Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 394
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35181608

ABSTRACT

Dynamic biomaterials excel at recapitulating the reversible interlocking and remoldable structure of the extracellular matrix (ECM), particularly in manipulating cell behaviors and adapting to tissue morphogenesis. While strategies based on dynamic chemistries have been extensively studied for ECM-mimicking dynamic biomaterials, biocompatible molecular means with biogenicity are still rare. Here, we report a nature-derived strategy for fabrication of dynamic biointerface as well as a three-dimensional (3D) hydrogel structure based on reversible receptor-ligand interaction between the glycopeptide antibiotic vancomycin and dipeptide d-Ala-d-Ala. We demonstrate the reversible regulation of multiple cell types with the dynamic biointerface and successfully implement the dynamic hydrogel as a functional antibacterial 3D scaffold to treat tissue repair. In view of the biogenicity and high applicability, this nature-derived reversible molecular strategy will bring opportunities for malleable biomaterial design with great potential in biomedicine.


Subject(s)
Extracellular Matrix/chemistry , Extracellular Matrix/physiology , Protein Engineering/methods , Alanine/chemistry , Alanine/metabolism , Biocompatible Materials/chemistry , Biomimetics/methods , Dipeptides/metabolism , Humans , Hydrogels/chemistry , Ligands , Vancomycin/chemistry , Vancomycin/metabolism
2.
J Pharmacol Exp Ther ; 388(1): 218-227, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38050132

ABSTRACT

Although vancomycin (VCM)-frequently used to treat drug-resistant bacterial infections-often induces acute kidney injury (AKI), discontinuation of the drug is the only effective treatment; therefore, analysis of effective avoidance methods is urgently needed. Here, we report the differences in the induction of AKI by VCM in 1/2-nephrectomized mice depending on the time of administration. Despite the lack of difference in the accumulation of VCM in the kidney between the light (ZT2) and dark (ZT14) phases, the expression of AKI markers due to VCM was observed only in the ZT2 treatment. Genomic analysis of the kidney suggested that the time of administration was involved in VCM-induced changes in monocyte and macrophage activity, and VCM had time-dependent effects on renal macrophage abundance, ATP activity, and interleukin (IL)-1ß expression. Furthermore, the depletion of macrophages with clodronate abolished the induction of IL-1ß and AKI marker expression by VCM administration at ZT2. This study provides evidence of the need for time-dependent pharmacodynamic considerations in the prevention of VCM-induced AKI as well as the potential for macrophage-targeted AKI therapy. SIGNIFICANCE STATEMENT: There is a time of administration at which vancomycin (VCM)-induced renal injury is more and less likely to occur, and macrophages are involved in this difference. Therefore, there is a need for time-dependent pharmacodynamic considerations in the prevention of VCM-induced acute kidney injury as well as the potential for macrophage-targeted acute kidney injury therapy.


Subject(s)
Acute Kidney Injury , Vancomycin , Mice , Animals , Vancomycin/pharmacology , Vancomycin/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Kidney , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Macrophages
3.
Int J Toxicol ; 43(2): 177-183, 2024.
Article in English | MEDLINE | ID: mdl-37989131

ABSTRACT

Septic arthritis as a complication of orthopaedic joint surgery can have catastrophic outcomes for patients. To minimise infection risk associated with elective orthopaedics, topical vancomycin during surgery has become increasingly common. Evidence suggests that high concentrations of vancomycin, following direct application of the drug to the joint, are toxic towards various local cell types in the joint, including chondrocytes. However, the mechanism of this vancomycin tissue toxicity is yet to be determined. The aim of this study was to evaluate the toxicity of vancomycin on chondrocytes and the mechanisms of cell death involved. Human primary knee chondrocytes were exposed to vancomycin (1.25-10 mg/mL) for 24 h and their viability assessed using the resazurin reduction assay in vitro. Specific cell death mechanisms and their contributors, including reactive oxygen species (ROS) production and apoptosis, were measured. This study showed that high concentrations of vancomycin (5 and 10 mg/mL) were toxic towards human primary knee chondrocyte cells, while lower concentrations (1.25 and 2.5 mg/mL) were not. Cell death studies found that this occurred through an apoptotic pathway. This study provides additional support that vancomycin in high doses is toxic towards chondrocytes and preliminary evidence that this toxicity occurs via apoptotic cell death mechanisms.


Subject(s)
Chondrocytes , Vancomycin , Humans , Vancomycin/toxicity , Vancomycin/metabolism , Chondrocytes/metabolism , Apoptosis , Cell Death , Reactive Oxygen Species/metabolism , Cells, Cultured
4.
Bioorg Chem ; 130: 106232, 2023 01.
Article in English | MEDLINE | ID: mdl-36371819

ABSTRACT

Bacterial two-component systems (TCSs), which typically consist of a sensor histidine kinase (HK) and a response regulator (RR), have been investigated as attractive antibacterial drug targets. Unfortunately, current HK activity assays based on the quantification of autophosphorylated HKs are hampered by the instability of the phosphohistidine (pHis) product, rendering them ill-suited for high-throughput screenings. To address this challenge, we developed a simple HK activity assay using readily available reagents, which we have termed AUDECY (AUtophosphorylation-DEphosphorylation CYcle assay). Instead of trying to preserve the fragile pHis, we deliberately decomposed it with a pHis-specific phosphatase to constitute an ATPase-like cycle for convenient colorimetric measurements. This kinetic assay was successfully employed for the kinetic characterization of E. coli EnvZ and for high-throughput inhibitor screening of vancomycin-resistant Enterococcus faecium (VRE) VanS, of which histidine kinase activity was hardly detectable with conventional methods. Through the screening, we identified OSU-03012, a potent VanS HK inhibitor, which sensitized VRE toward vancomycin, highlighting the potential of AUDECY in HK inhibitor discovery.


Subject(s)
Escherichia coli , Vancomycin , Histidine Kinase/metabolism , Vancomycin/metabolism , Vancomycin/pharmacology , Escherichia coli/metabolism , Protein Kinases/metabolism , High-Throughput Screening Assays , Transcription Factors/metabolism , Bacterial Proteins/metabolism
5.
Bioconjug Chem ; 33(5): 767-772, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35499914

ABSTRACT

Bacterial cell walls represent one of the most prominent targets of antibacterial agents. These agents include natural products (e.g., vancomycin) and proteins stemming from the innate immune system (e.g., peptidoglycan-recognition proteins and lysostaphin). Among bacterial pathogens that infect humans, Staphylococcus aureus (S. aureus) continues to impose a tremendous healthcare burden across the globe. S. aureus has evolved countermeasures that can directly restrict the accessibility of innate immune proteins, effectively protecting itself from threats that target key cell well components. We recently described a novel assay that directly reports on the accessibility of molecules to the peptidoglycan layer within the bacterial cell wall of S. aureus. The assay relies on site-specific chemical remodeling of the peptidoglycan with a biorthogonal handle. Here, we disclose the application of our assay to a screen of a nonredundant transposon mutant library for susceptibility of the peptidoglycan layer with the goal of identifying genes that contribute to the control of cell surface accessibility. We discovered several genes that resulted in higher accessibility levels to the peptidoglycan layer and showed that these genes modulate sensitivity to lysostaphin. These results indicate that this assay platform can be leveraged to gain further insight into the biology of bacterial cell surfaces.


Subject(s)
Lysostaphin , Staphylococcus aureus , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Cell Wall/chemistry , Humans , Lysostaphin/chemistry , Lysostaphin/metabolism , Lysostaphin/pharmacology , Peptidoglycan/chemistry , Vancomycin/metabolism
6.
Int J Med Sci ; 19(4): 740-752, 2022.
Article in English | MEDLINE | ID: mdl-35582415

ABSTRACT

Background: Oxidative stress-related apoptosis is considered as the key mechanism implicated in the pathophysiology of nephrotoxicity with vancomycin (VCM) therapy. We evaluated the possible effects of N-acetylcysteine (NAC) on VCM-induced nephrotoxicity and the underlying mechanism. Methods: VCM-induced nephrotoxicity was established using HK-2 cells and SD rats and observed by measuring cell survival, kidney histological changes, renal function and kidney injury related markers (KIM-1 and NGAL). Oxidative stress, renal cell apoptosis and the involved signaling pathways were also evaluated. Results: In model rats, NAC could protect against VCM-induced acute kidney injury with histological damage, renal dysfunction, and increased Cre and BUN levels. In HK-2 cells, VCM-induced decreased cell viability was restored by NAC. In addition, increased expression of caspase-3, KIM-1 and NGAL suffering from VCM was also reversed by NAC in vivo and in vitro. NAC inhibited ROS production, decreased cell apoptosis by decreasing the Bax/Bcl-2 ratio and caspase-3 expression in HK-2 cells and regulated oxidative stress indicators in the kidney by decreasing GSH, SOD and CAT activity and increasing MDA levels. Furthermore, NAC could effectively reverse VCM-associated increased P38 MAPK/JNK phosphorylation. Conclusions: The results demonstrated that NAC had a protective effect against nephrotoxicity from VCM by inhibiting oxidative stress and apoptosis via P38 MAPK/JNK.


Subject(s)
Acute Kidney Injury , Vancomycin , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Animals , Anti-Bacterial Agents/adverse effects , Apoptosis , Caspase 3/metabolism , Kidney/pathology , Lipocalin-2/metabolism , Lipocalin-2/pharmacology , Oxidative Stress , Rats , Rats, Sprague-Dawley , Rats, Wistar , Vancomycin/adverse effects , Vancomycin/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Acc Chem Res ; 53(11): 2587-2599, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33138354

ABSTRACT

Since its discovery, vancomycin has been used in the clinic for >60 years. Because of their durability, vancomycin and related glycopeptides serve as the antibiotics of last resort for the treatment of protracted bacterial infections of resistant Gram-positive pathogens, including methicillin-resistant Staphylococcus aureus (MRSA) and multidrug-resistant (MDR) Streptococcus pneumoniae. After 30 years of use, vancomycin resistance was first observed and is now widespread in enterococci and more recently in S. aureus. The widespread prevalence of vancomycin-resistant enterococci (VRE) and the emergence of vancomycin-resistant S. aureus (VRSA) represent a call to focus on the challenge of resistance, highlight the need for new therapeutics, and provide the inspiration for the design of more durable antibiotics less prone to bacterial resistance than even vancomycin.Herein we summarize progress on efforts to overcome vancomycin resistance, first addressing recovery of its original durable mechanism of action and then introducing additional independent mechanisms of action intended to increase the potency and durability beyond that of vancomycin itself. The knowledge of the origin of vancomycin resistance and an understanding of the molecular basis of the loss of binding affinity between vancomycin and the altered target ligand d-Ala-d-Lac provided the basis for the subtle and rational redesign of the vancomycin binding pocket to remove the destabilizing lone-pair repulsion or reintroduce a lost H-bond while not impeding binding to the unaltered ligand d-Ala-d-Ala. Preparation of the modified glycopeptide core structure was conducted by total synthesis, providing binding pocket-modified vancomycin aglycons with dual d-Ala-d-Ala/d-Lac binding properties that directly address the intrinsic mechanism of resistance to vancomycin. Fully glycosylated pocket-modified vancomycin analogues were generated through a subsequent two-step enzymatic glycosylation, providing a starting point for peripheral modifications used to introduce additional mechanisms of action. A well-established vancosamine N-(4-chlorobiphenyl)methyl (CBP) modification as well as newly discovered C-terminal trimethylammonium cation (C1) or guanidine modifications were introduced, providing two additional synergistic mechanisms of action independent of d-Ala-d-Ala/d-Lac binding. The CBP modification provides an additional stage for inhibition of cell wall synthesis that results from direct competitive inhibition of transglycosylase, whereas the C1/guanidine modification induces bacteria cell permeablization. The synergistic behavior of the three independent mechanisms of action combined in a single molecule provides ultrapotent antibiotics (MIC = 0.01-0.005 µg/mL against VanA VRE). Beyond the remarkable antimicrobial activity, the multiple mechanisms of action suppress the rate at which resistance may be selected, where any single mechanism of action is protected by the action of others. The results detailed herein show that rational targeting of durable vancomycin-derived antibiotics has generated compounds with a "resistance against resistance", provided new candidate antibiotics, and may serve as a generalizable strategy to combat antibacterial resistance.


Subject(s)
Anti-Bacterial Agents/chemistry , Drug Design , Vancomycin/analogs & derivatives , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Binding Sites , Dipeptides/chemistry , Dipeptides/metabolism , Glycopeptides/chemistry , Glycopeptides/metabolism , Guanidine/chemistry , Half-Life , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Staphylococcus aureus/drug effects , Vancomycin/chemical synthesis , Vancomycin/metabolism , Vancomycin/pharmacology , Vancomycin Resistance/drug effects
8.
Anesthesiology ; 132(6): 1516-1527, 2020 06.
Article in English | MEDLINE | ID: mdl-32053565

ABSTRACT

BACKGROUND: Intravenous vancomycin is used to treat ventilator-associated pneumonia caused by methicillin-resistant Staphylococcus aureus, but achieves high rates of failure. Vancomycin nebulization may be efficient to provide high vancomycin lung tissue concentrations. The aim of this study was to compare lung tissue and serum concentrations of vancomycin administered intravenously and by aerosol in mechanically ventilated and anesthetized healthy piglets. METHODS: Twelve female piglets received a single intravenous dose of vancomycin (15 mg/kg) and were killed 1 (n = 6) or 12 h (n = 6) after the end of administration. Twelve piglets received a single nebulized dose of vancomycin (37.5 mg/kg) and were killed 1 (n = 6) or 12 h (n = 6) after the end of the aerosol administration. In each group, vancomycin lung tissue concentrations were assessed on postmortem lung specimens using high-performance liquid chromatography. Blood samples were collected for serum vancomycin concentration measurement 30 min and 1, 2, 4, 6, 8, and 12 h after the end of vancomycin administration. Pharmacokinetics was analyzed by nonlinear mixed effect modeling. RESULTS: One hour after vancomycin administration, lung tissue concentrations in the aerosol group were 13 times the concentrations in the intravenous group (median and interquartile range: 161 [71, 301] µg/g versus 12 [4, 42] µg/g; P < 0.0001). Twelve hours after vancomycin administration, lung tissue concentrations in the aerosol group were 63 (23, 119) µg/g and 0 (0, 19) µg/g in the intravenous group (P < 0.0001). A two-compartment weight-scaled allometric model with first-order absorption and elimination best fit serum pharmacokinetics after both routes of administration. Area under the time-concentration curve from 0 to 12 h was lower in the aerosol group in comparison to the intravenous group (56 [8, 70] mg · h · l vs. 121 [103, 149] mg · h · l, P = 0.002). Using a population model, vancomycin bioavailability was 13% (95% CI, 6 to 69; coefficient of variation = 85%) and absorption rate was slow (absorption half life = 0.3 h). CONCLUSIONS: Administration of vancomycin by nebulization resulted in higher lung tissue concentrations than the intravenous route.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Lung/metabolism , Nebulizers and Vaporizers , Respiration, Artificial/methods , Vancomycin/administration & dosage , Administration, Inhalation , Administration, Intravenous , Animals , Anti-Bacterial Agents/metabolism , Female , Models, Animal , Swine , Vancomycin/metabolism
9.
J Sep Sci ; 43(21): 3987-3994, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32845063

ABSTRACT

A rapid and simple ultra high performance liquid chromatography with tandem mass spectrometry method was developed and validated for the simultaneous separation and determination of vancomycin and its crystalline degradation products in human serum. Vancomycin and two isomers of the degradants were extracted from human serum with a protein precipitation method. The compounds were separated on an Acquity BEH C18 column (2.1 × 50 mm, 1.7 µm) eluted with a gradient mixture of acetonitrile and 0.1% formic acid as the mobile phase. Norvancomycin was used as the internal standard. The linear ranges of vancomycin and two degradant isomers were 1.057-105.7, 0.1437-14.37, and 0.2540-25.40 µg/mL, respectively. The established methods were validated and successfully applied to a therapeutic drug monitoring study of vancomycin in patients with renal insufficiency.


Subject(s)
Drug Monitoring , Vancomycin/blood , Chromatography, High Pressure Liquid , Humans , Molecular Structure , Tandem Mass Spectrometry , Vancomycin/isolation & purification , Vancomycin/metabolism
10.
J Appl Toxicol ; 40(7): 897-907, 2020 07.
Article in English | MEDLINE | ID: mdl-32079046

ABSTRACT

Vancomycin is a first-line treatment for invasive infections caused by multidrug-resistant gram-positive bacteria. However, vancomycin-induced nephrotoxicity is an increasing burden, particularly in patients with complex life-threatening conditions. Vancomycin-induced nephrotoxicity associated with clinically relevant exposure on the target site has not been well defined. This study aimed to acquire the concentration of vancomycin in the renal tubules and kidneys in humans using physiologically based pharmacokinetic (PBPK) modeling and simulation. Based upon the exposure of vancomycin in the renal tubule, the toxicity of vancomycin in human renal proximal tubular epithelial cells was examined with the XTT assay and in vitro metabolomics analysis. A rat PBPK model predicting plasma and kidney concentration-time profiles of vancomycin matched the observed behavior after a single administration of 10 mg/kg. The concentration of vancomycin in renal tubules was about 40-50 times higher than that in plasma. The human PBPK model transferred from the rat model predicted renal tubule concentrations of vancomycin as 316.1-2136.6 µg/mL at 500 mg every 6 hours, and 199.0-3932.5 µg/mL at 1000 mg every 12 hours. Vancomycin showed significant nephrotoxicity at 4 mg/mL in XTT assessment. In total, 11 lysophosphatidylcholines and one lysophosphatidylethanolamine were identified by metabolomics analysis. The concentration-dependent increase was evident in the release of lysophospholipids after vancomycin treatment (0.125-4 mg/mL) for 24 hours. Our study revealed the relationship between the exposure of vancomycin in the kidney and toxicity of vancomycin at clinically relevant concentrations achieved from a mechanical PBPK model. A series of lysophospholipids as potential metabolic markers of renal toxicity were identified.


Subject(s)
Kidney Diseases/chemically induced , Kidney Diseases/diagnosis , Kidney Diseases/metabolism , Kidney Tubules/drug effects , Kidney/drug effects , Vancomycin/metabolism , Vancomycin/pharmacokinetics , Vancomycin/toxicity , Adult , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/toxicity , Cells, Cultured/drug effects , Female , Humans , Kidney/chemistry , Kidney Tubules/chemistry , Male , Metabolomics , Models, Animal , Toxicity Tests , Vancomycin/blood , Young Adult
11.
J Korean Med Sci ; 35(37): e306, 2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32959542

ABSTRACT

BACKGROUND: The objective of this study was to compare the performance of cystatin C- and creatinine-based estimated glomerular filtration rate (eGFR) equations in predicting the clearance of vancomycin. METHODS: MEDLINE and Embase databases were searched from inception up to September 2019 to identify all studies that compared the predictive performance of cystatin C- and/or creatinine-based eGFR in predicting the clearance of vancomycin. The prediction errors (PEs) (the value of eGFR equations minus vancomycin clearance) were quantified for each equation and were pooled using a random-effects model. The root mean squared errors were also quantified to provide a metric for imprecision. RESULTS: This meta-analysis included evaluations of seven different cystatin C- and creatinine-based eGFR equations in total from 26 studies and 1,234 patients. The mean PE (MPE) for cystatin C-based eGFR was 4.378 mL min-1 (95% confidence interval [CI], -29.425, 38.181), while the creatinine-based eGFR provided an MPE of 27.617 mL min-1 (95% CI, 8.675, 46.560) in predicting clearance of vancomycin. This indicates the presence of unbiased results in vancomycin clearance prediction by the cystatin C-based eGFR equations. Meanwhile, creatinine-based eGFR equations demonstrated a statistically significant positive bias in vancomycin clearance prediction. CONCLUSION: Cystatin C-based eGFR equations are better than creatinine-based eGFR equations in predicting the clearance of vancomycin. This suggests that utilising cystatin C-based eGFR equations could result in better accuracy and precision to predict vancomycin pharmacokinetic parameters.


Subject(s)
Creatinine/blood , Cystatin C/blood , Glomerular Filtration Rate/physiology , Vancomycin/metabolism , Bayes Theorem , Databases, Factual , Humans
12.
Mol Microbiol ; 104(2): 319-333, 2017 04.
Article in English | MEDLINE | ID: mdl-28118510

ABSTRACT

Peptidoglycan (PG), the major component of the bacterial cell wall, is one large macromolecule. To allow for the different curvatures of PG at cell poles and division sites, there must be local differences in PG architecture and eventually also chemistry. Here we report such local differences in the Gram-positive rod-shaped model organism Bacillus subtilis. Single-cell analysis after antibiotic treatment and labeling of the cell wall with a fluorescent analogue of vancomycin or the fluorescent D-amino acid analogue (FDAA) HCC-amino-D-alanine revealed that PG at the septum contains muropeptides with unprocessed stem peptides (pentapeptides). Whereas these pentapeptides are normally shortened after incorporation into PG, this activity is reduced at division sites indicating either a lower local degree of PG crosslinking or a difference in PG composition, which could be a topological marker for other proteins. The pentapeptides remain partially unprocessed after division when they form the new pole of a cell. The accumulation of unprocessed PG at the division site is not caused by the activity of the cell division specific penicillin-binding protein 2B. To our knowledge, this is the first indication of local differences in the chemical composition of PG in Gram-positive bacteria.


Subject(s)
Peptidoglycan/chemistry , Peptidoglycan/metabolism , Amino Acid Sequence , Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Cell Division , Cell Wall/metabolism , Penicillin-Binding Proteins/metabolism , Single-Cell Analysis , Vancomycin/metabolism
13.
Mol Microbiol ; 106(5): 719-741, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28922502

ABSTRACT

When the cell envelope integrity is compromised, bacteria trigger signaling cascades resulting in the production of proteins that counteract these extracytoplasmic stresses. Here, we show that the two-component system EsrSR regulates a cell envelope stress response in the Actinobacterium Corynebacterium glutamicum. The sensor kinase EsrS possesses an amino-terminal phage shock protein C (PspC) domain, a property that sets EsrSR apart from all other two-component systems characterized so far. An integral membrane protein, EsrI, whose gene is divergently transcribed to the esrSR gene locus and which interestingly also possesses a PspC domain, acts as an inhibitor of EsrSR under non-stress conditions. The resulting EsrISR three-component system is activated among others by antibiotics inhibiting the lipid II cycle, such as bacitracin and vancomycin, and it orchestrates a broad regulon including the esrI-esrSR gene locus itself, genes encoding heat shock proteins, ABC transporters, and several putative membrane-associated or secreted proteins of unknown function. Among those, the ABC transporter encoded by cg3322-3320 was shown to be directly involved in bacitracin resistance of C. glutamicum. Since similar esrI-esrSR loci are present in a large number of actinobacterial genomes, EsrISR represents a novel type of stress-responsive system whose components are highly conserved in the phylum Actinobacteria.


Subject(s)
Bacterial Proteins/metabolism , Corynebacterium glutamicum/metabolism , Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Bacitracin/pharmacology , Base Sequence , Binding Sites , Cell Wall/metabolism , Corynebacterium glutamicum/genetics , Gene Expression Profiling , Gene Expression Regulation, Bacterial/drug effects , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Sigma Factor/metabolism , Stress, Physiological , Transcription, Genetic/drug effects , Vancomycin/metabolism
14.
J Am Soc Nephrol ; 28(6): 1723-1728, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28082518

ABSTRACT

Vancomycin is a widely prescribed antibiotic, but the exact nature of vancomycin-associated nephrotoxicity is unclear, in particular when considering the frequent coadministration of aminoglycosides. We describe here the initial case of a 56-year-old woman with normal renal function developing unexplained ARF without hypovolemia after administration of vancomycin without coadministration of aminoglycosides. Studying the patient's renal biopsy specimen, we ascertained that obstructive tubular casts composed of noncrystal nanospheric vancomycin aggregates entangled with uromodulin explained the vancomycin-associated ARF. We developed in parallel a new immunohistologic staining technique to detect vancomycin in renal tissue and confirmed retrospectively that deleterious vancomycin-associated casts existed in eight additional patients with acute tubular necrosis in the absence of hypovolemia. Concomitant high vancomycin trough plasma levels had been observed in each patient. We also reproduced experimentally the toxic and obstructive nature of vancomycin-associated cast nephropathy in mice, which we detected using different in vivo imaging techniques. In conclusion, the interaction of uromodulin with nanospheric vancomycin aggregates represents a new mode of tubular cast formation, revealing the hitherto unsuspected mechanism of vancomycin-associated renal injury.


Subject(s)
Anti-Bacterial Agents/adverse effects , Kidney Diseases/chemically induced , Vancomycin/adverse effects , Anti-Bacterial Agents/metabolism , Female , Humans , Kidney Diseases/pathology , Middle Aged , Uromodulin/metabolism , Vancomycin/metabolism
15.
J Antimicrob Chemother ; 72(3): 801-804, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27999035

ABSTRACT

Objectives: The objectives of this observational study were to investigate plasma protein binding and to evaluate target attainment rates of vancomycin therapy in critically ill children. Patients and methods: Paediatric ICU patients, in whom intravenous intermittent dosing (ID) or continuous dosing (CD) with vancomycin was indicated, were included. Covariates on unbound vancomycin fraction and concentration were tested using a linear mixed model analysis and attainment of currently used pharmacokinetic/pharmacodynamic (PK/PD) targets was evaluated. Clinicaltrials.gov: NCT02456974. Results: One hundred and eighty-eight plasma samples were collected from 32 patients. The unbound vancomycin fraction (median = 71.1%; IQR = 65.4%-79.7%) was highly variable within and between patients and significantly correlated with total protein and albumin concentration, which were both decreased in our population. Total trough concentration (ID) and total concentration (CD) were within the aimed target concentrations in 8% of patients. The targets of AUC/MIC ≥400 and f AUC/MIC ≥200 were achieved in 54% and 83% of patients, respectively. Unbound vancomycin concentrations were adequately predicted using the following equation: unbound vancomycin concentration (mg/L) = 5.38 + [0.71 × total vancomycin concentration (mg/L)] - [0.085 × total protein concentration (g/L)]. This final model was externally validated using 51 samples from another six patients. Conclusions: The protein binding of vancomycin in our paediatric population was lower than reported in non-critically ill adults and exhibited large variability. Higher target attainment rates were achieved when using PK/PD indices based on unbound concentrations, when compared with total concentrations. These results highlight the need for protein binding assessment in future vancomycin PK/PD research.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Blood Proteins/metabolism , Critical Illness/therapy , Vancomycin/pharmacokinetics , Adolescent , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/therapeutic use , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Intensive Care Units , Linear Models , Male , Microbial Sensitivity Tests , Prospective Studies , Protein Binding , Vancomycin/blood , Vancomycin/metabolism , Vancomycin/therapeutic use
16.
J Antimicrob Chemother ; 72(9): 2454-2460, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28595277

ABSTRACT

Objectives: We previously reported the first case of vancomycin treatment failure due to development of vancomycin-intermediate resistance in a patient with an MRSA of ST72, a community genotype in Korea. We investigated two isogenic MRSA strains from this patient, who experienced treatment failure with vancomycin and rifampicin. Methods: We tracked the genetic alterations that confer reduced susceptibility to vancomycin on those two isogenic MRSA strains by WGS. Results: Five non-synonymous mutations were identified, including rpoB (H481Y), dprA (G196C), femA (F92C), vraR (E127K) and agrC (E391stop). We further studied the role of a mutation of vraR in reduced susceptibility to vancomycin. Introduction of the mutated vraR (E127K) into a vancomycin-susceptible Staphylococcus aureus strain resulted in an increase in vraSR mRNA expression and vancomycin MIC and development of the hetero-VISA phenotype, which was confirmed by the population analysis profile (PAP)/AUC. Electron microscopy showed increased cell wall thickness in the strains with mutated vraR. Conclusions: Based on the genomic data, molecular experiments and PAP and cell wall analyses, we propose that a single mutation of vraR is associated with the reduced susceptibility to vancomycin in MRSA and further treatment failure.


Subject(s)
Anti-Bacterial Agents/pharmacology , Community-Acquired Infections/microbiology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Staphylococcal Infections/microbiology , Vancomycin/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Proteins/genetics , Cell Wall/drug effects , Cell Wall/genetics , Cell Wall/ultrastructure , Community-Acquired Infections/drug therapy , DNA-Binding Proteins/genetics , Genotype , Humans , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/metabolism , Microbial Sensitivity Tests , Microscopy, Electron , Mutation , Phenotype , Staphylococcal Infections/drug therapy , Treatment Failure , Vancomycin/metabolism
17.
Bioconjug Chem ; 28(2): 353-361, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27959504

ABSTRACT

Many bacterial pathogens have now acquired resistance toward commonly used antibiotics, such as the glycopeptide antibiotic vancomycin. In this study, we show that immobilization of vancomycin onto a nanometer-scale solid surface with controlled local density can potentiate antibiotic action and increase target affinity of the drug. Magnetic nanoparticles were conjugated with vancomycin and used as a model system to investigate the relationship between surface density and drug potency. We showed remarkable improvement in minimum inhibitory concentration against vancomycin-resistant strains with values of 13-28 µg/mL for conjugated vancomycin compared to 250-4000 µg/mL for unconjugated vancomycin. Higher surface densities resulted in enhanced affinity toward the bacterial target compared to that of unconjugated vancomycin, as measured by a competition experiment using a surrogate ligand for bacterial Lipid II, N-Acetyl-l-Lys-d-Ala-d-Ala. High density vancomycin nanoparticles required >64 times molar excess of ligand (relative to the vancomycin surface density) to abrogate antibacterial activity compared to only 2 molar excess for unconjugated vancomycin. Further, the drug-nanoparticle conjugates caused rapid permeabilization of the bacterial cell wall within 2 h, whereas no effect was seen with unconjugated vancomycin, suggesting additional modes of action for the nanoparticle-conjugated drug. Hence, immobilization of readily available antibiotics on nanocarriers may present a general strategy for repotentiating drugs that act on bacterial membranes or membrane-bound targets but have lost effectiveness against resistant bacterial strains.


Subject(s)
Bacteria/drug effects , Cell Membrane Permeability , Nanoparticles/chemistry , Vancomycin Resistance/drug effects , Vancomycin/chemistry , Vancomycin/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Azides/chemistry , Ligands , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Surface Properties , Vancomycin/metabolism
18.
Biochim Biophys Acta Gen Subj ; 1861(8): 1951-1959, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28511809

ABSTRACT

A-type resistance towards "last-line" glycopeptide antibiotic vancomycin in the leading hospital acquired infectious agent, the enterococci, is the most common in the UK. Resistance is regulated by the VanRASA two-component system, comprising the histidine sensor kinase VanSA and the partner response regulator VanRA. The nature of the activating ligand for VanSA has not been identified, therefore this work sought to identify and characterise ligand(s) for VanSA. In vitro approaches were used to screen the structural and activity effects of a range of potential ligands with purified VanSA protein. Of the screened ligands (glycopeptide antibiotics vancomycin and teicoplanin, and peptidoglycan components N-acetylmuramic acid, D-Ala-D-Ala and Ala-D-y-Glu-Lys-D-Ala-D-Ala) only glycopeptide antibiotics vancomycin and teicoplanin were found to bind VanSA with different affinities (vancomycin 70µM; teicoplanin 30 and 170µM), and were proposed to bind via exposed aromatic residues tryptophan and tyrosine. Furthermore, binding of the antibiotics induced quicker, longer-lived phosphorylation states for VanSA, proposing them as activators of type A vancomycin resistance in the enterococci.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Enterococcus/drug effects , Protein Kinases/metabolism , Teicoplanin/metabolism , Transcription Factors/metabolism , Vancomycin Resistance , Vancomycin/metabolism , Bacterial Proteins/chemistry , Enterococcus/metabolism , Phosphorylation , Protein Conformation , Protein Kinases/chemistry , Transcription Factors/chemistry
19.
J Am Chem Soc ; 138(47): 15397-15404, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27797504

ABSTRACT

The concurrence of enzymatic reaction and ligand-receptor interactions is common for proteins, but rare for small molecules and has yet to be explored. Here we show that ligand-receptor interaction modulates the morphology of molecular assemblies formed by enzyme-instructed assembly of small molecules. While the absence of ligand-receptor interaction allows enzymatic dephosphorylation of a precursor to generate the hydrogelator that self-assembles to form long nanofibers, the presence of the ligand-receptor interaction biases the pathway to form precipitous aggregates containing short nanofibers. While the hydrogelators self-assemble to form nanofibers or nanoribbons that are unable to bind with the ligand (i.e., vancomycin), the addition of surfactant breaks up the assemblies to restore the ligand-receptor interaction. In addition, an excess amount of the ligands can disrupt the nanofibers and result in the precipitates. As the first example of the use of ligand-receptor interaction to modulate the kinetics of enzymatic self-assembly, this work not only provides a solution to evaluate the interaction between aggregates and target molecules but also offers new insight for understanding the emergent behavior of sophisticated molecular systems having multiple and parallel processes.


Subject(s)
Alkaline Phosphatase/metabolism , Small Molecule Libraries/metabolism , Vancomycin/metabolism , Alkaline Phosphatase/chemistry , Biocatalysis , Kinetics , Ligands , Particle Size , Small Molecule Libraries/chemistry , Surface Properties , Surface-Active Agents/chemistry , Surface-Active Agents/metabolism , Vancomycin/chemistry
20.
J Biol Regul Homeost Agents ; 30(4 Suppl 1): 229-235, 2016.
Article in English | MEDLINE | ID: mdl-28002924

ABSTRACT

The purpose of this study is to investigate the best preparation method of the cement powder mixture, solvent and antibiotic in order to obtain the greatest amount of antibiotic in the joint for the longest time as possible. At time T0 the three samples, packed in a sterile environment in different formulations, were placed in sterile tubes, adding to each one 5 ml of saline phosphate buffer solution (PBS) and put in a stove at 37°C for 24 h. A sample of PBS without cement (T control) was also created. Qualitative and quantitative assessment of the incubated liquid with cement was performed along with biochemical analysis with High Performance Liquid Chromatography (HPLC). The analysis of the raw data demonstrated that at T1 there was a prevalence of antibiotic release from sample , compared to sample 2 and 3. This difference was maintained until the T20; from T21 the antibiotic release gradually leveled in 3 samples. The elution of the antibiotic remained detectable up to T60. Our work shows that the sample preparation is decisive on the quantity of released antibiotic. These results are confirmed by microbiological tests. It is useful to know the actual kinetics of antibiotics in articulation. Further studies are necessary to determine the effectiveness of antibiotic against micro-organisms and how long it acts.


Subject(s)
Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bone Cements/chemistry , Bone Cements/metabolism , Drug Liberation , Vancomycin/metabolism , Vancomycin/pharmacology , Microbial Sensitivity Tests
SELECTION OF CITATIONS
SEARCH DETAIL