Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Genome Res ; 31(7): 1258-1268, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34108268

ABSTRACT

Neisseria meningitidis (the meningococcus) is a major human pathogen with a history of high invasive disease burden, particularly in sub-Saharan Africa. Our current understanding of the evolution of meningococcal genomes is limited by the rarity of large-scale genomic population studies and lack of in-depth investigation of the genomic events associated with routine pathogen transmission. Here, we fill this knowledge gap by a detailed analysis of 2839 meningococcal genomes obtained through a carriage study of over 50,000 samples collected systematically in Burkina Faso, West Africa, before, during, and after the serogroup A vaccine rollout, 2009-2012. Our findings indicate that the meningococcal genome is highly dynamic, with highly recombinant loci and frequent gene sharing across deeply separated lineages in a structured population. Furthermore, our findings illustrate how population structure can correlate with genome flexibility, as some lineages in Burkina Faso are orders of magnitude more recombinant than others. We also examine the effect of selection on the population, in particular how it is correlated with recombination. We find that recombination principally acts to prevent the accumulation of deleterious mutations, although we do also find an example of recombination acting to speed the adaptation of a gene. In general, we show the importance of recombination in the evolution of a geographically expansive population with deep population structure in a short timescale. This has important consequences for our ability to both foresee the outcomes of vaccination programs and, using surveillance data, predict when lineages of the meningococcus are likely to become a public health concern.

2.
BJU Int ; 133(6): 680-689, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38469686

ABSTRACT

BACKGROUND: Older men (aged ≥75 years) with high risk, non-metastatic prostate cancer (PCa) are increasingly treated with curative therapy (surgery or radiotherapy). However, it is unclear if curative therapy prolongs life and improves health-related quality of life (HRQoL) in this age group compared to conservative therapy, which has evolved considerably during the last decade. STUDY DESIGN: The Scandinavian Prostate Cancer Group (SPCG) 19/Norwegian Get-Randomized Research Group-Prostate (GRand-P) is a randomised, two-armed, controlled, multicentre, phase III trial carried out at study centres in Norway, Denmark, Finland, and Sweden. ENDPOINTS: The primary endpoints are overall survival and HRQoL (burden of disease scale, European Organisation for the Research and Treatment of Cancer [EORTC] Elderly Cancer patients). Secondary endpoints are PCa-specific survival, metastasis-free survival, role-functioning scale (EORTC quality of life questionnaire 30-item core), urinary irritative/obstructive scale (26-item Expanded Prostate Cancer Index Composite [EPIC-26]), bowel scale (EPIC-26), intervention-free survival, PCa morbidity, use of secondary and tertiary systemic therapies, mean quality-adjusted life-years (QALYs), and mean total healthcare costs. PATIENTS AND METHODS: A total of 980 men (aged ≥75 years) with non-metastatic, high-risk PCa will initially be screened with Geriatric 8 (G8) health status screening tool and Mini-COG© brief cognitive test. Participants identified by G8 as 'fit' or 'frail' will be randomised (ratio 1:1) to either immediate curative therapy (radiotherapy or prostatectomy) or conservative therapy (endocrine therapy or observation). Participants who are unable or unwilling to participate in randomisation will be enrolled in a separate observation group. Randomised patients will be followed for 10 years. TRIAL REGISTRATION: Ethics approval has been granted in Norway (457593), Denmark (H-22051998), Finland (R23043) and Sweden (Dnr 2023-05296-01). The trial is registered on Clinicaltrials.org (NCT05448547).


Subject(s)
Conservative Treatment , Prostatic Neoplasms , Quality of Life , Aged , Aged, 80 and over , Humans , Male , Clinical Trials, Phase III as Topic , Prostatectomy , Prostatic Neoplasms/therapy , Prostatic Neoplasms/pathology , Randomized Controlled Trials as Topic , Multicenter Studies as Topic
3.
Mol Biol Evol ; 39(1)2022 01 07.
Article in English | MEDLINE | ID: mdl-34662416

ABSTRACT

The soil bacterium Burkholderia pseudomallei is the causative agent of melioidosis and a significant cause of human morbidity and mortality in many tropical and subtropical countries. The species notoriously survives harsh environmental conditions but the genetic architecture for these adaptations remains unclear. Here we employed a powerful combination of genome-wide epistasis and co-selection studies (2,011 genomes), condition-wide transcriptome analyses (82 diverse conditions), and a gene knockout assay to uncover signals of "co-selection"-that is a combination of genetic markers that have been repeatedly selected together through B. pseudomallei evolution. These enabled us to identify 13,061 mutation pairs under co-selection in distinct genes and noncoding RNA. Genes under co-selection displayed marked expression correlation when B. pseudomallei was subjected to physical stress conditions, highlighting the conditions as one of the major evolutionary driving forces for this bacterium. We identified a putative adhesin (BPSL1661) as a hub of co-selection signals, experimentally confirmed a BPSL1661 role under nutrient deprivation, and explored the functional basis of co-selection gene network surrounding BPSL1661 in facilitating the bacterial survival under nutrient depletion. Our findings suggest that nutrient-limited conditions have been the common selection pressure acting on this species, and allelic variation of BPSL1661 may have promoted B. pseudomallei survival during harsh environmental conditions by facilitating bacterial adherence to different surfaces, cells, or living hosts.


Subject(s)
Biological Evolution , Burkholderia pseudomallei , Adhesins, Bacterial , Alleles , Burkholderia pseudomallei/genetics , Burkholderia pseudomallei/physiology , Selection, Genetic , Stress, Physiological
4.
Eur J Neurol ; 30(5): 1352-1363, 2023 05.
Article in English | MEDLINE | ID: mdl-36786305

ABSTRACT

BACKGROUND AND PURPOSE: There are currently no biomarkers to select cryptogenic stroke (CS) patients for monitoring with insertable cardiac monitors (ICMs), the most effective tool for diagnosing atrial fibrillation (AF) in CS. The purpose of this study was to assess clinically available biomarkers as predictors of AF. METHODS: Eligible CS and cryptogenic transient ischaemic attack patients underwent 12-month monitoring with ICMs, clinical follow-up and biomarker sampling. Levels of cardiac and thromboembolic biomarkers, taken within 14 days from symptom onset, were compared between patients diagnosed with AF (n = 74) during monitoring and those without AF (n = 185). Receiver operating characteristic curves were created. Biomarkers reaching area under the receiver operating characteristic curve ≥ 0.7 were dichotomized by finding optimal cut-off values and were used in logistic regression establishing their predictive value for increased risk of AF in unadjusted and adjusted models. RESULTS: B-type natriuretic peptide (BNP), N-terminal pro-brain natriuretic peptide (NT-proBNP), creatine kinase, D-dimer and high-sensitivity cardiac troponin I and T were significantly higher in the AF than non-AF group. BNP and NT-proBNP reached the predefined area under the curve level, 0.755 and 0.725 respectively. Optimal cut-off values were 33.5 ng/l for BNP and 87 ng/l for NT-proBNP. Regression analysis showed that NT-proBNP was a predictor of AF in both unadjusted (odds ratio 7.72, 95% confidence interval 3.16-18.87) and age- and sex-adjusted models (odds ratio 4.82, 95% confidence interval 1.79-12.96). CONCLUSION: Several clinically established biomarkers were associated with AF. NT-proBNP performed best as AF predictor and could be used for selecting patients for long-term monitoring with ICMs.


Subject(s)
Atrial Fibrillation , Ischemic Attack, Transient , Ischemic Stroke , Stroke , Humans , Atrial Fibrillation/complications , Atrial Fibrillation/diagnosis , Stroke/complications , Biomarkers , Natriuretic Peptide, Brain , Ischemic Attack, Transient/complications , Ischemic Stroke/complications , Peptide Fragments
5.
Nucleic Acids Res ; 47(18): e112, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31361894

ABSTRACT

Covariance-based discovery of polymorphisms under co-selective pressure or epistasis has received considerable recent attention in population genomics. Both statistical modeling of the population level covariation of alleles across the chromosome and model-free testing of dependencies between pairs of polymorphisms have been shown to successfully uncover patterns of selection in bacterial populations. Here we introduce a model-free method, SpydrPick, whose computational efficiency enables analysis at the scale of pan-genomes of many bacteria. SpydrPick incorporates an efficient correction for population structure, which adjusts for the phylogenetic signal in the data without requiring an explicit phylogenetic tree. We also introduce a new type of visualization of the results similar to the Manhattan plots used in genome-wide association studies, which enables rapid exploration of the identified signals of co-evolution. Simulations demonstrate the usefulness of our method and give some insight to when this type of analysis is most likely to be successful. Application of the method to large population genomic datasets of two major human pathogens, Streptococcus pneumoniae and Neisseria meningitidis, revealed both previously identified and novel putative targets of co-selection related to virulence and antibiotic resistance, highlighting the potential of this approach to drive molecular discoveries, even in the absence of phenotypic data.


Subject(s)
Computational Biology/methods , Epistasis, Genetic , Genome, Bacterial/genetics , Genomics , Drug Resistance, Microbial/genetics , Humans , Metagenomics/methods , Neisseria meningitidis/genetics , Neisseria meningitidis/pathogenicity , Streptococcus pneumoniae/genetics , Virulence/genetics
6.
Appl Environ Microbiol ; 86(6)2020 03 02.
Article in English | MEDLINE | ID: mdl-31900307

ABSTRACT

Listeria monocytogenes causes the severe foodborne illness listeriosis and survives in food-associated environments due to its high stress tolerance. A data assembly and analysis protocol for microbial growth experiments was compiled to elucidate the strain variability of L. monocytogenes stress tolerance. The protocol includes measurement of growth ability under stress (step 1), selection of a suitable method for growth parameter calculation (step 2), comparison of growth patterns between strains (step 3), and biological interpretation of the discovered differences (step 4). In step 1, L. monocytogenes strains (n = 388) of various serovars and origins grown on media with 9.0% NaCl were measured using a Bioscreen C microbiology reader. Technical variability of the growth measurements was assessed and eliminated. In step 2, the growth parameters determined by Gompertz, modified-Gompertz, logistic, and Richards models and model-free splines were compared, illustrating differences in the suitability of these methods to describe the experimental data. In step 3, hierarchical clustering was used to describe the NaCl tolerance of L. monocytogenes measured by strain-specific variation in growth ability; tolerant strains had higher growth rates and maximum optical densities and shorter lag phases than susceptible strains. The spline parameter area under the curve best classified "poor," "average," and "good" growers. In step 4, the tested L. monocytogenes lineage I strains (serovars 4b and 1/2b) proved to be significantly more tolerant toward 9.0% NaCl than lineage II strains (serovars 1/2a, 1/2c, and 3a). Our protocol provides systematic tools to gain comparable data for investigating strain-specific variation of bacterial growth under stress.IMPORTANCE The pathogen Listeria monocytogenes causes the foodborne disease listeriosis, which can be fatal in immunocompromised individuals. L. monocytogenes tolerates several environmental stressors and can persist in food-processing environments and grow in foodstuffs despite traditional control measures such as high salt content. Nonetheless, L. monocytogenes strains differ in their ability to withstand stressors. Elucidating the intraspecies strain variability of L. monocytogenes stress tolerance is crucial for the identification of particularly tolerant strains. To enhance reliable identification of variability in bacterial stress tolerance phenotypes, we compiled a large-scale protocol for the entire data assembly and analysis of microbial growth experiments, providing a systematic approach and checklist for experiments on strain-specific growth ability. Our study illustrated the diversity and strain-specific variation of L. monocytogenes stress tolerance with an unprecedented scope and discovered biologically relevant serovar- and lineage-dependent phenotypes of NaCl tolerance.


Subject(s)
Listeria monocytogenes/physiology , Salt Stress/genetics , Sodium Chloride/adverse effects , High-Throughput Screening Assays , Listeria monocytogenes/genetics , Phenotype , Serotyping
7.
PLoS Genet ; 13(2): e1006508, 2017 02.
Article in English | MEDLINE | ID: mdl-28207813

ABSTRACT

Recent advances in the scale and diversity of population genomic datasets for bacteria now provide the potential for genome-wide patterns of co-evolution to be studied at the resolution of individual bases. Here we describe a new statistical method, genomeDCA, which uses recent advances in computational structural biology to identify the polymorphic loci under the strongest co-evolutionary pressures. We apply genomeDCA to two large population data sets representing the major human pathogens Streptococcus pneumoniae (pneumococcus) and Streptococcus pyogenes (group A Streptococcus). For pneumococcus we identified 5,199 putative epistatic interactions between 1,936 sites. Over three-quarters of the links were between sites within the pbp2x, pbp1a and pbp2b genes, the sequences of which are critical in determining non-susceptibility to beta-lactam antibiotics. A network-based analysis found these genes were also coupled to that encoding dihydrofolate reductase, changes to which underlie trimethoprim resistance. Distinct from these antibiotic resistance genes, a large network component of 384 protein coding sequences encompassed many genes critical in basic cellular functions, while another distinct component included genes associated with virulence. The group A Streptococcus (GAS) data set population represents a clonal population with relatively little genetic variation and a high level of linkage disequilibrium across the genome. Despite this, we were able to pinpoint two RNA pseudouridine synthases, which were each strongly linked to a separate set of loci across the chromosome, representing biologically plausible targets of co-selection. The population genomic analysis method applied here identifies statistically significantly co-evolving locus pairs, potentially arising from fitness selection interdependence reflecting underlying protein-protein interactions, or genes whose product activities contribute to the same phenotype. This discovery approach greatly enhances the future potential of epistasis analysis for systems biology, and can complement genome-wide association studies as a means of formulating hypotheses for targeted experimental work.


Subject(s)
Epistasis, Genetic , Selection, Genetic/genetics , Streptococcus pneumoniae/genetics , Streptococcus pyogenes/genetics , beta-Lactam Resistance/genetics , Aminoacyltransferases/genetics , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/genetics , Gene Regulatory Networks/genetics , Genetics, Population , Genome, Bacterial/genetics , Genomics , Genotype , Humans , Microbial Sensitivity Tests , Penicillin-Binding Proteins/chemistry , Penicillin-Binding Proteins/genetics , Peptidyl Transferases/genetics , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/pathogenicity , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/pathogenicity , beta-Lactams/metabolism
8.
Environ Manage ; 56(5): 1199-213, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26141223

ABSTRACT

The proportion of beef cattle in relation to the total number of cattle has increased in Europe, which has led to a higher contribution of beef cattle in the management of semi-natural grasslands. Changes in vegetation caused by this change in grazers are virtually unexplored so far. In the present study, the impacts of beef and dairy cattle on vegetation structure and composition were compared on Bothnian Bay coastal meadows. Vegetation parameters were measured in seven beef cattle, six dairy heifer pastures, and in six unmanaged meadows. Compared to unmanaged meadows, vegetation in grazed meadows was significantly lower in height and more frequently colonized by low-growth species. As expected, vegetation grazed by beef cattle was more open than that on dairy heifer pastures where litter cover and proportion of bare ground were in the same level as in the unmanaged meadows. However, the observed differences may have in part arisen from the higher cattle densities in coastal meadows grazed by beef cattle than by dairy heifers. The frequencies of different species groups and the species richness values of vegetation did not differ between the coastal meadows grazed by the two cattle types. One reason for this may be the relatively short management history of the studied pastures. The potential differences in grazing impacts of the two cattle types on vegetation structure can be utilized in the management of coastal meadows for species with divergent habitat requirements.


Subject(s)
Agriculture/methods , Feeding Behavior , Grassland , Animals , Biodiversity , Cattle , Conservation of Natural Resources , Ecosystem , Europe , Female , Male
9.
Front Cell Infect Microbiol ; 14: 1326730, 2024.
Article in English | MEDLINE | ID: mdl-38333035

ABSTRACT

Introduction: Improved understanding of Staphylococcus aureus throat colonization in the presence of other co-existing microbes is important for mapping S. aureus adaptation to the human throat, and recurrence of infection. Here, we explore the responses triggered by the encounter between two common throat bacteria, S. aureus and Streptococcus anginosus, to identify genes in S. aureus that are important for colonization in the presence of human tonsillar epithelial cells and S. anginosus, and further compare this transcriptome with the genes expressed in S. aureus as only bacterium. Methods: We performed an in vitro co-culture experiment followed by RNA sequencing to identify interaction-induced transcriptional alterations and differentially expressed genes (DEGs), followed by gene enrichment analysis. Results and discussion: A total of 332 and 279 significantly differentially expressed genes with p-value < 0.05 and log2 FoldChange (log2FC) ≥ |2| were identified in S. aureus after 1 h and 3 h co-culturing, respectively. Alterations in expression of various S. aureus survival factors were observed when co-cultured with S. anginosus and tonsillar cells. The serine-aspartate repeat-containing protein D (sdrD) involved in adhesion, was for example highly upregulated in S. aureus during co-culturing with S. anginosus compared to S. aureus grown in the absence of S. anginosus, especially at 3 h. Several virulence genes encoding secreted proteins were also highly upregulated only when S. aureus was co-cultured with S. anginosus and tonsillar cells, and iron does not appear to be a limiting factor in this environment. These findings may be useful for the development of interventions against S. aureus throat colonization and could be further investigated to decipher the roles of the identified genes in the host immune response in context of a throat commensal landscape.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Transcriptome , Streptococcus anginosus/genetics , Coculture Techniques , Staphylococcal Infections/microbiology
10.
J Neurol ; 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38904781

ABSTRACT

BACKGROUND: Whether specific imaging aspects can be used to identify cryptogenic stroke (CS) patients with high risk of underlying atrial fibrillation (AF) remains unclear. The purpose of this study was to evaluate brain-imaging features in CS patients and their utility as AF predictors. METHODS: The Nordic Atrial Fibrillation and Stroke study was a prospective observational study of CS and transient ischemic attack patients undergoing 12-month cardiac-rhythm monitoring, biomarker and clinical assessments. In this imaging sub-study, brain magnetic resonance imaging and computed tomography scans from 106 patients were assessed for acute and chronic ischemic lesions in relation to AF occurrence and included in a score to predict AF. Receiver operating characteristics (ROC) curve was used to evaluate the discriminative ability of the score and for its dichotomization for predictive model. RESULTS: Age, periventricular white-matter hyperintensities (PVWMH), acute lesion size, and vessel occlusion were significantly associated with AF. Acute and chronic cortical infarcts as well as chronic cerebellar infarcts were numerically more frequent in the AF group than the non-AF group. A score consisting of six features (0-6 points) was proposed (age ≥ 65 years, chronic cortical or cerebellar lesions, acute cortical lesions, PVWMH ≥ 2 in Fazekas scale, vessel occlusion, and acute lesion size ≥ 10 mm). Area under ROC curve was 0.735 and a score of ≥ 3 points was a predictor of AF. CONCLUSIONS: The suggested score was shown to identify CS patients with an increased risk of underlying AF.

11.
Clin Nutr ESPEN ; 60: 17-23, 2024 04.
Article in English | MEDLINE | ID: mdl-38479906

ABSTRACT

BACKGROUND AND AIMS: Low muscle strength, low muscle mass, and sarcopenia have a negative impact on health outcomes in colorectal cancer (CRC) patients. Different diagnostic modalities are used to identify these conditions but it is unknown how well the modalities agree. The aim of this study was to compare different diagnostic modalities by means of calculating the proportion of low muscle strength, low muscle mass, and sarcopenia in CRC patients, and to investigate the agreement for sarcopenia between the various modalities. METHODS: Men and women participating in the Norwegian Dietary Guidelines and colorectal cancer Survival (CRC-NORDIET) study were included in the analyses. Cut-off values for low muscle strength, low muscle mass, and sarcopenia were defined according to the second consensus set by the European Working Group on Sarcopenia in Older People (EWGSOP2). The diagnostic modalities used to assess muscle strength were handgrip strength and the sit-to-stand test. For muscle mass, computed tomography, dual-energy X-ray absorptiometry (DXA), multi-frequency bioelectrical impedance analysis (MF-BIA), and single-frequency BIA (SF-BIA) were applied. Cohen's kappa was calculated to determine the agreement for low muscle strength and confirmed sarcopenia between diagnostic modalities. RESULTS: Five hundred and three men and women (54 % men, mean age of 66 (range 50-80) years old) were included in the analysis. As much as 99 % (n = 70) of the population was identified with low muscle mass by MF-BIA, while the other modalities identified 9-49 % as having low muscle mass. Handgrip strength identified a lower proportion of low muscle strength as compared with the sit-to-stand test (4 % vs. 8 %). When applying various combinations of diagnostic modalities for low muscle strength and low muscle mass, the proportion of sarcopenia was found to be between 0.3 and 11.4 %. There was relatively poor agreement between the different diagnostic modalities with Cohen's Kappa ranging from 0.0 to 0.55, except for the agreement between SF-BIASergi and MF-BIASergi, which was 1. CONCLUSION: The proportion of low muscle strength, low muscle mass, and sarcopenia in CRC patients varied considerably depending on the diagnostic modalities used. Further studies are needed to provide modality-specific cut-off values, adjusted to sex, age and body size.


Subject(s)
Colorectal Neoplasms , Sarcopenia , Male , Humans , Female , Aged , Middle Aged , Aged, 80 and over , Sarcopenia/diagnosis , Sarcopenia/epidemiology , Hand Strength/physiology , Muscle, Skeletal/pathology , Electric Impedance , Muscle Strength , Colorectal Neoplasms/complications , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology
12.
NPJ Vaccines ; 9(1): 105, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38866805

ABSTRACT

Campylobacter is a leading cause of food-borne gastroenteritis worldwide, linked to the consumption of contaminated poultry meat. Targeting this pathogen at source, vaccines for poultry can provide short-term caecal reductions in Campylobacter numbers in the chicken intestine. However, this approach is unlikely to reduce Campylobacter in the food chain or human incidence. This is likely as vaccines typically target only a subset of the high genomic strain diversity circulating among chicken flocks, and rapid evolution diminishes vaccine efficacy over time. To address this, we used a genomic approach to develop a whole-cell autogenous vaccine targeting isolates harbouring genes linked to survival outside of the host. We hyper-immunised a whole major UK breeder farm to passively target offspring colonisation using maternally-derived antibody. Monitoring progeny, broiler flocks revealed a near-complete shift in the post-vaccination Campylobacter population with an ~50% reduction in isolates harbouring extra-intestinal survival genes and a significant reduction of Campylobacter cells surviving on the surface of meat. Based on these findings, we developed a logistic regression model that predicted that vaccine efficacy could be extended to target 65% of a population of clinically relevant strains. Immuno-manipulation of poultry microbiomes towards less harmful commensal isolates by competitive exclusion, has major potential for reducing pathogens in the food production chain.

13.
Lancet Microbe ; 5(2): e142-e150, 2024 02.
Article in English | MEDLINE | ID: mdl-38219757

ABSTRACT

BACKGROUND: The effect of antibiotic usage on the success of multidrug-resistant (MDR) clones in a population remains unclear. With this genomics-based molecular epidemiology study, we aimed to investigate the contribution of antibiotic use to Escherichia coli clone success, relative to intra-strain competition for colonisation and infection. METHODS: We sequenced all the available E coli bloodstream infection isolates provided by the British Society for Antimicrobial Chemotherapy (BSAC) from 2012 to 2017 (n=718) and combined these with published data from the UK (2001-11; n=1090) and Norway (2002-17; n=3254). Defined daily dose (DDD) data from the European Centre for Disease Prevention and Control (retrieved on Sept 21, 2021) for major antibiotic classes (ß-lactam, tetracycline, macrolide, sulfonamide, quinolone, and non-penicillin ß-lactam) were used together with sequence typing, resistance profiling, regression analysis, and non-neutral Wright-Fisher simulation-based modelling to enable systematic comparison of resistance levels, clone success, and antibiotic usage between the UK and Norway. FINDINGS: Sequence type (ST)73, ST131, ST95, and ST69 accounted for 892 (49·3%) of 1808 isolates in the BSAC collection. In the UK, the proportion of ST69 increased between 2001-10 and 2011-17 (p=0·0004), whereas the proportions of ST73 and ST95 did not vary between periods. ST131 expanded quickly after its emergence in 2003 and its prevalence remained consistent throughout the study period (apart from a brief decrease in 2009-10). The extended-spectrum ß-lactamase (ESBL)-carrying, globally disseminated MDR clone ST131-C2 showed overall greater success in the UK (154 [56·8%] of 271 isolates in 2003-17) compared with Norway (51 [18·3%] of 278 isolates in 2002-17; p<0·0001). DDD data indicated higher total use of antimicrobials in the UK, driven mainly by the class of non-penicillin ß-lactams, which were used between 2·7-times and 5·1-times more in the UK per annum (ratio mean 3·7 [SD 0·8]). This difference was associated with the higher success of the MDR clone ST131-C2 (pseudo-R2 69·1%). A non-neutral Wright-Fisher model replicated the observed expansion of non-MDR and MDR sequence types under higher DDD regimes. INTERPRETATION: Our study indicates that resistance profiles of contemporaneously successful clones can vary substantially, warranting caution in the interpretation of correlations between aggregate measures of resistance and antibiotic usage. Our study further suggests that in countries with low-to-moderate use of antibiotics, such as the UK and Norway, the extent of non-penicillin ß-lactam use modulates rather than determines the success of widely disseminated MDR ESBL-carrying E coli clones. Detailed understanding of underlying causal drivers of success is important for improved control of resistant pathogens. FUNDING: Trond Mohn Foundation, Marie Sklodowska-Curie Actions, European Research Council, Royal Society, and Wellcome Trust.


Subject(s)
Escherichia coli Infections , Escherichia coli , Humans , Escherichia coli/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cohort Studies , beta-Lactamases/genetics , beta-Lactamases/pharmacology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/epidemiology , Genomics , beta-Lactams/pharmacology
14.
BMJ Open ; 13(3): e064311, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36997259

ABSTRACT

OBJECTIVES: This study aimed to characterise participants lost to follow-up and identify possible factors associated with non-participation in a prospective population-based study of respiratory health in Norway. We also aimed to analyse the impact of potentially biased risk estimates associated with a high proportion of non-responders. DESIGN: Prospective 5-year follow-up study. SETTING: Randomly selected inhabitants from the general population of Telemark County in south-eastern Norway were invited to fill in a postal questionnaire in 2013. Responders in 2013 were followed-up in 2018. PARTICIPANTS: 16 099 participants aged 16-50 years completed the baseline study. 7958 responded at the 5-year follow-up, while 7723 did not. MAIN OUTCOME MEASURES: χ2 test was performed to compare demographic and respiratory health-related characteristics between those who participated in 2018 and those who were lost to follow-up. Adjusted multivariable logistic regression models were used to assess the relationship between loss to follow-up, background variables, respiratory symptoms, occupational exposure and interactions, and to analyse whether loss to follow-up leads to biased risk estimates. RESULTS: 7723 (49%) participants were lost to follow-up. Loss to follow-up was significantly higher for male participants, those in the youngest age group (16-30 years), those in lowest education level category and among current smokers (all p<0.001). In multivariable logistic regression analysis, loss to follow-up was significantly associated with unemployment (OR 1.34, 95% CI 1.22 to 1.46), reduced work ability (1.48, 1.35 to 1.60), asthma (1.22, 1.10 to 1.35), being woken by chest tightness (1.22, 1.11 to 1.34) and chronic obstructive pulmonary disease (1.81, 1.30 to 2.52). Participants with more respiratory symptoms and exposure to vapour, gas, dust and fumes (VGDF) (1.07 to 1.00-1.15), low-molecular weight (LMW) agents (1.19, 1.00 to 1.41) and irritating agents (1.15, 1.05 to 1.26) were more likely to be lost to follow-up. We found no statistically significant association of wheezing and exposure to LMW agents for all participants at baseline (1.11, 0.90 to 1.36), responders in 2018 (1.12, 0.83 to 1.53) and those lost to follow-up (1.07, 0.81 to 1.42). CONCLUSION: The risk factors for loss to 5-year follow-up were comparable to those reported in other population-based studies and included younger age, male gender, current smoking, lower educational level and higher symptom prevalence and morbidity. We found that exposure to VGDF, irritating and LMW agents can be risk factors associated with loss to follow-up. Results suggest that loss to follow-up did not affect estimates of occupational exposure as a risk factor for respiratory symptoms.


Subject(s)
Asthma , Occupational Exposure , Pulmonary Disease, Chronic Obstructive , Humans , Male , Follow-Up Studies , Prospective Studies , Risk Factors , Pulmonary Disease, Chronic Obstructive/epidemiology , Asthma/epidemiology , Occupational Exposure/adverse effects , Gases/adverse effects
15.
PLoS One ; 17(5): e0268822, 2022.
Article in English | MEDLINE | ID: mdl-35622848

ABSTRACT

BACKGROUND: In March 2020, WHO announced the COVID-19 a pandemic and a major global public health emergency. Mortality from COVID-19 is rapidly increasing globally, with acute respiratory failure as the predominant cause of death. Many patients experience severe hypoxia and life-threatening respiratory failure often requiring mechanical ventilation. To increase safety margins during emergency anaesthesia and rapid sequence intubation (RSI), patients are preoxygenated with a closed facemask with high-flow oxygen and positive end-expiratory pressure (PEEP). Due to the high shunt fraction of deoxygenated blood through the lungs frequently described in COVID-19 however, these measures may be insufficient to avoid harmful hypoxemia. Preoxygenation with inhaled nitric oxide (iNO) potentially reduces the shunt fraction and may thus allow for the necessary margins of safety during RSI. METHODS AND DESIGN: The INOCOV protocol describes a phase II pharmacological trial of inhaled nitric oxide (iNO) as an adjunct to standard of care with medical oxygen in initial airway and ventilation management of patients with known or suspected COVID-19 in acute respiratory failure. The trial is parallel two-arm, randomized, controlled, blinded trial. The primary outcome measure is the change in oxygen saturation (SpO2), and the null hypothesis is that there is no difference in the change in SpO2 following initiation of iNO. TRIAL REGISTRATION: EudraCT number 2020-001656-18; WHO UTN: U1111-1250-1698. Protocol version: 2.0 (June 25th, 2021).


Subject(s)
COVID-19 , Respiratory Distress Syndrome , Respiratory Insufficiency , Administration, Inhalation , Humans , Hypoxia/drug therapy , Nitric Oxide/therapeutic use , Oxygen , Randomized Controlled Trials as Topic , Respiratory Insufficiency/complications
16.
Nat Commun ; 13(1): 7417, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36456554

ABSTRACT

Opportunistic bacterial pathogen species and their strains that colonise the human gut are generally understood to compete against both each other and the commensal species colonising this ecosystem. Currently we are lacking a population-wide quantification of strain-level colonisation dynamics and the relationship of colonisation potential to prevalence in disease, and how ecological factors might be modulating these. Here, using a combination of latest high-resolution metagenomics and strain-level genomic epidemiology methods we performed a characterisation of the competition and colonisation dynamics for a longitudinal cohort of neonatal gut microbiomes. We found strong inter- and intra-species competition dynamics in the gut colonisation process, but also a number of synergistic relationships among several species belonging to genus Klebsiella, which includes the prominent human pathogen Klebsiella pneumoniae. No evidence of preferential colonisation by hospital-adapted pathogen lineages in either vaginal or caesarean section birth groups was detected. Our analysis further enabled unbiased assessment of strain-level colonisation potential of extra-intestinal pathogenic Escherichia coli (ExPEC) in comparison with their propensity to cause bloodstream infections. Our study highlights the importance of systematic surveillance of bacterial gut pathogens, not only from disease but also from carriage state, to better inform therapies and preventive medicine in the future.


Subject(s)
Cesarean Section , Ecosystem , Female , Pregnancy , Infant, Newborn , Humans , Klebsiella , Metagenomics , Parturition , Escherichia coli/genetics
17.
BMJ Open ; 11(6): e048541, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34168032

ABSTRACT

INTRODUCTION: In the multisystem inflammatory disorder systemic sclerosis (SSc), gastrointestinal tract (GIT) affliction is highly prevalent. There are no known disease modifying therapies and the negative impact is substantial. Aiming for a new therapeutic principle, and inspired by recent work showing associations between gut microbiota changes and GIT symptoms in SSc, we performed a pilot study on faecal microbiota transplantation (FMT) with the single-donor bacterial culture 'Anaerobic Cultivated Human Intestinal Microbiome (ACHIM)'. Motivated by positive pilot study signals, we designed the ReSScue trial as a phase II multicentre, placebo-controlled, randomised 20-week trial to evaluate safety and efficacy on lower GIT symptoms of FMT by ACHIM in SSc. METHODS AND ANALYSES: We aim to include 70 SSc participants with moderate to severe lower GIT symptoms, defined by the validated patient-reported University of California Los Angeles Scleroderma Clinical Trial Consortium GIT 2.0 2.0 questionnaire. The trial includes three parts. In part A1 (induction phase) lasting from week 0 to week 12, participants will be randomised 1:1 to repeat infusions of 30 mL ACHIM or placebo at week 0 and 2 by gastroduodenoscopy. In part A2, which is an 8-week subsequent maintenance phase, all study participants will receive 30 mL ACHIM at week 12 and followed until week 20 on continued blind. In part B, which will last until the last participant completes part A2, the participants will be followed through a maximum 16-week extended monitoring period, for longer-term data on safety and intervention effects. Primary endpoint is change from baseline to week 12 in UCLA GIT subscale scores of diarrhoea or bloating, depending on the worst symptom at baseline evaluated separately for each patient. Secondary endpoints are safety measures and changes in UCLA GIT scores (total, diarrhoea and bloating). ETHICS AND DISSEMINATION: This protocol was approved by the Northern Norwegian Committee for Medical Ethics. Study findings will be published. TRIAL REGISTRATION NUMBER: NCT04300426; Pre-results. PROTOCOL VERSION: V.3.1.


Subject(s)
Gastrointestinal Microbiome , Scleroderma, Systemic , Anaerobiosis , Clinical Trials, Phase II as Topic , Double-Blind Method , Fecal Microbiota Transplantation , Humans , Los Angeles , Multicenter Studies as Topic , Pilot Projects , Randomized Controlled Trials as Topic , Scleroderma, Systemic/therapy , Treatment Outcome
18.
Nat Commun ; 12(1): 1523, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33750782

ABSTRACT

Enterococcus faecalis is a commensal and nosocomial pathogen, which is also ubiquitous in animals and insects, representing a classical generalist microorganism. Here, we study E. faecalis isolates ranging from the pre-antibiotic era in 1936 up to 2018, covering a large set of host species including wild birds, mammals, healthy humans, and hospitalised patients. We sequence the bacterial genomes using short- and long-read techniques, and identify multiple extant hospital-associated lineages, with last common ancestors dating back as far as the 19th century. We find a population cohesively connected through homologous recombination, a metabolic flexibility despite a small genome size, and a stable large core genome. Our findings indicate that the apparent hospital adaptations found in hospital-associated E. faecalis lineages likely predate the "modern hospital" era, suggesting selection in another niche, and underlining the generalist nature of this nosocomial pathogen.


Subject(s)
Cross Infection/microbiology , Enterococcus faecalis/genetics , Animals , Anti-Bacterial Agents , Birds , Drug Resistance, Bacterial/genetics , Enterococcus faecalis/drug effects , Enterococcus faecalis/isolation & purification , Genes, MDR/genetics , Genome, Bacterial , Gram-Positive Bacterial Infections/microbiology , Hospitals , Host Specificity , Humans , Phylogeny , Virulence Factors , Whole Genome Sequencing
19.
Microb Genom ; 6(12)2020 12.
Article in English | MEDLINE | ID: mdl-33253085

ABSTRACT

Enterococcus faecium is a gut commensal of the gastro-digestive tract, but also known as nosocomial pathogen among hospitalized patients. Population genetics based on whole-genome sequencing has revealed that E. faecium strains from hospitalized patients form a distinct clade, designated clade A1, and that plasmids are major contributors to the emergence of nosocomial E. faecium. Here we further explored the adaptive evolution of E. faecium using a genome-wide co-evolution study (GWES) to identify co-evolving single-nucleotide polymorphisms (SNPs). We identified three genomic regions harbouring large numbers of SNPs in tight linkage that are not proximal to each other based on the completely assembled chromosome of the clade A1 reference hospital isolate AUS0004. Close examination of these regions revealed that they are located at the borders of four different types of large-scale genomic rearrangements, insertion sites of two different genomic islands and an IS30-like transposon. In non-clade A1 isolates, these regions are adjacent to each other and they lack the insertions of the genomic islands and IS30-like transposon. Additionally, among the clade A1 isolates there is one group of pet isolates lacking the genomic rearrangement and insertion of the genomic islands, suggesting a distinct evolutionary trajectory. In silico analysis of the biological functions of the genes encoded in three regions revealed a common link to a stress response. This suggests that these rearrangements may reflect adaptation to the stringent conditions in the hospital environment, such as antibiotics and detergents, to which bacteria are exposed. In conclusion, to our knowledge, this is the first study using GWES to identify genomic rearrangements, suggesting that there is considerable untapped potential to unravel hidden evolutionary signals from population genomic data.


Subject(s)
Enterococcus faecium/classification , Gram-Positive Bacterial Infections/microbiology , Polymorphism, Single Nucleotide , Whole Genome Sequencing/methods , Cross Infection/microbiology , DNA Transposable Elements , Enterococcus faecium/genetics , Evolution, Molecular , Genomic Islands , High-Throughput Nucleotide Sequencing , Humans , Phylogeny , Plasmids/genetics
20.
Nat Genet ; 51(3): 548-559, 2019 03.
Article in English | MEDLINE | ID: mdl-30778225

ABSTRACT

Streptococcus pyogenes causes 700 million human infections annually worldwide, yet, despite a century of intensive effort, there is no licensed vaccine against this bacterium. Although a number of large-scale genomic studies of bacterial pathogens have been published, the relationships among the genome, transcriptome, and virulence in large bacterial populations remain poorly understood. We sequenced the genomes of 2,101 emm28 S. pyogenes invasive strains, from which we selected 492 phylogenetically diverse strains for transcriptome analysis and 50 strains for virulence assessment. Data integration provided a novel understanding of the virulence mechanisms of this model organism. Genome-wide association study, expression quantitative trait loci analysis, machine learning, and isogenic mutant strains identified and confirmed a one-nucleotide indel in an intergenic region that significantly alters global transcript profiles and ultimately virulence. The integrative strategy that we used is generally applicable to any microbe and may lead to new therapeutics for many human pathogens.


Subject(s)
Genome, Bacterial/genetics , Streptococcus pyogenes/genetics , Transcriptome/genetics , Virulence/genetics , Gene Expression Regulation, Bacterial/genetics , Genome-Wide Association Study/methods , Genomics/methods , Phylogeny , Quantitative Trait Loci/genetics
SELECTION OF CITATIONS
SEARCH DETAIL