Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 200
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38732074

ABSTRACT

Early diagnosis of infections in young infants remains a clinical challenge. Young infants are particularly vulnerable to infection, and it is often difficult to clinically distinguish between bacterial and viral infections. Urinary tract infection (UTI) is the most common bacterial infection in young infants, and the incidence of associated bacteremia has decreased in the recent decades. Host RNA expression signatures have shown great promise for distinguishing bacterial from viral infections in young infants. This prospective study included 121 young infants admitted to four pediatric emergency care departments in the capital region of Denmark due to symptoms of infection. We collected whole blood samples and performed differential gene expression analysis. Further, we tested the classification performance of a two-gene host RNA expression signature approaching clinical implementation. Several genes were differentially expressed between young infants with UTI without bacteremia and viral infection. However, limited immunological response was detected in UTI without bacteremia compared to a more pronounced response in viral infection. The performance of the two-gene signature was limited, especially in cases of UTI without bloodstream involvement. Our results indicate a need for further investigation and consideration of UTI in young infants before implementing host RNA expression signatures in clinical practice.


Subject(s)
Urinary Tract Infections , Humans , Urinary Tract Infections/genetics , Infant , Prospective Studies , Female , Male , Transcriptome , Infant, Newborn , Gene Expression Profiling/methods , Bacteremia/genetics , RNA/genetics , Virus Diseases/genetics
2.
Am J Trop Med Hyg ; 110(5): 994-998, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38507807

ABSTRACT

Melioidosis, infection caused by Burkholderia pseudomallei, is characterized by robust innate immune responses. We have previously reported associations of TLR1 single nucleotide missense variant rs76600635 with mortality and of TLR5 nonsense variant rs5744168 with both bacteremia and mortality in single-center studies of patients with melioidosis in northeastern Thailand. The objective of this study was to externally validate the associations of rs76600635 and rs5744168 with bacteremia and mortality in a large multicenter cohort of melioidosis patients. We genotyped rs76600635 and rs5744168 in 1,338 melioidosis patients enrolled in a prospective parent cohort study conducted at nine hospitals in northeastern Thailand. The genotype frequencies of rs76600635 did not differ by bacteremia status (P = 0.27) or 28-day mortality (P = 0.84). The genotype frequencies of rs5744168 did not differ by either bacteremia status (P = 0.46) or 28-day mortality (P = 0.10). Assuming a dominant genetic model, there was no association of the rs76600635 variant with bacteremia (adjusted odds ratio [OR], 0.75; 95% CI, 0.54-1.04, P = 0.08) or 28-day mortality (adjusted OR, 0.96; 95% CI, 0.71-1.28, P = 0.77). There was no association of the rs5744168 variant with bacteremia (adjusted OR, 1.24; 95% CI, 0.76-2.03, P = 0.39) or 28-day mortality (adjusted OR, 1.22; 95% CI, 0.83-1.79, P = 0.21). There was also no association of either variant with 1-year mortality. We conclude that in a large multicenter cohort of patients hospitalized with melioidosis in northeastern Thailand, neither TLR1 missense variant rs76600635 nor TLR5 nonsense variant rs5744168 is associated with bacteremia or mortality.


Subject(s)
Bacteremia , Melioidosis , Toll-Like Receptor 1 , Toll-Like Receptor 5 , Humans , Melioidosis/mortality , Melioidosis/genetics , Melioidosis/microbiology , Male , Female , Toll-Like Receptor 1/genetics , Thailand/epidemiology , Middle Aged , Bacteremia/mortality , Bacteremia/microbiology , Bacteremia/genetics , Toll-Like Receptor 5/genetics , Adult , Cohort Studies , Polymorphism, Single Nucleotide , Genotype , Burkholderia pseudomallei/genetics , Prospective Studies , Aged , Genetic Predisposition to Disease
3.
Cell Rep ; 42(9): 113069, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37703880

ABSTRACT

Outcomes of severe bacterial infections are determined by the interplay between host, pathogen, and treatments. While human genomics has provided insights into host factors impacting Staphylococcus aureus infections, comparatively little is known about S. aureus genotypes and disease severity. Building on the hypothesis that bacterial pathoadaptation is a key outcome driver, we developed a genome-wide association study (GWAS) framework to identify adaptive mutations associated with treatment failure and mortality in S. aureus bacteremia (1,358 episodes). Our research highlights the potential of vancomycin-selected mutations and vancomycin minimum inhibitory concentration (MIC) as key explanatory variables to predict infection severity. The contribution of bacterial variation was much lower for clinical outcomes (heritability <5%); however, GWASs allowed us to identify additional, MIC-independent candidate pathogenesis loci. Using supervised machine learning, we were able to quantify the predictive potential of these adaptive signatures. Our statistical genomics framework provides a powerful means to capture adaptive mutations impacting severe bacterial infections.


Subject(s)
Bacteremia , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Humans , Vancomycin/pharmacology , Vancomycin/therapeutic use , Staphylococcus aureus/genetics , Anti-Bacterial Agents/pharmacology , Genome-Wide Association Study , Staphylococcal Infections/drug therapy , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Bacteremia/drug therapy , Bacteremia/genetics , Bacteremia/microbiology , Microbial Sensitivity Tests , Treatment Outcome
4.
Am J Physiol Gastrointest Liver Physiol ; 325(5): G471-G491, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37697947

ABSTRACT

The liver plays a significant role in regulating a wide range of metabolic, homeostatic, and host-defense functions. However, the impact of liver injury on the host's ability to control bacteremia and morbidity in sepsis is not well understood. Leukocyte recruitment and activation lead to cytokine and chemokine release, which, in turn, trigger hepatocellular injury and elevate nucleotide levels in the extracellular milieu. P2Y2 purinergic receptors, G protein-coupled and activated by extracellular ATP/UTP, are expressed at the cell surface of hepatocytes and nonparenchymal cells. We sought to determine whether P2Y2 purinergic receptor function is necessary for the maladaptive host response to bacterial infection and endotoxin-mediated inflammatory liver injury and mortality in mice. We report that P2Y2 purinergic receptor knockout mice (P2Y2-/-) had attenuated inflammation and liver injury, with improved survival in response to LPS/galactosamine (LPS/GalN; inflammatory liver injury) and cecal ligation and puncture (CLP; polymicrobial sepsis). P2Y2-/- livers had attenuated c-Jun NH2-terminal kinase activation, matrix metallopeptidase-9 expression, and hepatocyte apoptosis in response to LPS/GalN and attenuated inducible nitric oxide synthase and nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain containing 3 protein expression in response to CLP. Implicating liver injury in the disruption of amino acid homeostasis, CLP led to lower serum arginine and higher bacterial load and morbidity in the WT mice, whereas serum arginine levels were comparable to sham-operated controls in P2Y2-/- mice, which had attenuated bacteremia and improved survival. Collectively, our studies highlight the pathophysiological relevance of P2Y2 purinergic receptor function in inflammatory liver injury and dysregulation of systemic amino acid homeostasis with implications for sepsis-associated immune dysfunction and morbidity in mice.NEW & NOTEWORTHY Our studies provide experimental evidence for P2Y2 purinergic receptor-mediated potentiation of inflammatory liver injury, morbidity, and mortality, in two well-established animal models of inflammatory liver injury. Our findings highlight the potential to target P2Y2 purinergic signaling to attenuate the induction of "cytokine storm" and prevent its deleterious consequences on liver function, systemic amino acid homeostasis, host response to bacterial infection, and sepsis-associated morbidity and mortality.


Subject(s)
Bacteremia , Bacterial Infections , Sepsis , Mice , Animals , Lipopolysaccharides/pharmacology , Gene Deletion , Liver , Cytokines/genetics , Bacteremia/complications , Bacteremia/genetics , Nucleotides , Arginine , Receptors, Purinergic , Amino Acids , Mice, Inbred C57BL , Receptors, Purinergic P2Y2/genetics , Mice, Knockout
5.
PLoS Pathog ; 19(7): e1011233, 2023 07.
Article in English | MEDLINE | ID: mdl-37463183

ABSTRACT

Gram-negative bacteremia is a major cause of global morbidity involving three phases of pathogenesis: initial site infection, dissemination, and survival in the blood and filtering organs. Klebsiella pneumoniae is a leading cause of bacteremia and pneumonia is often the initial infection. In the lung, K. pneumoniae relies on many factors like capsular polysaccharide and branched chain amino acid biosynthesis for virulence and fitness. However, mechanisms directly enabling bloodstream fitness are unclear. Here, we performed transposon insertion sequencing (TnSeq) in a tail-vein injection model of bacteremia and identified 58 K. pneumoniae bloodstream fitness genes. These factors are diverse and represent a variety of cellular processes. In vivo validation revealed tissue-specific mechanisms by which distinct factors support bacteremia. ArnD, involved in Lipid A modification, was required across blood filtering organs and supported resistance to soluble splenic factors. The purine biosynthesis enzyme PurD supported liver fitness in vivo and was required for replication in serum. PdxA, a member of the endogenous vitamin B6 biosynthesis pathway, optimized replication in serum and lung fitness. The stringent response regulator SspA was required for splenic fitness yet was dispensable in the liver. In a bacteremic pneumonia model that incorporates initial site infection and dissemination, splenic fitness defects were enhanced. ArnD, PurD, DsbA, SspA, and PdxA increased fitness across bacteremia phases and each demonstrated unique fitness dynamics within compartments in this model. SspA and PdxA enhanced K. pnuemoniae resistance to oxidative stress. SspA, but not PdxA, specifically resists oxidative stress produced by NADPH oxidase Nox2 in the lung, spleen, and liver, as it was a fitness factor in wild-type but not Nox2-deficient (Cybb-/-) mice. These results identify site-specific fitness factors that act during the progression of Gram-negative bacteremia. Defining K. pneumoniae fitness strategies across bacteremia phases could illuminate therapeutic targets that prevent infection and sepsis.


Subject(s)
Bacteremia , Klebsiella Infections , Pneumonia , Mice , Animals , Klebsiella pneumoniae/genetics , Lung , Bacteremia/genetics , Oxidative Stress , Klebsiella Infections/genetics
6.
Sci Rep ; 13(1): 6571, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37085513

ABSTRACT

We investigated the clinical features of bloodstream infections (BSIs) caused by Klebsiella pneumoniae harboring rmpA and molecular characteristics of the bacteria. We retrospectively investigated adult patients with K. pneumoniae BSI from January 2010 to March 2021 at Nagasaki University Hospital. A matched case-control study in a 1:3 ratio was conducted to clarify the clinical and bacterial characteristics of BSI caused by rmpA-positive K. pneumoniae compared with those caused by rmpA-negative isolates. Antimicrobial susceptibility testing and multilocus sequence typing (MLST) were performed for rmpA-positive isolates. The rmpA was detected in 36 (13.4%) of the 268 isolates. Of these 36 isolates, 31 (86.1%) harbored iucA and 35 (97.2%) each possessed peg-344 and iroB; capsular types were identified as K1 in 9 (25.0%) and K2 in 10 isolates (27.8%). Contrarily, of the 108 rmpA-negative isolates, which were matched for case-control studies, 5 isolates (4.6%) harbored iucA and 1 (0.9%) each possessed peg-344 and iroB; 2 (1.9%) and 3 isolates (2.8%) had K1 and K2 capsular types, respectively. Among the rmpA-positive isolates, ST23/K1 (eight isolates) was the most frequent, followed by ST412/non-K1/K2 (seven isolates), ST86/K2 (five isolates), and ST268/non-K1/K2 (four isolates). In a multivariate analysis using clinical factors, liver abscess positively correlated with rmpA-positive isolates, whereas biliary tract infection and use of anticancer drugs negatively correlated with rmpA-positive isolates in patients with K. pneumoniae BSI. Considering the correlation between rmpA-positive isolates and clinical features, rmpA can be used as a marker for understanding the pathophysiology of K. pneumoniae BSI.


Subject(s)
Bacteremia , Bacterial Proteins , Klebsiella Infections , Klebsiella pneumoniae , Adult , Humans , Bacteremia/diagnosis , Bacteremia/genetics , Bacteremia/microbiology , Bacteremia/physiopathology , Bacterial Proteins/blood , Bacterial Proteins/genetics , Case-Control Studies , East Asian People , Japan , Klebsiella Infections/drug therapy , Klebsiella Infections/genetics , Klebsiella Infections/microbiology , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/isolation & purification , Klebsiella pneumoniae/pathogenicity , Multilocus Sequence Typing , Retrospective Studies , Sepsis/diagnosis , Sepsis/genetics , Sepsis/microbiology , Sepsis/physiopathology , Virulence Factors/genetics , Virulence Factors/isolation & purification
7.
Front Cell Infect Microbiol ; 13: 1075255, 2023.
Article in English | MEDLINE | ID: mdl-36844412

ABSTRACT

The occurrence and transmission of carbapenemase-producing-Enterobacterales (CPE) on a global scale has become a major issue. Clinical reports are rarely providing information on the genomic and plasmid features of carbapenem-resistant Serratia marcescens. Our objective was to investigate the resistance and transmission dynamics of two carbapenem-resistant S. marcescens that are resistant to carbapenem and have caused bacteremia in China. Blood specimens were taken from two individuals with bacteremia. Multiplex PCR was employed to identify genes that code for carbapenemase. Antimicrobial susceptibility tests and plasmid analysis were conducted on S. marcescens isolates SM768 and SM4145. The genome of SM768 and SM4145 were completely sequenced using NovaSeq 6000-PE150 and PacBio RS II platforms. Antimicrobial resistance genes (ARGs) were predicted using the ResFinder tool. S1 nuclease pulsed-field gel electrophoresis (S1-PFGE) and southern blotting were employed to analyze plasmids. Two S. marcescens that produced KPC-2 were identified from bloodstream infections. The antimicrobial susceptibility testing demonstrated that both of the isolates had a resistance to various antibiotics. The whole-genome sequence (WGS) and plasmid analysis revealed the presence of bla KPC-2-bearing IncR plasmids and multiple plasmid-borne antimicrobial resistance genes in the isolates. Our comparative plasmid analysis suggested that the two IncR plasmids identified in this study could be derived from a common ancestor. Our findings revealed the emergence of bla KPC-2-bearing IncR plasmid in China, which could be a hindrance to the transmission of KPC-2-producing S. marcescens in clinical settings.


Subject(s)
Anti-Bacterial Agents , Bacteremia , Drug Resistance, Bacterial , Serratia Infections , Serratia marcescens , beta-Lactamases , Humans , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Bacteremia/genetics , Bacteremia/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , beta-Lactamases/genetics , beta-Lactamases/metabolism , Carbapenems/pharmacology , Genomics , Klebsiella Infections , Klebsiella pneumoniae/genetics , Microbial Sensitivity Tests , Plasmids/genetics , Serratia marcescens/genetics , Serratia Infections/drug therapy , Serratia Infections/genetics , Serratia Infections/metabolism , Serratia Infections/microbiology , Drug Resistance, Bacterial/genetics , Drug Resistance, Bacterial/physiology , China , Genome, Bacterial
8.
Infect Immun ; 91(2): e0055922, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36651775

ABSTRACT

Healthcare-acquired infections are a leading cause of disease in patients that are hospitalized or in long-term-care facilities. Klebsiella pneumoniae (Kp) is a leading cause of bacteremia, pneumonia, and urinary tract infections in these settings. Previous studies have established that the ter operon, a genetic locus that confers tellurite oxide (K2TeO3) resistance, is associated with infection in colonized patients. Rather than enhancing fitness during infection, the ter operon increases Kp fitness during gut colonization; however, the biologically relevant function of this operon is unknown. First, using a murine model of urinary tract infection, we demonstrate a novel role for the ter operon protein TerC as a bladder fitness factor. To further characterize TerC, we explored a variety of functions, including resistance to metal-induced stress, resistance to radical oxygen species-induced stress, and growth on specific sugars, all of which were independent of TerC. Then, using well-defined experimental guidelines, we determined that TerC is necessary for tolerance to ofloxacin, polymyxin B, and cetylpyridinium chloride. We used an ordered transposon library constructed in a Kp strain lacking the ter operon to identify the genes that are required to resist K2TeO3-induced and polymyxin B-induced stress, which suggested that K2TeO3-induced stress is experienced at the bacterial cell envelope. Finally, we confirmed that K2TeO3 disrupts the Kp cell envelope, though these effects are independent of ter. Collectively, the results from these studies indicate a novel role for the ter operon as a stress tolerance factor, thereby explaining its role in enhancing fitness in the gut and bladder.


Subject(s)
Bacteremia , Klebsiella Infections , Urinary Tract Infections , Humans , Animals , Mice , Klebsiella pneumoniae/genetics , Polymyxin B/pharmacology , Operon , Urinary Tract Infections/genetics , Bacteremia/genetics , Klebsiella Infections/microbiology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism
9.
Am J Trop Med Hyg ; 108(1): 181-186, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36509047

ABSTRACT

Class 1 and Class 2 integrons are mobilizable elements able to carry a variety of antibiotic resistance determinants. In the present study, Class 1 and 2 integrons present in 355 pathogenic Escherichia coli (285 diarrheagenic, of these 129 were enteropathogenic, 90 enteroaggregative, 66 enterotoxigenic, and 70 bacteremic) isolated from healthy and ill children under age 5 from periurban areas of Lima, Peru, were characterized. The presence of integrase 1 and 2 was established by polymerase chain reaction (PCR), and variable regions were grouped by PCR-restriction fragment length polymorphism and subsequent sequencing. Antimicrobial resistance was established by disk diffusion. Ninety-seven isolates (27.3%) presented integrase 1, and 16 (4.5%) presented integrase 2 (P < 0.0001); in addition, seven (2.0%) isolates, six diarrheagenic and one bacteremic, presented both integrase genes. The presence of integrase 1 was more frequent among bacteremic isolates (P = 0.0004). Variable regions were amplified in 76/120 (63.3%) isolates with up to 14 gene arrangements. The most prevalent gene cassettes were those encoding dihydrofolate reductases as well as aminoglycoside modifying enzymes. Of note, Class 1 integrons tended to be associated with the presence of extended-spectrum ß-lactamases (ESBLs). A variety of Class 1 and 2 integrons were detected in diarrheagenic and bacteremic E. coli, demonstrating the heterogeneity of variable regions circulating in the area. The association of integrons with ESBLs is worrisome and has an impact on the development of multidrug resistance.


Subject(s)
Bacteremia , Diarrhea , Escherichia coli Infections , Escherichia coli , Integrons , Child , Child, Preschool , Humans , Anti-Bacterial Agents/pharmacology , Bacteremia/epidemiology , Bacteremia/genetics , Bacteremia/microbiology , Diarrhea/epidemiology , Diarrhea/genetics , Diarrhea/microbiology , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/isolation & purification , Escherichia coli Infections/epidemiology , Escherichia coli Infections/genetics , Integrases/genetics , Integrons/genetics , Microbial Sensitivity Tests , Peru/epidemiology
10.
Genes (Basel) ; 13(9)2022 08 25.
Article in English | MEDLINE | ID: mdl-36140695

ABSTRACT

Persistent methicillin-resistant Staphylococcus aureus (MRSA) endovascular infections represent a significant subset of S. aureus infections and correlate with exceptionally high mortality. We have recently demonstrated that the lysogenization of prophage ϕSA169 from a clinical persistent MRSA bacteremia isolate (300-169) into a clinical resolving bacteremia MRSA isolate (301-188) resulted in the acquisition of well-defined in vitro and in vivo phenotypic and genotypic profiles related to persistent outcome. However, the underlying mechanism(s) of this impact is unknown. In the current study, we explored the genetic mechanism that may contribute to the ϕSA169-correlated persistence using RNA sequencing. Transcriptomic analyses revealed that the most significant impacts of ϕSA169 were: (i) the enhancement of fatty acid biosynthesis and purine and pyrimidine metabolic pathways; (ii) the repression of galactose metabolism and phosphotransferase system (PTS); and (iii) the down-regulation of the mutual prophage genes in both 300-169 and 301-188 strains. In addition, the influence of different genetic backgrounds between 300-169 and 301-188 might also be involved in the persistent outcome. These findings may provide targets for future studies on the persistence of MRSA.


Subject(s)
Bacteremia , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Bacteremia/genetics , Fatty Acids , Galactose , Gene Expression Profiling , Humans , Methicillin-Resistant Staphylococcus aureus/genetics , Phosphotransferases , Prophages/genetics , Purines , Pyrimidines , Staphylococcal Infections/genetics , Staphylococcus aureus/genetics
11.
Microb Genom ; 8(8)2022 08.
Article in English | MEDLINE | ID: mdl-35921144

ABSTRACT

Insertion sequences (ISs) and other transposable elements are associated with the mobilization of antibiotic resistance determinants and the modulation of pathogenic characteristics. In this work, we aimed to investigate the association between ISs and antibiotic resistance genes, and their role in the dissemination and modification of the antibiotic-resistant phenotype. To that end, we leveraged fully resolved Enterococcus faecium and Enterococcus faecalis genomes of isolates collected over 5 days from an inpatient with prolonged bacteraemia. Isolates from both species harboured similar IS family content but showed significant species-dependent differences in copy number and arrangements of ISs throughout their replicons. Here, we describe two inter-specific IS-mediated recombination events and IS-mediated excision events in plasmids of E. faecium isolates. We also characterize a novel arrangement of the ISs in a Tn1546-like transposon in E. faecalis isolates likely implicated in a vancomycin genotype-phenotype discrepancy. Furthermore, an extended analysis revealed a novel association between daptomycin resistance mutations in liaSR genes and a putative composite transposon in E. faecium, offering a new paradigm for the study of daptomycin resistance and novel insights into its dissemination. In conclusion, our study highlights the role ISs and other transposable elements play in the rapid adaptation and response to clinically relevant stresses such as aggressive antibiotic treatment in enterococci.


Subject(s)
Bacteremia , Daptomycin , Gram-Positive Bacterial Infections , Anti-Bacterial Agents/pharmacology , Bacteremia/genetics , DNA Transposable Elements/genetics , Drug Resistance, Microbial , Enterococcus/genetics , Humans , Inpatients , Microbial Sensitivity Tests
12.
Infect Immun ; 90(7): e0022422, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35762751

ABSTRACT

Klebsiella pneumoniae is a leading cause of Gram-negative bacteremia, which is a major source of morbidity and mortality worldwide. Gram-negative bacteremia requires three major steps: primary site infection, dissemination to the blood, and bloodstream survival. Because K. pneumoniae is a leading cause of health care-associated pneumonia, the lung is a common primary infection site leading to secondary bacteremia. K. pneumoniae factors essential for lung fitness have been characterized, but those required for subsequent bloodstream infection are unclear. To identify K. pneumoniae genes associated with dissemination and bloodstream survival, we combined previously and newly analyzed insertion site sequencing (InSeq) data from a murine model of bacteremic pneumonia. This analysis revealed the gene gmhB as important for either dissemination from the lung or bloodstream survival. In Escherichia coli, GmhB is a partially redundant enzyme in the synthesis of ADP-heptose for the lipopolysaccharide (LPS) core. To characterize its function in K. pneumoniae, an isogenic knockout strain (ΔgmhB) and complemented mutant were generated. During pneumonia, GmhB did not contribute to lung fitness and did not alter normal immune responses. However, GmhB enhanced bloodstream survival in a manner independent of serum susceptibility, specifically conveying resistance to spleen-mediated killing. In a tail-vein injection of murine bacteremia, GmhB was also required by K. pneumoniae, E. coli, and Citrobacter freundii for optimal fitness in the spleen and liver. Together, this study identifies GmhB as a conserved Gram-negative bacteremia fitness factor that acts through LPS-mediated mechanisms to enhance fitness in blood-filtering organs.


Subject(s)
Bacteremia , Klebsiella Infections , Adenosine Diphosphate , Animals , Bacteremia/genetics , Escherichia coli/genetics , Heptoses , Klebsiella pneumoniae/genetics , Lipopolysaccharides , Mice
13.
PLoS Genet ; 18(3): e1010112, 2022 03.
Article in English | MEDLINE | ID: mdl-35324915

ABSTRACT

Escherichia coli is an important cause of bloodstream infections (BSI), which is of concern given its high mortality and increasing worldwide prevalence. Finding bacterial genetic variants that might contribute to patient death is of interest to better understand infection progression and implement diagnostic methods that specifically look for those factors. E. coli samples isolated from patients with BSI are an ideal dataset to systematically search for those variants, as long as the influence of host factors such as comorbidities are taken into account. Here we performed a genome-wide association study (GWAS) using data from 912 patients with E. coli BSI from hospitals in Paris, France. We looked for associations between bacterial genetic variants and three patient outcomes (death at 28 days, septic shock and admission to intensive care unit), as well as two portals of entry (urinary and digestive tract), using various clinical variables from each patient to account for host factors. We did not find any association between genetic variants and patient outcomes, potentially confirming the strong influence of host factors in influencing the course of BSI; we however found a strong association between the papGII operon and entrance of E. coli through the urinary tract, which demonstrates the power of bacterial GWAS when applied to actual clinical data. Despite the lack of associations between E. coli genetic variants and patient outcomes, we estimate that increasing the sample size by one order of magnitude could lead to the discovery of some putative causal variants. Given the wide adoption of bacterial genome sequencing of clinical isolates, such sample sizes may be soon available.


Subject(s)
Bacteremia , Escherichia coli Infections , Sepsis , Bacteremia/epidemiology , Bacteremia/genetics , Bacteremia/microbiology , Bacteria , Escherichia coli/genetics , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Genome-Wide Association Study , Humans
14.
Microbiol Spectr ; 9(3): e0191021, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34937173

ABSTRACT

Due to their phylogenetic proximity to humans, nonhuman primates (NHPs) are considered an adequate choice for a basic and preclinical model of sepsis. Gram-negative bacteria are the primary causative of sepsis. During infection, bacteria continuously release the potent toxin lipopolysaccharide (LPS) into the bloodstream, which triggers an uncontrolled systemic inflammatory response leading to death. Our previous research has demonstrated in vitro and in vivo using a mouse model of septic shock that Fh15, a recombinant variant of the Fasciola hepatica fatty acid binding protein, acts as an antagonist of Toll-like receptor 4 (TLR4) suppressing the LPS-induced proinflammatory cytokine storm. The present communication is a proof-of concept study aimed to demonstrate that a low-dose of Fh15 suppresses the cytokine storm and other inflammatory markers during the early phase of sepsis induced in rhesus macaques by intravenous (i.v.) infusion with lethal doses of live Escherichia coli. Fh15 was administered as an isotonic infusion 30 min prior to the bacterial infusion. Among the novel findings reported in this communication, Fh15 (i) significantly prevented bacteremia, suppressed LPS levels in plasma, and the production of C-reactive protein and procalcitonin, which are key signatures of inflammation and bacterial infection, respectively; (ii) reduced the production of proinflammatory cytokines; and (iii) increased innate immune cell populations in blood, which suggests a role in promoting a prolonged steady state in rhesus macaques even in the presence of inflammatory stimuli. This report is the first to demonstrate that a F. hepatica-derived molecule possesses potential as an anti-inflammatory drug against sepsis in an NHP model. IMPORTANCE Sepsis caused by Gram-negative bacteria affects 1.7 million adults annually in the United States and is one of the most important causes of death at intensive care units. Although the effective use of antibiotics has resulted in improved prognosis of sepsis, the pathological and deathly effects have been attributed to the persistent inflammatory cascade. There is a present need to develop anti-inflammatory agents that can suppress or neutralize the inflammatory responses and prevent the lethal consequences of sepsis. We demonstrated here that a small molecule of 14.5 kDa can suppress the bacteremia, endotoxemia, and many other inflammatory markers in an acute Gram-negative sepsis rhesus macaque model. These results reinforce the notion that Fh15 constitutes an excellent candidate for drug development against sepsis.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Bacteremia/drug therapy , Fasciola hepatica/metabolism , Fatty Acid-Binding Proteins/administration & dosage , Gram-Negative Bacteria/physiology , Helminth Proteins/administration & dosage , Animals , Anti-Inflammatory Agents/metabolism , Bacteremia/genetics , Bacteremia/immunology , Bacteremia/microbiology , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Fasciola hepatica/chemistry , Fasciola hepatica/genetics , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Gram-Negative Bacteria/classification , Gram-Negative Bacteria/genetics , Helminth Proteins/genetics , Helminth Proteins/metabolism , Humans , Macaca mulatta , Male , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
15.
Dis Markers ; 2021: 6166492, 2021.
Article in English | MEDLINE | ID: mdl-34691288

ABSTRACT

Melioidosis is a serious infectious disease caused by the environmental Gram-negative bacillus Burkholderia pseudomallei. It has been shown that the host immune system, mainly comprising various types of immune cells, fights against the disease. The present study was to specify correlation between septicemic melioidosis and the levels of multiple immune cells. First, the genes with differential expression patterns between patients with septicemic melioidosis (B. pseudomallei) and health donors (control/healthy) were identified. These genes being related to cytokine binding, cell adhesion molecule binding, and MHC relevant proteins may influence immune response. The Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed 23 enriched immune response pathways. We further leveraged the microarray data to investigate the relationship between immune response and septicemic melioidosis, using the CIBERSORT analysis. Comparison of the percentages of 22 immune cell types in B. pseudomallei vs. control/healthy revealed that those of CD4 memory resting cells, CD8+ T cells, B memory cells, and CD4 memory activated cells were low, whereas those of M0 macrophages, neutrophils, and gamma delta T cells were high. The multivariate logistic regression analysis further revealed that CD8+ T cells, M0 macrophages, neutrophils, and naive CD4+ cells were strongly associated with the onset of septicemic melioidosis, and M2 macrophages and neutrophils were associated with the survival in septicemic melioidosis. Taken together, these data point to a complex role of immune cells on the development and progression of melioidosis.


Subject(s)
Bacteremia/immunology , Bacteremia/mortality , Blood Proteins/genetics , Melioidosis/immunology , Melioidosis/mortality , Bacteremia/blood , Bacteremia/genetics , Blood/immunology , Blood Physiological Phenomena , Blood Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Case-Control Studies , Gene Expression Profiling , Gene Ontology , Humans , Macrophages/immunology , Macrophages/physiology , Melioidosis/blood , Melioidosis/genetics
16.
Sci Rep ; 11(1): 20328, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34645857

ABSTRACT

Epidemiological studies of communicable diseases increasingly use large whole-genome sequencing (WGS) datasets to explore the transmission of pathogens. It is important to obtain an initial overview of datasets and identify closely related isolates, but this can be challenging with large numbers of isolates and imperfect sequencing. We used an ad hoc whole-genome multi locus sequence typing method to summarise data from a longitudinal study of Staphylococcus aureus in a primary school in New Zealand. Each pair of isolates was compared and the number of genes where alleles differed between isolates was tallied to produce a matrix of "allelic differences". We plotted histograms of the number of allelic differences between isolates for: all isolate pairs; pairs of isolates from different individuals; and pairs of isolates from the same individual. 340 sequenced isolates were included, and the ad hoc shared genome contained 445 genes. There were between 0 and 420 allelic differences between isolate pairs and the majority of pairs had more than 260 allelic differences. We found many genetically closely related S. aureus isolates from single individuals and a smaller number of closely-related isolates from separate individuals. Multiple S. aureus isolates from the same individual were usually very closely related or identical over the ad hoc shared genome. Siblings carried genetically similar, but not identical isolates. An ad hoc shared genome approach to WGS analysis can accommodate imperfect sequencing of the included isolates, and can provide insights into relationships between isolates in epidemiological studies with large WGS datasets containing diverse isolates.


Subject(s)
Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Whole Genome Sequencing , Alleles , Bacteremia/genetics , Child , Computational Biology , Female , Genetic Variation , Genome , Genome, Bacterial , Humans , Longitudinal Studies , Male , Methicillin-Resistant Staphylococcus aureus/genetics , Multilocus Sequence Typing , New Zealand , Schools , Students
17.
J Immunol ; 207(6): 1566-1577, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34433620

ABSTRACT

Nontypeable Haemophilus influenzae (NTHi) is a Gram-negative human pathogen that causes infections mainly in the upper and lower respiratory tract. The bacterium is associated with bronchitis and exacerbations in patients suffering from chronic obstructive pulmonary disease and frequently causes acute otitis media in preschool children. We have previously demonstrated that the binding of C4b binding protein (C4BP) is important for NTHi complement evasion. In this study, we identified outer membrane protein 5 (P5) of NTHi as a novel ligand of C4BP. Importantly, we observed significantly lower C4BP binding and decreased serum resistance in P5-deficient NTHi mutants. Surface expression of recombinant P5 on Escherichia coli conferred C4BP binding and consequently increased serum resistance. Moreover, P5 expression was positively correlated with C4BP binding in a series of clinical isolates. We revealed higher levels of P5 surface expression and consequently more C4BP binding in isolates from the lower respiratory tract of chronic obstructive pulmonary disease patients and tonsil specimens compared with isolates from the upper respiratory tract and the bloodstream (invasive strains). Our results highlight P5 as an important protein for protecting NTHi against complement-mediated killing.


Subject(s)
Bacteremia/immunology , Bacterial Outer Membrane Proteins/metabolism , Complement C4b-Binding Protein/metabolism , Haemophilus Infections/immunology , Haemophilus influenzae/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , Tonsillitis/immunology , Aged , Aged, 80 and over , Bacteremia/genetics , Bacterial Outer Membrane Proteins/genetics , Child , Complement System Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Haemophilus Infections/microbiology , Haemophilus influenzae/genetics , Humans , Ligands , Male , Middle Aged , Organisms, Genetically Modified , Protein Binding/genetics , Pulmonary Disease, Chronic Obstructive/microbiology , Recombinant Proteins/metabolism , Signal Transduction/genetics , Tonsillitis/microbiology
18.
PLoS One ; 16(8): e0256768, 2021.
Article in English | MEDLINE | ID: mdl-34437647

ABSTRACT

White adipocytes store energy differently than brown and brite adipocytes which dissipate energy under the form of heat. Studies have shown that adipocytes are able to respond to bacteria thanks to the presence of Toll-like receptors at their surface. Despite this, little is known about the involvement of each class of adipocytes in the infectious response. We treated mice for one week with a ß3-adrenergic receptor agonist to induce activation of brown adipose tissue and brite adipocytes within white adipose tissue. Mice were then injected intraperitoneally with E. coli to generate acute infection. The metabolic, infectious and inflammatory parameters of the mice were analysed during 48 hours after infection. Our results shown that in response to bacteria, thermogenic activity promoted a discrete and local anti-inflammatory environment in white adipose tissue characterized by the increase of the IL-1RA secretion. More generally, activation of brown and brite adipocytes did not modify the host response to infection including no additive effect with fever and an equivalent bacteria clearance and inflammatory response. In conclusion, these results suggest an IL-1RA-mediated immunomodulatory activity of thermogenic adipocytes in response to acute bacterial infection and open a way to characterize their effect along more chronic infection as septicaemia.


Subject(s)
Bacteremia/drug therapy , Inflammation/drug therapy , Interleukin 1 Receptor Antagonist Protein/genetics , Receptors, Adrenergic, beta-3/genetics , Thermogenesis/drug effects , Adipocytes, Beige/drug effects , Adipocytes, Beige/metabolism , Adipocytes, White/drug effects , Adipocytes, White/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adrenergic Agonists/pharmacology , Animals , Bacteremia/genetics , Bacteremia/metabolism , Bacteremia/microbiology , Dioxoles/pharmacology , Disease Models, Animal , Energy Metabolism/drug effects , Escherichia coli/pathogenicity , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/microbiology , Mice , Toll-Like Receptors/genetics
19.
PLoS Pathog ; 17(4): e1009537, 2021 04.
Article in English | MEDLINE | ID: mdl-33930099

ABSTRACT

Klebsiella pneumoniae (Kp) is an important cause of healthcare-associated infections, which increases patient morbidity, mortality, and hospitalization costs. Gut colonization by Kp is consistently associated with subsequent Kp disease, and patients are predominantly infected with their colonizing strain. Our previous comparative genomics study, between disease-causing and asymptomatically colonizing Kp isolates, identified a plasmid-encoded tellurite (TeO3-2)-resistance (ter) operon as strongly associated with infection. However, TeO3-2 is extremely rare and toxic to humans. Thus, we used a multidisciplinary approach to determine the biological link between ter and Kp infection. First, we used a genomic and bioinformatic approach to extensively characterize Kp plasmids encoding the ter locus. These plasmids displayed substantial variation in plasmid incompatibility type and gene content. Moreover, the ter operon was genetically independent of other plasmid-encoded virulence and antibiotic resistance loci, both in our original patient cohort and in a large set (n = 88) of publicly available ter operon-encoding Kp plasmids, indicating that the ter operon is likely playing a direct, but yet undescribed role in Kp disease. Next, we employed multiple mouse models of infection and colonization to show that 1) the ter operon is dispensable during bacteremia, 2) the ter operon enhances fitness in the gut, 3) this phenotype is dependent on the colony of origin of mice, and 4) antibiotic disruption of the gut microbiota eliminates the requirement for ter. Furthermore, using 16S rRNA gene sequencing, we show that the ter operon enhances Kp fitness in the gut in the presence of specific indigenous microbiota, including those predicted to produce short chain fatty acids. Finally, administration of exogenous short-chain fatty acids in our mouse model of colonization was sufficient to reduce fitness of a ter mutant. These findings indicate that the ter operon, strongly associated with human infection, encodes factors that resist stress induced by the indigenous gut microbiota during colonization. This work represents a substantial advancement in our molecular understanding of Kp pathogenesis and gut colonization, directly relevant to Kp disease in healthcare settings.


Subject(s)
Gastrointestinal Microbiome/genetics , Intestines/microbiology , Klebsiella/genetics , Plasmids/genetics , Animals , Bacteremia/genetics , Bacterial Proteins/genetics , Female , Genetic Fitness/physiology , Genetic Loci/physiology , Genome, Bacterial , Host-Pathogen Interactions/genetics , Kanamycin Resistance/genetics , Klebsiella Infections/microbiology , Male , Mice , Mice, Inbred C57BL , Operon/genetics , Organ Specificity/genetics , Virulence/genetics , beta-Lactamases/genetics
20.
Sci Rep ; 11(1): 6314, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33737590

ABSTRACT

Bacteremia resulting from dental surgery is increasingly recognized as a health risk, especially in older and immunocompromised patients. Dentistry-associated bacteremia can lead to remote infections, as exemplified by valvular endocarditis. Emerging evidence points to a novel role played by oral cavity commensals in the pathogenesis of diabetes, respiratory disease, cardiovascular disease, and adverse pregnancy outcomes. Whether dental extraction, a commonly undertaken procedure in old horses, causes bacteremia has not been reported extensively. In a prospective clinical study using next generation sequencing (based on bacterial 16S rRNA), the circulating blood microbiome was characterized before and at 1 h following extraction of incisor, canine or cheek teeth from 29 adult horses with dental disease. 16S rRNA gene sequencing results from the blood microbiome were compared with those from gingival swab samples obtained prior to extraction at the location of the diseased tooth. Bacteremia associated with translocated gingival commensals was demonstrated in horses undergoing exodontia and was, in some cases, still evident one hour post-operatively.


Subject(s)
Bacteremia/genetics , Horse Diseases/microbiology , Tooth/microbiology , Animals , Bacteremia/complications , Bacteremia/microbiology , Bacteremia/veterinary , High-Throughput Nucleotide Sequencing , Horse Diseases/genetics , Horses , Humans , RNA, Ribosomal, 16S/genetics , Tooth/pathology , Tooth/surgery , Tooth Extraction/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL
...