Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Front Med (Lausanne) ; 8: 687415, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34746164

RESUMO

Objective: To analyze the availability and access to the hospital for the patients with intra-abdominal infections (IAIs) by Escherichia coli (E. coli) as a result of the coronavirus disease 2019 (COVID-19) pandemic and the impact of these changes in the diagnosis and their effects on the death of these patients. Methods: Two prospective observational cohorts of the patients with IAI by E. coli were conducted in 2016 (the pre-COVID-19, n = 108) and in 2020 (during the COVID-19, n = 96) at the University Hospital of Seville, Spain. The demographic and clinical variables of the patients were collected and analyzed. The patients were followed-up for 120 days, until the hospital discharge or death. The bivariate and multivariate analyses were performed. Results: Both the cohorts were homogeneous according to age, sex, emergency surgery cause, immunosuppression, neutropenia, acquisition type, and previous intervention. The patients attended during the COVID-19 had significantly higher Charlson comorbidity index and the more McCabe score, required more emergency surgery, had more severe infections with the higher rates of septic shock and sepsis, and the presence of additional care support such as a nasogastric tube. They were diagnosed later; the time intervals between the symptoms onset (SO) to the first medical contact or surgical intervention (SI) and between the first medical contact to the admission or SI were significantly higher. The death rates during the COVID-19 and the pre-COVID-19 were 16.7 and 6.5%, respectively (p = 0.02). Finally, the multivariate analysis in both the cohorts together identified the patients diagnosed during the COVID-19, the longer period from SO to SI, septic shock, and the Charlson comorbidity index as the independent factors associated with death. Conclusion: This study showed the impact of the COVID-19 pandemic on the clinical outcome and death due to IAI with an extension of the time between SO and SI.

3.
BMJ Open ; 10(7): e034570, 2020 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-32690735

RESUMO

INTRODUCTION: Haematopoietic stem cell transplantation (HSCT) is a life-saving treatment for a number of haematological diseases. Graft versus host disease (GVHD) is its main complication and hampers survival. There is strong evidence that intestinal microbiota diversity of the recipient may increase the risk of GVHD worsening survival. Antibiotic regimens used during the early phase of the transplant may influence clinical outcomes by reducing intestinal microbiota diversity. Present guidelines of European Conference on Infections in Leukaemia exhort to optimising antibiotic use in haematological patients including HSCT recipients. The present study aims to investigate if, in HSCT recipients, the optimisation of antibacterial use may preserve intestinal microbiota composition reducing the incidence and severity of acute GVHD and improving relevant clinical outcomes. METHODS AND ANALYSIS: This is a prospective longitudinal observational study of two cohorts of HSCT recipients: (1) the intervention cohort includes patients treated in centres in which a predefined strategy of antibiotherapy optimisation is implemented, with the objective of optimising and reducing antibiotic administration according to clinical criteria and (2) the control cohort includes patients treated in centres in which a classic permissive strategy of antibiotic prophylaxis and treatment is used. Adult patient receiving a first HSCT as a treatment for any haematological condition are included. Clinical variables are prospectively recorded and up to five faecal samples are collected for microbiota characterisation at prestablished peritransplant time points. Patients are followed since the preconditioning phase throughout 1-year post-transplant and four follow-up visits are scheduled. Faecal microbiota composition and diversity will be compared between both cohorts along with acute GVHD incidence and severity, severe infections rate, mortality and overall and disease-free survival. ETHICS AND DISSEMINATION: The study was approved between 2017 and 2018 by the Ethical Committees of participant centres. Study results will be disseminated through peer-reviewed journals and national and international scientific conferences. TRIAL REGISTRATION NUMBER: NCT03727113.


Assuntos
Antibioticoprofilaxia , Gestão de Antimicrobianos , Microbioma Gastrointestinal , Transplante de Células-Tronco Hematopoéticas , Estudos Observacionais como Assunto , Transplantados , Estudos de Casos e Controles , Fezes/microbiologia , Doença Enxerto-Hospedeiro , Humanos , Estudos Longitudinais , Estudos Multicêntricos como Assunto , Estudos Prospectivos , Projetos de Pesquisa
4.
J Infect Dis ; 222(7): 1204-1212, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32324853

RESUMO

Acinetobacter baumannii is a successful pathogen responsible for infections with high mortality rate. During the course of infection it can be found in microaerobic environments, which influences virulence factor expression. From a previous transcriptomic analysis of A. baumannii ATCC 17978 under microaerobiosis, we know the gene pstS is overexpressed under microaerobiosis. Here, we studied its role in A. baumannii virulence. pstS loss significantly decreased bacterial adherence and invasion into A549 cells and increased A549 cell viability. pstS loss also reduced motility and biofilm-forming ability of A. baumannii. In a peritoneal sepsis murine model, the minimum lethal dose required by A. baumannii ATCC 17978 ΔpstS was lower compared to the wild type (4.3 vs 3.2 log colony forming units/mL, respectively), and the bacterial burden in tissues and fluids was lower. Thus, the loss of the phosphate sensor PstS produced a decrease in A. baumannii pathogenesis, supporting its role as a virulence factor.


Assuntos
Acinetobacter baumannii/genética , Acinetobacter baumannii/patogenicidade , Proteínas de Bactérias/genética , Proteínas de Ligação a Fosfato/genética , Células A549 , Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/crescimento & desenvolvimento , Aerobiose , Animais , Aderência Bacteriana/genética , Biofilmes , Morte Celular , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Feminino , Regulação Bacteriana da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/farmacologia , Peritonite/microbiologia , Virulência/genética , Fatores de Virulência/genética
5.
Infect Immun ; 86(10)2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30082478

RESUMO

Hypoxia modulates bacterial virulence and the inflammation response through hypoxia-inducible factor 1α (HIF-1α). Here we study the influence of hypoxia on Acinetobacter baumannii and Pseudomonas aeruginosa infections. In vitro, hypoxia increases the bactericidal activities of epithelial cells against A. baumannii and P. aeruginosa, reducing extracellular bacterial concentrations to 50.5% ± 7.5% and 90.8% ± 13.9%, respectively, at 2 h postinfection. The same phenomenon occurs in macrophages (67.6% ± 18.2% for A. baumannii at 2 h and 50.3% ± 10.9% for P. aeruginosa at 24 h). Hypoxia decreases the adherence of A. baumannii to epithelial cells (42.87% ± 8.16% at 2 h) and macrophages (52.0% ± 18.7% at 24 h), as well as that of P. aeruginosa (24.9% ± 4.5% in epithelial cells and 65.7% ± 5.5% in macrophages at 2 h). Moreover, hypoxia decreases the invasion of epithelial cells (48.6% ± 3.8%) and macrophages (8.7% ± 6.9%) by A. baumannii at 24 h postinfection and by P. aeruginosa at 2 h postinfection (75.0% ± 16.3% and 63.4% ± 5.4%, respectively). In vivo, hypoxia diminishes bacterial loads in fluids and tissues in animal models of infection by both pathogens. In contrast, mouse survival time was shorter under hypoxia (23.92 versus 36.42 h) with A. baumannii infection. No differences in the production of cytokines or HIF-1α were found between hypoxia and normoxia in vitro or in vivo We conclude that hypoxia increases the bactericidal activities of host cells against both pathogens and reduces the interaction of pathogens with host cells. Moreover, hypoxia accelerates the rate at which animals die despite the lower bacterial concentrations in vivo.


Assuntos
Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/patogenicidade , Hipóxia/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade , Infecções por Acinetobacter/genética , Infecções por Acinetobacter/metabolismo , Acinetobacter baumannii/crescimento & desenvolvimento , Acinetobacter baumannii/fisiologia , Animais , Aderência Bacteriana , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Macrófagos/metabolismo , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , Oxigênio/metabolismo , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/fisiologia , Virulência
6.
mSphere ; 3(2)2018.
Artigo em Inglês | MEDLINE | ID: mdl-29600279

RESUMO

Acinetobacter baumannii is a significant human pathogen associated with hospital-acquired infections. While adhesion, an initial and important step in A. baumannii infection, is well characterized, the intracellular trafficking of this pathogen inside host cells remains poorly studied. Here, we demonstrate that transcription factor EB (TFEB) is activated after A. baumannii infection of human lung epithelial cells (A549). We also show that TFEB is required for the invasion and persistence inside A549 cells. Consequently, lysosomal biogenesis and autophagy activation were observed after TFEB activation which could increase the death of A549 cells. In addition, using the Caenorhabditis elegans infection model by A. baumannii, the TFEB orthologue HLH-30 was required for survival of the nematode to infection, although nuclear translocation of HLH-30 was not required. These results identify TFEB as a conserved key factor in the pathogenesis of A. baumannii. IMPORTANCE Adhesion is an initial and important step in Acinetobacter baumannii infections. However, the mechanism of entrance and persistence inside host cells is unclear and remains to be understood. In this study, we report that, in addition to its known role in host defense against Gram-positive bacterial infection, TFEB also plays an important role in the intracellular trafficking of A. baumannii in host cells. TFEB was activated shortly after A. baumannii infection and is required for its persistence within host cells. Additionally, using the C. elegans infection model by A. baumannii, the TFEB orthologue HLH-30 was required for survival of the nematode to infection, although nuclear translocation of HLH-30 was not required.


Assuntos
Infecções por Acinetobacter/metabolismo , Acinetobacter baumannii/fisiologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Interações Hospedeiro-Patógeno , Células A549 , Acinetobacter baumannii/patogenicidade , Animais , Autofagia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Transporte Biológico , Caenorhabditis elegans/microbiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Humanos , Lisossomos/metabolismo
7.
Sci Rep ; 7(1): 14683, 2017 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-29089624

RESUMO

Preventing the adhesion of pathogens to host cells provides an innovative approach to tackling multidrug-resistant bacteria. In this regard, the identification of outer membrane protein A (OmpA) as a key bacterial virulence factor has been a major breakthrough. The use of virtual screening helped us to identify a cyclic hexapeptide AOA-2 that inhibits the adhesion of Acinetobacter baumannii, Pseudomonas aeruginosa and Escherichia coli to host cells and the formation of biofilm, thereby preventing the development of infection in vitro and in a murine sepsis peritoneal model. Inhibition of OmpA offers a strategy as monotherapy to address the urgent need for treatments for infections caused by Gram-negative bacilli.


Assuntos
Infecções por Acinetobacter/imunologia , Acinetobacter baumannii/fisiologia , Células Epiteliais Alveolares/fisiologia , Aderência Bacteriana/efeitos dos fármacos , Proteínas da Membrana Bacteriana Externa/antagonistas & inibidores , Infecções por Escherichia coli/imunologia , Escherichia coli/fisiologia , Peptídeos/metabolismo , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/fisiologia , RNA Helicases/metabolismo , Sepse/imunologia , Fatores de Virulência/antagonistas & inibidores , Infecções por Acinetobacter/tratamento farmacológico , Animais , Antibacterianos/uso terapêutico , Biofilmes , Linhagem Celular , DNA Helicases , Modelos Animais de Doenças , Infecções por Escherichia coli/tratamento farmacológico , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Enzimas Multifuncionais , Peptídeos/genética , Peptídeos/uso terapêutico , Infecções por Pseudomonas/tratamento farmacológico , RNA Helicases/genética , RNA Helicases/uso terapêutico
8.
Antimicrob Agents Chemother ; 59(7): 3920-4, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25896698

RESUMO

Due to the significant increase in antimicrobial resistance of Acinetobacter baumannii, immune system stimulation to block infection progression may be a therapeutic adjuvant to antimicrobial treatment. Lysophosphatidylcholine (LPC), a major component of phospholipids in eukaryotic cells, is involved in immune cell recruitment and modulation. The aim of this study was to show if LPC could be useful for treating infections caused by A. baumannii. A. baumannii ATCC 17978 was used in this study. Levels of serum LPC and levels of the inflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), IL-1ß, and IL-10 were determined by spectrophotometric assay and enzyme-linked immunosorbent assay (ELISA), respectively, using a murine peritoneal sepsis model in which mice were inoculated with 5.3 log CFU/ml of A. baumannii. The therapeutic efficacy of LPC against A. baumannii in murine peritoneal sepsis and pneumonia models was assessed for 48 h after bacterial infection. At early time points in the murine model of peritoneal sepsis caused by A. baumannii, LPC was depleted and was associated with an increase of inflammatory cytokine release. Preemptive therapy with LPC in murine peritoneal sepsis and pneumonia models markedly enhanced spleen and lung bacterial clearance and reduced the numbers of positive blood cultures and the mouse mortality rates. Moreover, treatment with LPC reduced proinflammatory cytokine production. These data demonstrate that LPC is efficacious as a preemptive treatment in experimental models of peritoneal sepsis and pneumonia caused by A. baumannii.


Assuntos
Infecções por Acinetobacter/tratamento farmacológico , Acinetobacter baumannii , Lisofosfatidilcolinas/uso terapêutico , Infecções por Acinetobacter/microbiologia , Infecções por Acinetobacter/mortalidade , Animais , Contagem de Colônia Microbiana , Citocinas/sangue , Farmacorresistência Bacteriana , Feminino , Pulmão/microbiologia , Lisofosfatidilcolinas/sangue , Lisofosfatidilcolinas/farmacocinética , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Pneumonia/microbiologia , Sepse/microbiologia , Baço/microbiologia
9.
Int J Antimicrob Agents ; 43(2): 135-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24315790

RESUMO

This study was designed to evaluate the potential role of fosfomycin as a therapeutic agent in human listeriosis. The in vitro activity of fosfomycin against 154 Listeria monocytogenes clinical isolates under conditions that mimic the induction of prfA expression was determined and was correlated with fosfomycin intracellular antimicrobial activity. In vitro, partial induction of prfA expression is achieved through bacterial growth in brain-heart infusion agar supplemented with activated charcoal (BHIC). A fosfomycin pharmacokinetic/pharmacodynamic breakpoint of ≤64 mg/L was estimated using a Monte Carlo simulation to assess the success of an intravenous fosfomycin dose of 300 mg/kg/day over 5000 individuals. Eighty strains (51.9%) were susceptible to fosfomycin in BHIC, with minimum inhibitory concentrations (MICs) of ≤64 mg/L; 13 strains (8.4%) had the epidemic clone (EC) marker. In addition, 27 strains (17.5%) had a three doubling dilutions reduction in the MIC from ≥1024 mg/L to 128 mg/L (96-128 mg/L by Etest). The fosfomycin modal MIC is lower under prfA expression. However, this effect is smaller in terms of clinical categorisation of isolates and can be influenced by the serotype and clonal type. In A549 cells, the reductions in bacterial inocula of the two susceptible isolates studied after 1h and 24h of incubation with fosfomycin at 0.5× the human maximum serum concentration (Cmax) were 45.8% and 46.6%, and 93.8% and 99.1%, respectively. Slightly higher reductions were found with fosfomycin at 1× Cmax. The resistant strain tested showed significantly lower reductions in all assays.


Assuntos
Antibacterianos/farmacologia , Fosfomicina/farmacologia , Listeria monocytogenes/efeitos dos fármacos , Proteínas de Bactérias/biossíntese , Linhagem Celular , Meios de Cultura/química , Células Epiteliais/microbiologia , Humanos , Testes de Sensibilidade Microbiana , Fatores de Terminação de Peptídeos/biossíntese , Ativação Transcricional
10.
J Infect Dis ; 208(10): 1561-70, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23908480

RESUMO

Outer membrane protein 33 (Omp33) is an outer membrane porin of Acinetobacter baumannii associated with carbapenem resistance. However, the role of Omp33 in the fitness and virulence of A. baumannii remains unknown. In the present study, we investigated the role of Omp33 in fitness and virulence of A. baumannii by using an isogenic knockout strain deficient in the omp33 gene (JPAB02), derived from the ATCC 17978 wild-type (wt). Both in vitro and in vivo defect in the growth rate was found in the JPAB02 strain in competition with the ATCC 17978 wt, highlighting the effect of Omp33 on the metabolic fitness. A significant reduction was observed both in adherence and invasion of human lung epithelial cells and in cytotoxicity of these cells and macrophages with JPAB02. In a murine peritoneal sepsis model, the JPAB02 strain exhibited lower lethal dose 0 (LD0), LD50, and LD100, and dissemination in mice, with reduced bacterial concentration in spleen and lungs. From these data, we concluded that Omp33 plays an important role for fitness and virulence of A. baumannii.


Assuntos
Acinetobacter baumannii/genética , Acinetobacter baumannii/patogenicidade , Proteínas da Membrana Bacteriana Externa/genética , Aptidão Genética , Infecções por Acinetobacter/microbiologia , Infecções por Acinetobacter/mortalidade , Acinetobacter baumannii/crescimento & desenvolvimento , Animais , Aderência Bacteriana/genética , Biofilmes , Morte Celular , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Técnicas de Inativação de Genes , Humanos , Camundongos , Peritonite/microbiologia , Peritonite/mortalidade , Fenótipo , Virulência/genética
11.
J Infect Dis ; 203(8): 1110-9, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21357946

RESUMO

OBJECTIVE: Recently, it has become apparent that rifampin can act on eukaryotic cells modulating production of host mediators. We aimed to study the cytoprotective effect of rifampin against multidrug- and pandrug-resistant Acinetobacter baumannii-induced cell death using human lung epithelial cells. METHODS: We pretreated A549 cells with rifampin and infected them with 3 different A. baumannii strains (susceptible, multidrug-resistant, and pandrug-resistant) that induce cell death. Cellular viability, apoptosis and host mediators, free radicals, and proinflammatory cytokines associated with A. baumannii pathogenesis were studied. Moreover, bacterial concentrations in A549 cells culture were determined. RESULTS: Rifampin-pretreated A549 cells demonstrated decreases in apoptosis and cell death induced by A. baumannii. The oxidative stress and proinflammatory responses to A. baumannii were reduced in rifampin-pretreated A549 cells, as shown by decreased superoxide anion, tumor necrosis factor-α, and interleukin-6. Furthermore, bacterial count performed in A549 cell culture medium showed that rifampin did not reduce significantly the bacterial concentrations. CONCLUSION: These data demonstrate that rifampin is able to attenuate the cellular damage induced by multidrug- and pandrug-resistant A. baumannii clinical isolates without being relevantly bactericidal. Indeed, the cytoprotective effect of rifampin was observed on the decrease of dead cells induced by A. baumannii by reducing oxidative stress and proinflammatory cytokines release.


Assuntos
Acinetobacter baumannii/patogenicidade , Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla , Células Epiteliais/microbiologia , Pulmão/citologia , Rifampina/farmacologia , Apoptose , Linhagem Celular Tumoral , Meios de Cultura , Humanos , Estresse Oxidativo
12.
Microb Pathog ; 50(5): 224-32, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21288481

RESUMO

A growing body of evidence supports the notion that susceptible Acinetobacter baumannii strain ATCC 19606 induces human epithelial cells death. However, most of the cellular and molecular mechanisms associated with this cell death remain unknown, and also the degree of the cytotoxic effects of a clinical panresistant strain compared with a susceptible strain has never been studied. Due to the role of proinflammatory cytokine release, oxidative stress and cytosolic calcium increase in the cell death-induced by other Gram-negative bacteria, we investigated whether these intracellular targets were involved in the cell death induced by clinical panresistant 113-16 and susceptible ATCC 19606 strains. Data presented here show that 113-16 and ATCC 19606 induce time-dependent cell death of lung epithelial cells involving a perturbation of cytosolic calcium homeostasis with subsequent calpain and caspase-3 activation. Prevention of this cell death by TNF-α and interleukin-6 blockers and antioxidant highlights the involvement of proinflammatory cytokines and oxidative stress in this phenomenon. These results demonstrate the involvement of calpain calcium-dependent in cell death induced by A. baumannii and the impact of proinflammatory cytokines and oxidative stress in this cell death; it is noteworthy to stress that some mechanisms are less induced by the panresistant strain.


Assuntos
Acinetobacter baumannii/patogenicidade , Cálcio/metabolismo , Morte Celular , Células Epiteliais/microbiologia , Inflamação/patologia , Estresse Oxidativo , Calpaína/metabolismo , Caspase 3/metabolismo , Humanos , Pulmão/microbiologia , Pulmão/patologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA