Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 299(3): 102912, 2023 03.
Article in English | MEDLINE | ID: mdl-36649910

ABSTRACT

Daptomycin (DAP) is an antibiotic frequently used as a drug of last resort against vancomycin-resistant enterococci. One of the major challenges when using DAP against vancomycin-resistant enterococci is the emergence of resistance, which is mediated by the cell-envelope stress system LiaFSR. Indeed, inhibition of LiaFSR signaling has been suggested as a strategy to "resensitize" enterococci to DAP. In the absence of LiaFSR, alternative pathways mediating DAP resistance have been identified, including adaptive mutations in the enolpyruvate transferase MurAA (MurAAA149E), which catalyzes the first committed step in peptidoglycan biosynthesis; however, how these mutations confer resistance is unclear. Here, we investigated the biochemical basis for MurAAA149E-mediated adaptation to DAP to determine whether such an alternative pathway would undermine the potential efficacy of therapies that target the LiaFSR pathway. We found cells expressing MurAAA149E had increased susceptibility to glycoside hydrolases, consistent with decreased cell wall integrity. Furthermore, structure-function studies of MurAA and MurAAA149E using X-ray crystallography and biochemical analyses indicated only a modest decrease in MurAAA149E activity, but a 16-fold increase in affinity for MurG, which performs the last intracellular step of peptidoglycan synthesis. Exposure to DAP leads to mislocalization of cell division proteins including MurG. In Bacillus subtilis, MurAA and MurG colocalize at division septa and, thus, we propose MurAAA149E may contribute to DAP nonsusceptibility by increasing the stability of MurAA-MurG interactions to reduce DAP-induced mislocalization of these essential protein complexes.


Subject(s)
Daptomycin , Enterococcus faecium , Transferases , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Daptomycin/metabolism , Daptomycin/pharmacology , Drug Resistance, Bacterial , Enterococcus faecium/drug effects , Enterococcus faecium/metabolism , Microbial Sensitivity Tests , Peptidoglycan/metabolism , Transferases/metabolism
2.
Antimicrob Agents Chemother ; 68(3): e0106923, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38289081

ABSTRACT

Daptomycin (DAP) is often used as a first-line therapy to treat vancomycin-resistant Enterococcus faecium infections, but emergence of DAP non-susceptibility threatens the effectiveness of this antibiotic. Moreover, current methods to determine DAP minimum inhibitory concentrations (MICs) have poor reproducibility and accuracy. In enterococci, DAP resistance is mediated by the LiaFSR cell membrane stress response system, and deletion of liaR encoding the response regulator results in hypersusceptibility to DAP and antimicrobial peptides. The main genes regulated by LiaR are a cluster of three genes, designated liaXYZ. In Enterococcus faecalis, LiaX is surface-exposed with a C-terminus that functions as a negative regulator of cell membrane remodeling and an N-terminal domain that is released to the extracellular medium where it binds DAP. Thus, in E. faecalis, LiaX functions as a sentinel molecule recognizing DAP and controlling the cell membrane response, but less is known about LiaX in E. faecium. Here, we found that liaX is essential in E. faecium with an activated LiaFSR system. Unlike E. faecalis, E. faecium LiaX is not detected in the extracellular milieu and does not appear to alter phospholipid architecture. We further postulated that LiaX could be used as a surrogate marker for cell envelope activation and non-susceptibility to DAP. For this purpose, we developed and optimized a LiaX enzyme-linked immunosorbent assay (ELISA). We then assessed 86 clinical E. faecium bloodstream isolates for DAP MICs and used whole genome sequencing to assess for substitutions in LiaX. All DAP-resistant clinical strains of E. faecium exhibited elevated LiaX levels. Strikingly, 73% of DAP-susceptible isolates by standard MIC determination also had elevated LiaX ELISAs compared to a well-characterized DAP-susceptible strain. Phylogenetic analyses of predicted amino acid substitutions showed 12 different variants of LiaX without a specific association with DAP MIC or LiaX ELISA values. Our findings also suggest that many E. faecium isolates that test DAP susceptible by standard MIC determination are likely to have an activated cell stress response that may predispose to DAP failure. As LiaX appears to be essential for the cell envelope response to DAP, its detection could prove useful to improve the accuracy of susceptibility testing by anticipating therapeutic failure.


Subject(s)
Daptomycin , Enterococcus faecium , Gram-Positive Bacterial Infections , Humans , Daptomycin/pharmacology , Daptomycin/therapeutic use , Phylogeny , Reproducibility of Results , Drug Resistance, Bacterial/genetics , Anti-Bacterial Agents/therapeutic use , Cell Membrane , Biomarkers/metabolism , Microbial Sensitivity Tests , Enterococcus faecalis , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/metabolism
3.
J Antimicrob Chemother ; 79(4): 801-809, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38334390

ABSTRACT

OBJECTIVES: To investigate the genomic diversity and ß-lactam susceptibilities of Enterococcus faecalis collected from patients with infective endocarditis (IE). METHODS: We collected 60 contemporary E. faecalis isolates from definite or probable IE cases identified between 2018 and 2021 at the University of Pittsburgh Medical Center. We used whole-genome sequencing to study bacterial genomic diversity and employed antibiotic checkerboard assays and a one-compartment pharmacokinetic-pharmacodynamic (PK/PD) model to investigate bacterial susceptibility to ampicillin and ceftriaxone both alone and in combination. RESULTS: Genetically diverse E. faecalis were collected, however, isolates belonging to two STs, ST6 and ST179, were collected from 21/60 (35%) IE patients. All ST6 isolates encoded a previously described mutation upstream of penicillin-binding protein 4 (pbp4) that is associated with pbp4 overexpression. ST6 isolates had higher ceftriaxone MICs and higher fractional inhibitory concentration index values for ampicillin and ceftriaxone (AC) compared to other isolates, suggesting diminished in vitro AC synergy against this lineage. Introduction of the pbp4 upstream mutation found among ST6 isolates caused increased ceftriaxone resistance in a laboratory E. faecalis isolate. PK/PD testing showed that a representative ST6 isolate exhibited attenuated efficacy of AC combination therapy at humanized antibiotic exposures. CONCLUSIONS: We find evidence for diminished in vitro AC activity among a subset of E. faecalis IE isolates with increased pbp4 expression. These findings suggest that alternate antibiotic combinations against diverse contemporary E. faecalis IE isolates should be evaluated.


Subject(s)
Endocarditis, Bacterial , Endocarditis , Gram-Positive Bacterial Infections , Humans , Ceftriaxone/pharmacology , Ceftriaxone/therapeutic use , Enterococcus faecalis , Ampicillin/pharmacology , Ampicillin/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Endocarditis, Bacterial/drug therapy , Endocarditis, Bacterial/microbiology , Endocarditis/drug therapy , Microbial Sensitivity Tests , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/microbiology , Drug Therapy, Combination
4.
Clin Infect Dis ; 77(Suppl 4): S295-S304, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37843115

ABSTRACT

The Antibacterial Resistance Leadership Group (ARLG) has prioritized infections caused by gram-positive bacteria as one of its core areas of emphasis. The ARLG Gram-positive Committee has focused on studies responding to 3 main identified research priorities: (1) investigation of strategies or therapies for infections predominantly caused by gram-positive bacteria, (2) evaluation of the efficacy of novel agents for infections caused by methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci, and (3) optimization of dosing and duration of antimicrobial agents for gram-positive infections. Herein, we summarize ARLG accomplishments in gram-positive bacterial infection research, including studies aiming to (1) inform optimal vancomycin dosing, (2) determine the role of dalbavancin in MRSA bloodstream infection, (3) characterize enterococcal bloodstream infections, (4) demonstrate the benefits of short-course therapy for pediatric community-acquired pneumonia, (5) develop quality of life measures for use in clinical trials, and (6) advance understanding of the microbiome. Future studies will incorporate innovative methodologies with a focus on interventional clinical trials that have the potential to change clinical practice for difficult-to-treat infections, such as MRSA bloodstream infections.


Subject(s)
Gram-Positive Bacterial Infections , Methicillin-Resistant Staphylococcus aureus , Sepsis , Humans , Child , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Leadership , Quality of Life , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacteria , Sepsis/drug therapy
5.
Antimicrob Agents Chemother ; 67(1): e0128422, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36541772

ABSTRACT

In the United States, vanB-mediated resistance in enterococci is rare. We characterized three sequence type (ST) 6, vancomycin-resistant Enterococcus faecalis isolates causing bacteremia in unique patients in spatiotemporally distinct settings. Isolates were recovered between 2018 and 2020 in two cities in the United States (Houston, TX; Miami, FL). The isolates harbored the vanB operon on a chromosomally located Tn1549 transposon, and epidemiological data suggested multiple introductions of the vanB gene cluster into ST6 E. faecalis.


Subject(s)
Enterococcus faecium , Gram-Positive Bacterial Infections , Vancomycin-Resistant Enterococci , Humans , Enterococcus faecalis/genetics , Vancomycin Resistance/genetics , Florida/epidemiology , Texas/epidemiology , Vancomycin-Resistant Enterococci/genetics , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/epidemiology , Bacterial Proteins/genetics , Anti-Bacterial Agents/pharmacology
6.
Antimicrob Agents Chemother ; 66(6): e0233321, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35543524

ABSTRACT

Infections caused by vancomycin-resistant Enterococcus faecium (VREfm) are an important public health threat. VREfm isolates have become increasingly resistant to the front-line antibiotic daptomycin (DAP). As such, the use of DAP combination therapies with other antibiotics like fosfomycin (FOS) has received increased attention. Antibiotic combinations could extend the efficacy of currently available antibiotics and potentially delay the onset of further resistance. We investigated the potential for E. faecium HOU503, a clinical VREfm isolate that is DAP and FOS susceptible, to develop resistance to a DAP-FOS combination. Of particular interest was whether the genetic drivers for DAP-FOS resistance might be epistatic and, thus, potentially decrease the efficacy of a combinatorial approach in either inhibiting VREfm or in delaying the onset of resistance. We show that resistance to DAP-FOS could be achieved by independent mutations to proteins responsible for cell wall synthesis for FOS and in altering membrane dynamics for DAP. However, we did not observe genetic drivers that exhibited substantial cross-drug epistasis that could undermine the DAP-FOS combination. Of interest was that FOS resistance in HOU503 was largely mediated by changes in phosphoenolpyruvate (PEP) flux as a result of mutations in pyruvate kinase (pyk). Increasing PEP flux could be a readily accessible mechanism for FOS resistance in many pathogens. Importantly, we show that HOU503 was able to develop DAP resistance through a variety of biochemical mechanisms and was able to employ different adaptive strategies. Finally, we showed that the addition of FOS can prolong the efficacy of DAP and slow down DAP resistance in vitro.


Subject(s)
Anti-Bacterial Agents , Daptomycin , Drug Resistance, Bacterial , Enterococcus faecium , Fosfomycin , Anti-Bacterial Agents/pharmacology , Daptomycin/pharmacology , Drug Resistance, Bacterial/genetics , Enterococcus faecium/genetics , Evolution, Molecular , Fosfomycin/pharmacology , Microbial Sensitivity Tests , Vancomycin-Resistant Enterococci/genetics
7.
Antimicrob Agents Chemother ; 66(6): e0261420, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35475634

ABSTRACT

The long-acting lipoglycopeptides (LGPs) dalbavancin and oritavancin are semisynthetic antimicrobials with broad and potent activity against Gram-positive bacterial pathogens. While they are approved by the Food and Drug Administration for acute bacterial skin and soft tissue infections, their pharmacological properties suggest a potential role of these agents for the treatment of deep-seated and severe infections, such as bloodstream and bone and joint infections. The use of these antimicrobials is particularly appealing when prolonged therapy, early discharge, and avoidance of long-term intravascular catheter access are desirable or when multidrug-resistant bacteria are suspected. This review describes the current evidence for the use of oritavancin and dalbavancin in the treatment of invasive infections, as well as the hurdles that are preventing their optimal use. Moreover, this review discusses the current knowledge gaps that need to be filled to understand the potential role of LGPs in highly needed clinical scenarios and the ongoing clinical studies that aim to address these voids in the upcoming years.


Subject(s)
Anti-Infective Agents , Gram-Positive Bacterial Infections , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Infective Agents/pharmacology , Anti-Infective Agents/therapeutic use , Glycopeptides/chemistry , Glycopeptides/pharmacology , Glycopeptides/therapeutic use , Gram-Positive Bacterial Infections/drug therapy , Humans , Lipoglycopeptides/therapeutic use , Teicoplanin/pharmacology , Teicoplanin/therapeutic use
8.
Eur J Clin Microbiol Infect Dis ; 41(10): 1207-1213, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36002777

ABSTRACT

Patients with invasive candidiasis (IC) have complex medical and infectious disease problems that often require continued care after discharge. This study aimed to assess echinocandin use at hospital discharge and develop a transition of care (TOC) model to facilitate discharge for patients with IC. This was a mixed method study design that used epidemiologic assessment to better understand echinocandin use at hospital discharge TOC. Using grounded theory methodology focused on patients given echinocandins during their last day of hospitalization, a TOC model for patients with IC, the invasive candidiasis [I Can] discharge model was developed to better understand discharge barriers. A total of 33% (1405/4211) echinocandin courses were continued until the last day of hospitalization. Of 536 patients chosen for in-depth review, 220 (41%) were discharged home, 109 (20%) were transferred, and 207 (39%) died prior to discharge. Almost half (46%, 151/329) of patients discharged alive received outpatient echinocandin therapy. Independent predictors for outpatient echinocandin use were osteomyelitis (OR, 4.1; 95% CI, 1.1-15.7; p = 0.04), other deep-seated infection (OR, 4.4; 95% CI, 1.7-12.0; p = 0.003), and non-home discharge location (OR, 3.9, 95% CI, 2.0-7.7; p < 0.001). The I Can discharge model was developed encompassing four distinct themes which was used to identify potential barriers to discharge. Significant echinocadin use occurs at hospital discharge TOC. The I Can discharge model may help clinical, policy, and research decision-making processes to facilitate smoother and earlier hospital discharges.


Subject(s)
Candidiasis, Invasive , Patient Discharge , Antifungal Agents/therapeutic use , Candidiasis , Candidiasis, Invasive/diagnosis , Candidiasis, Invasive/drug therapy , Candidiasis, Invasive/microbiology , Echinocandins/therapeutic use , Humans
9.
Proc Natl Acad Sci U S A ; 116(52): 26925-26932, 2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31818937

ABSTRACT

Bacteria have developed several evolutionary strategies to protect their cell membranes (CMs) from the attack of antibiotics and antimicrobial peptides (AMPs) produced by the innate immune system, including remodeling of phospholipid content and localization. Multidrug-resistant Enterococcus faecalis, an opportunistic human pathogen, evolves resistance to the lipopeptide daptomycin and AMPs by diverting the antibiotic away from critical septal targets using CM anionic phospholipid redistribution. The LiaFSR stress response system regulates this CM remodeling via the LiaR response regulator by a previously unknown mechanism. Here, we characterize a LiaR-regulated protein, LiaX, that senses daptomycin or AMPs and triggers protective CM remodeling. LiaX is surface exposed, and in daptomycin-resistant clinical strains, both LiaX and the N-terminal domain alone are released into the extracellular milieu. The N-terminal domain of LiaX binds daptomycin and AMPs (such as human LL-37) and functions as an extracellular sentinel that activates the cell envelope stress response. The C-terminal domain of LiaX plays a role in inhibiting the LiaFSR system, and when this domain is absent, it leads to activation of anionic phospholipid redistribution. Strains that exhibit LiaX-mediated CM remodeling and AMP resistance show enhanced virulence in the Caenorhabditis elegans model, an effect that is abolished in animals lacking an innate immune pathway crucial for producing AMPs. In conclusion, we report a mechanism of antibiotic and AMP resistance that couples bacterial stress sensing to major changes in CM architecture, ultimately also affecting host-pathogen interactions.

10.
Article in English | MEDLINE | ID: mdl-33468468

ABSTRACT

LiaFSR signaling plays a major role in mediating daptomycin (DAP) resistance in enterococci, and the lack of a functional LiaFSR pathway leads to DAP hypersusceptibility. Using in vitro experimental evolution, we evaluated how Enterococcus faecium with a liaR response regulator gene deletion evolved DAP resistance. We found that knocking out LiaFSR signaling significantly delayed the onset of resistance, but resistance could emerge eventually through various alternate mechanisms that were influenced by the environment.


Subject(s)
Daptomycin , Enterococcus faecium , Gram-Positive Bacterial Infections , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Daptomycin/pharmacology , Drug Resistance, Bacterial/genetics , Enterococcus faecium/genetics , Humans , Microbial Sensitivity Tests
11.
Antimicrob Agents Chemother ; 65(10): e0114621, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34339274

ABSTRACT

Clinical cases of C. auris noted during a COVID-19 surge led to an epidemiological, clinical, and genomic investigation. Evaluation identified a close genetic relationship but inconclusive epidemiologic link between all cases. Prolonged hospitalization due to critical illness from COVID-19 and use of antimicrobials may have contributed to clinical infections.


Subject(s)
COVID-19 , Candidiasis, Invasive , Antifungal Agents/therapeutic use , Candida/genetics , Candidiasis, Invasive/drug therapy , Humans , SARS-CoV-2
12.
J Antimicrob Chemother ; 76(1): 179-183, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33035321

ABSTRACT

OBJECTIVES: Reduced in vitro ß-lactam activity against a dense bacterial population is well recognized. It is commonly attributed to the presence of ß-lactamase(s) and it is unknown whether the inoculum effect could be diminished by a ß-lactamase inhibitor. We evaluated different ß-lactam/ß-lactamase inhibitor combinations in suppressing a high inoculum of ESBL-producing bacteria. METHODS: Three clinical isolates expressing representative ESBLs (CTX-M-15 and SHV-12) were examined. The impact of escalating ß-lactamase inhibitor (tazobactam or avibactam) concentrations on ß-lactam (piperacillin or ceftazidime) MIC reduction was characterized by an inhibitory sigmoid Emax model. The effect of various dosing regimens of ß-lactam/ß-lactamase inhibitor combinations was predicted using %T>MICi and selected exposures were experimentally validated in a hollow-fibre infection model over 120 h. The threshold exposure to suppress bacterial regrowth was identified using recursive partitioning. RESULTS: A concentration-dependent reduction in ß-lactam MIC was observed (r2 ≥0.93). Regrowth could be suppressed in all six experiments using %T>MICi ≥73.6%, but only one out of six experiments below the threshold (P = 0.015). The exposures to suppress regrowth might be attained using the clinical dose of avibactam, but a much higher dose than the standard dose would be needed for tazobactam. CONCLUSIONS: A dense population of ESBL-producing bacteria could be suppressed by an optimized dosing regimen of selected ß-lactam/ß-lactamase inhibitor combinations. The reversibility of enzyme inhibition could play an important role in diminishing the inoculum effect. In vivo investigations to validate these findings are warranted.


Subject(s)
Lactams , beta-Lactamase Inhibitors , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria , Microbial Sensitivity Tests , beta-Lactamase Inhibitors/pharmacology , beta-Lactamases
13.
J Infect Dis ; 222(9): 1531-1539, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32514561

ABSTRACT

BACKGROUND: The combination of daptomycin (DAP) plus ampicillin (AMP), ertapenem (ERT), or ceftaroline has been demonstrated to be efficacious against a DAP-tolerant Enterococcus faecium strain (HOU503). However, the mechanism for the efficacy of these combinations against DAP-resistant (DAP-R) E. faecium strains is unknown. METHODS: We investigated the efficacy of DAP in combination with AMP, ERT, ceftaroline, ceftriaxone, or amoxicillin against DAP-R E. faecium R497 using established in vitro and in vivo models. We evaluated pbp expression, levels of penicillin-binding protein (PBP) 5 (PBP5) and ß-lactam binding affinity in HOU503 versus R497. RESULTS: DAP plus AMP was the only efficacious regimen against DAP-R R497 and prevented emergence of resistance. DAP at 8, 6, and 4 mg/kg in combination with AMP was efficacious but showed delayed killing compared with 10 mg/kg. PBP5 of HOU503 exhibited amino acid substitutions in the penicillin-binding domain relative to R497. No difference in pbp mRNA or PBP5 levels was detected between HOU503 and R497. labeling of PBPs with Bocillin FL, a fluorescent penicillin derivative, showed increased ß-lactam binding affinity of PBP5 of HOU503 compared with that of R497. CONCLUSIONS: Only DAP (10 mg/kg) plus AMP or amoxicillin was efficacious against a DAP-R E. faecium strain, and pbp5 alleles may be important contributors to efficacy of DAP plus ß-lactam therapy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Daptomycin/pharmacology , Enterococcus faecium/drug effects , beta-Lactams/pharmacology , Ampicillin/administration & dosage , Ampicillin/pharmacology , Animals , Anti-Bacterial Agents/administration & dosage , Cephalosporins/administration & dosage , Cephalosporins/therapeutic use , Daptomycin/administration & dosage , Disease Models, Animal , Drug Resistance, Bacterial , Drug Therapy, Combination , Endocarditis, Bacterial/drug therapy , Enterococcus faecium/genetics , Ertapenem/administration & dosage , Ertapenem/pharmacology , Gram-Positive Bacterial Infections/drug therapy , Microbial Sensitivity Tests , Rats , Sequence Alignment , Transcriptome , beta-Lactams/administration & dosage , Ceftaroline
14.
Mol Microbiol ; 111(3): 811-824, 2019 03.
Article in English | MEDLINE | ID: mdl-30582877

ABSTRACT

The lipopeptide antibiotic daptomycin (DAP) is a key drug against serious enterococcal infections, but the emergence of resistance in the clinical setting is a major concern. The LiaFSR system plays a prominent role in the development of DAP resistance (DAP-R) in enterococci, and blocking this stress response system has been proposed as a novel therapeutic strategy. In this work, we identify LiaR-independent pathways in Enterococcus faecalis that regulate cell membrane adaptation in response to antibiotics. We adapted E. faecalis OG1RF (a laboratory strain) and S613TM (a clinical strain) lacking liaR to increasing concentrations of DAP, leading to the development of DAP-R and elevated MICs to bacitracin and ceftriaxone. Whole genome sequencing identified changes in the YxdJK two-component regulatory system and a putative fatty acid kinase (dak) in both DAP-R strains. Deletion of the gene encoding the YxdJ response regulator in both the DAP-R mutant and wild-type OG1RF decreased MICs to DAP, even when a functional LiaFSR system was present. Mutations in dak were associated with slower growth, decreased membrane fluidity and alterations of cell morphology. These findings suggest that overlapping stress response pathways can provide protection against antimicrobial peptides in E. faecalis at a significant cost in bacterial fitness.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Daptomycin/pharmacology , Drug Resistance, Bacterial , Enterococcus faecalis/drug effects , Mutation , Adaptation, Biological , Bacitracin/pharmacology , Ceftriaxone/pharmacology , Enterococcus faecalis/genetics , Enterococcus faecalis/growth & development , Gene Expression Regulation, Bacterial , Microbial Sensitivity Tests , Serial Passage , Whole Genome Sequencing
15.
Article in English | MEDLINE | ID: mdl-31871075

ABSTRACT

We report a 15 year-old Nigerian adolescent male with chronic osteomyelitis caused by an extensively drug-resistant (XDR) Pseudomonas aeruginosa strain of sequence type 773 (ST773) carrying blaNDM-1 and an extended spectrum ß-lactamase (ESBL)-producing Klebsiella pneumoniae strain. The patient developed neurological side effects in the form of circumoral paresthesia with polymyxin B and asymptomatic elevation of transaminases with aztreonam (used in combination with ceftazidime-avibactam). Cefiderocol treatment for 14 weeks plus bone implantation resulted in apparent cure and avoided amputation.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Cephalosporins/therapeutic use , Compassionate Use Trials/methods , Klebsiella pneumoniae/drug effects , Pseudomonas aeruginosa/drug effects , Surgical Wound Infection/drug therapy , Adolescent , Drug Resistance, Multiple, Bacterial/genetics , Humans , Male , Microbial Sensitivity Tests , Nigeria , Osteomyelitis/drug therapy , Osteomyelitis/microbiology , Surgical Wound Infection/microbiology , beta-Lactam Resistance/genetics , beta-Lactamases/genetics , Cefiderocol
16.
J Infect Dis ; 220(3): 494-504, 2019 07 02.
Article in English | MEDLINE | ID: mdl-30938438

ABSTRACT

Daptomycin resistance in enterococci is often mediated by the LiaFSR system, which orchestrates the cell membrane stress response. Activation of LiaFSR through the response regulator LiaR generates major changes in cell membrane function and architecture (membrane adaptive response), permitting the organism to survive the antibiotic attack. Here, using a laboratory strain of Enterococcus faecalis, we developed a novel Caenorhabditis elegans model of daptomycin therapy and showed that disrupting LiaR-mediated cell membrane adaptation restores the in vivo activity of daptomycin. The LiaR effect was also seen in a clinical strain of daptomycin-resistant Enterococcus faecium, using a murine model of peritonitis. Furthermore, alteration of the cell membrane response increased the ability of human polymorphonuclear neutrophils to readily clear both E. faecalis and multidrug-resistant E. faecium. Our results provide proof of concept that targeting the cell membrane adaptive response restores the in vivo activity of antibiotics, prevents resistance, and enhances the ability of the innate immune system to kill infecting bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Membrane/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , Enterococcus faecalis/drug effects , Enterococcus faecium/drug effects , Gram-Positive Bacterial Infections/drug therapy , Neutrophils/drug effects , Animals , Bacterial Proteins , Cell Membrane/microbiology , Gram-Positive Bacterial Infections/microbiology , Humans , Mice , Microbial Sensitivity Tests/methods , Neutrophils/microbiology
17.
Clin Infect Dis ; 69(2): 259-265, 2019 07 02.
Article in English | MEDLINE | ID: mdl-30339217

ABSTRACT

BACKGROUND: Vancomycin-resistant enterococci are an important cause of healthcare-associated infections and are inherently resistant to many commonly used antibiotics. Linezolid is the only drug currently approved by the US Food and Drug Administration to treat vancomycin-resistant enterococci; however, resistance to this antibiotic appears to be increasing. Although outbreaks of linezolid- and vancomycin-resistant Enterococcus faecium (LR-VRE) in solid organ transplant recipients remain uncommon, they represent a major challenge for infection control and hospital epidemiology. METHODS: We describe a cluster of 4 LR-VRE infections among a group of liver and multivisceral transplant recipients in a single intensive care unit. Failure of treatment with linezolid in 2 cases led to a review of standard clinical laboratory methods for susceptibility determination. Testing by alternative methods including whole genome sequencing (WGS) and a comprehensive outbreak investigation including sampling of staff members and surfaces was performed. RESULTS: Review of laboratory testing methods revealed a limitation in the VITEK 2 system with regard to reporting resistance to linezolid. Linezolid resistance in all cases was confirmed by E-test method. The use of WGS identified a resistant subpopulation with the G2376C mutation in the 23S ribosomal RNA. Sampling of staff members' dominant hands as well as sampling of surfaces in the unit identified no contaminated sources for transmission. CONCLUSIONS: This cluster of LR-VRE in transplant recipients highlights the possible shortcomings of standard microbiology laboratory methods and underscores the importance of WGS to identify resistance mechanisms that can inform patient care, as well as infection control and antibiotic stewardship measures.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Enterococcus faecium/drug effects , Gram-Positive Bacterial Infections/microbiology , Linezolid/pharmacology , Transplant Recipients , Vancomycin-Resistant Enterococci/drug effects , Aged , Antimicrobial Stewardship , Disease Management , Disease Outbreaks , Enterococcus faecium/genetics , Enterococcus faecium/isolation & purification , Gram-Positive Bacterial Infections/epidemiology , Humans , Infection Control/methods , Intensive Care Units , Male , Middle Aged , Point Mutation , RNA, Ribosomal, 23S/genetics , Sequence Analysis, DNA , Vancomycin-Resistant Enterococci/genetics , Vancomycin-Resistant Enterococci/isolation & purification , Whole Genome Sequencing
18.
Article in English | MEDLINE | ID: mdl-31235559

ABSTRACT

Weekly oritavancin plus ampicillin continuous infusion combination therapy was used to successfully treat a deep spine vancomycin-resistant Enterococcus faecium infection associated with hardware. Checkerboard and time-kill assays confirmed synergy between these two antibiotics. Further synergies of oritavancin and ampicillin with rifampin or the endogenous human antimicrobial peptide cathelicidin LL-37 were demonstrated.


Subject(s)
Ampicillin/therapeutic use , Drug Resistance, Multiple, Bacterial/drug effects , Enterococcus faecium/drug effects , Gram-Positive Bacterial Infections/drug therapy , Lipoglycopeptides/therapeutic use , Osteomyelitis/drug therapy , Vancomycin-Resistant Enterococci/drug effects , Anti-Bacterial Agents/therapeutic use , Drug Synergism , Gram-Positive Bacterial Infections/microbiology , Humans , Male , Microbial Sensitivity Tests/methods , Middle Aged , Vancomycin/therapeutic use
19.
Article in English | MEDLINE | ID: mdl-31332078

ABSTRACT

Daptomycin binds to bacterial cell membranes and disrupts essential cell envelope processes, leading to cell death. Bacteria respond to daptomycin by altering their cell envelopes to either decrease antibiotic binding to the membrane or by diverting binding away from septal targets. In Enterococcus faecalis, daptomycin resistance is typically coordinated by the three-component cell envelope stress response system, LiaFSR. Here, studying a clinical strain of multidrug-resistant Enterococcus faecium containing alleles associated with activation of the LiaFSR signaling pathway, we found that specific environments selected for different evolutionary trajectories, leading to high-level daptomycin resistance. Planktonic environments favored pathways that increased cell surface charge via yvcRS upregulation of dltABCD and mprF, causing a reduction in daptomycin binding. Alternatively, environments favoring complex structured communities, including biofilms, evolved both diversion and repulsion strategies via divIVA and oatA mutations, respectively. Both environments subsequently converged on cardiolipin synthase (cls) mutations, suggesting the importance of membrane modification across strategies. Our findings indicate that E. faecium can evolve diverse evolutionary trajectories to daptomycin resistance that are shaped by the environment to produce a combination of resistance strategies. The accessibility of multiple and different biochemical pathways simultaneously suggests that the outcome of daptomycin exposure results in a polymorphic population of resistant phenotypes, making E. faecium a recalcitrant nosocomial pathogen.


Subject(s)
Daptomycin/pharmacology , Drug Resistance, Bacterial/genetics , Enterococcus faecium/drug effects , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Cell Membrane/microbiology , Enterococcus faecalis/drug effects , Enterococcus faecium/genetics , Membrane Proteins/genetics , Microbial Sensitivity Tests/methods , Mutation/genetics , Plankton/microbiology , Transferases (Other Substituted Phosphate Groups)/genetics
20.
Article in English | MEDLINE | ID: mdl-30962347

ABSTRACT

The viridans group streptococci (VGS) are a heterogeneous group of organisms which are important components of the normal human oral flora. Among the VGS, the Streptococcus mitis/oralis subgroup is one of the most common causes of infective endocarditis (IE). Daptomycin (DAP) is a potential alternative therapeutic option for invasive S. mitis infections, given high rates of ß-lactam resistance and vancomycin tolerance in such strains. However, the ability of these strains to rapidly evolve high-level and durable DAP resistance (DAP-R) is problematic. Recent data suggest that combination DAP-ß-lactam therapy circumvents this issue. Human-simulated dose-escalating DAP-alone dose regimens (6, 8, 10, or 12 mg/kg/day times 4 days) versus DAP (6 mg/kg/day) plus ceftriaxone (CRO) (2 g once daily times 4 days or 0.5 g, single dose) were assessed against two prototypical DAP-susceptible (DAP-S) S. mitis/oralis strains (SF100 and 351), as measured by a pharmacokinetic/pharmacodynamic (PK/PD) model of simulated endocardial vegetations (SEVs). No DAP-alone regimen was effective, with regrowth of high-level DAP-R isolates observed for both strains over 96-h exposures. Combinations of DAP-CRO with either single- or multidose regimens yielded significant reductions in log10 CFU/g amounts within SEVs for both strains (∼6 log10 CFU/g) within 24 h. In addition, no DAP-R strains were detected in either DAP-CRO combination regimens over the 96-h exposure. In contrast to prior in vitro studies, no perturbations in two key cardiolipin biosynthetic genes (cdsA and pgsA) were identified in DAP-R SEV isolates emerging from strain 351, despite defective phospholipid production. The combination of DAP-CRO warrants further investigation for treatment of IE due to S. mitis/oralis.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Ceftriaxone/administration & dosage , Daptomycin/administration & dosage , Endocarditis, Bacterial/drug therapy , Streptococcus mitis/drug effects , Streptococcus oralis/drug effects , Drug Resistance, Bacterial/drug effects , Drug Therapy, Combination/methods , Endocarditis/drug therapy , Endocarditis/microbiology , Endocarditis, Bacterial/microbiology , Humans , Microbial Sensitivity Tests/methods , Streptococcus mitis/metabolism , Streptococcus oralis/metabolism , Vancomycin/administration & dosage , beta-Lactams/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL