Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Article in English | MEDLINE | ID: mdl-38656811

ABSTRACT

Pseudomonas aeruginosa causes chronic lung infection in cystic fibrosis (CF), resulting in structural lung damage and progressive pulmonary decline. P. aeruginosa in the CF lung undergoes numerous changes, adapting to host-specific airway pressures while establishing chronic infection. P. aeruginosa undergoes lipid A structural modification during CF chronic infection, not seen in any other disease state. Lipid A, the membrane anchor of lipopolysaccharide (i.e., endotoxin), comprises the majority of the outer membrane of Gram-negative bacteria and is a potent toll-like receptor (TLR)4 agonist. The structure of P. aeruginosa lipid A is intimately linked with its recognition by TLR4, and subsequent immune response. Prior work has identified P. aeruginosa strains with altered lipid A structures that arise during chronic CF lung infection; however, the impact of P. aeruginosa lipid A structure on airway disease has not been investigated. Here, we show that P. aeruginosa lipid A lacks PagL-mediated deacylation during human airway infection using a direct-from-sample mass spectrometry approach on human bronchoalveolar lavage fluid. This structure triggers increased pro-inflammatory cytokine production by primary human macrophages. Furthermore, alterations in lipid A 2-hydroxylation impact cytokine response in a site-specific manner, independent of CFTR function. Interestingly, there is a CF-specific reduction in IL-8 secretion within the epithelial-cell compartment that only occurs in CF bronchial epithelial cells when infected with CF-adapted P. aeruginosa that lack PagL-mediated lipid A deacylation. Taken together, we show that P. aeruginosa alters its lipid A structure during acute lung infection and that this lipid A structure induces stronger signaling through TLR4.

2.
mBio ; 15(2): e0282323, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38131669

ABSTRACT

Pseudomonas aeruginosa can survive in a myriad of environments, partially due to modifications of its lipid A, the membrane anchor of lipopolysaccharide. We previously demonstrated that divergent late acyltransferase paralogs, HtrB1 and HtrB2, add acyloxyacyl laurate to lipid A 2- and 2'-acyl chains, respectively. The genome of P. aeruginosa also has genes which encode two dioxygenase enzymes, LpxO1 and LpxO2, that individually hydroxylate a specific secondary laurate. LpxO1 acts on the 2'-acyloxyacyl laurate (added by HtrB2), whereas LpxO2 acts on the 2-acyloxyacyl laurate (added by HtrB1) in a site-specific manner. Furthermore, while both enzyme pairs are evolutionarily linked, phylogenomic analysis suggests the LpxO1/HtrB2 enzyme pair as being of ancestral origin, present throughout the Pseudomonas lineage, whereas the LpxO2/HtrB1 enzyme pair likely arose via horizontal gene transfer and has been retained in P. aeruginosa over time. Using a murine pulmonary infection model, we showed that both LpxO1 and LpxO2 enzymes are functional in vivo, as direct analysis of in vivo lipid A structure from bronchoalveolar lavage fluid revealed 2-hydroxylated lipid A. Gene expression analysis reveals increased lpxO2 but unchanged lpxO1 expression in vivo, suggesting differential regulation of these enzymes during infection. We also demonstrate that loss-of-function mutations arise in lpxO1 and lpxO2 during chronic lung infection in people with cystic fibrosis (CF), indicating a potential role for pathogenesis and airway adaptation. Collectively, our study characterizes lipid A 2-hydroxylation during P. aeruginosa airway infection that is regulated by two distinct lipid A dioxygenase enzymes.IMPORTANCEPseudomonas aeruginosa is an opportunistic pathogen that causes severe infection in hospitalized and chronically ill individuals. During infection, P. aeruginosa undergoes adaptive changes to evade host defenses and therapeutic interventions, increasing mortality and morbidity. Lipid A structural alteration is one such change that P. aeruginosa isolates undergo during chronic lung infection in CF. Investigating genetic drivers of this lipid A structural variation is crucial in understanding P. aeruginosa adaptation during infection. Here, we describe two lipid A dioxygenases with acyl-chain site specificity, each with different evolutionary origins. Further, we show that loss of function in these enzymes occurs in CF clinical isolates, suggesting a potential pathoadaptive phenotype. Studying these bacterial adaptations provides insight into selection pressures of the CF airway on P. aeruginosa phenotypes that persist during chronic infection. Understanding these adaptive changes may ultimately provide clinicians better control over bacterial populations during chronic infection.


Subject(s)
Cystic Fibrosis , Dioxygenases , Pseudomonas Infections , Humans , Animals , Mice , Pseudomonas aeruginosa/metabolism , Lipid A/metabolism , Persistent Infection , Laurates/metabolism , Hydroxylation , Cystic Fibrosis/microbiology , Pseudomonas Infections/microbiology , Dioxygenases/metabolism
3.
Food Chem ; 404(Pt B): 134693, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36323033

ABSTRACT

This work aimed to study the effects of dietary lipid composition and content on cecal and fecal microbiota of mice fed the following diets for 8 weeks: palm olein (PO)-based low-fat diet, PO-based high-fat diet, palm stearin (PS)-based low-fat diet, and PS-based high-fat diet. Increasing the dietary PS level favored the growth of Firmicutes over Bacteroidetes in the cecum and feces. In addition, it significantly elevated the total lipid (p < 0.01) and bile acid content (p < 0.01) in feces, resulting in the enrichment of fat-degrading and bile-acid tolerant genera within the families Ruminococcaceae and Lachnospiraceae. Although increasing the PO intake also caused obesity in mice, it did not affect the microbial structure. When fat intake is constant, only at a high-fat level can PS (vs PO) induce the above-mentioned microbial shifts. These results highlighted the combined roles of lipid quality and quantity on the gut microbiota.


Subject(s)
Dietary Fats , Microbiota , Mice , Animals , Mice, Inbred C57BL , Dietary Fats/pharmacology , Cecum , Diet, High-Fat/adverse effects , Feces , Bile Acids and Salts , Palm Oil
4.
Vaccines (Basel) ; 10(6)2022 May 24.
Article in English | MEDLINE | ID: mdl-35746441

ABSTRACT

Many microbes of concern to human health remain without vaccines. We have developed a whole-microbe inactivation technology that enables us to rapidly inactivate large quantities of a pathogen while retaining epitopes that were destroyed by previous inactivation methods. The method that we call UVC-MDP inactivation can be used to make whole-cell vaccines with increased potency. We and others are exploring the possibility of using improved irradiation-inactivation technologies to develop whole-cell vaccines for numerous antibiotic-resistant microbes. Here, we apply UVC-MDP to produce candidate MRSA vaccines which we test in a stringent tibia implant model of infection challenged with a virulent MSRA strain. We report high levels of clearance in the model and observe a pattern of protection that correlates with the immunogen protein profile used for vaccination.

5.
J Bone Joint Surg Am ; 104(6): 497-503, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35041629

ABSTRACT

BACKGROUND: Next-generation DNA sequencing (NGS) detects bacteria-specific DNA corresponding to the 16S ribosomal RNA gene and can identify bacterial presence with greater accuracy than traditional culture methods. The clinical relevance of these findings is unknown. The purpose of the present study was to compare the results from bacterial culture and NGS in order to characterize the potential use of NGS in orthopaedic trauma patients. METHODS: A prospective cohort study was performed at a single academic, level-I trauma center. Three patient groups were enrolled: (1) patients undergoing surgical treatment of acute closed fractures (presumed to have no bacteria), (2) patients undergoing implant removal at the site of a healed fracture without infection, and (3) patients undergoing a first procedure for the treatment of a fracture nonunion who might or might not have subclinical infection. Surgical site tissue was sent for culture and NGS. The proportions of culture and NGS positivity were compared among the groups. The agreement between culture and NGS results was assessed with use of the Cohen kappa statistic. RESULTS: Bacterial cultures were positive in 9 of 111 surgical sites (110 patients), whereas NGS was positive in 27 of 111 surgical sites (110 patients). Significantly more cases were positive on NGS as compared with culture (24% vs. 8.1%; p = 0.001), primarily in the acute closed fracture group. No difference was found in terms of the percent positivity of NGS when comparing the acute closed fracture, implant removal, and nonunion groups. With respect to bacterial identification, culture and NGS agreed in 73% of cases (κ = 0.051; 95% confidence interval, -0.12 to 0.22) indicating only slight agreement compared with expected chance agreement of 50%. CONCLUSIONS: NGS identified bacterial presence more frequently than culture, but with only slight agreement between culture and NGS. It is possible that the increased frequency of bacterial detection with molecular methods is reflective of biofilm presence on metal or colonization with nonpathogenic bacteria, as culture methods have selection pressure posed by restrictive, artificial growth conditions and there are low metabolic activity and replication rates of bacteria in biofilms. Our data suggest that NGS should not currently substitute for or complement conventional culture in orthopaedic trauma cases with low suspicion of infection. LEVEL OF EVIDENCE: Diagnostic Level II. See Instructions for Authors for a complete description of levels of evidence.


Subject(s)
Fractures, Closed , Orthopedics , Bacteria/genetics , DNA, Bacterial/genetics , Humans , Prospective Studies , Sequence Analysis, DNA
6.
Food Microbiol ; 98: 103785, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33875213

ABSTRACT

Cronobacter sakazakii is an emerging opportunistic foodborne pathogen causing rare but severe infections in neonates. Furthermore, the formation of biofilm allows C. sakazakii to persist in different environments. We have demonstrated that the mutator phenotype ascribed to deficiency of the pmrA gene results in more biomass in the first 24 h but less during the post maturation stage (7-14 d) compared with BAA 894. The present study aimed to investigate the regulatory mechanism modulating biofilm formation due to pmrA mutation. The transcriptomic analyses of BAA 894 and s-3 were performed by RNA-sequencing on planktonic and biofilm cells collected at different time points. According to the results, when comparing biofilm to planktonic cells, expression of genes encoding outer membrane proteins, lysozyme, etc. were up-regulated, with LysR family transcriptional regulators, periplasmic proteins, etc. down-regulated. During biofilm formation, cellulose synthase operon genes, flagella-related genes, etc. played essential roles in different stages. Remarkably, pmrA varies the expression of a number of genes related to motility, biofilm formation, and antimicrobial resistance, including srfB, virK, mviM encoding virulence factor, flgF, fliN, etc. encoding flagellar assembly, and marA, ramA, etc. encoding AraC family transcriptional regulators in C. sakazakii. This study provides valuable insights into transcriptional regulation of C. sakazakii pmrA mutant during biofilm formation.


Subject(s)
Bacterial Proteins/metabolism , Biofilms , Cronobacter sakazakii/genetics , Plankton/genetics , Transcriptome , Bacterial Proteins/genetics , Cronobacter sakazakii/growth & development , Cronobacter sakazakii/physiology , Gene Expression Regulation, Bacterial , Plankton/growth & development , Plankton/physiology , Transcription, Genetic , Virulence Factors/genetics , Virulence Factors/metabolism
7.
Crit Rev Microbiol ; 47(3): 386-396, 2021 May.
Article in English | MEDLINE | ID: mdl-33663335

ABSTRACT

The coordination of single or multiple microorganisms are required for the manufacture of traditional fermented foods, improving the flavour and nutrition of the food materials. However, both the additional economic benefits and safety concerns have been raised by microbiotas in fermented products. Among the fermented products, Lactobacillus and Saccharomyces cerevisiae are one of the stable microbiotas, suggesting their interaction is mediated by coexistence-relevant mechanisms and prevent to be excluded by other microbial species. Thus, aiming to guide the manufacture of fermented foods, this review will focus on interactions of coexistence-relevant mechanisms between Lactobacillus and S. cerevisiae, including metabolites communications, aggregation, and polymicrobial biofilm. Also, the molecular regulatory network of the coexistence-relevant mechanisms is discussed according to omics researches.


Subject(s)
Lactobacillus/physiology , Saccharomyces cerevisiae/physiology , Fermented Foods/microbiology , Food Microbiology , Lactobacillus/genetics , Microbial Interactions , Saccharomyces cerevisiae/genetics
8.
J Orthop Trauma ; 35(1): 35-40, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32516196

ABSTRACT

OBJECTIVES: To evaluate the efficacy of intraoperative tobramycin powder in preventing surgical site infection (SSI) and implant colonization with Enterobacter cloacae in a rabbit fixation model. Gram-negative rods, particularly Enterobacter species, comprise an increasing percentage of SSI at our institution. METHODS: Eighteen New Zealand White rabbits underwent surgical fixation of the left tibia with implantation of a plate and screws. The surgical site and implant were inoculated with 1 × 107 CFUs E. cloacae. The selected E. cloacae isolate was resistant to tobramycin and capable of forming biofilms. Nine rabbits received 125 mg tobramycin powder directly into the surgical site, overlying the implant. The control group was untreated. Fourteen days postinfection, the tibiae and implants were explanted. Radiographs were taken with and without the implants in place. One tibia from each group was examined after hematoxylin and eosin staining. The remaining tibiae and implants were morselized or sonicated, respectively, and plated on agar to determine infection burden. Data were analyzed with Fisher exact tests and Mann-Whitney U tests. RESULTS: No bone infection or implant colonization occurred in the tobramycin-treated group. In the control group, 7 of 8 rabbits developed bone infections (P = 0.001), and 4 of 8 implants were colonized (P = 0.07). No gross disruption of the normal bone architecture was observed in either group. CONCLUSIONS: Intraoperative tobramycin powder applied at the time of contamination prevented bone infection with E. cloacae in this rabbit fixation model. The results are encouraging because the E. cloacae isolate was tobramycin-resistant, demonstrating the utility of intraoperative powdered antibiotics.


Subject(s)
Surgical Wound Infection , Tobramycin , Animals , Anti-Bacterial Agents/therapeutic use , Enterobacter cloacae , Powders , Rabbits , Surgical Wound Infection/drug therapy , Surgical Wound Infection/prevention & control
9.
mBio ; 11(4)2020 07 07.
Article in English | MEDLINE | ID: mdl-32636245

ABSTRACT

Bacterial biofilms are responsible for a variety of serious human infections and are notoriously difficult to treat due to their recalcitrance to antibiotics. Further work is necessary to elicit a full understanding of the mechanism of this antibiotic tolerance. The arginine deiminase (ADI) pathway is responsible for bacterial pH maintenance and is highly expressed during biofilm growth in multiple bacterial species. Using the group A Streptococcus (GAS) as a model human pathogen, the ADI pathway was demonstrated to contribute to biofilm growth. The inability of antibiotics to reduce GAS populations when in a biofilm was demonstrated by in vitro studies and a novel animal model of nasopharyngeal infection. However, disruption of the ADI pathway returned GAS biofilms to planktonic levels of antibiotic sensitivity, suggesting the ADI pathway is influential in biofilm-related antibiotic treatment failure and provides a new strategic target for the treatment of biofilm infections in GAS and potentially numerous other bacterial species.IMPORTANCE Biofilm-mediated bacterial infections are a major threat to human health because of their recalcitrance to antibiotic treatment. Through the study of Streptococcus pyogenes, a significant human pathogen that is known to form antibiotic-tolerant biofilms, we demonstrated the role that a bacterial pathway known for responding to acid stress plays in biofilm growth and antibiotic tolerance. This not only provides some insight into antibiotic treatment failure in S. pyogenes infections but also, given the widespread nature of this pathway, provides a potentially broad target for antibiofilm therapies. This discovery has the potential to impact the treatment of many different types of recalcitrant biofilm infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Biofilms/growth & development , Hydrolases/metabolism , Streptococcus pyogenes/drug effects , Animals , Biofilms/drug effects , Female , Gene Expression Regulation, Bacterial , Humans , Male , Metabolic Networks and Pathways , Mice, Inbred C57BL , Streptococcus pyogenes/enzymology
10.
Appl Environ Microbiol ; 86(19)2020 09 17.
Article in English | MEDLINE | ID: mdl-32709721

ABSTRACT

Periprosthetic joint infection (PJI) occurring after artificial joint replacement is a major clinical issue requiring multiple surgeries and antibiotic interventions. Staphylococcus aureus is the bacterium most commonly responsible for PJI. Recent in vitro research has shown that staphylococcal strains rapidly form aggregates in the presence of synovial fluid (SF). We hypothesize that these aggregates provide early protection to bacteria entering the wound site, allowing them time to attach to the implant surface, leading to biofilm formation. Thus, understanding the attachment kinetics of these aggregates is critical in understanding their adhesion to various biomaterial surfaces. In this study, the number, size, and surface area coverage of aggregates as well as of single cells of S. aureus were quantified under various conditions on different orthopedic materials relevant to orthopedic surgery: stainless steel (316L), titanium (Ti), hydroxyapatite (HA), and polyethylene (PE). It was observed that, regardless of the material type, SF-induced aggregation resulted in reduced aggregate surface attachment and greater aggregate size than the single-cell populations under various shear stresses. Additionally, the surface area coverage of bacterial aggregates on PE was relatively high compared to that on other materials, which could potentially be due to the rougher surface of PE. Furthermore, increasing shear stress to 78 mPa decreased aggregate attachment to Ti and HA while increasing the aggregates' average size. Therefore, this study demonstrates that SF induced inhibition of aggregate attachment to all materials, suggesting that biofilm formation is initiated by lodging of aggregates on the surface features of implants and host tissues.IMPORTANCE Periprosthetic joint infection occurring after artificial joint replacement is a major clinical issue that require repeated surgeries and antibiotic interventions. Unfortunately, 26% of patients die within 5 years of developing these infections. Staphylococcus aureus is the bacterium most commonly responsible for this problem and can form biofilms to provide protection from antibiotics as well as the immune system. Although biofilms are evident on the infected implants, it is unclear how these are attached to the surface in the first place. Recent in vitro investigations have shown that staphylococcal strains rapidly form aggregates in the presence of synovial fluid and provide protection to bacteria, thus allowing them time to attach to the implant surface, leading to biofilm formation. In this study, we investigated the attachment kinetics of Staphylococcus aureus aggregates on different orthopedic materials. The information presented in this article will be useful in surgical management and implant design.


Subject(s)
Orthopedic Equipment/microbiology , Shear Strength , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Stress, Mechanical , Synovial Fluid/microbiology
11.
Infect Immun ; 88(9)2020 08 19.
Article in English | MEDLINE | ID: mdl-32631918

ABSTRACT

The opportunistic pathogen Pseudomonas aeruginosa is responsible for much of the morbidity and mortality associated with cystic fibrosis (CF), a condition that predisposes patients to chronic lung infections. P. aeruginosa lung infections are difficult to treat because P. aeruginosa adapts to the CF lung, can develop multidrug resistance, and can form biofilms. Despite the clinical significance of P. aeruginosa, modeling P. aeruginosa infections in CF has been challenging. Here, we characterize Scnn1b-transgenic (Tg) BALB/c mice as P. aeruginosa lung infection models. Scnn1b-Tg mice overexpress the epithelial Na+ channel (ENaC) in their lungs, driving increased sodium absorption that causes lung pathology similar to CF. We intranasally infected Scnn1b-Tg mice and wild-type littermates with the laboratory P. aeruginosa strain PAO1 and CF clinical isolates and then assessed differences in bacterial clearance, cytokine responses, and histological features up to 12 days postinfection. Scnn1b-Tg mice carried higher bacterial burdens when infected with biofilm-grown rather than planktonic PAO1; Scnn1b-Tg mice also cleared infections more slowly than their wild-type littermates. Infection with PAO1 elicited significant increases in proinflammatory and Th17-linked cytokines on day 3. Scnn1b-Tg mice infected with nonmucoid early CF isolates maintained bacterial burdens and mounted immune responses similar to those of PAO1-infected Scnn1b-Tg mice. In contrast, Scnn1b-Tg mice infected with a mucoid CF isolate carried high bacterial burdens, produced significantly more interleukin 1ß (IL-1ß), IL-13, IL-17, IL-22, and KC, and showed severe immune cell infiltration into the bronchioles. Taken together, these results show the promise of Scnn1b-Tg mice as models of early P. aeruginosa colonization in the CF lung.


Subject(s)
Cystic Fibrosis/genetics , Disease Models, Animal , Epithelial Sodium Channels/genetics , Opportunistic Infections/genetics , Pseudomonas Infections/genetics , Pseudomonas aeruginosa/immunology , Animals , Bacterial Load , Biofilms/growth & development , Cystic Fibrosis/immunology , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Epithelial Sodium Channels/immunology , Female , Gene Expression Regulation , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-8/genetics , Interleukin-8/immunology , Interleukins/genetics , Interleukins/immunology , Ion Transport , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Opportunistic Infections/immunology , Opportunistic Infections/microbiology , Opportunistic Infections/pathology , Plankton/growth & development , Plankton/immunology , Plankton/pathogenicity , Pseudomonas Infections/immunology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/pathogenicity , Sodium/metabolism , Interleukin-22
12.
J Agric Food Chem ; 68(26): 7062-7071, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32496800

ABSTRACT

The impacts of lipid physical state and content on lipid digestion behavior were investigated using 4 and 20% palm olein-in-water emulsions (4% PO and 20% PO) and 4 and 20% palm stearin-in-water emulsions (4% PS and 20% PS). The changes of lipid physical state, particle size, and microstructure during gastrointestinal digestion; the free fatty acid (FFA) released in the intestinal phase; and the fatty acid composition of micellar phases were investigated. After gastric digestion, all emulsions underwent flocculation and coalescence, with 20% PS showing the most extensive aggregation. During intestinal digestion, the FFA release rate and level decreased as the lipid content increased from 4 to 20%, with 4% PO presenting the highest digestion rate and extent. Besides, the solid fat in 4% PS and 20% PS decreased and increased the maximum lipid digestibility, respectively. These results highlighted the combined roles of lipid physical state and content in modulating dietary lipid digestion.


Subject(s)
Intestinal Mucosa/metabolism , Palm Oil/metabolism , Digestion , Emulsions/chemistry , Emulsions/metabolism , Fatty Acids, Nonesterified/chemistry , Fatty Acids, Nonesterified/metabolism , Humans , Palm Oil/chemistry , Particle Size , Water/chemistry
13.
Microb Pathog ; 147: 104258, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32422334

ABSTRACT

Streptococcus agalactiae is considered as a leading case of bacterial infection among neonates. Although relative protection strategies have been performed in many high-income countries, resulting in a massive reduction in the occurrences of early-onset GBS disease, the late-onset disease has not affected. Here, the whole genome of S. agalactiae Guangzhou-SAG036 was sequenced by the Pacific Biosciences Sequel using the P4-C2 chemistry and the continuous long reads were used for de novo assembly using HGAP. Besides, genes prediction and multiply annotation were performed by comparing it with diverse databases. The whole genome has a length of 2,206,504 bp and contains 2162 predicted genes with an average G + C content of 35.85%. Based on the whole genome sequence, 2 large prophages, 20 virulence factors genes, and 8 antibiotic resistant genes were identified. MLST analysis revealed S. agalactiae Guangzhou-SAG036 was identified as ST-17. The virulence factors genes were identified with different functions including adherence, antiphagocytosis, spreading factor, immune evasion, invasion, toxin. Besides, the antibiotic-resistant genes may provide S. agalactiae with resistance to multi-drugs including erythromycin, streptomycin, azithromycin, spiramycin, ampicillin, kanamycin, cationic peptides, and tetracycline. Therefore, the infection of S. agalactiae Guangzhou-SAG036 ST-17 strain maybe caused by the complex virulence factors and multi-drugs resistance. These results contribute to further understand GBS epidemiology and surveillance targets.


Subject(s)
Streptococcal Infections , Streptococcus agalactiae , Erythromycin/pharmacology , Humans , Infant, Newborn , Multilocus Sequence Typing , Streptococcus agalactiae/genetics , Virulence Factors/genetics
14.
Microb Pathog ; 145: 104186, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32272213

ABSTRACT

BACKGROUND: Lactobacillus brevis is a major contaminant of spoiled beer. And it was able to enter VBNC state and cause false negative detection, which poses a major challenge to the brewing industry. METHODS: The genomic DNA of L. brevis BM-LB13908 was extracted and purified to form a sequencing library that meets the quality requirements and was sequenced. The sequencing results were then screened and assembled to obtain the entire genome sequence of L. brevis. Predicted genes were annotated by GO database, KEGG pathway database and COG functional classification system. RESULTS: The final assembly yielded 275 scaffolds of a total length of 2 840 080 bp with a G + C content of 53.35%. There were 2357, 701, 1519 predicted genes with corresponding GO functional, COG functional, and KEGG biological pathway annotations, respectively. The genome of L. brevis BM-LB13908 contains hop resistance gene horA and multiple genes related to the formation of VBNC state. CONCLUSIONS: This report describes the draft genome sequence of L. brevis BM-LB13908, a spoilage strain isolated from finished beer sample. This study may support further study on L. brevis and other beer spoilage bacteria, and prevent and control beer spoilage caused by microorganisms.


Subject(s)
Levilactobacillus brevis , Bacteria , Beer , Food Microbiology , Genomics , Levilactobacillus brevis/genetics
15.
J Clin Microbiol ; 58(5)2020 04 23.
Article in English | MEDLINE | ID: mdl-32051263

ABSTRACT

Prosthetic joint infections are difficult to diagnose and treat due to biofilm formation by the causative pathogens. Pathogen identification relies on microbial culture that requires days to weeks, and in the case of chronic biofilm infections, lacks sensitivity. Diagnosis of infection is often delayed past the point of effective treatment such that only the removal of the implant is curative. Early diagnosis of an infection based on antibody detection might lead to less invasive, early interventions. Our study examined antibody-based assays against the Staphylococcus aureus biofilm-upregulated antigens SAOCOL0486 (a lipoprotein), glucosaminidase (a domain of SACOL1062), and SACOL0688 (the manganese transporter MntC) for detection of chronic S. aureus infection. We evaluated these antigens by enzyme-linked immunosorbent assay (ELISA) using sera from naive rabbits and rabbits with S. aureus-mediated osteomyelitis, and then we validated a proof of concept for the lateral flow assay (LFA). The SACOL0688 LFA demonstrated 100% specificity and 100% sensitivity. We demonstrated the clinical diagnostic utility of the SACOL0688 antigen using synovial fluid (SF) from humans with orthopedic implant infections. Elevated antibody levels to SACOL0688 in clinical SF specimens correlated with 91% sensitivity and 100% specificity for the diagnosis of S. aureus infection by ELISA. We found measuring antibodies levels to SACOL0688 in SF using ELISA or LFA provides a tool for the sensitive and specific diagnosis of S. aureus prosthetic joint infection. Development of the LFA diagnostic modality is a desirable, cost-effective option, potentially providing rapid readout in minutes for chronic biofilm infections.


Subject(s)
Osteomyelitis , Staphylococcal Infections , Animals , Antigens, Bacterial , Biofilms , Rabbits , Staphylococcal Infections/diagnosis , Staphylococcus aureus
16.
Infect Immun ; 88(1)2019 12 17.
Article in English | MEDLINE | ID: mdl-31712267

ABSTRACT

Staphylococcus aureus is a causative agent of chronic biofilm-associated infections that are recalcitrant to resolution by the immune system or antibiotics. To combat these infections, an antistaphylococcal, biofilm-specific quadrivalent vaccine against an osteomyelitis model in rabbits has previously been developed and shown to be effective at eliminating biofilm-embedded bacterial populations. However, the addition of antibiotics was required to eradicate remaining planktonic populations. In this study, a planktonic upregulated antigen was combined with the quadrivalent vaccine to remove the need for antibiotic therapy. Immunization with this pentavalent vaccine followed by intraperitoneal challenge of BALB/c mice with S. aureus resulted in 16.7% and 91.7% mortality in pentavalent vaccine and control groups, respectively (P < 0.001). Complete bacterial elimination was found in 66.7% of the pentavalent cohort, while only 8.3% of the control animals cleared the infection (P < 0.05). Further protective efficacy was observed in immunized rabbits following intramedullary challenge with S. aureus, where 62.5% of the pentavalent cohort completely cleared the infection, versus none of the control animals (P < 0.05). Passive immunization of BALB/c mice with serum IgG against the vaccine antigens prior to intraperitoneal challenge with S. aureus prevented mortality in 100% of mice and eliminated bacteria in 33.3% of the challenged mice. These results demonstrate that targeting both the planktonic and biofilm stages with the pentavalent vaccine or the IgG elicited by immunization can effectively protect against S. aureus infection.


Subject(s)
Antigens, Bacterial/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/administration & dosage , Antibodies, Bacterial/immunology , Disease Models, Animal , Immunization, Passive , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Mice, Inbred BALB C , Rabbits , Staphylococcal Vaccines/administration & dosage , Survival Analysis , Treatment Outcome
17.
Microbiologyopen ; 8(9): e00853, 2019 09.
Article in English | MEDLINE | ID: mdl-31179649

ABSTRACT

The inhibition of microbial biofilms is a significant concern in food safety. In the present study, the inhibitory effect of sodium citrate and cinnamic aldehyde on biofilm formation at minimum inhibitory concentrations (MICs) and sub-MICs was investigated for Escherichia coli O157:H7 and Staphylococcus aureus. The biofilm inhibition rate was measured to evaluate the effect of sodium citrate on S. aureus biofilms at 24, 48, 72, and 96 hr. According to the results, an antibiofilm effect was shown by both food additives, with 10 mg/ml of sodium citrate exhibiting the greatest inhibition of S. aureus biofilms at 24 hr (inhibition rate as high as 77.51%). These findings strongly suggest that sodium citrate exhibits a pronounced inhibitory effect on biofilm formation with great potential in the extension of food preservation and storage.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Escherichia coli O157/drug effects , Food Additives/pharmacology , Staphylococcus aureus/drug effects , Acrolein/analogs & derivatives , Acrolein/pharmacology , Microbial Sensitivity Tests , Sodium Citrate/pharmacology , Time Factors
18.
mBio ; 10(3)2019 06 04.
Article in English | MEDLINE | ID: mdl-31164467

ABSTRACT

Candida albicans and Staphylococcus aureus are among the most prevalent nosocomial pathogens that are responsible for severe morbidity and mortality, even with appropriate treatment. Using a murine model of polymicrobial intra-abdominal infection (IAI), we have previously shown that coinfection with these pathogens results in synergistic lethality that is partially dependent on exacerbated prostaglandin signaling, while monomicrobial infection is nonlethal. Therefore, the objective of this study was to identify staphylococcal virulence determinants that drive lethal synergism during polymicrobial IAI. Using the toxigenic S. aureus strain JE2, we observed that coinfection with C. albicans led to a striking 80 to 100% mortality rate within 20 h postinoculation (p.i.) while monomicrobial infections were nonlethal. Use of a green fluorescent protein (GFP)-P3 promoter S. aureus reporter strain revealed enhanced activation of the staphylococcal agr quorum sensing system during in vitro polymicrobial versus monomicrobial growth. Analyses by quantitative real-time PCR (qPCR), Western blot, and toxin functional assays confirmed enhanced agr-associated gene transcription and increases in secreted alpha- and delta-toxins. C. albicans-mediated elevated toxin production and hemolytic activity were determined to be agrA dependent, and genetic knockout and complementation of hla identified alpha-toxin as the key staphylococcal virulence factor driving lethal synergism. Analysis of mono- and polymicrobial infections 8 h p.i. demonstrated equivalent bacterial burdens in the peritoneal cavity but significantly elevated levels of alpha-toxin (3-fold) and the eicosanoid prostaglandin E2 (PGE2) (4-fold) during coinfection. Importantly, prophylactic passive immunization using the monoclonal anti-alpha-toxin antibody MEDI4893* led to significantly improved survival rates compared to those following treatment with isotype control antibody. Collectively, these results define alpha-toxin as an essential virulence determinant during C. albicans-S. aureus IAI and describe a novel mechanism by which a human-pathogenic fungus can augment the virulence of a highly pathogenic bacterium in vivoIMPORTANCE Relatively little is known about the complex interactions and signaling events that occur between microbes and even less so about how microbial "cross talk" shapes human health and disease. Candida albicans (a fungus) and Staphylococcus aureus (a bacterium) are formidable human nosocomial pathogens, causing severe morbidity and mortality. Moreover, they are frequently coisolated from central venous catheters and deep-seated infections, including intra-abdominal sepsis. In this work, we have shown that coinfection with C. albicans and S. aureus is highly lethal, leading to >80% mortality by day 1 postinfection, whereas monoinfection with C. albicans or S. aureus does not cause mortality. This infectious synergism is dependent on the expression of staphylococcal alpha-toxin, and secretion of this potent virulence factor is actually augmented by C. albicans via an agr-dependent mechanism. Moreover, prophylactic neutralization of alpha-toxin with a monoclonal antibody is sufficient to elicit protection during coinfection. Therefore, we have demonstrated that a pathogenic fungus can enhance virulence determinants of a bacterium in vivo with devastating consequences to the host. These results have important implications in the surveillance and treatment of polymicrobial disease and highlight the dynamic intersection of environment, pathogens, and host.


Subject(s)
Bacterial Proteins/genetics , Candida albicans/physiology , Coinfection/microbiology , Quorum Sensing , Staphylococcus aureus/pathogenicity , Trans-Activators/genetics , Animals , Bacterial Toxins/genetics , Candidiasis/microbiology , Coinfection/mortality , Gastrointestinal Diseases/microbiology , Hemolysin Proteins/genetics , Mice , Microbial Interactions , Staphylococcal Infections/microbiology , Virulence , Virulence Factors/genetics
19.
Sci Rep ; 9(1): 1312, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718527

ABSTRACT

This study aimed to evaluate the Staphylococcus aureus biofilm formation and Nε-carboxymethyl-lysine generation ability under food heat processing conditions including pH (5.0-9.0), temperature (25 °C, 31 °C, 37 °C, 42 °C and 65 °C), NaCl concentration (10%, 15% and 20%, w/v) and glucose concentration (0.5%, 1%, 2%, 3%, 5%, 10%, w/v). S. aureus biofilm genetic character was obtained by PCR detecting atl, ica operon, sasG and agr. Biofilm biomass and metabolic activity were quantified with crystal violet and methyl thiazolyl tetrazolium staining methods. S. aureus biofilm was sensitive to food heat processing conditions with 37 °C, pH 7.0, 2% glucose concentration (w/v) and 10% NaCl concentration (w/v) were favorable conditions. Besides, free and bound Nε-carboxymethyl-lysine level in weak, moderate and strong biofilm were detected by optimized high performance liquid chromatography tandem mass spectrometry. Nε-carboxymethyl-lysine level in S. aureus biofilm possessed a significant gap between strong, moderate and weak biofilm strains. This investigation revealed the biological and chemical hazard of Staphylococcus aureus biofilm to food processing environment.


Subject(s)
Biofilms/growth & development , Food Handling , Food Microbiology , Staphylococcus aureus/physiology , Hydrogen-Ion Concentration , Staphylococcus aureus/ultrastructure , Temperature
20.
Front Microbiol ; 9: 2413, 2018.
Article in English | MEDLINE | ID: mdl-30425687

ABSTRACT

Staphylococcus aureus is one of the representative foodborne pathogens which forms biofilm. Antibiotics are widely applied in livestock husbandry to maintain animal health and productivity, thus contribute to the dissemination of antimicrobial resistant livestock and human pathogens, and pose a significant public health threat. Effect of antibiotic pressure on S. aureus biofilm formation, as well as the mechanism, remains unclear. In this study, the regulatory mechanism of low concentration of ampicillin on S. aureus biofilm formation was elucidated. The viability and biomass of biofilm with and without 1/4 MIC ampicillin treatment for 8 h were determined by XTT and crystal violet straining assays, respectively. Transcriptomics analysis on ampicillin-induced and non-ampicillin-induced biofilms were performed by RNA-sequencing, differentially expressed genes identification and annotation, GO functional and KEGG pathway enrichment. The viability and biomass of ampicillin-induced biofilm showed dramatical increase compared to the non-ampicillin-induced biofilm. A total of 530 differentially expressed genes (DEGs) with 167 and 363 genes showing up- and down-regulation, respectively, were obtained. Upon GO functional enrichment, 183, 252, and 21 specific GO terms in biological process, molecular function and cellular component were identified, respectively. Eight KEGG pathways including "Microbial metabolism in diverse environments", "S. aureus infection", and "Monobactam biosynthesis" were significantly enriched. In addition, "beta-lactam resistance" pathway was also highly enriched. In ampicillin-induced biofilm, the significant up-regulation of genes encoding multidrug resistance efflux pump AbcA, penicillin binding proteins PBP1, PBP1a/2, and PBP3, and antimicrobial resistance proteins VraF, VraG, Dlt, and Aur indicated the positive response of S. aureus to ampicillin. The up-regulation of genes encoding surface proteins ClfB, IsdA, and SasG and genes (cap5B and cap5C) which promote the adhesion of S. aureus in ampicillin induced biofilm might explain the enhanced biofilm viability and biomass.

SELECTION OF CITATIONS
SEARCH DETAIL
...