Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.049
Filter
2.
Trop Biomed ; 41(2): 157-159, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-39154267

ABSTRACT

Vibrio vulnificus is a halophilic gram-negative bacillus that can cause fulminant septicaemia in immunocompromised patients. A 67-year-old man who was immunosuppressed as a result of cytotoxic chemotherapy presented with a brief history of fever, lethargy, myalgia, and reduced oral intake. He had recently travelled to the beach to consume seafood. His blood pressure was 81/47 mm Hg, necessitating fluid resuscitation followed by inotropic support and admission to the intensive care unit. His blood culture was positive for curved gram-negative bacilli. The isolate was oxidase-positive and produced an acid butt with an alkaline slant in triple sugar iron agar. Matrix-assisted laser desorption ionization-time of flight mass spectrometry conclusively identified the isolate as V. vulnificus. Intravenous ceftazidime plus ciprofloxacin were administered, and by the fifth day of admission, he was successfully transferred out to the general ward. In total, the patient completed a 14-day course of antibiotic therapy.


Subject(s)
Anti-Bacterial Agents , Sepsis , Vibrio Infections , Vibrio vulnificus , Humans , Male , Aged , Vibrio vulnificus/isolation & purification , Anti-Bacterial Agents/therapeutic use , Sepsis/microbiology , Immunocompromised Host , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Ciprofloxacin/therapeutic use , Ceftazidime/therapeutic use
3.
Brief Bioinform ; 25(5)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39133098

ABSTRACT

Pseudomonas aeruginosa is a complex nosocomial infectious agent responsible for numerous illnesses, with its growing resistance variations complicating treatment development. Studies have emphasized the importance of virulence factors OprE and OprF in pathogenesis, highlighting their potential as vaccine candidates. In this study, B-cell, MHC-I, and MHC-II epitopes were identified, and molecular linkers were active to join these epitopes with an appropriate adjuvant to construct a vaccine. Computational tools were employed to forecast the tertiary framework, characteristics, and also to confirm the vaccine's composition. The potency was weighed through population coverage analysis and immune simulation. This project aims to create a multi-epitope vaccine to reduce P. aeruginosa-related illness and mortality using immunoinformatics resources. The ultimate complex has been determined to be stable, soluble, antigenic, and non-allergenic upon inspection of its physicochemical and immunological properties. Additionally, the protein exhibited acidic and hydrophilic characteristics. The Ramachandran plot, ProSA-web, ERRAT, and Verify3D were employed to ensure the final model's authenticity once the protein's three-dimensional structure had been established and refined. The vaccine model showed a significant binding score and stability when interacting with MHC receptors. Population coverage analysis indicated a global coverage rate of 83.40%, with the USA having the highest coverage rate, exceeding 90%. Moreover, the vaccine sequence underwent codon optimization before being cloned into the Escherichia coli plasmid vector pET-28a (+) at the EcoRI and EcoRV restriction sites. Our research has developed a vaccine against P. aeruginosa that has strong binding affinity and worldwide coverage, offering an acceptable way to mitigate nosocomial infections.


Subject(s)
Computational Biology , Pseudomonas Infections , Pseudomonas aeruginosa , Sepsis , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Humans , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Sepsis/prevention & control , Sepsis/immunology , Sepsis/microbiology , Computational Biology/methods , Epitopes/immunology , Epitopes/chemistry , Pneumonia/prevention & control , Pneumonia/immunology , Pneumonia/microbiology , Pseudomonas Vaccines/immunology , Bacterial Vaccines/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics
4.
Vet Res ; 55(1): 102, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152462

ABSTRACT

In Chile, Piscirickettsia salmonis contains two genetically isolated genogroups, LF-89 and EM-90. However, the impact of a potential co-infection with these two variants on Salmonid Rickettsial Septicemia (SRS) in Atlantic salmon (Salmo salar) remains largely unexplored. In our study, we evaluated the effect of P. salmonis LF-89-like and EM-90-like co-infection on post-smolt Atlantic salmon after an intraperitoneal challenge to compare changes in disease dynamics and host immune response. Co-infected fish had a significantly lower survival rate (24.1%) at 21 days post-challenge (dpc), compared with EM-90-like single-infected fish (40.3%). In contrast, all the LF-89-like single-infected fish survived. In addition, co-infected fish presented a higher presence of clinical lesions than any of the single-infected fish. The gene expression of salmon immune-related biomarkers evaluated in the head kidney, spleen, and liver showed that the EM-90-like isolate and the co-infection induced the up-regulation of cytokines (e.g., il-1ß, ifnγ, il8, il10), antimicrobial peptides (hepdicin) and pattern recognition receptors (PRRs), such as TLR5s. Furthermore, in serum samples from EM-90-like and co-infected fish, an increase in the total IgM level was observed. Interestingly, specific IgM against P. salmonis showed greater detection of EM-90-like antigens in LF-89-like infected fish serum (cross-reaction). These data provide evidence that P. salmonis LF-89-like and EM-90-like interactions can modulate SRS disease dynamics in Atlantic salmon, causing a synergistic effect that increases the severity of the disease and the mortality rate of the fish. Overall, this study contributes to achieving a better understanding of P. salmonis population dynamics.


Subject(s)
Coinfection , Fish Diseases , Piscirickettsia , Piscirickettsiaceae Infections , Salmo salar , Animals , Piscirickettsia/physiology , Fish Diseases/microbiology , Fish Diseases/immunology , Piscirickettsiaceae Infections/veterinary , Piscirickettsiaceae Infections/microbiology , Coinfection/veterinary , Coinfection/microbiology , Coinfection/immunology , Chile , Sepsis/veterinary , Sepsis/microbiology , Sepsis/immunology
5.
BMC Pediatr ; 24(1): 491, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090628

ABSTRACT

BACKGROUND: Sepsis is one of the major causes of morbidity and mortality among pediatric patients throughout the world. The varying microbiological pattern of sepsis warrants the need for researches on the causative organisms and their antimicrobial susceptibility pattern. The epidemiology of neonatal and pediatric sepsis in Ethiopia is under-research. The objective of this study was to evaluate the burden of bacterial pathogens and their antimicrobial susceptibility patterns among children suspected of sepsis. METHODS: An institutional-based prospective cross-sectional study was conducted on 370 pediatric(age birth-15 years) patients suspected of sepsis at the University of Gondar Comprehensive Specialized hospital from December 2020 to November 2021. Blood samples were collected aseptically and inoculated into Tryptone Soya Broth for culture. The organisms grown were identified by standard microbiological methods and subjected to antibiotic susceptibility testing by modified Kirby-Bauer disk diffusion method recommended by Clinical laboratory and standard institute. Methicillin resistance was confirmed using Cefoxitin disk diffusion method. Data entry and analysis were done using Statistical Package for Social Sciences (SPSS) version 26 software. A p-value less than 0.05 at 95% confidence interval was considered statically significant. RESULTS: Out of the total 370 study subjects, 21.6% (80/370) of them were culture positive. Of these, 43 (53.8%) and 37 (46.3%) were Gram-positive and Gram-negative bacterial pathogens, respectively. The most prevalent Gram-positive bacterial isolate was Staphylococcus aureus (n = 24; 30%) and coagulase negative staphylococci (n = 7; 8.8%). Among the Gram-negative bacterial isolates, the leading bacteria was Klebsiella pneumoniae (n = 20; 25%) followed by Escherichia coli (n = 7; 8.8%). Clindamycin, Chloramphenicol, Gentamicin and Ciprofloxacin were the most effective antibiotics against Gram-positive bacterial isolates while Amikacin, Meropenem and Chloramphenicol were effective against Gram-negative pathogens. Methicillin resistance was detected in 45.8% of Staphylococcus aureus. Multi-drug resistance (MDR) antimicrobial susceptibility pattern was observed in 76% of the bacterial isolates. CONCLUSION: Gram positive bacteria were the predominant isolates among pediatric sepsis cases and most of the bacterial isolates showed MDR. Staphylococcus aureus and Klebsiella pneumoniae were frequently isolated bacteria. The high prevalence of drug resistance warrants rational use of antibiotics and the need for regular antibiotic susceptibility surveillance studies.


Subject(s)
Microbial Sensitivity Tests , Sepsis , Humans , Ethiopia/epidemiology , Child , Child, Preschool , Infant , Cross-Sectional Studies , Adolescent , Sepsis/microbiology , Sepsis/epidemiology , Sepsis/drug therapy , Male , Prospective Studies , Female , Infant, Newborn , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Hospitals, University , Drug Resistance, Multiple, Bacterial
6.
Antimicrob Resist Infect Control ; 13(1): 84, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39113089

ABSTRACT

BACKGROUND: Endoscopic retrograde cholangiopancreatography (ERCP) has become a routine endoscopic procedure that is essential for diagnosing and managing various conditions, including gallstone extraction and the treatment of bile duct and pancreatic tumors. Despite its efficacy, post-ERCP infections - particularly those caused by carbapenem-resistant Enterobacterales (CRE) - present significant risks. These risks highlight the need for accurate predictive models to enhance postprocedural care, reduce the mortality risk associated with post-ERCP CRE sepsis, and improve patient outcomes in the context of increasing antibiotic resistance. OBJECTIVE: This study aimed to examine the risk factors for 30-day mortality in patients with CRE sepsis following ERCP and to develop a nomogram for accurately predicting 30-day mortality risk. METHODS: Data from 195 patients who experienced post-ERCP CRE sepsis between January 2010 and December 2022 were analyzed. Variable selection was optimized via the least absolute shrinkage and selection operator (LASSO) regression model. Multivariate logistic regression analysis was then employed to develop a predictive model, which was evaluated in terms of discrimination, calibration, and clinical utility. Internal validation was achieved through bootstrapping. RESULTS: The nomogram included the following predictors: age > 80 years (hazard ratio [HR] 2.61), intensive care unit (ICU) admission within 90 days prior to ERCP (HR 2.64), hypoproteinemia (HR 4.55), quick Pitt bacteremia score ≥ 2 (HR 2.61), post-ERCP pancreatitis (HR 2.52), inappropriate empirical therapy (HR 3.48), delayed definitive therapy (HR 2.64), and short treatment duration (< 10 days) (HR 5.03). The model demonstrated strong discrimination and calibration. CONCLUSIONS: This study identified significant risk factors associated with 30-day mortality in patients with post-ERCP CRE sepsis and developed a nomogram to accurately predict this risk. This tool enables healthcare practitioners to provide personalized risk assessments and promptly administer appropriate therapies against CRE, thereby reducing mortality rates.


Subject(s)
Cholangiopancreatography, Endoscopic Retrograde , Enterobacteriaceae Infections , Nomograms , Sepsis , Humans , Cholangiopancreatography, Endoscopic Retrograde/adverse effects , Male , Female , Retrospective Studies , Risk Factors , Aged , Middle Aged , Sepsis/mortality , Sepsis/microbiology , Enterobacteriaceae Infections/mortality , Enterobacteriaceae Infections/drug therapy , Carbapenem-Resistant Enterobacteriaceae/isolation & purification , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Aged, 80 and over
7.
J Trop Pediatr ; 70(5)2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39164190

ABSTRACT

The objective of this study was to determine the risk factors associated with Elizabethkingia anophelis infection in neonates admitted to a tertiary care neonatal intensive care unit (NICU). A case-control study was undertaken as part of the outbreak investigation for E. anophelis sepsis in a tertiary care NICU in South India. Thirty-eight neonates with E. anophelis bloodstream infection (BSI) between January 2021 and February 2022 were enrolled as cases, and 38 neonates symptomatic with other BSIs, were selected as controls, and risk factors analysed. The 38 cases were relatively stable neonates, likely to be admitted to level 1 and level 2 NICU, unlike the controls, who were sicker and required level 3 NICU care. Only a third of neonates with Elizabethkingia sepsis had traditional risk factors like central lines, need for respiratory support or perinatal risk factors. Multiple logistic regression analysis revealed that neonates with E. anophelis infection were more likely to be stable and on only enteral feeds, cared in level 1 or 2 of the NICU. This observation, combined with isolation of Elizabethkingia meningosepticum from breast pumps earlier, led us to autoclave the feeding vessels and milk containers along with provision of hot water for cleaning breast pumps, and adoption of general infection control measures, after which incident cases declined. Sanger sequencing of 10 representative isolates obtained from the neonates showed 100% sequence identity to E. anophelis. Infection due to E. anophelis affects relatively stable neonates without traditional risk factors for sepsis. Adherence to asepsis routines and housekeeping protocols helps to prevent the spread of infection.


Elizabethkingia anophelis is an emerging pathogen causing infection in neonates. In the present case­control study, we found that E. anophelis was more likely to infect otherwise healthy neonates, on enteral nutrition, without the traditional risk factors for sepsis. Mortality was 23.7% (9/38). About 55.3% (21/38) had meningitis and 23.8% (9/38) had hydrocephalus. Additionally, 76% isolates were multi-drug resistant, with the isolates showing highest susceptibility to minocycline (100%) and levofloxacin (97.8%). Source identification was not possible even after multiple rounds of extensive environmental testing, but it is possibly related to contamination of water and/or milk sources. Interventions addressing the same led to a dramatic decline in the infection rates, though occasional infection without clustering continues to occur. Sanger sequencing of 10 representative isolates confirmed sequence identity to E. anophelis.


Subject(s)
Disease Outbreaks , Flavobacteriaceae Infections , Flavobacteriaceae , Intensive Care Units, Neonatal , Tertiary Care Centers , Humans , Infant, Newborn , Case-Control Studies , Flavobacteriaceae/isolation & purification , Flavobacteriaceae/genetics , Risk Factors , Male , Female , Flavobacteriaceae Infections/epidemiology , Flavobacteriaceae Infections/microbiology , India/epidemiology , Cross Infection/epidemiology , Cross Infection/microbiology , Sepsis/epidemiology , Sepsis/microbiology
8.
Future Microbiol ; 19(11): 963-970, 2024.
Article in English | MEDLINE | ID: mdl-39109653

ABSTRACT

Non-diphtheroid Corynebacterium sepsis is rare and has affected only immunocompromised or particularly predisposed patients so far. We present the first case of urosepsis caused by Corynebacterium aurimucosum in a 67-year-old woman, without any known immunodeficiencies and in absence of any immunosuppressive therapy, admitted to the hospital for fever and acute dyspnea. This work suggests a new approach in evaluating the isolation of Corynebacteria, especially if isolated from blood. In particular, it highlights the potential infectious role of C. aurimucosum (often considered a contaminant and only rarely identified as an etiological agent of infections) and its clinical consequences, detailing also interesting aspects about its microbiological diagnosis and relative therapy and clarifying contrasting data of literature.


[Box: see text].


Subject(s)
Corynebacterium Infections , Corynebacterium , Sepsis , Urinary Tract Infections , Humans , Corynebacterium/isolation & purification , Corynebacterium/genetics , Corynebacterium/pathogenicity , Corynebacterium/classification , Aged , Female , Corynebacterium Infections/microbiology , Corynebacterium Infections/diagnosis , Corynebacterium Infections/drug therapy , Sepsis/microbiology , Sepsis/drug therapy , Urinary Tract Infections/microbiology , Urinary Tract Infections/drug therapy , Urinary Tract Infections/diagnosis , Anti-Bacterial Agents/therapeutic use
9.
Bull Exp Biol Med ; 177(2): 243-247, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39090462

ABSTRACT

The influence of non-opsonized and opsonized S. aureus 2879M and E. coli 321 strains on the total strength of interaction between the endothelial cell and neutrophil during the docking process was studied using in vitro model of experimental septicemia. We observed a decrease in the force and work of adhesion between receptors of neutrophils and endothelial cells under the influence of non-opsonized strains and further decrease in the affinity of single interactions between cells under the influence of opsonized S. aureus, which was compensated by an increase in the number of contacts, as well as an increase in the force of adhesion under the influence of opsonized E. coli compared to non-opsonized bacteria, which remained below the control level, while adhesion work reaches the control level. Thus, opsonization of S. aureus aggravates the "immunological uncoupling" between neutrophils and endothelial cells, while opsonization of E. coli reduces the pathological effect compared to non-opsonized bacteria.


Subject(s)
Endothelial Cells , Escherichia coli , Neutrophils , Sepsis , Staphylococcus aureus , Neutrophils/immunology , Neutrophils/metabolism , Escherichia coli/immunology , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Sepsis/immunology , Sepsis/microbiology , Sepsis/metabolism , Sepsis/pathology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Humans , Phagocytosis , Cell Adhesion/immunology , Opsonin Proteins/metabolism , Opsonin Proteins/immunology , Bacterial Adhesion , Animals
10.
PLoS One ; 19(8): e0308471, 2024.
Article in English | MEDLINE | ID: mdl-39106284

ABSTRACT

BACKGROUND: Evidence associating body mass index (BMI) with the prognosis of Staphylococcus aureus sepsis remains scarce. OBJECTIVE: To explore the association between BMI and clinical outcomes in intensive care units patients with Staphylococcus aureus sepsis. METHODS: A retrospective analysis of patients with Staphylococcus aureus sepsis was conducted using the MIMIC-IV database from the Critical Care Medicine Information. Data were collected within the first 24 hours of intensive care units admission. The primary endpoint was 28-day mortality. The association between BMI and 28-day all-cause mortality was assessed using multivariable logistic regression, subgroup analyses, restricted cubic spline curves and Kaplan-Meier survival analysis. RESULTS: The study included 2,295 patients with an average age of 63.5 (16.1) years, 60.2% of whom were male. Multivariate analysis revealed that each 1 kg/m2 increase in BMI was linked to a 2.8% decrease in the risk of 28-day mortality (adjusted OR = 0.972, 95% CI: 0.955-0.990, P = 0.002). Patients in the medium and high BMI categories had significantly lower risks of 28-day mortality compared to those in the low BMI group (OR [95% CI] 0.650 [0.474-0.891]; OR [95% CI] 0.516 [0.378-0.705]; P trend < 0.0001). The RCS model showed a non-linear association between BMI and 28-day mortality (P = 0.014). Kaplan-Meier analysis showed that patients with elevated BMI had lower 28-day mortality (P < 0.0001). Notably, significant interactions between AKI and SOFA with BMI were observed (P<0.05). CONCLUSION: Increased BMI is associated with a reduced risk of 28-day all-cause mortality in patients with Staphylococcus aureus sepsis.


Subject(s)
Body Mass Index , Intensive Care Units , Sepsis , Staphylococcal Infections , Staphylococcus aureus , Humans , Male , Female , Middle Aged , Retrospective Studies , Staphylococcal Infections/mortality , Staphylococcal Infections/microbiology , Aged , Sepsis/mortality , Sepsis/microbiology , Kaplan-Meier Estimate , Prognosis , Hospital Mortality , Critical Care
11.
Medicina (B Aires) ; 84(4): 649-655, 2024.
Article in Spanish | MEDLINE | ID: mdl-39172564

ABSTRACT

INTRODUCTION: Delay in initiating appropriate antimicrobial therapy prolongs hospitalization, increases in-hospital mortality, and raises economic costs. Currently, the identification and susceptibility testing of bacteria in positive blood cultures require a considerable amount of time. The objective of this study was to assess the impact of the BCID2 FilmArray® (FA) panel on the timing of appropriate antimicrobial therapy and potential antimicrobial costs. METHODS: This is a retrospective observational study focused on positive blood cultures in hospitalized patients. FA processing was conducted concurrently with routine sample processing. Changes in antibiotic treatments based on FA results were evaluated, and the reduction in antimicrobial therapy duration and associated cost savings were calculated. RESULTS: Eighty-seven bacteremia episodes were analysed. In 42 (48%) of them antimicrobial therapy was de-escalated to narrower spectrum agents, while in 7 (8%) therapy was escalated to broader spectrum antimicrobials. Additionally, in 8 (9%) antimicrobials were switched without changing spectrum and in 30 (34%) no changes were made based on FA results. Antimicrobial changes were made 2.3 days faster than with routine sample processing resulting in calculated potential savings of US$ 7408. CONCLUSION: The implementation of FA facilitated a faster administration of appropriate antimicrobial therapy, leading to a reduction in the duration of broadspectrum empirical antimicrobial therapy and subsequent economic savings.


Introducción: Los retrasos en el tratamiento antimicrobiano adecuado de las bacteriemias prolongan la estadía hospitalaria, aumentan la mortalidad e incrementan los costos. Aún hoy en día se requiere un tiempo considerable para obtener la identificación y antibiograma de los microorganismos en los hemocultivos positivos. El objetivo fue evaluar el impacto de la implementación del panel BCID2 de FilmArray® (FA) sobre el tiempo de inicio de tratamientos antimicrobianos adecuados y sobre los costos potenciales de los mismos. Métodos: Estudio observacional retrospectivo de los hemocultivos positivos de pacientes hospitalizados, procesados por FA y por metodología tradicional. Se evaluaron los cambios de antimicrobianos en base a los resultados del FA. Se calcularon los días de reducción de tratamiento antimicrobiano y el ahorro potencial en el uso de los mismos, teniendo en cuenta también los costos del FA. Resultados: Se analizaron 87 episodios de bacteriemia. En 42 (48.3%) de ellos se desescaló el tratamiento a antimicrobianos de menor espectro, en 7 (8%) se escaló a antimicrobianos de mayor espectro, en 8 (9.2%) se cambió el antimicrobiano sin variar el espectro y en 30 (34.5%) no se realizaron cambios con los resultados del FA. Los cambios de antimicrobianos se realizaron en promedio 2.3 días más rápido que con los métodos convencionales. Se calculó un ahorro potencial de US$ 7408. Conclusión: La implementación del panel BCID2 de FilmArray® permitió adecuar los tratamientos antimicrobianos más rápidamente acortando la duración de los tratamientos empíricos de amplio espectro, lo cual resultó costo-efectivo.


Subject(s)
Anti-Bacterial Agents , Bacteremia , Humans , Retrospective Studies , Male , Bacteremia/diagnosis , Bacteremia/drug therapy , Bacteremia/microbiology , Female , Anti-Bacterial Agents/therapeutic use , Middle Aged , Aged , Tertiary Care Centers , Microbial Sensitivity Tests , Sepsis/diagnosis , Sepsis/drug therapy , Sepsis/microbiology , Adult , Blood Culture/methods , Molecular Diagnostic Techniques/methods , Molecular Diagnostic Techniques/economics , Aged, 80 and over
12.
Diagn Microbiol Infect Dis ; 110(2): 116476, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111106

ABSTRACT

We present a case of a 34-year-old patient with abdominal sepsis caused by an infrequent species: Chimaeribacter arupi. Genomic analysis confirmed the identification which is difficult to achieve by other methods so far. To our knowledge, this represents the first case of infection by this species reported in Argentina.


Subject(s)
Sepsis , Humans , Adult , Sepsis/microbiology , Sepsis/diagnosis , Male , Argentina , RNA, Ribosomal, 16S/genetics , Phylogeny , Anti-Bacterial Agents/therapeutic use , DNA, Bacterial/genetics , Fusobacteriaceae Infections/microbiology , Fusobacteriaceae Infections/diagnosis , Sequence Analysis, DNA
13.
Elife ; 132024 Aug 20.
Article in English | MEDLINE | ID: mdl-39163101

ABSTRACT

Sepsis causes millions of deaths per year worldwide and is a current global health priority declared by the WHO. Sepsis-related deaths are a result of dysregulated inflammatory immune responses indicating the need to develop strategies to target inflammation. An important mediator of inflammation is extracellular adenosine triphosphate (ATP) that is released by inflamed host cells and tissues, and also by bacteria in a strain-specific and growth-dependent manner. Here, we investigated the mechanisms by which bacteria release ATP. Using genetic mutant strains of Escherichia coli (E. coli), we demonstrate that ATP release is dependent on ATP synthase within the inner bacterial membrane. In addition, impaired integrity of the outer bacterial membrane notably contributes to ATP release and is associated with bacterial death. In a mouse model of abdominal sepsis, local effects of bacterial ATP were analyzed using a transformed E. coli bearing an arabinose-inducible periplasmic apyrase hydrolyzing ATP to be released. Abrogating bacterial ATP release shows that bacterial ATP suppresses local immune responses, resulting in reduced neutrophil counts and impaired survival. In addition, bacterial ATP has systemic effects via its transport in outer membrane vesicles (OMV). ATP-loaded OMV are quickly distributed throughout the body and upregulated expression of genes activating degranulation in neutrophils, potentially contributing to the exacerbation of sepsis severity. This study reveals mechanisms of bacterial ATP release and its local and systemic roles in sepsis pathogenesis.


Sepsis is a severe condition often caused by the body's immune system overreacting to bacterial infections. This can lead to excessive inflammation which damages organs and requires urgent medical care. With sepsis claiming millions of lives each year, new and improved ways to treat this condition are urgently needed. One potential strategy for treating sepsis is to target the underlying mechanisms controlling inflammation. Inflamed and dying cells release molecules called ATP (the energy carrier of all living cells), which strongly influence the immune system, including during sepsis. In the early stages of an infection, ATP acts as a danger signal warning the body that something is wrong. However, over time, it can worsen infections by disturbing the immune response. Similar to human cells, bacteria release their own ATP, which can have different impacts depending on the type of bacteria and where they are located in the body. However, it is not well understood how bacterial ATP influences severe infections like sepsis. To investigate this question, Spari et al analysed how ATP is released from Escherichia coli, a type of bacteria that causes severe infections. This revealed that the bacteria secrete ATP directly in to their environment and via small membrane-bound structures called vesicles. Spari et al. then probed a mouse model of abdominal sepsis which had been infected with E. coli that release either normal or low levels of ATP. They found that the ATP released from E. coli impaired the mice's survival and lowered the number of neutrophils (immune cells which are important for defending against bacteria) at the site of the infection. The ATP secreted via vesicles also altered the role of neutrophils but in more distant regions, and it is possible that these changes may be contributing to the severity of sepsis. These findings provide a better understanding of how ATP released from bacteria impacts the immune system during sepsis. While further investigation is needed, these findings may offer new therapeutic targets for treating sepsis.


Subject(s)
Adenosine Triphosphate , Escherichia coli , Inflammation , Sepsis , Animals , Adenosine Triphosphate/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Sepsis/microbiology , Sepsis/metabolism , Mice , Inflammation/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Escherichia coli Infections/microbiology , Escherichia coli Infections/immunology
14.
Front Cell Infect Microbiol ; 14: 1398756, 2024.
Article in English | MEDLINE | ID: mdl-39176264

ABSTRACT

Background: Gut microbiota is closely related to the occurrence and development of sepsis. However, the causal effects between the gut microbiota and sepsis, and whether circulating inflammatory proteins act as mediators, remain unclear. Methods: Gut microbiota, circulating inflammatory proteins, and four sepsis-related outcomes were identified from large-scale genome wide association studies (GWAS) summary data. Inverse Variance Weighted (IVW) was the primary statistical method. Additionally, we investigated whether circulating inflammatory proteins play a mediating role in the pathway from gut microbiota to the four sepsis-related outcomes. Results: There were 14 positive and 15 negative causal effects between genetic liability in the gut microbiota and four sepsis-related outcomes. Additionally, eight positive and four negative causal effects were observed between circulating inflammatory proteins and the four sepsis-related outcomes. Circulating inflammatory proteins do not act as mediators. Conclusions: Gut microbiota and circulating inflammatory proteins were causally associated with the four sepsis-related outcomes. However, circulating inflammatory proteins did not appear to mediate the pathway from gut microbiota to the four sepsis-related outcomes.


Subject(s)
Gastrointestinal Microbiome , Genome-Wide Association Study , Mendelian Randomization Analysis , Sepsis , Sepsis/microbiology , Sepsis/blood , Humans , Gastrointestinal Microbiome/genetics , Inflammation/blood , Polymorphism, Single Nucleotide
15.
Br J Hosp Med (Lond) ; 85(7): 1-16, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39078906

ABSTRACT

Aims/Background Adult-onset Still's disease (AOSD) shares similar clinical symptoms with sepsis. Thus, differentiating between AOSD and sepsis presents a great challenge while making diagnosis. This study aimed to analyse the changes in blood microbiota related to AOSD and sepsis using metagenomic next-generation sequencing (mNGS), identify potential biomarkers that distinguish AOSD from sepsis, and explore the diagnostic value of mNGS in differentiation between these two pathological conditions. Methods Clinical data of four AOSD patients and four sepsis patients treated in the Department of Rheumatology and Immunology, The Affiliated Hospital of Xuzhou Medical University between October 2021 and February 2022 were collected. The mNGS diagnostic records of these patients were analysed for microbial correlations in terms of species taxonomic structure and beta diversity by comparing blood microbiota between AOSD and sepsis. The biomarkers with the strongest capability in distinguishing the subgroups were screened using a random forest algorithm. Results There was no statistically significant differences between AOSD patients and sepsis controls in terms of gender and age (p > 0.05). A total of 91 operational taxonomic units (OTUs) were obtained. At the level of phylum, Proteobacteria, Ascomycota and Basidiomycota were present in high abundances in both groups (79.76%, 14.18% and 3.30% vs 54.03%, 32.77% and 5.81%). At the genus level, the abundances of Parainfluenzae, Aspergillus and Ralstonia were the top three highest in the AOSD group (73.88%, 10.92% and 5.48%), while Ralstonia, Aspergillus and Malassezia were ranked as the top three in the sepsis group in term of abundance (48.69%, 27.36% and 5.52%). In beta-diversity analysis, there were advances shown in visual principal coordinates analysis (PCoA) and non-metric multidimensional scaling (NMDS) between the AOSD group and sepsis group (p < 0.05), with little significant differences in the analysis of similarities (Anosim) (p > 0.05). Linear discriminant analysis effect size (LEfSe) showed that Mucoromycota, Saccharomycetes, Moraxellales, Mucorales, Xanthomonadales, Saccharomycetales, Acinetobacter, Stenotrophomonas, Yarrowia, Apophysomyces, Acinetobacter johnson, Yarrowia lipolytica, Apophysomyces variabilis and Stenotrophomonas maltophilia were more enriched in sepsis group (p < 0.05). The top five variables with the strongest capability in distinguishing between AOSD and sepsis were Acinetobacter johnsonii, Apophysomyces variabilis, Propionibacterium acnes, Stenotrophomonas maltophilia and Yarrowia lipolytica. Conclusion The blood microorganisms in AOSD were different from sepsis, and mNGS was potential to distinguish between AOSD and sepsis.


Subject(s)
High-Throughput Nucleotide Sequencing , Metagenomics , Sepsis , Still's Disease, Adult-Onset , Humans , Sepsis/microbiology , Sepsis/blood , Sepsis/diagnosis , Male , Female , Still's Disease, Adult-Onset/blood , Still's Disease, Adult-Onset/microbiology , Still's Disease, Adult-Onset/diagnosis , Adult , Middle Aged , Metagenomics/methods , Microbiota/genetics , Diagnosis, Differential , Biomarkers/blood
16.
BMC Med Genomics ; 17(1): 183, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982318

ABSTRACT

BACKGROUND AND PURPOSE: The association of water loading with several infections remains unclear. Observational studies are hard to investigate definitively due to potential confounders. In this study, we employed Mendelian randomization (MR) analysis to assess the association between genetically predicted whole body water mass (BWM) and several infections. METHODS: BWM levels were predicted among 331,315 Europeans in UK Biobank using 418 SNPs associated with BWM. For outcomes, we used genome-wide association data from the UK Biobank and FinnGen consortium, including sepsis, pneumonia, intestinal infections, urinary tract infections (UTIs) and skin and soft tissue infections (SSTIs). Inverse-variance weighted MR analyses as well as a series of sensitivity analyses were conducted. RESULTS: Genetic prediction of BWM is associated with an increased risk of sepsis (OR 1.34; 95% CI 1.19 to 1.51; P = 1.57 × 10- 6), pneumonia (OR: 1.17; 95% CI 1.08 to 1.29; P = 3.53 × 10- 4), UTIs (OR: 1.26; 95% CI 1.16 to 1.37; P = 6.29 × 10- 8), and SSTIs (OR: 1.57; 95% CI 1.25 to 1.96; P = 7.35 × 10- 5). In the sepsis and pneumonia subgroup analyses, the relationship between BWM and infection was observed in bacterial but not in viral infections. Suggestive evidence suggests that BWM has an effect on viral intestinal infections (OR: 0.86; 95% CI 0.75 to 0.99; P = 0.03). There is limited evidence of an association between BWM levels and bacteria intestinal infections, and genitourinary tract infection (GUI) in pregnancy. In addition, MR analyses supported the risk of BWM for several edematous diseases. However, multivariable MR analysis shows that the associations of BWM with sepsis, pneumonia, UTIs and SSTIs remains unaffected when accounting for these traits. CONCLUSIONS: In this study, the causal relationship between BWM and infectious diseases was systematically investigated. Further prospective studies are necessary to validate these findings.


Subject(s)
Bacterial Infections , Genome-Wide Association Study , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Humans , Bacterial Infections/genetics , Female , Risk Factors , Male , Urinary Tract Infections/genetics , Urinary Tract Infections/microbiology , Sepsis/genetics , Sepsis/microbiology
17.
BMC Microbiol ; 24(1): 272, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39039501

ABSTRACT

BACKGROUND: Late-onset sepsis (LOS) and pneumonia are common infectious diseases, with high morbidity and mortality in neonates. This study aimed to investigate the differences in the gut microbiota among preterm infants with LOS, or pneumonia, and full-term infants. Furthermore, this study aimed to determine whether there is a correlation between intestinal pathogenic colonization and LOS. METHODS: In a single-center case‒control study, 16 S rRNA gene sequencing technology was used to compare gut microbiota characteristics and differences among the LOS group, pneumonia group, and control group. RESULTS: Our study revealed that the gut microbiota in the control group was more diverse than that in the LOS group and pneumonia group (P < 0.05). No significant differences in diversity were detected between the LOS and pneumonia groups (P > 0.05). Compared with the control group, the abundances of Akkermansia, Escherichia/Shigella, and Enterococcus increased, while the abundances of Bacteroides and Stenotrophomonas decreased in the LOS and pneumonia groups. The pathogenic bacteria in infants with LOS were consistent with the distribution of the main bacteria in the intestinal microbiota. An increase in Escherichia/Shigella abundance may predict a high risk of LOS occurrence, with an area under the curve (AUC) of 0.773. CONCLUSION: Changes in the gut microbiota composition were associated with an increased risk of LOS and pneumonia. The dominant bacteria in the gut microbiota of the LOS group were found to be associated with the causative pathogen of LOS. Moreover, preterm infants exhibiting an elevated abundance of Escherichia/Shigella may be considered potential candidates for predicting the onset of LOS.


Subject(s)
Bacteria , Gastrointestinal Microbiome , Infant, Premature , Pneumonia , RNA, Ribosomal, 16S , Sepsis , Humans , Case-Control Studies , Infant, Newborn , Male , Female , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , RNA, Ribosomal, 16S/genetics , Sepsis/microbiology , Pilot Projects , Pneumonia/microbiology , Feces/microbiology
18.
Front Immunol ; 15: 1424332, 2024.
Article in English | MEDLINE | ID: mdl-39026673

ABSTRACT

Background: The protective role of gut microbiota and its metabolites against intestinal damage in sepsis patients remain unclear. Methods: Fecal samples were acquired from patients categorized into sepsis and non-sepsis groups for analysis of microbial composition via 16S rRNA sequencing and untargeted metabolomics analysis. We assessed the impact of gut microbiota from sepsis patients on intestinal barriers in antibiotic-treated mice. Furthermore, We conducted spearman's correlation analysis to examine the relationship between metabolites and the severity of sepsis. Additionally, we performed animal experiments to validate the functionality of identified metabolites. Results: The diversity of intestinal flora is decreased in patients with sepsis compared to the control group. Through fecal microbiota transplantation experiments, it was discovered that the gut microbiota derived from sepsis patients could induce intestinal damage in antibiotic-treated mice. Metabolomics analysis of the microbiota revealed a significant enrichment of the Valine, leucine, and isoleucine biosynthesis pathway. Further analysis showed a significant decrease in the abundance of L-valine in sepsis patients, which was negatively correlated with APACHE-II and SOFA scores. In sepsis mouse experiments, it was found that L-valine could alleviate sepsis-induced intestinal damage. Conclusion: Alterations in microbial and metabolic features in the gut can affect the severity of sepsis. Furthermore, L-valine can protect against sepsis-induced intestinal injury.


Subject(s)
Gastrointestinal Microbiome , Sepsis , Valine , Gastrointestinal Microbiome/drug effects , Sepsis/microbiology , Animals , Mice , Humans , Valine/pharmacology , Valine/therapeutic use , Male , Female , Middle Aged , Fecal Microbiota Transplantation , Severity of Illness Index , Metabolomics/methods , Aged , Feces/microbiology , Disease Models, Animal , Mice, Inbred C57BL , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/microbiology , RNA, Ribosomal, 16S/genetics
19.
Int J Mol Sci ; 25(14)2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39063011

ABSTRACT

Sepsis-induced multiple organ dysfunction arises from the highly complex pathophysiology encompassing the interplay of inflammation, oxidative stress, endothelial dysfunction, mitochondrial damage, cellular energy failure, and dysbiosis. Over the past decades, numerous studies have been dedicated to elucidating the underlying molecular mechanisms of sepsis in order to develop effective treatments. Current research underscores liver and cardiac dysfunction, along with acute lung and kidney injuries, as predominant causes of mortality in sepsis patients. This understanding of sepsis-induced organ failure unveils potential therapeutic targets for sepsis treatment. Various novel therapeutics, including melatonin, metformin, palmitoylethanolamide (PEA), certain herbal extracts, and gut microbiota modulators, have demonstrated efficacy in different sepsis models. In recent years, the research focus has shifted from anti-inflammatory and antioxidative agents to exploring the modulation of energy metabolism and gut microbiota in sepsis. These approaches have shown a significant impact in preventing multiple organ damage and mortality in various animal sepsis models but require further clinical investigation. The accumulation of this knowledge enriches our understanding of sepsis and is anticipated to facilitate the development of effective therapeutic strategies in the future.


Subject(s)
Multiple Organ Failure , Sepsis , Humans , Sepsis/complications , Sepsis/metabolism , Sepsis/drug therapy , Sepsis/microbiology , Multiple Organ Failure/etiology , Multiple Organ Failure/metabolism , Animals , Gastrointestinal Microbiome , Oxidative Stress , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology
20.
Diagn Microbiol Infect Dis ; 110(1): 116412, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39018932

ABSTRACT

Rapid and reliable identification of the causal organism in bloodstream infections and sepsis is crucial for both individual patient care and public health. We have implemented a rapid in-house identification protocol (with 10 % Triton) using MALDI-TOF MS for identifying the causative organism in positive blood cultures without prior culture. Our objective was to retrospectively analyze data collected over a four-year period while implementing this rapid in-house identification protocol and to develop a guide for evaluating and reporting the obtained results. Overall, our method utilizing MALDI-TOF MS for rapid in-house identification, demonstrated comparable results to other commercially available and in-house methods reported in the literature. Over the past four years, direct identification has facilitated the distinction between clinically relevant positive blood cultures and irrelevant ones, guiding rapid focus control and appropriate antibiotic treatment. The established guide can serve as a valuable tool in reporting positive blood cultures and associated antibiotic treatments.


Subject(s)
Bacteremia , Blood Culture , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Workflow , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Humans , Blood Culture/methods , Retrospective Studies , Bacteremia/diagnosis , Bacteremia/microbiology , Bacteria/isolation & purification , Bacteria/classification , Octoxynol , Sepsis/diagnosis , Sepsis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL