prevent the spread of MDROs and regional hubs have a critical role to play in achieving this outcome. Furthermore, bacteria have no borders, consequently, cooperation networks should be extended between countries as a crucial strategy for achieving the success of infection control. Euregions, which are a specific form of cooperation between local authorities of two or more bordering European countries, can help solve common problems and improve the lives of people living on both sides of the border. Regional collaboration strategies can enhance infection control and build resilience against antimicrobial resistance. This review identifies risk factors and the correct approaches to infection prevention and control, including education and awareness programs for healthcare professionals, appropriate prescribing practices, and infection prevention control measures. These measures can help reduce the incidence of antimicrobial resistance in the region and save lives. It is therefore essential to take concrete actions and foster the creation of more effective regional and cross-border centers to ensure the success of infection control policies and the management of healthcare-associated infections. This work sheds light on the issue of MDRO infections within healthcare settings, while also acknowledging the crucial role of the One Health concept in understanding the broader context of these infections. By recognizing the interdependence of human and animal health and the environment, we can take constructive steps toward mitigating the risks of these infections and promoting better health outcomes for all.


Asunto(s)
Antiinfecciosos , Infección Hospitalaria , Animales , Humanos , Infección Hospitalaria/microbiología , Control de Infecciones , Europa (Continente)/epidemiología , Factores de Riesgo
15.
BMC Microbiol ; 24(1): 184, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802754

RESUMEN

BACKGROUND: Pseudomonas aeruginosa is an opportunistic pathogen responsible for complicated UTIs and exhibits high antibiotic resistance, leading to increased mortality rates, especially in cases of multidrug-resistant strains. This study aimed to investigate the antibiotic susceptibility patterns and genomic characterization of XDR strains identified in end-stage liver disease patients who underwent liver transplants. METHODS: In this study, a number of 30 individuals who underwent liver transplants were registered. Ninety urine and 60 wound site swab samples were collected and processed for culturing, identification, and antimicrobial sensitivity. Extensively drug-resistant strain EMARA01 was confirmed through Sanger sequencing and was then processed for whole genome sequencing to characterize the genomic pattern. Sequencing data were processed for de novo assembly using various tools and databases, including genome annotation, serotype identification, virulence factor genes, and antimicrobial resistance gene. Pangenome analysis of randomly selected 147 reference strains and EMAR01 sequenced strain was performed using the Bacterial Pan Genome Analysis (BPGA) software. RESULTS: Of these total examined samples, nosocomial infection due to P. aeruginosa was detected in twelve patients' samples. AST analysis showed that P. aeruginosa strains exhibit resistance to tobramycin, erythromycin, and gentamicin, followed by piperacillin and ofloxacin, and no strains exhibit resistance to meropenem and imipenem. The CARD database identified 59 AMR genes similar to the EMAR01 strain genome and mostly belong to the family involved in the resistance-nodulation-cell division (RND) antibiotic efflux pump. Five genes; nalC, nalD, MexR, MexA, and MexB, exhibit resistance to 14 classes of antibiotics, while two AMR; CpxR, and OprM, exhibit resistance to 15 classes of drugs. Pangenome analysis revealed that the pan-genome remained open, suggesting the potential for acquiring accessory and unique genes. Notably, the genes predominantly involved in amino acid transport metabolism were identified using the KEGG database. CONCLUSIONS: This study provides valuable insights into the antimicrobial resistance profile, genetic features, and genomic evolution of P. aeruginosa strains causing UTIs in liver transplant patients. The findings emphasize the significance of comprehending AMR mechanisms and genetic diversity in P. aeruginosa for developing effective treatment strategies and infection control measures.


Asunto(s)
Antibacterianos , Farmacorresistencia Bacteriana Múltiple , Trasplante de Hígado , Pruebas de Sensibilidad Microbiana , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Infecciones Urinarias , Secuenciación Completa del Genoma , Humanos , Farmacorresistencia Bacteriana Múltiple/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/aislamiento & purificación , Trasplante de Hígado/efectos adversos , Egipto , Infecciones Urinarias/microbiología , Infecciones por Pseudomonas/microbiología , Antibacterianos/farmacología , Masculino , Femenino , Genoma Bacteriano/genética , Adulto , Infección Hospitalaria/microbiología , Persona de Mediana Edad , Factores de Virulencia/genética
16.
BMC Microbiol ; 24(1): 215, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38890594

RESUMEN

BACKGROUND: A multidrug-resistant lineage of Staphylococcus epidermidis named ST215 is a common cause of prosthetic joint infections and other deep surgical site infections in Northern Europe, but is not present elsewhere. The increasing resistance among S. epidermidis strains is a global concern. We used whole-genome sequencing to characterize ST215 from healthcare settings. RESULTS: We completed the genome of a ST215 isolate from a Swedish hospital using short and long reads, resulting in a circular 2,676,787 bp chromosome and a 2,326 bp plasmid. The new ST215 genome was placed in phylogenetic context using 1,361 finished public S. epidermidis reference genomes. We generated 10 additional short-read ST215 genomes and 11 short-read genomes of ST2, which is another common multidrug-resistant lineage at the same hospital. We studied recombination's role in the evolution of ST2 and ST215, and found multiple recombination events averaging 30-50 kb. By comparing the results of antimicrobial susceptibility testing for 31 antimicrobial drugs with the genome content encoding antimicrobial resistance in the ST215 and ST2 isolates, we found highly similar resistance traits between the isolates, with 22 resistance genes being shared between all the ST215 and ST2 genomes. The ST215 genome contained 29 genes that were historically identified as virulence genes of S. epidermidis ST2. We established that in the nucleotide sequence stretches identified as recombination events, virulence genes were overrepresented in ST215, while antibiotic resistance genes were overrepresented in ST2. CONCLUSIONS: This study features the extensive antibiotic resistance and virulence gene content in ST215 genomes. ST215 and ST2 lineages have similarly evolved, acquiring resistance and virulence through genomic recombination. The results highlight the threat of new multidrug-resistant S. epidermidis lineages emerging in healthcare settings.


Asunto(s)
Antibacterianos , Infección Hospitalaria , Farmacorresistencia Bacteriana Múltiple , Genoma Bacteriano , Filogenia , Infecciones Estafilocócicas , Staphylococcus epidermidis , Secuenciación Completa del Genoma , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/aislamiento & purificación , Staphylococcus epidermidis/patogenicidad , Farmacorresistencia Bacteriana Múltiple/genética , Genoma Bacteriano/genética , Humanos , Infecciones Estafilocócicas/microbiología , Infección Hospitalaria/microbiología , Antibacterianos/farmacología , Pruebas de Sensibilidad Microbiana , Suecia , Plásmidos/genética , Recombinación Genética
17.
BMC Microbiol ; 24(1): 152, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702660

RESUMEN

BACKGROUND: Pseudomonas aeruginosa is a common cause of nosocomial infections. However, the emergence of multidrug-resistant strains has complicated the treatment of P. aeruginosa infections. While polymyxins have been the mainstay for treatment, there is a global increase in resistance to these antibiotics. Therefore, our study aimed to determine the prevalence and molecular details of colistin resistance in P. aeruginosa clinical isolates collected between June 2019 and May 2023, as well as the genetic linkage of colistin-resistant P. aeruginosa isolates. RESULTS: The resistance rate to colistin was 9% (n = 18) among P. aeruginosa isolates. All 18 colistin-resistant isolates were biofilm producers and carried genes associated with biofilm formation. Furthermore, the presence of genes encoding efflux pumps, TCSs, and outer membrane porin was observed in all colistin-resistant P. aeruginosa strains, while the mcr-1 gene was not detected. Amino acid substitutions were identified only in the PmrB protein of multidrug- and colistin-resistant strains. The expression levels of mexA, mexC, mexE, mexY, phoP, and pmrA genes in the 18 colistin-resistant P. aeruginosa strains were as follows: 88.8%, 94.4%, 11.1%, 83.3%, 83.3%, and 38.8%, respectively. Additionally, down-regulation of the oprD gene was observed in 44.4% of colistin-resistant P. aeruginosa strains. CONCLUSION: This study reports the emergence of colistin resistance with various mechanisms among P. aeruginosa strains in Ardabil hospitals. We recommend avoiding unnecessary use of colistin to prevent potential future increases in colistin resistance.


Asunto(s)
Antibacterianos , Proteínas Bacterianas , Colistina , Pruebas de Sensibilidad Microbiana , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Factores de Transcripción , Colistina/farmacología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/aislamiento & purificación , Antibacterianos/farmacología , Humanos , Proteínas Bacterianas/genética , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/epidemiología , Prevalencia , Farmacorresistencia Bacteriana Múltiple/genética , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Hospitales , Farmacorresistencia Bacteriana/genética , Infección Hospitalaria/microbiología , Infección Hospitalaria/epidemiología , Proteínas de Transporte de Membrana/genética , Porinas/genética
18.
BMC Microbiol ; 24(1): 225, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926687

RESUMEN

BACKGROUND: The incidence of hospital-acquired infections in extensively drug-resistant Pseudomonas aeruginosa (XDR-PA) has been increasing worldwide and is frequently associated with an increase in mortality and morbidity rates. The aim of this study was to characterize clinical XDR-PA isolates recovered during six months at three different hospitals in Egypt. RESULTS: Seventy hospital-acquired clinical isolates of P. aeruginosa were classified into multidrug-resistant (MDR), extensively drug-resistant (XDR) and pandrug-resistant (PDR), according to their antimicrobial resistance profile. In addition, the possession of genes associated with mobile genetic elements and genes encoding antimicrobial resistance determinants among isolates were detected using polymerase chain reaction. As a result, a significant percentage of the isolates (75.7%) were XDR, while 18.5% were MDR, however only 5.7% of the isolates were non-MDR. The phenotypic detection of carbapenemases, extended-spectrum ß-lactamases (ESBLs) and metallo ß-lactamase (MBL) enzymes showed that 73.6% of XDR-PA isolates were carbapenemases producers, whereas 75.5% and 88.7% of XDR-PA isolates produced ESBLs and MBL respectively. In addition, PCR screening showed that oxa gene was the most frequently detected gene of carbapenemases (91.4%), while aac(6')-lb gene was mostly detected (84.3%) among the screened aminoglycosides-resistance genes. Furthermore, the molecular detection of the colistin resistance gene showed that 12.9% of isolates harbored mcr-1 gene. Concerning mobile genetic element markers (intI, traA, tnp513, and merA), intI was the highest detected gene as it was amplified in 67 isolates (95.7%). Finally, phylogenetic and molecular typing of the isolates via ERIC-PCR analysis revealed 10 different ERIC fingerprints. CONCLUSION: The present study revealed a high prevalence of XDR-PA in hospital settings which were resistant to a variety of antibiotics due to several mechanisms. In addition, 98% of the XDR-PA clinical isolates contained at least one gene associated with movable genetic elements, which could have aided the evolution of these XDR-PA strains. To reduce spread of drug resistance, judicious use of antimicrobial agents and strict infection control measures are therefore essential.


Asunto(s)
Antibacterianos , Infección Hospitalaria , Farmacorresistencia Bacteriana Múltiple , Pruebas de Sensibilidad Microbiana , Infecciones por Pseudomonas , Pseudomonas aeruginosa , beta-Lactamasas , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/aislamiento & purificación , Humanos , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/epidemiología , Farmacorresistencia Bacteriana Múltiple/genética , Infección Hospitalaria/microbiología , Infección Hospitalaria/epidemiología , Egipto/epidemiología , beta-Lactamasas/genética , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Hospitales/estadística & datos numéricos , Secuencias Repetitivas Esparcidas/genética , Reacción en Cadena de la Polimerasa
19.
Microb Pathog ; 190: 106637, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38570103

RESUMEN

We seek to investigate the multifaceted factors influencing secondary infections in patients with multidrug-resistant Gram-negative bacteria (MDR-GNB) colonization or infection post-hospitalization. A total of 100 patients with MDR-GNB colonization or infection were retrospectively reviewed, encompassing those admitted to both the general ward and intensive care unit of our hospital from August 2021 to December 2022. Patients were categorized into the control group (non-nosocomial infection, n = 56) and the observation group (nosocomial infection, n = 44) based on the occurrence of nosocomial infection during hospitalization. Clinical data were compared between the two groups, including the distribution and antibiotic sensitivity of MDR-GNB before nosocomial infection. Significant differences were observed between the two groups in terms of age, underlying diseases, immune status, length of stay, and invasive medical procedures (P < 0.05). The observation group also had fewer patients practicing optimized hygiene, strict isolation, and antibiotic control than the control group (P < 0.05). Factors influencing the risk of secondary infection after hospitalization in patients colonized or infected with MDR-GNB included patient age, underlying diseases, immune status, length of hospitalization, medical invasive procedures, optimized hygiene, strict isolation, and antibiotic control (P < 0.05). The length of hospitalization and treatment cost in the observation group were higher than those in the control group (P < 0.05). This study comprehensively analyzes the intricate mechanisms of secondary infections in patients with MDR-GNB infections post-hospitalization. Key factors influencing infection risk include patient age, underlying diseases, immune status, length of hospitalization, medical invasive procedures, optimized hygiene, strict isolation, and antibiotic control.


Asunto(s)
Antibacterianos , Infección Hospitalaria , Farmacorresistencia Bacteriana Múltiple , Bacterias Gramnegativas , Infecciones por Bacterias Gramnegativas , Hospitalización , Humanos , Masculino , Femenino , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/epidemiología , Infección Hospitalaria/microbiología , Infección Hospitalaria/epidemiología , Estudios Retrospectivos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/aislamiento & purificación , Persona de Mediana Edad , Hospitalización/estadística & datos numéricos , Anciano , Factores de Riesgo , Antibacterianos/uso terapéutico , Antibacterianos/farmacología , Tiempo de Internación , Adulto , Anciano de 80 o más Años , Unidades de Cuidados Intensivos/estadística & datos numéricos
20.
Eur J Clin Microbiol Infect Dis ; 43(1): 187-194, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37971537

RESUMEN

This study aimed to map MDRO carriage and potential transmission within and between three Flemish tertiary care hospitals and their neighbouring nursing homes. A cross-sectional MDRO prevalence survey was organized between October 2017 and February 2019. Perianal swabs were cultured for detection of MDRO. Determination of clonal relatedness based on wgMLST allelic profiles was performed. The prevalence of MDRO in Belgian hospitals and NHs is on the rise, compared to previous studies, and transmission in and between institutions is observed. These results re-emphasize the need for a healthcare network-wide infection prevention strategy in which WGS of MDRO strains can be supportive.


Asunto(s)
Infección Hospitalaria , Casas de Salud , Humanos , Bélgica/epidemiología , Centros de Atención Terciaria , Estudios Transversales , Farmacorresistencia Bacteriana Múltiple , Bacterias , Tipificación Molecular , Infección Hospitalaria/epidemiología , Infección Hospitalaria/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA