129 results on '"Tony Velkov"'
Search Results
2. Mutations in the Vicinity of the IRAK3 Guanylate Cyclase Center Impact Its Subcellular Localization and Ability to Modulate Inflammatory Signaling in Immortalized Cell Lines
- Author
-
Ilona Turek, Trang H. Nguyen, Charles Galea, Isaiah Abad, Lubna Freihat, David T. Manallack, Tony Velkov, and Helen Irving
- Subjects
interleukin-1 receptor-associated kinase 3 (IRAK3 or IRAK-M) ,guanylate cyclase ,cyclic guanosine monophosphate (cGMP) ,human Toll-like receptor 4 (hTLR4) ,lipopolysaccharide ,nuclear factor kappa-light-chain-enhancer of activated B (NFκB) ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Interleukin-1 receptor-associated kinase 3 (IRAK3) modulates the magnitude of cellular responses to ligands perceived by interleukin-1 receptors (IL-1Rs) and Toll-like receptors (TLRs), leading to decreases in pro-inflammatory cytokines and suppressed inflammation. The molecular mechanism of IRAK3’s action remains unknown. IRAK3 functions as a guanylate cyclase, and its cGMP product suppresses lipopolysaccharide (LPS)-induced nuclear factor kappa-light-chain-enhancer of activated B cell (NFκB) activity. To understand the implications of this phenomenon, we expanded the structure–function analyses of IRAK3 through site-directed mutagenesis of amino acids known or predicted to impact different activities of IRAK3. We verified the capacity of the mutated IRAK3 variants to generate cGMP in vitro and revealed residues in and in the vicinity of its GC catalytic center that impact the LPS-induced NFκB activity in immortalized cell lines in the absence or presence of an exogenous membrane-permeable cGMP analog. Mutant IRAK3 variants with reduced cGMP generating capacity and differential regulation of NFκB activity influence subcellular localization of IRAK3 in HEK293T cells and fail to rescue IRAK3 function in IRAK3 knock-out THP-1 monocytes stimulated with LPS unless the cGMP analog is present. Together, our results shed new light on the mechanism by which IRAK3 and its enzymatic product control the downstream signaling, affecting inflammatory responses in immortalized cell lines.
- Published
- 2023
- Full Text
- View/download PDF
3. An Efficient Approach for the Design and Synthesis of Antimicrobial Peptide-Peptide Nucleic Acid Conjugates
- Author
-
Nitin A. Patil, Varsha J. Thombare, Rong Li, Xiaoji He, Jing Lu, Heidi H. Yu, Hasini Wickremasinghe, Kavya Pamulapati, Mohammad A. K. Azad, Tony Velkov, Kade D. Roberts, and Jian Li
- Subjects
antisense oligonucleotides ,peptide nucleic acids ,antimicrobial agents ,cell-penetrating peptides ,conjugation ,Chemistry ,QD1-999 - Abstract
Peptide-Peptide Nucleic Acid (PNA) conjugates targeting essential bacterial genes have shown significant potential in developing novel antisense antimicrobials. The majority of efforts in this area are focused on identifying different PNA targets and the selection of peptides to deliver the peptide-PNA conjugates to Gram-negative bacteria. Notably, the selection of a linkage strategy to form peptide-PNA conjugate plays an important role in the effective delivery of PNAs. Recently, a unique Cysteine- 2-Cyanoisonicotinamide (Cys-CINA) click chemistry has been employed for the synthesis of cyclic peptides. Considering the high selectivity of this chemistry, we investigated the efficiency of Cys-CINA conjugation to synthesize novel antimicrobial peptide-PNA conjugates. The PNA targeting acyl carrier protein gene (acpP), when conjugated to the membrane-active antimicrobial peptides (polymyxin), showed improvement in antimicrobial activity against multidrug-resistant Gram-negative Acinetobacter baumannii. Thus, indicating that the Cys-CINA conjugation is an effective strategy to link the antisense oligonucleotides with antimicrobial peptides. Therefore, the Cys-CINA conjugation opens an exciting prospect for antimicrobial drug development.
- Published
- 2022
- Full Text
- View/download PDF
4. Lipidomic Analysis of the Outer Membrane Vesicles from Paired Polymyxin-Susceptible and -Resistant Klebsiella pneumoniae Clinical Isolates
- Author
-
Raad Jasim, Mei-Ling Han, Yan Zhu, Xiaohan Hu, Maytham H. Hussein, Yu-Wei Lin, Qi (Tony) Zhou, Charlie Yao Da Dong, Jian Li, and Tony Velkov
- Subjects
outer membrane vesicles ,lipidomics ,Gram-negative ,polymyxin ,extremely drug resistant ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Gram-negative bacteria produce outer membrane vesicles (OMVs) as delivery vehicles for nefarious bacterial cargo such as virulence factors, which are antibiotic resistance determinants. This study aimed to investigate the impact of polymyxin B treatment on the OMV lipidome from paired polymyxin-susceptible and -resistant Klebsiella pneumoniae isolates. K. pneumoniae ATCC 700721 was employed as a reference strain in addition to two clinical strains, K. pneumoniae FADDI-KP069 and K. pneumoniae BM3. Polymyxin B treatment of the polymyxin-susceptible strains resulted in a marked reduction in the glycerophospholipid, fatty acid, lysoglycerophosphate and sphingolipid content of their OMVs. Conversely, the polymyxin-resistant strains expressed OMVs richer in all of these lipid species, both intrinsically and increasingly under polymyxin treatment. The average diameter of the OMVs derived from the K. pneumoniae ATCC 700721 polymyxin-susceptible isolate, measured by dynamic light scattering measurements, was ~90.6 nm, whereas the average diameter of the OMVs isolated from the paired polymyxin-resistant isolate was ~141 nm. Polymyxin B treatment (2 mg/L) of the K. pneumoniae ATCC 700721 cells resulted in the production of OMVs with a larger average particle size in both the susceptible (average diameter ~124 nm) and resistant (average diameter ~154 nm) strains. In light of the above, we hypothesize that outer membrane remodelling associated with polymyxin resistance in K. pneumoniae may involve fortifying the membrane structure with increased glycerophospholipids, fatty acids, lysoglycerophosphates and sphingolipids. Putatively, these changes serve to make the outer membrane and OMVs more impervious to polymyxin attack.
- Published
- 2018
- Full Text
- View/download PDF
5. A novel chemical biology and computational approach to expedite the discovery of new-generation polymyxins against life-threatening Acinetobacter baumannii
- Author
-
Kai Yang, Hasini Wickremasinghe, Lushan Wang, Xukai Jiang, Nitin A. Patil, Bin Gong, Lin Wan, Wendong Ma, Tony Velkov, Mengyao Li, Bing Yuan, Philip E. Thompson, Falk Schreiber, Jinxin Zhao, Xinru Zhang, Jian Li, Mohammad Abul Kalam Azad, Kade D. Roberts, and Heidi Yu
- Subjects
Chemistry ,biology ,medicine.drug_class ,Polymyxin ,Chemical biology ,medicine ,General Chemistry ,Computational biology ,Bacterial outer membrane ,biology.organism_classification ,Acinetobacter baumannii - Abstract
Multidrug-resistant Gram-negative bacteria represent a major medical challenge worldwide. New antibiotics are desperately required with ‘old’ polymyxins often being the only available therapeutic option. Here, we systematically investigated the structure–activity relationship (SAR) of polymyxins using a quantitative lipidomics-informed outer membrane (OM) model of Acinetobacter baumannii and a series of chemically synthesized polymyxin analogs. By integrating chemical biology and all-atom molecular dynamics simulations, we deciphered how each residue of the polymyxin molecule modulated its conformational folding and specific interactions with the bacterial OM. Importantly, a novel designed polymyxin analog FADDI-287 with predicted stronger OM penetration showed improved in vitro antibacterial activity. Collectively, our study provides a novel chemical biology and computational strategy to expedite the discovery of new-generation polymyxins against life-threatening Gram-negative ‘superbugs’., Multidrug-resistant Gram-negative bacteria have been an urgent threat to global public health. Novel antibiotics are desperately needed to combat these 'superbugs'.
- Published
- 2021
6. Simulations of octapeptin–outer membrane interactions reveal conformational flexibility is linked to antimicrobial potency
- Author
-
Falk Schreiber, James D. Swarbrick, Bing Yuan, Nitin A. Patil, Xukai Jiang, Jian Li, Lushan Wang, Lin Wan, Kade D. Roberts, Bin Gong, Kai Yang, and Tony Velkov
- Subjects
Acinetobacter baumannii ,0301 basic medicine ,Conformational change ,Gram-negative bacteria ,Antimicrobial peptides ,Molecular Dynamics Simulation ,Biochemistry ,Lipopeptides ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Structure–activity relationship ,Molecular Biology ,030102 biochemistry & molecular biology ,biology ,Cell Membrane ,Computational Biology ,Lipopeptide ,Cell Biology ,biology.organism_classification ,030104 developmental biology ,chemistry ,octapeptin, polymyxin, antimicrobial resistance, Gram-negative bacteria, molecular dynamics, conformational transition, antibiotic action, antibiotic resistance, drug design, conformational change ,Biophysics ,ddc:004 ,Umbrella sampling ,Bacterial outer membrane ,Acyl group - Abstract
The octapeptins are lipopeptide antibiotics that are structurally similar to polymyxins yet retain activity against polymyxin-resistant Gram-negative pathogens, suggesting they might be used to treat recalcitrant infections. However, the basis of their unique activity is unclear because of the difficulty in generating high-resolution experimental data of the interaction of antimicrobial peptides with lipid membranes. To elucidate these structure-activity relationships, we employed all-atom molecular dynamics simulations with umbrella sampling to investigate the conformational and energetic landscape of octapeptins interacting with bacterial outer membrane (OM). Specifically, we examined the interaction of octapeptin C4 and FADDI-115, lacking a single hydroxyl group compared with octapeptin C4, with the lipid A-phosphoethanolamine modified OM of Acinetobacter baumannii Octapeptin C4 and FADDI-115 both penetrated into the OM hydrophobic center but experienced different conformational transitions from an unfolded to a folded state that was highly dependent on the structural flexibility of their respective N-terminal fatty acyl groups. The additional hydroxyl group present in the fatty acyl group of octapeptin C4 resulted in the molecule becoming trapped in a semifolded state, leading to a higher free energy barrier for OM penetration. The free energy barrier for the translocation through the OM hydrophobic layer was ∼72 kcal/mol for octapeptin C4 and 62 kcal/mol for FADDI-115. Our results help to explain the lower antimicrobial activity previously observed for octapeptin C4 compared with FADDI-115 and more broadly improve our understanding of the structure-function relationships of octapeptins. These findings may facilitate the discovery of next-generation octapeptins against polymyxin-resistant Gram-negative 'superbugs.' published
- Published
- 2020
7. Outer Membranes of Polymyxin-Resistant Acinetobacter baumannii with Phosphoethanolamine-Modified Lipid A and Lipopolysaccharide Loss Display Different Atomic-Scale Interactions with Polymyxins
- Author
-
Falk Schreiber, Lushan Wang, Jian Li, Tony Velkov, Mei-Ling Han, Xukai Jiang, Bin Gong, Bing Yuan, Jingliang Li, and Kai Yang
- Subjects
Acinetobacter baumannii ,Lipopolysaccharides ,0301 basic medicine ,Lipopolysaccharide ,medicine.drug_class ,Polymyxin ,030106 microbiology ,Article ,Microbiology ,Lipid A ,03 medical and health sciences ,chemistry.chemical_compound ,Antibiotic resistance ,Lipidomics ,medicine ,Polymyxins ,biology ,biology.organism_classification ,Anti-Bacterial Agents ,030104 developmental biology ,Infectious Diseases ,chemistry ,Ethanolamines ,Colistin ,lipids (amino acids, peptides, and proteins) ,Bacterial outer membrane ,medicine.drug - Abstract
Resistance to the last-line polymyxins is increasingly reported in multidrug-resistant Gram-negative pathogens, including Acinetobacter baumannii, which develops resistance via either lipid A modification (e.g., with phosphoethanolamine [pEtN]) or even lipopolysaccharide (LPS) loss in the outer membrane (OM). Considering these two different mechanisms, quantitative membrane lipidomics data were utilized to develop three OM models representing polymyxin-susceptible and -resistant A. baumannii strains. Through all-atom molecular simulations with enhanced sampling techniques, the effect of lipid A-pEtN modification and LPS loss on the action of colistin (i.e., polymyxin E) was examined for the first time, with a focus on the dynamics and energetics of colistin penetration into these OMs. Lipid A-pEtN modification improved the OM stability, impeding the penetration of colistin into the OM; this differed from the current literature that lipid A-pEtN modification confers resistance by diminishing the initial interaction with polymyxins. In contrast, the LPS deficiency significantly reduced the negative charges on the OM surface, diminishing the binding of colistin. Moreover, both lipid A-pEtN modification and LPS loss also constituted colistin resistance through disturbing the conformational transitions of the colistin molecule. Collectively, atomic-scale interactions between polymyxins and different bacterial OMs are very different and the findings may facilitate the discovery of new-generation polymyxins against Gram-negative 'superbugs'.
- Published
- 2020
8. Polymyxin B combinations with FDA-approved non-antibiotic phenothiazine drugs targeting multi-drug resistance of Gram-negative pathogens
- Author
-
Olivia K. A. Paulin, Tony Velkov, Elena K Schneider-Futschik, Xiaohan Hu, Yan Zhu, Qi Tony Zhou, Simon Crawford, Jian Li, Mark Baker, and Maytham Hussein
- Subjects
medicine.drug_class ,lcsh:Biotechnology ,Polymyxin ,Antibiotics ,Biophysics ,Drug resistance ,Antimicrobial resistance ,Biochemistry ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Antibiotic resistance ,Structural Biology ,Phenothiazines ,lcsh:TP248.13-248.65 ,MDR ,Genetics ,medicine ,Metabolomics ,030304 developmental biology ,ComputingMethodologies_COMPUTERGRAPHICS ,0303 health sciences ,biology ,Chemistry ,biology.organism_classification ,Gram-negative ,Computer Science Applications ,Acinetobacter baumannii ,030220 oncology & carcinogenesis ,Membrane biogenesis ,Colistin ,Antimicrobial peptides ,Polymyxin B ,Biotechnology ,medicine.drug ,Research Article - Abstract
Graphical abstract, The status quo for combating uprising antibacterial resistance is to employ synergistic combinations of antibiotics. Nevertheless, the currently available combination therapies are fast becoming untenable. Combining antibiotics with various FDA-approved non-antibiotic drugs has emerged as a novel strategy against otherwise untreatable multi-drug resistant (MDR) pathogens. The apex of this study was to investigate the mechanisms of antibacterial synergy of the combination of polymyxin B with the phenothiazines against the MDR Gram-negative pathogens Acinetobacter baumannii, Klebsiella pneumoniae and Pseudomonas aeruginosa. The synergistic antibacterial effects were tested using checkerboard and static time-kill assays. Electron microscopy (EM) and untargeted metabolomics were used to ascertain the mechanism(s) of the antibacterial synergy. The combination of polymyxin B and the phenothiazines showed synergistic antibacterial activity in checkerboard and static time-kill assays at clinically relevant concentrations against both polymyxin-susceptible and polymyxin-resistant isolates. EM revealed that the polymyxin B-prochlorperazine combination resulted in greater damage to the bacterial cell compared to each drug monotherapy. In metabolomics, at 0.5 h, polymyxin B monotherapy and the combination (to a greatest extent) disorganised the bacterial cell envelope as manifested by a major perturbation in bacterial membrane lipids (glycerophospholipids and fatty acids), peptidoglycan and lipopolysaccharide (LPS) biosynthesis. At the late time exposure (4 h), the aforementioned effects (except LPS biosynthesis) perpetuated mainly with the combination therapy, indicating the disorganising bacterial membrane biogenesis is potentially behind the mechanisms of antibacterial synergy. In conclusion, the study highlights the potential usefulness of the combination of polymyxin B with phenothiazines for the treatment of polymyxin-resistant Gram-negative infections (e.g. CNS infections).
- Published
- 2020
9. Effective Strategy Targeting Polymyxin-Resistant Gram-Negative Pathogens: Polymyxin B in Combination with the Selective Serotonin Reuptake Inhibitor Sertraline
- Author
-
Yan Zhu, Qi Tony Zhou, Simon Crawford, Olivia K. A. Paulin, Rafah Allobawi, Adil Hanif, Jian Li, Mark Baker, Maytham Hussein, Elena K Schneider-Futschik, and Tony Velkov
- Subjects
0301 basic medicine ,medicine.drug_class ,Polymyxin ,Serotonin reuptake inhibitor ,030106 microbiology ,Drug Synergism ,Microbial Sensitivity Tests ,Pharmacology ,Antimicrobial ,Multiple drug resistance ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Infectious Diseases ,chemistry ,Sertraline ,medicine ,Fatty acid elongation ,Polymyxins ,Peptidoglycan ,Bacterial outer membrane ,Selective Serotonin Reuptake Inhibitors ,Polymyxin B ,medicine.drug - Abstract
This study aimed to investigate synergistic antibacterial activity of polymyxin B in combination with the selective serotonin reuptake inhibitor, sertraline, against the Gram-negative pathogens Acinetobacter baumannii, Klebsiella pneumoniae, and Pseudomonas aeruginosa. The combination of polymyxin B and sertraline showed synergistic antibacterial activity in checkerboard and static time-kill assays at clinically relevant concentrations against both polymyxin-susceptible and polymyxin-resistant isolates. The potential antimicrobial mode of action of the combination was investigated against P. aeruginosa FADDI-PA024 using untargeted metabolomics alongside scanning and transmission electron microscopy (EM). Scanning and transmission EM revealed that the polymyxin B and sertraline combination resulted in greater damage to the bacterial cell compared to each drug alone. Metabolomics results showed that the combination significantly affected the bacterial ability to remodel its outer membrane. This was reflected by the major perturbation of glycerophospholipids and fatty acids and the pantothenate and coenzyme A (CoA) pathways, which feed fatty acid elongation (e.g., trans-hexadec-2-enoyl-CoA) as well as inhibit the biosynthesis of lipopolysaccharide and peptidoglycan. The combination also inhibited the polymyxin resistance phosphoethanolamine (pEtN) lipid A modification pathway, indicated by the declined levels of phosphoethanolamine. In summary, the present study highlights the potential possibilities of a polymyxin-sertraline combination for the treatment of infections caused by multidrug resistant Gram-negative bacteria such as central nervous system (CNS) infections via direct intraventricular/intrathecal delivery.
- Published
- 2020
10. Structure of micelle bound cationic peptides by NMR spectroscopy using a lanthanide shift reagent
- Author
-
Jian Li, James D. Swarbrick, John A. Karas, and Tony Velkov
- Subjects
Lanthanide ,Protein Conformation ,Chemistry ,Ligand ,Metals and Alloys ,Cationic polymerization ,General Chemistry ,Nuclear magnetic resonance spectroscopy ,Nuclear Overhauser effect ,Lanthanoid Series Elements ,Micelle ,Catalysis ,Surfaces, Coatings and Films ,Electronic, Optical and Magnetic Materials ,NMR spectra database ,Crystallography ,Reagent ,Materials Chemistry ,Ceramics and Composites ,Indicators and Reagents ,Peptides ,Nuclear Magnetic Resonance, Biomolecular ,Micelles ,Antimicrobial Cationic Peptides ,Polymyxin B - Abstract
[Tm(DPA)3]3- was used to generate multiple, paramagnetic nuclear Overhauser effect NMR spectra of cationic peptides when weakly bound to a lipopolysaccharide micelle. Increased spectral resolution combined with a marked increase in the number of distance restraints yielded high resolution structures of polymyxin and MSI-594 in the liposaccharide bound state.
- Published
- 2020
11. Polymyxin-Induced Metabolic Perturbations in Human Lung Epithelial Cells
- Author
-
Tony Velkov, Maizbha Uddin Ahmed, Mei-Ling Han, Kim H. Chan, Qi Tony Zhou, Jian Li, Mengyao Li, Darren J. Creek, Mohammad Abul Kalam Azad, and Fanfan Zhou
- Subjects
Taurine ,medicine.drug_class ,Polymyxin ,Hypotaurine ,Pharmacology ,chemistry.chemical_compound ,Tandem Mass Spectrometry ,medicine ,Humans ,Pharmacology (medical) ,Polymyxins ,Lung ,Polymyxin B ,A549 cell ,Colistin ,Epithelial Cells ,Glutathione ,Anti-Bacterial Agents ,Infectious Diseases ,chemistry ,Toxicity ,Chromatography, Liquid ,medicine.drug - Abstract
Inhaled polymyxins are associated with toxicity in human lung epithelial cells that involves multiple apoptotic pathways. However, the mechanism of polymyxin-induced pulmonary toxicity remains unclear. This study aims to investigate polymyxin-induced metabolomic perturbations in human lung epithelial A549 cells. A549 cells were treated with 0.5 or 1.0 mM polymyxin B or colistin for 1, 4, and 24 h. Cellular metabolites were analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS), and significantly perturbed metabolites (log(2) fold change [log(2)FC] ≥ 1; false-discovery rate [FDR] ≤ 0.2) and key pathways were identified relative to untreated control samples. At 1 and 4 h, very few significant changes in metabolites were observed relative to the untreated control cells. At 24 h, taurine (log(2)FC = −1.34 ± 0.64) and hypotaurine (log(2)FC = −1.20 ± 0.27) were significantly decreased by 1.0 mM polymyxin B. The reduced form of glutathione (GSH) was significantly depleted by 1.0 mM polymyxin B at 24 h (log(2)FC = −1.80 ± 0.42). Conversely, oxidized glutathione (GSSG) was significantly increased by 1.0 mM both polymyxin B (log(2)FC = 1.38 ± 0.13 at 4 h and 2.09 ± 0.20 at 24 h) and colistin (log(2)FC = 1.33 ± 0.24 at 24 h). l-Carnitine was significantly decreased by 1.0 mM of both polymyxins at 24 h, as were several key metabolites involved in biosynthesis and degradation of choline and ethanolamine (log(2)FC ≤ −1); several phosphatidylserines were also increased (log(2)FC ≥ 1). Polymyxins perturbed key metabolic pathways that maintain cellular redox balance, mitochondrial β-oxidation, and membrane lipid biogenesis. These mechanistic findings may assist in developing new pharmacokinetic/pharmacodynamic strategies to attenuate the pulmonary toxicities of inhaled polymyxins and in the discovery of new-generation polymyxins.
- Published
- 2021
12. Insights Into Patient Variability During Ivacaftor-Lumacaftor Therapy in Cystic Fibrosis
- Author
-
Patrick O. Hanafin, Isabelle Sermet-Gaudelus, Matthias Griese, Matthias Kappler, Helmut Ellemunter, Carsten Schwarz, John Wilson, Marsha Tan, Tony Velkov, Gauri G. Rao, and Elena K. Schneider-Futschik
- Subjects
cytochrome interactions ,Metabolite ,Cmax ,RM1-950 ,Pharmacology ,030226 pharmacology & pharmacy ,Cystic fibrosis ,drug- drug interactions ,Ivacaftor ,cystic fibrosis ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,medicine ,pharmacodynamics ,Pharmacology (medical) ,pharmacokinetic ,Original Research ,biology ,business.industry ,lumacaftor ,Lumacaftor ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,chemistry ,Pharmacodynamics ,biology.protein ,ivacaftor ,Therapeutics. Pharmacology ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Background: The advent of cystic fibrosis transmembrane conductance regulator protein (CFTR) modulators like ivacaftor have revolutionised the treatment of cystic fibrosis (CF). However, due to the plethora of variances in disease manifestations in CF, there are inherent challenges in unified responses under CFTR modulator treatment arising from variability in patient outcomes. The pharmacokinetic (PK) data available for ivacaftor-lumacaftor cystic fibrosis (CF) transmembrane conductance regulator (CFTR) modulator drug combination is limited.Methods: Secondary objectives were to identify (1) patient characteristics and (2) the interactions between ivacaftor-lumacaftor responsible for interindividual variability (IIV).Results: Peak plasma concentrations (Cmax) of ivacaftor - lumacaftor were >10 fold lower than expected compared to label information. The one-way ANOVA indicated that the patient site had an effect on Cmax values of ivacaftor metabolites ivacaftor-M1, ivacaftor-M6, and lumacaftor (p < 0.001, p < 0.001, and p < 0.001, respectively). The Spearman’s rho test indicated that patient weight and age have an effect on the Cmax of lumacaftor (p = 0.003 and p < 0.001, respectively) and ivacaftor metabolite M1 (p = 0.020 and p < 0.001, respectively). Age (p < 0.001) was found to effect on Cmax of ivacaftor M6 and on Tmax of ivacaftor M1 (p = 0.026). A large impact of patient characteristics on the IIV of PK parameters Cmax and Tmax, was observed among the CF patients.Conclusion: Understanding the many sources of variability can help reduce this individual patient variability and ensure consistent patient outcomes.
- Published
- 2021
13. Inhibition of Oxidative Stress and ALOX12 and NF-κB Pathways Contribute to the Protective Effect of Baicalein on Carbon Tetrachloride-Induced Acute Liver Injury
- Author
-
Chongshan Dai, Tony Velkov, Jianzhong Shen, Hui Li, Yang Wang, and Shusheng Tang
- Subjects
0301 basic medicine ,Programmed cell death ,baicalein ,Physiology ,Clinical Biochemistry ,Inflammation ,RM1-950 ,Pharmacology ,acute liver injury ,medicine.disease_cause ,Biochemistry ,digestive system ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,ALOX12 pathway ,medicine ,oxidative stress ,Molecular Biology ,Nrf2 pathway ,Cell Biology ,Glutathione ,Malondialdehyde ,digestive system diseases ,ferroptosis ,Baicalein ,Heme oxygenase ,030104 developmental biology ,chemistry ,Apoptosis ,030220 oncology & carcinogenesis ,Therapeutics. Pharmacology ,medicine.symptom ,Oxidative stress - Abstract
This study investigates the protective effect of baicalein on carbon tetrachloride (CCl4)-induced acute liver injury and the underlying molecular mechanisms. Mice were orally administrated baicalein at 25 and 100 mg/kg/day for 7 consecutive days or ferrostatin-1 (Fer-1) at 10 mg/kg was i.p. injected in mice at 2 and 24 h prior to CCl4 injection or the vehicle. Our results showed that baicalein or Fer-1 supplementation significantly attenuated CCl4 exposure-induced elevations of serum alanine aminotransferase and aspartate aminotransferase, and malondialdehyde levels in the liver tissues and unregulated glutathione levels. Baicalein treatment inhibited the nuclear factor kappa-B (NF-κB) pathway, activated the erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway in liver tissues, and markedly improved CCl4-induced apoptosis, inflammation and ferroptosis in liver tissues exposed with CCl4. In vitro, baicalein treatment improved CCl4 -induced decreases of cell viabilities and knockdown of Nrf2 and arachidonate 12-lipoxygenase (ALOX12) genes partly abolished the protective effect of baicalein on CCl4 -induced cytotoxicity in HepG2 cells. In conclusion, our results reveal that baicalein supplementation ameliorates CCl4-induced acute liver injury in mice by upregulating the antioxidant defense pathways and downregulating oxidative stress, apoptosis, inflammation and ferroptosis, which involved the activation of Nrf2 pathway and the inhibition of ALOX12 and NF-κB pathways.
- Published
- 2021
14. An optimised Cu(0)-RDRP approach for the synthesis of lipidated oligomeric vinyl azlactone: toward a versatile antimicrobial materials screening platform
- Author
-
John F. Quinn, Véronique Montembault, Cornelia B. Landersdorfer, Nghia P. Truong, Alysha G. Elliott, Tony Velkov, Sagrario Pascual, Michael R. Whittaker, James L. Grace, Thomas P. Davis, Janet C. Reid, Maite Amado, Kristian Kempe, Laurent Fontaine, Mark E. Cooper, Dalhousie University [Halifax], Institut des Molécules et Matériaux du Mans (IMMM), Le Mans Université (UM)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), Department of Haematology, University College London Cancer Institute, and Monash University [Melbourne]
- Subjects
Lysis ,Tertiary amine ,Biomedical Engineering ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Oxazolone ,Structure-Activity Relationship ,chemistry.chemical_compound ,Imidazole ,General Materials Science ,Polyvinyl Chloride ,Guanidine ,ComputingMilieux_MISCELLANEOUS ,Molecular Structure ,[CHIM.ORGA]Chemical Sciences/Organic chemistry ,Cationic polymerization ,General Chemistry ,General Medicine ,021001 nanoscience & nanotechnology ,Antimicrobial ,Combinatorial chemistry ,6. Clean water ,Anti-Bacterial Agents ,0104 chemical sciences ,[CHIM.POLY]Chemical Sciences/Polymers ,chemistry ,Amine gas treating ,0210 nano-technology - Abstract
This report details the synthesis of lipidated 2-vinyl-4,4-dimethyl-5-oxazolone (VDM) oligomers via an optimised Cu(0)-mediated reversible-deactivation radical polymerisation approach, and the use of these oligomers as a versatile functional platform for the rapid generation of antimicrobial materials. The relative amounts of CuBr2 and Me6TREN were optimised to allow the fast and controlled polymerisation of VDM. These conditions were then used with the initiators ethyl 2-bromoisobutyrate, dodecyl 2-bromoisobutyrate, and (R)-3-((2-bromo-2-methylpropanoyl)oxy)propane-1,2-diyl didodecanoate to synthesise a library of oligo(VDM) (degree of polymerisation = 10) with ethyl, dodecyl or diglyceride end-groups. Subsequently, ring-opening of the pendant oxazolone group with various amines (i.e., 2-(2-aminoethyl)-1,3-di-Boc-guanidine, 1-(3-aminopropyl)imidazole, N-Boc-ethylenediamine, or N,N-dimethylethylenediamine) expanded the library to give 12 functional oligomers incorporating different cationic and lipid elements. The antimicrobial activities of these oligomers were assessed against a palette of bacteria and fungi: i.e. Staphylococcus aureus, Escherichia coli, Candida albicans, and Cryptococcus neoformans. The oligomers generally exhibited the greatest activity against the fungus, C. neoformans, with a minimum inhibitory concentration of 1 μg mL-1 (comparable to the clinically approved antifungal fluconazole). To assess haemocompatibility, the oligomers were assayed against erythrocytes, with the primary amine or guanidine containing C12 and 2C12 oligomers exhibiting greater lysis against the red blood cells (HC10 values between 7.1 and 43 μg mL-1) than their imidazole and tertiary amine counterparts (HC10 of >217 μg mL-1). Oligomers showed the greatest selectivity for C. neoformans, with the C12- and 2C12-tertiary amine and C12-imidazole oligomers possessing the greatest selectivity of >54-109. These results demonstrate the utility of reactive oligomers for rapidly assessing structure-property relationships for antibacterial and antifungal materials.
- Published
- 2019
15. Coarse-grained simulations uncover Gram-negative bacterial defense against polymyxins by the outer membrane
- Author
-
Xukai Jiang, Bing Yuan, Tony Velkov, Yuliang Sun, Kai Yang, Jian Li, and Lushan Wang
- Subjects
LPS ,Lipopolysaccharide ,medicine.drug_class ,Antibiotic resistance ,Polymyxin ,Biophysics ,Biochemistry ,Lipid A ,chemistry.chemical_compound ,Structural Biology ,Genetics ,medicine ,ComputingMethodologies_COMPUTERGRAPHICS ,biology ,Molecular dynamics simulations ,Inner core ,Antimicrobial ,biology.organism_classification ,Computer Science Applications ,Outer membrane ,Membrane ,chemistry ,lipids (amino acids, peptides, and proteins) ,Bacterial outer membrane ,Bacteria ,TP248.13-248.65 ,Biotechnology ,Research Article - Abstract
Graphical abstract, Highlights • A structural model of bacterial outer membrane (OM) was developed with Ra LPS. • Free energy landscape was revealed for polymyxin interactions with the OM. • LPS core sugars and calcium ions confer intrinsic resistance to antibiotics., The outer membrane (OM) of Gram-negative bacteria is a formidable barrier against antibiotics. Understanding the structure and function of the OM is essential for the discovery of novel membrane-acting agents against multidrug-resistant Gram-negative pathogens. However, it remains challenging to obtain three-dimensional structure of bacterial membranes using crystallographic approaches, which has significantly hindered the elucidation of its interaction with antibiotics. Here, we developed an asymmetric OM model consisting of rough lipopolysaccharide (LPS) and three key types of phospholipids. Using coarse-grained molecular dynamics simulations, we investigated the interaction dynamics of LPS-containing OM with the polymyxins, a last-line class of antibiotics against Gram-negative ‘superbugs’. We discovered that polymyxin molecules spontaneously penetrated the OM core sugar region where most were trapped before entering the lipid A region. Examination of the free energy profile of polymyxin penetration revealed a major free energy barrier at the LPS inner core and lipid A interface. Further analysis revealed calcium ions predominantly distributed in the inner core region and mediated extensive cross-linking interactions between LPS molecules, thereby inhibiting the penetration of polymyxins into the hydrophobic region of the OM. Collectively, our results provide novel mechanistic insights into an intrinsic defense of Gram-negative bacteria to polymyxins and may help identify new antimicrobial targets.
- Published
- 2021
16. Clinically Relevant Concentrations of Polymyxin B and Meropenem Synergistically Kill Multidrug-Resistant Pseudomonas aeruginosa and Minimize Biofilm Formation
- Author
-
Phillip J. Bergen, Qi Tony Zhou, Heidi H. Yu, Mohammad A. K. Azad, Yan Zhu, Jian Li, Jinxin Zhao, Tony Velkov, and Hasini Wickremasinghe
- Subjects
Microbiology (medical) ,Membrane permeability ,medicine.drug_class ,Polymyxin ,Antibiotics ,synergy ,medicine.disease_cause ,Biochemistry ,Microbiology ,Meropenem ,Article ,biofilm ,combination therapy ,Antibiotic resistance ,meropenem ,multidrug resistance ,medicine ,Pharmacology (medical) ,General Pharmacology, Toxicology and Pharmaceutics ,Chemistry ,Pseudomonas aeruginosa ,lcsh:RM1-950 ,polymyxin ,Multiple drug resistance ,lcsh:Therapeutics. Pharmacology ,Infectious Diseases ,Polymyxin B ,medicine.drug - Abstract
The emergence of antibiotic resistance has severely impaired the treatment of chronic respiratory infections caused by multidrug-resistant (MDR) Pseudomonas aeruginosa. Since the reintroduction of polymyxins as a last-line therapy against MDR Gram-negative bacteria, resistance to its monotherapy and recurrent infections continue to be reported and synergistic antibiotic combinations have been investigated. In this study, comprehensive in vitro microbiological evaluations including synergy panel screening, population analysis profiling, time-kill kinetics, anti-biofilm formation and membrane damage analysis studies were conducted to evaluate the combination of polymyxin B and meropenem against biofilm-producing, polymyxin-resistant MDR P. aeruginosa. Two phylogenetically unrelated MDR P. aeruginosa strains, FADDI-PA060 (MIC of polymyxin B [MICpolymyxin B], 64 mg/L, MICmeropenem, 64 mg/L) and FADDI-PA107 (MICpolymyxin B, 32 mg/L, MICmeropenem, 4 mg/L) were investigated. Genome sequencing identified 57 (FADDI-PA060) and 50 (FADDI-PA107) genes predicted to confer resistance to a variety of antimicrobials, as well as multiple virulence factors in each strain. The presence of resistance genes to a particular antibiotic class generally aligned with MIC results. For both strains, all monotherapies of polymyxin B failed with substantial regrowth and biofilm formation. The combination of polymyxin B (16 mg/L)/meropenem (16 mg/L) was most effective, enhancing initial bacterial killing of FADDI-PA060 by ~3 log10 CFU/mL, followed by a prolonged inhibition of regrowth for up to 24 h with a significant reduction in biofilm formation (* p <, 0.05). Membrane integrity studies revealed a substantial increase in membrane depolarization and membrane permeability in the surviving cells. Against FADDI-PA107, planktonic and biofilm bacteria were completely eradicated. In summary, the combination of polymyxin B and meropenem demonstrated synergistic bacterial killing while reinstating the efficacy of two previously ineffective antibiotics against difficult-to-treat polymyxin-resistant MDR P. aeruginosa.
- Published
- 2021
- Full Text
- View/download PDF
17. Synchrotron-based X-ray fluorescence microscopy reveals accumulation of polymyxins in single human alveolar epithelial cells
- Author
-
Qi Tony Zhou, Simon James, Martin D. de Jonge, Tony Velkov, Jing Fu, Shuo Zhang, Daryl L. Howard, Kade D. Roberts, Yeonuk Kim, Heidi H. Yu, Jiayao Li, Alex J. Fulcher, Jian Li, and Mohamad A K Azad
- Subjects
A549 cell ,Pharmacology ,0303 health sciences ,Inhalation ,030306 microbiology ,Chemistry ,medicine.drug_class ,Pulmonary toxicity ,Polymyxin ,Molecular biology ,Nephrotoxicity ,03 medical and health sciences ,Infectious Diseases ,Pharmacokinetics ,Toxicity ,medicine ,Pharmacology (medical) ,Intracellular ,030304 developmental biology - Abstract
Intravenous administration of the last-line polymyxins results in poor drug exposure in the lungs and potential nephrotoxicity, whereas inhalation therapy offers better pharmacokinetics/pharmacodynamics for pulmonary infections by delivering the antibiotic directly to the infection site. However, polymyxin inhalation therapy has not been optimized, and adverse effects can occur. This study aimed to quantitatively determine the intracellular accumulation and distribution of polymyxins in single human alveolar epithelial A549 cells. Cells were treated with the iodine-labeled polymyxin probe FADDI-096 (5.0 and 10.0 μM) for 1, 4, and 24 h. Concentrations of FADDI-096 in single A549 cells were determined by synchrotron-based X-ray fluorescence microscopy. Concentration- and time-dependent accumulation of FADDI-096 within A549 cells was observed. The intracellular concentrations (mean ± standard error of the mean [SEM], n ≥ 189) of FADDI-096 were 1.58 ± 0.11, 2.25 ± 0.10, and 2.46 ± 0.07 mM following 1, 4, and 24 h of treatment at 10 μM, respectively. The corresponding intracellular concentrations following the treatment at 5 μM were 0.05 ± 0.01, 0.24 ± 0.04, and 0.25 ± 0.02 mM (n ≥ 189). Over 24 h, FADDI-096 was mainly localized throughout the cytoplasm and nuclear region. The intracellular zinc concentration increased in a concentration- and time-dependent manner. This is the first study to quantitatively map the accumulation of polymyxins in human alveolar epithelial cells, and it provides crucial insights for deciphering the mechanisms of their pulmonary toxicity. Importantly, our results may shed light on the optimization of inhaled polymyxins in patients and the development of new-generation, safer polymyxins.
- Published
- 2021
18. Molecular dynamics simulations informed by membrane lipidomics reveal the structure-interaction relationship of polymyxins with the lipid A-based outer membrane of Acinetobacter baumannii
- Author
-
Tony Velkov, Jingliang Li, Kai Yang, Falk Schreiber, Jian Li, Bing Yuan, Nitin A. Patil, Kade D. Roberts, Lushan Wang, Robert E. W. Hancock, Mei-Ling Han, Xukai Jiang, Yan Zhu, and Bin Gong
- Subjects
0301 basic medicine ,Microbiology (medical) ,Acinetobacter baumannii ,Membrane permeability ,Polymyxin B1 ,medicine.drug_class ,Polymyxin ,Molecular Dynamics Simulation ,01 natural sciences ,03 medical and health sciences ,0103 physical sciences ,medicine ,Humans ,Pharmacology (medical) ,Polymyxins ,Original Research ,Pharmacology ,010304 chemical physics ,biology ,Chemistry ,Periplasmic space ,biology.organism_classification ,Anti-Bacterial Agents ,030104 developmental biology ,Infectious Diseases ,Lipid A ,Lipidomics ,Biophysics ,Colistin ,lipids (amino acids, peptides, and proteins) ,ddc:004 ,Bacterial outer membrane ,Polymyxin B ,medicine.drug - Abstract
Background MDR bacteria represent an urgent threat to human health globally. Polymyxins are a last-line therapy against life-threatening Gram-negative ‘superbugs’, including Acinetobacter baumannii. Polymyxins exert antimicrobial activity primarily via permeabilizing the bacterial outer membrane (OM); however, the mechanism of interaction between polymyxins and the OM remains unclear at the atomic level. Methods We constructed a lipid A-based OM model of A. baumannii using quantitative membrane lipidomics data and employed all-atom molecular dynamics simulations with umbrella sampling techniques to elucidate the structure–interaction relationship and thermodynamics governing the penetration of polymyxins [B1 and E1 (i.e. colistin A) representing the two clinically used polymyxins] into the OM. Results Polymyxin B1 and colistin A bound to the A. baumannii OM by the initial electrostatic interactions between the Dab residues of polymyxins and the phosphates of lipid A, competitively displacing the cations from the headgroup region of the OM. Both polymyxin B1 and colistin A formed a unique folded conformation upon approaching the hydrophobic centre of the OM, consistent with previous experimental observations. Polymyxin penetration induced reorientation of the headgroups of the OM lipids near the penetration site and caused local membrane disorganization, thereby significantly increasing membrane permeability and promoting the subsequent penetration of polymyxin molecules into the OM and periplasmic space. Conclusions The thermodynamics governing the penetration of polymyxins through the outer leaflet of the A. baumannii OM were examined and novel structure–interaction relationship information was obtained at the atomic and membrane level. Our findings will facilitate the discovery of novel polymyxins against MDR Gram-negative pathogens.
- Published
- 2020
19. Genome-Scale Metabolic Modeling Reveals Metabolic Alterations of Multidrug-Resistant Acinetobacter baumannii in a Murine Bloodstream Infection Model
- Author
-
Yan Zhu, Jiping Wang, Jiru Han, Ke Chen, Falk Schreiber, Michael Aichem, Jian Li, Yu-Wei Lin, Jinxin Zhao, and Tony Velkov
- Subjects
Microbiology (medical) ,Transposable element ,Acinetobacter baumannii ,genome-scale metabolic modeling ,Mutant ,Biology ,Microbiology ,Article ,Transcriptome ,03 medical and health sciences ,chemistry.chemical_compound ,transcriptomics ,bacteremia ,RNA-seq ,Virology ,lcsh:QH301-705.5 ,Pathogen ,Gene ,030304 developmental biology ,0303 health sciences ,030306 microbiology ,Metabolism ,biology.organism_classification ,lcsh:Biology (General) ,chemistry ,Peptidoglycan ,ddc:004 - Abstract
Multidrug-resistant (MDR) Acinetobacter baumannii is a critical threat to human health globally. We constructed a genome-scale metabolic model iAB5075 for the hypervirulent, MDR A. baumannii strain AB5075. Predictions of nutrient utilization and gene essentiality were validated using Biolog assay and a transposon mutant library. In vivo transcriptomics data were integrated with iAB5075 to elucidate bacterial metabolic responses to the host environment. iAB5075 contains 1530 metabolites, 2229 reactions, and 1015 genes, and demonstrated high accuracies in predicting nutrient utilization and gene essentiality. At 4 h post-infection, a total of 146 metabolic fluxes were increased and 52 were decreased compared to 2 h post-infection, these included enhanced fluxes through peptidoglycan and lipopolysaccharide biosynthesis, tricarboxylic cycle, gluconeogenesis, nucleotide and fatty acid biosynthesis, and altered fluxes in amino acid metabolism. These flux changes indicate that the induced central metabolism, energy production, and cell membrane biogenesis played key roles in establishing and enhancing A. baumannii bloodstream infection. This study is the first to employ genome-scale metabolic modeling to investigate A. baumannii infection in vivo. Our findings provide important mechanistic insights into the adaption of A. baumannii to the host environment and thus will contribute to the development of new therapeutic agents against this problematic pathogen.
- Published
- 2020
- Full Text
- View/download PDF
20. Polymyxin-Induced Cell Death of Human Macrophage-Like THP-1 and Neutrophil-Like HL-60 Cells Associated with the Activation of Apoptotic Pathways
- Author
-
Mohammad A. K. Azad, Qi Tony Zhou, Jian Li, Ahmed M. Fathalla, Seong Hoong Chow, Tony Velkov, and Thomas Naderer
- Subjects
Programmed cell death ,medicine.drug_class ,Neutrophils ,THP-1 Cells ,Polymyxin ,Apoptosis ,HL-60 Cells ,Mitochondrion ,Fas ligand ,03 medical and health sciences ,medicine ,Humans ,Pharmacology (medical) ,Polymyxins ,Caspase ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Innate immune system ,biology ,030306 microbiology ,Chemistry ,Macrophages ,Molecular biology ,Infectious Diseases ,biology.protein ,Polymyxin B ,medicine.drug - Abstract
Innate immunity is crucial for the host to defend against infections, and understanding the effect of polymyxins on innate immunity is important for optimizing their clinical use. In this study, we investigated the potential toxicity of polymyxins on human macrophage-like THP-1 and neutrophil-like HL-60 cells. Differentiated THP-1 human macrophages (THP-1-dMs) and HL-60 human neutrophils (HL-60-dNs) were employed. Flow cytometry was used to measure the concentration-dependent effects (100 to 2,500 μM for THP-1-dMs and 5 to 2,500 μM for HL-60-dNs) and time-dependent effects (1,000 μM for THP-1-dMs and 300 μM for HL-60-dNs) of polymyxin B over 24 h. Effects of polymyxin B on mitochondrial activity, activation of caspase-3, caspase-8, and caspase-9, and Fas ligand (FasL) expression in both cell lines were examined using fluorescence imaging, colorimetric, and fluorometric assays. In both cell lines, polymyxin B induced concentration- and time-dependent loss of viability at 24 h with 50% effective concentration (EC(50)) values of 751.8 μM (95% confidence interval [CI], 692.1 to 816.6 μM; Hill slope, 3.09 to 5.64) for THP-1-dM cells and 175.4 μM (95% CI, 154.8 to 198.7 μM; Hill slope, 1.42 to 2.21) for HL-60-dN cells. A concentration-dependent loss of mitochondrial membrane potential and generation of mitochondrial superoxide was also observed. Polymyxin B-induced apoptosis was associated with concentration-dependent activation of all three tested caspases. The death receptor apoptotic pathway activation was demonstrated by a concentration-dependent increase of FasL expression. For the first time, our results reveal that polymyxin B induced concentration- and time-dependent cell death in human macrophage-like THP-1 and neutrophil-like HL-60 cells associated with mitochondrial and death receptor apoptotic pathways.
- Published
- 2020
21. In vitro evaluation of drug delivery behavior for inhalable amorphous nanoparticle formulations in a human lung epithelial cell model
- Author
-
Chune Zhu, Jianting Chen, Shihui Yu, Weisan Pan, Maizbha Uddin Ahmed, Qi Tony Zhou, Jian Li, and Tony Velkov
- Subjects
Pharmaceutical Science ,Cystic Fibrosis Transmembrane Conductance Regulator ,02 engineering and technology ,Pharmacology ,medicine.disease_cause ,030226 pharmacology & pharmacy ,Cystic fibrosis ,Article ,Ivacaftor ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Lung ,Polypeptide antibiotic ,biology ,Pseudomonas aeruginosa ,Chemistry ,Epithelial Cells ,021001 nanoscience & nanotechnology ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,Pharmaceutical Preparations ,Drug delivery ,Toxicity ,Colistin ,biology.protein ,Nanoparticles ,0210 nano-technology ,medicine.drug - Abstract
Respiratory tract infections caused by multidrug-resistant (MDR) Gram-negative bacteria such as Pseudomonas aeruginosa are serious burdens to public health, especially in cystic fibrosis patients. The combination of colistin, a cationic polypeptide antibiotic, and ivacaftor, a cystic fibrosis transmembrane regulator (CFTR) protein modulator, displays a synergistic antibacterial effect against P. aeruginosa. The primary aim of the present study is to investigate the transport, accumulation and toxicity of a novel nanoparticle formulation containing colistin and ivacaftor in lung epithelial Calu-3 cells. The cell viability results demonstrated that ivacaftor alone or in combination with colistin in the physical mixture showed significant toxicity at an ivacaftor concentration of 10 μg/mL or higher. However, the cellular toxicity was significantly reduced in the nanoparticle formulation. Ivacaftor transport into the cells reached a plateau rapidly as compared to colistin. Colistin transport across the Calu-3 cell monolayer was less than ivacaftor. A substantial amount (46-83%) of ivacaftor, independent of dose, was accumulated in the cell monolayer following transport from the apical into the basal chamber, whereas the intracellular accumulation of colistin was relatively low (2-15%). The nanoparticle formulation significantly reduced the toxicity of colistin and ivacaftor to Calu-3 cells by reducing the accumulation of both drugs in the cell and potential protective effects by bovine serum albumin (BSA), which could be a promising safer option for the treatment of respiratory infections caused by MDR P. aeruginosa.
- Published
- 2020
22. Acinetobacter baumannii: Polymyxins Bind to the Cell Surface of Unculturable Acinetobacter baumannii and Cause Unique Dependent Resistance (Adv. Sci. 15/2020)
- Author
-
Yu-Wei Lin, Christopher K. Barlow, Lushan Wang, Heidi H. Yu, Nitin A. Patil, Mohammad Abul Kalam Azad, Rhys A. Dunstan, Yang Hu, Falk Schreiber, Björn Sommer, Ke Chen, Mei-Ling Han, Jiping Wang, Bin Gong, Xukai Jiang, Jian Li, Jing Fu, Trevor Lithgow, Darren J. Creek, Weifeng Li, Jinxin Zhao, Tony Velkov, John D. Boyce, Yan Zhu, Jing Lu, and Elena K Schneider-Futschik
- Subjects
biology ,medicine.drug_class ,Chemistry ,General Chemical Engineering ,Polymyxin ,Cell ,General Engineering ,General Physics and Astronomy ,Medicine (miscellaneous) ,biology.organism_classification ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Acinetobacter baumannii ,Microbiology ,medicine.anatomical_structure ,medicine ,Cover Picture ,General Materials Science ,lipids (amino acids, peptides, and proteins) - Abstract
In article number 2000704, Jian Li and co‐workers elucidate the novel mechanism of dependent resistance to the last‐line polymyxins in a high‐priority “superbug” Acinetobacter baumannii. Polymyxindependent resistance cannot be detected by conventional microbiological diagnosis due to the lack of growth on normal agar plates. These polymyxin‐dependent Acinetobacter baumannii isolates utilize the rigid polymyxin molecules as sticking plasters to stabilize their fragile lipopolysaccharide‐deficient and phosphatidylglycerol‐rich outer membrane. [Image: see text]
- Published
- 2020
23. Structure-Interaction Relationship of Polymyxins with the Membrane of Human Kidney Proximal Tubular Cells
- Author
-
Philip E. Thompson, Lin Wan, Shuo Zhang, Mohammad A. K. Azad, Kade D. Roberts, Tony Velkov, Jingliang Li, Kai Yang, Bing Yuan, Hemayet Uddin, Jian Li, Xukai Jiang, Jing Fu, Lushan Wang, and Bin Gong
- Subjects
0301 basic medicine ,Cellular membrane ,medicine.drug_class ,Polymyxin ,030106 microbiology ,Kidney ,Article ,Nephrotoxicity ,Cell membrane ,03 medical and health sciences ,Drug Resistance, Multiple, Bacterial ,medicine ,Humans ,In patient ,Polymyxins ,Chemistry ,Human kidney ,Epithelial Cells ,Anti-Bacterial Agents ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Membrane ,Colistin ,Biophysics ,lipids (amino acids, peptides, and proteins) ,medicine.drug - Abstract
Multidrug-resistant Gram-negative bacteria are a serious global threat to human health. Polymyxins are increasingly used in patients as a last-line therapy to treat infections caused by these life-threatening ‘superbugs’. Unfortunately, polymyxin-induced nephrotoxicity is the major dose-limiting factor and understanding its mechanism is crucial for the development of novel, safer polymyxins. Here, we undertook the first all-atom molecular dynamics simulations of the interaction between four naturally occurring polymyxins A(1), B(1), M(1) and colistin A (representative structural variations of the polymyxin core structure) and the membrane of human kidney proximal tubular cells. All polymyxins inserted spontaneously into the hydrophobic region of the membrane where they were retained, although their insertion abilities varied. Polymyxin A(1) completely penetrated into the hydrophobic region of the membrane with a unique folded conformation, whereas the other three polymyxins only inserted their fatty acyl tails into this region. Furthermore, local membrane defects and increased water penetration were induced by each polymyxin, which may represent the initial stage of cellular membrane damage. Finally, the structure-interaction relationship of polymyxins was investigated based on atomic interactions at the cell membrane level. The hydrophobicity at positions 6/7 and stereochemistry at position 3 regulated the interactions of polymyxins with the cell membrane. Collectively, our results provide new mechanistic insights into polymyxin-induced nephrotoxicity at the atomic level and will facilitate the development of new-generation polymyxins.
- Published
- 2020
24. The Killing Mechanism of Teixobactin against Methicillin-Resistant Staphylococcus aureus: an Untargeted Metabolomics Study
- Author
-
John A. Karas, Tony Velkov, Maytham Hussein, Olivia K. A. Paulin, Mark Baker, Jian Li, James D. Swarbrick, Daniel Hoyer, Yan Zhu, Fan Chen, and Elena K Schneider-Futschik
- Subjects
0301 basic medicine ,Modern medicine ,Physiology ,Teixobactin ,lcsh:QR1-502 ,methicillin-resistant Staphylococcus aureus ,medicine.disease_cause ,01 natural sciences ,Biochemistry ,Microbiology ,lcsh:Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,antimicrobial peptides ,Genetics ,medicine ,antimicrobial resistance ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Antibacterial agent ,Teichoic acid ,Lipid II ,010405 organic chemistry ,Therapeutics and Prevention ,metabolomics ,QR1-502 ,0104 chemical sciences ,Computer Science Applications ,030104 developmental biology ,chemistry ,Staphylococcus aureus ,Modeling and Simulation ,solid-phase peptide synthesis ,teixobactin ,Peptidoglycan ,Cell envelope ,Research Article - Abstract
Antimicrobial resistance is one of the greatest threats to the global health system. It is imperative that new anti-infective therapeutics be developed against problematic “superbugs.” The cyclic depsipeptide teixobactin holds much promise as a new class of antibiotics for highly resistant Gram-positive pathogens (e.g., methicillin-resistant Staphylococcus aureus [MRSA]). Understanding its molecular mechanism(s) of action could lead to the design of new compounds with a broader activity spectrum. Here, we describe the first metabolomics study to investigate the killing mechanism(s) of teixobactin against MRSA. Our findings revealed that teixobactin significantly disorganized the bacterial cell envelope, as reflected by a profound perturbation in the bacterial membrane lipids and cell wall biosynthesis (peptidoglycan and teichoic acid). Importantly, teixobactin significantly suppressed the main intermediate d-alanyl-d-lactate involved in the mechanism of vancomycin resistance in S. aureus. These novel results help explain the unique mechanism of action of teixobactin and its lack of cross-resistance with vancomycin., Antibiotics have served humankind through their use in modern medicine as effective treatments for otherwise fatal bacterial infections. Teixobactin is a first member of newly discovered natural antibiotics that was recently identified from a hitherto-unculturable soil bacterium, Eleftheria terrae, and recognized as a potent antibacterial agent against various Gram-positive bacteria, including methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci. The most distinctive characteristic of teixobactin as an effective antibiotic is that teixobactin resistance could not be evolved in a laboratory setting. It is purported that teixobactin’s “resistance-resistant” mechanism of action includes binding to the essential bacterial cell wall synthesis building blocks lipid II and lipid III. In the present study, metabolomics was used to investigate the potential metabolic pathways involved in the mechanisms of antibacterial activity of the synthetic teixobactin analogue Leu10-teixobactin against a MRSA strain, S. aureus ATCC 700699. The metabolomes of S. aureus ATCC 700699 cells 1, 3, and 6 h following treatment with Leu10-teixobactin (0.5 μg/ml, i.e., 0.5× MIC) were compared to those of the untreated controls. Leu10-teixobactin significantly perturbed bacterial membrane lipids (glycerophospholipids and fatty acids), peptidoglycan (lipid I and II) metabolism, and cell wall teichoic acid (lipid III) biosynthesis as early as after 1 h of treatment, reflecting an initial activity on the cell envelope. Concordant with its time-dependent antibacterial killing action, Leu10-teixobactin caused more perturbations in the levels of key intermediates in pathways of amino-sugar and nucleotide-sugar metabolism and their downstream peptidoglycan and teichoic acid biosynthesis at 3 and 6 h. Significant perturbations in arginine metabolism and the interrelated tricarboxylic acid cycle, histidine metabolism, pantothenate, and coenzyme A biosynthesis were also observed at 3 and 6 h. To conclude, this is the first study to provide novel metabolomics mechanistic information, which lends support to the development of teixobactin as an antibacterial drug for the treatment of multidrug-resistant Gram-positive infections. IMPORTANCE Antimicrobial resistance is one of the greatest threats to the global health system. It is imperative that new anti-infective therapeutics be developed against problematic “superbugs.” The cyclic depsipeptide teixobactin holds much promise as a new class of antibiotics for highly resistant Gram-positive pathogens (e.g., methicillin-resistant Staphylococcus aureus [MRSA]). Understanding its molecular mechanism(s) of action could lead to the design of new compounds with a broader activity spectrum. Here, we describe the first metabolomics study to investigate the killing mechanism(s) of teixobactin against MRSA. Our findings revealed that teixobactin significantly disorganized the bacterial cell envelope, as reflected by a profound perturbation in the bacterial membrane lipids and cell wall biosynthesis (peptidoglycan and teichoic acid). Importantly, teixobactin significantly suppressed the main intermediate d-alanyl-d-lactate involved in the mechanism of vancomycin resistance in S. aureus. These novel results help explain the unique mechanism of action of teixobactin and its lack of cross-resistance with vancomycin.
- Published
- 2020
25. Allosteric folding correction of F508del and rare CFTR mutants by elexacaftor-tezacaftor-ivacaftor (Trikafta) combination
- Author
-
Tony Velkov, Dillon F. Da Fonte, Saul Frenkiel, Elias Matouk, Guido Veit, Gergely L. Lukacs, Mark A. Hancock, Ariel Roldan, Maytham Hussein, and Haijin Xu
- Subjects
0301 basic medicine ,Protein Folding ,Indoles ,Cystic Fibrosis ,Pulmonology ,Pyridines ,Mutant ,Allosteric regulation ,Cystic Fibrosis Transmembrane Conductance Regulator ,Bronchi ,Aminophenols ,Cystic fibrosis ,Ivacaftor ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Benzodioxoles ,Cells, Cultured ,Chloride channels ,Chemistry ,General Medicine ,Cell Biology ,Potentiator ,medicine.disease ,Molecular biology ,Transmembrane protein ,3. Good health ,Drug Combinations ,030104 developmental biology ,Cyclic nucleotide-binding domain ,030220 oncology & carcinogenesis ,Mutation ,Chloride channel ,Quinolines ,Medicine ,Pyrazoles ,Drug therapy ,Epithelial transport of ions and water ,medicine.drug ,Research Article - Abstract
Based on its clinical benefits, Trikafta — the combination of folding correctors VX-661 (tezacaftor), VX-445 (elexacaftor), and the gating potentiator VX-770 (ivacaftor) — was FDA approved for treatment of patients with cystic fibrosis (CF) carrying deletion of phenylalanine at position 508 (F508del) of the CF transmembrane conductance regulator (CFTR) on at least 1 allele. Neither the mechanism of action of VX-445 nor the susceptibility of rare CF folding mutants to Trikafta are known. Here, we show that, in human bronchial epithelial cells, VX-445 synergistically restores F508del-CFTR processing in combination with type I or II correctors that target the nucleotide binding domain 1 (NBD1) membrane spanning domains (MSDs) interface and NBD2, respectively, consistent with a type III corrector mechanism. This inference was supported by the VX-445 binding to and unfolding suppression of the isolated F508del-NBD1 of CFTR. The VX-661 plus VX-445 treatment restored F508del-CFTR chloride channel function in the presence of VX-770 to approximately 62% of WT CFTR in homozygous nasal epithelia. Substantial rescue of rare misprocessing mutations (S13F, R31C, G85E, E92K, V520F, M1101K, and N1303K), confined to MSD1, MSD2, NBD1, and NBD2 of CFTR, was also observed in airway epithelia, suggesting an allosteric correction mechanism and the possible application of Trikafta for patients with rare misfolding mutants of CFTR., Trikafta, the combination of type I corrector VX-661, type III corrector VX-445, and the potentiator VX-770, may be applied for various CFTR folding mutants.
- Published
- 2020
26. Sialylation of Asparagine 612 inhibits Aconitase activity during mouse sperm capacitation; A possible mechanism for the switch from oxidative phosphorylation to glycolysis
- Author
-
Mark Baker, Louise Hetherington, Jacob Netherton, Rachel A Ogle, Tony Velkov, Ana Izabel Silva Balbin Villaverde, Vincenzo Carbone, and Peter J. Lewis
- Subjects
Citric acid cycle ,chemistry.chemical_compound ,Proteases ,chemistry ,Capacitation ,ACO2 ,Asparagine ,Oxidative phosphorylation ,Aconitase ,Sialic acid ,Cell biology - Abstract
After ejaculation, mammalian spermatozoa must undergo a process known as capacitation in order to successfully fertilize the oocyte. Several post-translational modifications occur during capacitation, including sialylation, which despite being limited to a few proteins, seems to be essential for proper sperm-oocyte interaction. Regardless of its importance, to date, no single study has ever identified nor quantified which glycoproteins bearing terminal sialic acid (Sia) are altered during capacitation. Here we characterize sialylation during mouse sperm capacitation. Using tandem mass spectrometry coupled with liquid chromatography (LC-MS/MS), we found 142 non-reductant peptides, with 9 of them showing potential modifications on their sialylated oligosaccharides during capacitation. As such, N-linked sialoglycopeptides from C4b-binding protein, endothelial lipase (EL), serine proteases 39 and 52, testis-expressed protein 101 and zonadhesin were reduced following capacitation. In contrast, mitochondrial aconitate hydratase (aconitase; ACO2) was the only protein to show an increase in Sia content during capacitation. Interestingly, while the loss of Sia within EL (N62) was accompanied by a reduction in its phospholipase A1activity, the increase of sialylation in the ACO2 (N612) also resulted in a decrease of the activity of this TCA cycle enzyme. The latter was confirmed by N612D recombinant protein with both His and GFP tag, in which the N612D mutant had no activity compared to WT when protein. Computer modelling show that N612 sits atop the catalytic site of ACO2. The introduction of sialic acid causes a large confirmation change in the alpha helix, essentially, distorting the active site, leading to complete loss of function. These findings suggest that the switch from oxidative phosphorylation, over to glycolysis that occurs during capacitation may come about through sialylation of ACO2.
- Published
- 2020
- Full Text
- View/download PDF
27. Sialylation of Asparagine 612 Inhibits Aconitase Activity during Mouse Sperm Capacitation; a Possible Mechanism for the Switch from Oxidative Phosphorylation to Glycolysis
- Author
-
Jacob Netherton, Tony Velkov, Mark Baker, Ana Izabel Silva Balbin Villaverde, Peter J. Lewis, Vincenzo Carbone, and Rachel A Ogle
- Subjects
Male ,Oxidative phosphorylation ,Biochemistry ,Aconitase ,Oxidative Phosphorylation ,Analytical Chemistry ,Serine ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Capacitation ,Tandem Mass Spectrometry ,Aspartic acid ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,Glycoproteins ,Aconitate Hydratase ,0303 health sciences ,Chemistry ,Research ,030302 biochemistry & molecular biology ,ACO2 ,Lipase ,Immunohistochemistry ,Spermatozoa ,N-Acetylneuraminic Acid ,Cell biology ,Sialic acid ,Citric acid cycle ,Molecular Docking Simulation ,HEK293 Cells ,Asparagine ,Acrosome ,Glycolysis ,Protein Processing, Post-Translational ,Sperm Capacitation ,Chromatography, Liquid - Abstract
After ejaculation, mammalian spermatozoa must undergo a process known as capacitation in order to successfully fertilize the oocyte. Several post-translational modifications occur during capacitation, including sialylation, which despite being limited to a few proteins, seems to be essential for proper sperm-oocyte interaction. Regardless of its importance, to date, no single study has ever identified nor quantified which glycoproteins bearing terminal sialic acid (Sia) are altered during capacitation. Here we characterize sialylation during mouse sperm capacitation. Using tandem MS coupled with liquid chromatography (LC–MS/MS), we found 142 nonreductant peptides, with 9 of them showing potential modifications on their sialylated oligosaccharides during capacitation. As such, N-linked sialoglycopeptides from C4b-binding protein, endothelial lipase (EL), serine proteases 39 and 52, testis-expressed protein 101 and zonadhesin were reduced following capacitation. In contrast, mitochondrial aconitate hydratase (aconitase; ACO2), a TCA cycle enzyme, was the only protein to show an increase in Sia content during capacitation. Interestingly, although the loss of Sia within EL (N62) was accompanied by a reduction in its phospholipase A(1) activity, a decrease in the activity of ACO2 (i.e. stereospecific isomerization of citrate to isocitrate) occurred when sialylation increased (N612). The latter was confirmed by N612D recombinant protein tagged with both His and GFP. The replacement of Sia for the negatively charged Aspartic acid in the N612D mutant caused complete loss of aconitase activity compared with the WT. Computer modeling show that N612 sits atop the catalytic site of ACO2. The introduction of Sia causes a large conformational change in the alpha helix, essentially, distorting the active site, leading to complete loss of function. These findings suggest that the switch from oxidative phosphorylation, over to glycolysis that occurs during capacitation may come about through sialylation of ACO2.
- Published
- 2020
28. Regulating polymyxin resistance in Gram-negative bacteria: roles of two-component systems PhoPQ and PmrAB
- Author
-
Jian Li, Yan Zhu, Jiayuan Huang, Lushan Wang, Chen Li, Jiangning Song, and Tony Velkov
- Subjects
Microbiology (medical) ,Gram-negative bacteria ,medicine.drug_class ,Polymyxin ,Drug resistance ,Review ,Microbiology ,Lipid A ,Bacterial Proteins ,Drug Resistance, Bacterial ,Gram-Negative Bacteria ,medicine ,Polymyxin B ,biology ,Chemistry ,Colistin ,biology.organism_classification ,Two-component regulatory system ,Anti-Bacterial Agents ,Mutation ,lipids (amino acids, peptides, and proteins) ,HAMP ,medicine.drug ,Transcription Factors - Abstract
Polymyxins (polymyxin B and colistin) are last-line antibiotics against multidrug-resistant Gram-negative pathogens. Polymyxin resistance is increasing worldwide, with resistance most commonly regulated by two-component systems such as PmrAB and PhoPQ. This review discusses the regulatory mechanisms of PhoPQ and PmrAB in mediating polymyxin resistance, from receiving an external stimulus through to activation of genes responsible for lipid A modifications. By analyzing the reported nonsynonymous substitutions in each two-component system, we identified the domains that are critical for polymyxin resistance. Notably, for PmrB 71% of resistance-conferring nonsynonymous mutations occurred in the HAMP (present in histidine kinases, adenylate cyclases, methyl accepting proteins and phosphatase) linker and DHp (dimerization and histidine phosphotransfer) domains. These results enhance our understanding of the regulatory mechanisms underpinning polymyxin resistance and may assist with the development of new strategies to minimize resistance emergence.
- Published
- 2020
29. Polymyxin Triple Combinations against Polymyxin-Resistant, Multidrug-Resistant, KPC-Producing Klebsiella pneumoniae
- Author
-
Ilias Karaiskos, Yu-Wei Lin, Brian T. Tsuji, Ke Chen, Su Mon Aye, Irene Galani, Jinxin Zhao, Hasini Wickremasinghe, Jian Li, Helen Giamarellou, Jiping Wang, Tony Velkov, Heidi Yu, and Phillip J. Bergen
- Subjects
Klebsiella pneumoniae ,medicine.drug_class ,Polymyxin ,Antibiotics ,Microbial Sensitivity Tests ,Meropenem ,beta-Lactamases ,Microbiology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Drug Resistance, Multiple, Bacterial ,medicine ,Animals ,Humans ,Pharmacology (medical) ,030212 general & internal medicine ,Polymyxins ,Polymyxin B ,Pharmacology ,0303 health sciences ,biology ,030306 microbiology ,Chemistry ,Drug Synergism ,Minocycline ,biology.organism_classification ,Anti-Bacterial Agents ,Multiple drug resistance ,Infectious Diseases ,Amikacin ,medicine.drug - Abstract
Resistance to polymyxin antibiotics is increasing. Without new antibiotic classes, combination therapy is often required. We systematically investigated bacterial killing with polymyxin-based combinations against multidrug-resistant (including polymyxin-resistant), carbapenemase-producing Klebsiella pneumoniae. Monotherapies and double- and triple-combination therapies were compared to identify the most efficacious treatment using static time-kill studies (24 h, six isolates), an in vitro pharmacokinetic/pharmacodynamic model (IVM; 48 h, two isolates), and the mouse thigh infection model (24 h, six isolates). In static time-kill studies, all monotherapies (polymyxin B, rifampin, amikacin, meropenem, or minocycline) were ineffective. Initial bacterial killing was enhanced with various polymyxin B-containing double combinations; however, substantial regrowth occurred in most cases by 24 h. Most polymyxin B-containing triple combinations provided greater and more sustained killing than double combinations. Standard dosage regimens of polymyxin B (2.5 mg/kg of body weight/day), rifampin (600 mg every 12 h), and amikacin (7.5 mg/kg every 12 h) were simulated in the IVM. Against isolate ATH 16, no viable bacteria were detected across 5 to 25 h with triple therapy, with regrowth to ∼2-log(10) CFU/ml occurring at 48 h. Against isolate BD 32, rapid initial killing of ∼3.5-log(10) CFU/ml at 5 h was followed by a slow decline to ∼2-log(10) CFU/ml at 48 h. In infected mice, polymyxin B monotherapy (60 mg/kg/day) generally was ineffective. With triple therapy (polymyxin B at 60 mg/kg/day, rifampin at 120 mg/kg/day, and amikacin at 300 mg/kg/day), at 24 h there was an ∼1.7-log(10) CFU/thigh reduction compared to the starting inoculum for all six isolates. Our results demonstrate that the polymyxin B-rifampin-amikacin combination significantly enhanced in vitro and in vivo bacterial killing, providing important information for the optimization of polymyxin-based combinations in patients.
- Published
- 2020
30. Chloroquine ameliorates carbon tetrachloride-induced acute liver injury in mice via the concomitant inhibition of inflammation and induction of apoptosis
- Author
-
Tony Velkov, Ying Wang, Chongshan Dai, Shusheng Tang, Xilong Xiao, Sun Tun, and Daowen Li
- Subjects
0301 basic medicine ,Cancer Research ,Immunology ,Aspartate transaminase ,CCL4 ,Stimulation ,Inflammation ,Apoptosis ,Pharmacology ,HMGB1 ,digestive system ,Article ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Mice ,parasitic diseases ,medicine ,Animals ,Humans ,HMGB1 Protein ,lcsh:QH573-671 ,Carbon Tetrachloride ,Liver injury ,biology ,business.industry ,Caspase 3 ,Tumor Necrosis Factor-alpha ,lcsh:Cytology ,NF-kappa B ,Chloroquine ,Cell Biology ,medicine.disease ,digestive system diseases ,3. Good health ,Oxidative Stress ,030104 developmental biology ,chemistry ,Gene Expression Regulation ,Liver ,biology.protein ,Carbon tetrachloride ,medicine.symptom ,Chemical and Drug Induced Liver Injury ,business ,Signal Transduction - Abstract
This is the first study to investigate the hepatoprotective effect of CQ on acute liver injury caused by carbon tetrachloride (CCl4) in a murine model and the underlying molecular mechanisms. Ninety-six mice were randomly divided into the control (n = 8), CQ (n = 8), CCl4 (n = 40), and CCl4 + CQ (n = 40) treatment groups. In the CCl4 group, mice were intraperitoneally (i.p) injected with 0.3% CCl4 (10 mL/kg, dissolved in olive oil); in the CCl4 + CQ group, mice were i.p injected with CQ at 50 mg/kg at 2, 24, and 48 h before CCl4 administration. The mice in the control and CQ groups were administered with an equal vehicle or CQ (50 mg/kg). Mice were killed at 2, 6, 12, 24, 48 h post CCl4 treatment and their livers were harvested for analysis. The results showed that CQ pre-treatment markedly inhibited CCl4-induced acute liver injury, which was evidenced by decreased serum transaminase, aspartate transaminase and lower histological scores of liver injury. CQ pretreatment downregulated the CCl4-induced hepatic tissue expression of high-mobility group box 1 (HMGB1) and the levels of serum HMGB1 as well as IL-6 and TNF-α. Furthermore, CQ pre-treatment inhibited autophagy, downregulated NF-kB expression, upregulated p53 expression, increased the ratio of Bax/Bcl-2, and increased the activation of caspase-3 in hepatic tissue. This is the first study to demonstrate that CQ ameliorates CCl4-induced acute liver injury via the inhibition of HMGB1-mediated inflammatory responses and the stimulation of pro-apoptotic pathways to modulate the apoptotic and inflammatory responses associated with progress of liver damage.
- Published
- 2018
31. Exploiting Macromolecular Design To Optimize the Antibacterial Activity of Alkylated Cationic Oligomers
- Author
-
John F. Quinn, Mark E. Cooper, Elena K Schneider-Futschik, Maite Amado, Michael R. Whittaker, Nghia P. Truong, Jian Li, Thomas P. Davis, Alysha G. Elliott, James L. Grace, and Tony Velkov
- Subjects
Alkylating Agents ,Alkylation ,Polymers and Plastics ,Macromolecular Substances ,Polymers ,Bioengineering ,Microbial Sensitivity Tests ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Oligomer ,Polymerization ,Biomaterials ,Structure-Activity Relationship ,chemistry.chemical_compound ,Cations ,Materials Chemistry ,Acrylate ,Molecular Structure ,Diethylene glycol ,Cationic polymerization ,021001 nanoscience & nanotechnology ,Combinatorial chemistry ,Anti-Bacterial Agents ,0104 chemical sciences ,Acrylates ,chemistry ,Ethyl acrylate ,0210 nano-technology ,Antibacterial activity ,Ethylene glycol ,Antimicrobial Cationic Peptides - Abstract
There is growing interest in synthetic polymers which co-opt the structural features of naturally occurring antimicrobial peptides. However, our understanding of how macromolecular architecture affects antibacterial activity remains limited. To address this, we investigated whether varying architectures of a series of block and statistical co-oligomers influenced antibacterial and hemolytic activity. Cu(0)-mediated polymerization was used to synthesize oligomers constituting 2-(Boc-amino)ethyl acrylate units and either diethylene glycol ethyl ether acrylate (DEGEEA) or poly(ethylene glycol) methyl ether acrylate units with varying macromolecular architecture; subsequent deprotection produced primary amine functional oligomers. Further guanylation provided an additional series of antimicrobial candidates. Both chemical composition and macromolecular architecture were shown to affect antimicrobial activity. A broad spectrum antibacterial oligomer (containing guanidine moieties and DEGEEA units) was identified that possessed promising activity (MIC = 2 μg mL-1) toward both Gram-negative and Gram-positive bacteria. Bacterial membrane permeabilization was identified as an important contributor to the mechanism of action.
- Published
- 2018
32. T-2 toxin-induced toxicity in neuroblastoma-2a cells involves the generation of reactive oxygen, mitochondrial dysfunction and inhibition of Nrf2/HO-1 pathway
- Author
-
Tony Velkov, Shusheng Tang, Ying Wang, Chongshan Dai, and Xiya Zhang
- Subjects
0301 basic medicine ,NF-E2-Related Factor 2 ,DNA damage ,Apoptosis ,Mitochondrion ,Pharmacology ,Toxicology ,medicine.disease_cause ,Mice ,Neuroblastoma ,03 medical and health sciences ,0404 agricultural biotechnology ,Cell Line, Tumor ,medicine ,Animals ,Humans ,chemistry.chemical_classification ,Reactive oxygen species ,Chemistry ,Toxin ,Neurotoxicity ,04 agricultural and veterinary sciences ,General Medicine ,medicine.disease ,040401 food science ,Mitochondria ,Oxidative Stress ,T-2 Toxin ,030104 developmental biology ,Cell culture ,Reactive Oxygen Species ,Heme Oxygenase-1 ,Oxidative stress ,Food Science - Abstract
The molecular mechanisms of T-2 mycotoxin induced neurotoxicity remains enigmatic. In the present study we show that T-2 toxin induced neurotoxicity in mouse neuroblastoma2a (N2a) cells is both dose- and time-dependent and is associated with oxidative stress, mitochondrial dysfunction and apoptosis. T-2 toxin treatment of N2a cells at 10, 20, 40 and 80 ng/mL for 24 h significantly up-regulated the mRNA expression of p53, Bax, and caspase-8 and down-regulated the expression of Nrf2 and HO-1 mRNA and protein expression. Activation of caspases-8, -9 and -3 was also evident in a concentration-dependent manner. Pre-treatment of the cells with the antioxidant N-acetyl-cysteine markedly suppressed T-2 toxin-induced neurotoxicity and caspase activation. Conversely, pre-treatment of the cells with the Nrf2 inhibitor brusatol or the HO-1 inhibitor zinc protoporphyrin IX, enhanced T-2 toxin induced neurotoxicity and increased the activation of caspase-9 and -3. Taken together, these novel findings suggest that T-2 toxin-induced neurotoxicity in N2a cells involves oxidative stress, mitochondrial dysfunction and apoptosis via the inhibition of the Nrf2/HO-1 and activation of p53 pathway. The present study highlights the potential of developing much needed pharmacological interventions to prevent T-2 toxin neurotoxicity.
- Published
- 2018
33. Sputum Active Polymyxin Lipopeptides: Activity against Cystic Fibrosis Pseudomonas aeruginosa Isolates and Their Interactions with Sputum Biomolecules
- Author
-
John A. Karas, Daniel Hoyer, Tony Velkov, Kade D. Roberts, Olivia K. A. Paulin, Elena K Schneider-Futschik, James Ziogas, Mark Baker, and Jian Li
- Subjects
0301 basic medicine ,Cystic Fibrosis ,medicine.drug_class ,Polymyxin ,030106 microbiology ,Microbial Sensitivity Tests ,polymyxins ,medicine.disease_cause ,biofilm ,Microbiology ,Surface-Active Agents ,03 medical and health sciences ,Minimum inhibitory concentration ,chemistry.chemical_compound ,Drug Resistance, Multiple, Bacterial ,medicine ,Tobramycin ,Humans ,mucoid “biofilm” growth ,Polymyxin B ,Pseudomonas aeruginosa ,Mucins ,Sputum ,Lipopeptide ,Featured Article ,P. aeruginosa ,lipopeptides ,Actins ,Anti-Bacterial Agents ,nonmucoid “planktonic” growth ,030104 developmental biology ,Infectious Diseases ,chemistry ,Biofilms ,Colistin ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,Protein Binding ,medicine.drug - Abstract
The mucoid biofilm mode of growth of Pseudomonas aeruginosa (P. aeruginosa) in the lungs of cystic fibrosis patients makes eradication of infections with antibiotic therapy very difficult. The lipopeptide antibiotics polymyxin B and colistin are currently the last-resort therapies for infections caused by multidrug-resistant P. aeruginosa. In the present study, we investigated the antibacterial activity of a series of polymyxin lipopeptides (polymyxin B, colistin, FADDI-003, octapeptin A3, and polymyxin A2) against a panel of polymyxin-susceptible and polymyxin-resistant P. aeruginosa cystic fibrosis isolates grown under planktonic or biofilm conditions in artificial sputum and their interactions with sputum component biomolecules. In sputum media under planktonic conditions, the lipopeptides FADDI-003 and octapeptin A3 displayed very promising activity against the polymyxin-resistant isolate FADDI-PA066 (polymyxin B minimum inhibitory concentration (MIC) = 32 mg/L), while retaining their activity against the polymyxin-sensitive strains FADDI-PA021 (polymyxin B MIC = 1 mg/L) and FADDI-PA020 (polymyxin B MIC = 2 mg/L). Polymyxin A2 was only effective against the polymyxin-sensitive isolates. However, under biofilm growth conditions, the hydrophobic lipopeptide FADDI-003 was inactive compared to the more hydrophilic lipopeptides, octapeptin A3, polymyxin A2, polymyxin B, and colistin. Transmission electron micrographs revealed octapeptin A3 caused reduction in the cell numbers in biofilm as well as biofilm disruption/“antibiofilm” activity. We therefore assessed the interactions of the lipopeptides with the component sputum biomolecules, mucin, deoxyribonucleic acid (DNA), surfactant, F-actin, lipopolysaccharide, and phospholipids. We observed the general trend that sputum biomolecules reduce lipopeptide antibacterial activity. Collectively, our data suggests that, in the airways, lipopeptide binding to component sputum biomolecules may reduce antibacterial efficacy and is dependent on the physicochemical properties of the lipopeptide.
- Published
- 2018
34. The inhibitory effects of eighteen front-line antibiotics on the substrate uptake mediated by human Organic anion/cation transporters, Organic anion transporting polypeptides and Oligopeptide transporters in in vitro models
- Author
-
Qi Tony Zhou, Xiaoxi Lu, Ling Zhu, Jian Li, Tony Velkov, Hak-Kim Chan, Fanfan Zhou, Ting Chan, and Xiaofeng Bao
- Subjects
Anions ,0301 basic medicine ,Organic anion transporter 1 ,medicine.drug_class ,Antibiotics ,Organic Anion Transporters ,Pharmaceutical Science ,030226 pharmacology & pharmacy ,Trimethoprim ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Sulfadiazine ,Ciprofloxacin ,Cations ,medicine ,Humans ,Drug Interactions ,Oligopeptide ,biology ,Chemistry ,Biological Transport ,Transporter ,In vitro ,Anti-Bacterial Agents ,Kinetics ,HEK293 Cells ,Methotrexate ,030104 developmental biology ,Biochemistry ,biology.protein ,Peptides ,Organic anion ,medicine.drug - Abstract
Human Organic anion/cation transporters (OATs/OCTs), Organic anion transporting polypeptides (OATPs) and proton-coupled Oligopeptide transporters (PepTs) are important membrane transporters responsible of the cellular influx of drugs in many human key tissues. Inhibitor(s) impacting on the cellular uptake of transporter drug substrates is one of the primary causes of drug-drug interactions that lead to unsatisfied therapeutic outcomes and/or unwanted side effects. In the current study, we selected eighteen antibiotic agents used in infectious disease treatment and comprehensively evaluated their inhibitory effects on the substrate uptake mediated through the essential OATs/OCTs, OATPs and PepTs isoforms. Transport functional assay, dose-response curve and kinetic analysis were performed on the HEK293 cells over-expressing each of these transporter genes. Our data revealed that nitrofurantoin, sulfadiazine and metronidazole significantly inhibited the transport activity of OAT3 (IC50 values of 6.23±1.33μM, 6.65±1.30μM and 6.51±0.99μM; Ki values of 5.86μM, 3.98μM and 6.48μM, respectively). Trimethoprim and ciprofloxacin potently decreased the substrate uptake mediated via OATP1A2 (IC50 values of 9.35±1.10μM and 9.25±1.18μM; Ki values of 8.19μM and 7.64μM, respectively). In addition, these antibiotic agents consistently decreased methotrexate influx via OAT3 and OATP1A2. In summary, our study is the first to show that nitrofurantoin, sulfadiazine and metronidazole are potent inhibitors of OAT3 and trimethoprim is a novel inhibitor of OATP1A2. Our study also provides new evidence for the drug-drug interactions of ciprofloxacin with OATP1A2 drug substrates like methotrexate. Therefore, precautions are required when co-administering these antibiotics with OAT3 or OATP1A2 drug substrates.
- Published
- 2018
35. Mechanistic Insights From Global Metabolomics Studies into Synergistic Bactericidal Effect of a Polymyxin B Combination With Tamoxifen Against Cystic Fibrosis MDR Pseudomonas aeruginosa
- Author
-
Yu-Wei Lin, Xiaohan Hu, Tony Velkov, Jian Li, Maytham Hussein, Darren J. Creek, Yan Zhu, Dovile Anderson, Elena K Schneider-Futschik, Daniel Hoyer, Qi Tony Zhou, and Mei-Ling Han
- Subjects
0301 basic medicine ,Modern medicine ,Combination therapy ,medicine.drug_class ,Polymyxin ,lcsh:Biotechnology ,030106 microbiology ,Antibiotics ,Biophysics ,Pharmacology ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,Structural Biology ,lcsh:TP248.13-248.65 ,Genetics ,medicine ,Metabolome ,Chemistry ,Pseudomonas aeruginosa ,3. Good health ,Computer Science Applications ,030104 developmental biology ,Colistin ,lipids (amino acids, peptides, and proteins) ,Polymyxin B ,Biotechnology ,medicine.drug - Abstract
Polymyxins are amongst the most important antibiotics in modern medicine, in recent times their clinical utility has been overshadowed by nosocomial outbreaks of polymyxin resistant MDR Gram-negative 'superbugs'. An effective strategy to surmount polymyxin resistance is combination therapy with FDA-approved non-antibiotic drugs. Herein we used untargeted metabolomics to investigate the mechanism(s) of synergy between polymyxin B and the selective estrogen receptor modulator (SERM) tamoxifen against a polymyxin-resistant MDR cystic fibrosis (CF) Pseudomonas aeruginosa FADDI-PA006 isolate (polymyxin B MIC=8 mg/L , it is an MDR polymyxin resistant P. aeruginosa isolated from the lungs of a CF patient). The metabolome of FADDI-PA006 was profiled at 15 min, 1 and 4 h following treatment with polymyxin B (2 mg/L), tamoxifen (8 mg/L) either as monotherapy or in combination. At 15 min, the combination treatment induced a marked decrease in lipids, primarily fatty acid and glycerophospholipid metabolites that are involved in the biosynthesis of bacterial membranes. In line with the polymyxin-resistant status of this strain, at 1 h, both polymyxin B and tamoxifen monotherapies produced little effect on bacterial metabolism. In contrast to the combination which induced extensive reduction (≥ 1.0-log2-fold, p ≤ 0.05; FDR ≤ 0.05) in the levels of essential intermediates involved in cell envelope biosynthesis. Overall, these novel findings demonstrate that the primary mechanisms underlying the synergistic bactericidal effect of the combination against the polymyxin-resistant P. aeruginosa CF isolate FADDI-PA006 involves a disruption of the cell envelope biogenesis and an inhibition of aminoarabinose LPS modifications that confer polymyxin resistance.
- Published
- 2018
36. Polymyxin-Induced Lipid A Deacylation in Pseudomonas aeruginosa Perturbs Polymyxin Penetration and Confers High-Level Resistance
- Author
-
Tony Velkov, Seong Hoong Chow, Mei-Ling Han, Anton P. Le Brun, Yan Zhu, Samuel M. Moskowitz, Jian Li, Kade D. Roberts, Hsin-Hui Shen, and Alina D. Gutu
- Subjects
0301 basic medicine ,Lipopolysaccharide ,medicine.drug_class ,Acylation ,Polymyxin ,030106 microbiology ,Biology ,medicine.disease_cause ,Biochemistry ,Microbiology ,Lipid A ,Cell membrane ,03 medical and health sciences ,chemistry.chemical_compound ,Bacterial Proteins ,Drug Resistance, Bacterial ,medicine ,Polymyxins ,Pseudomonas aeruginosa ,Cell Membrane ,Amino Sugars ,General Medicine ,Anti-Bacterial Agents ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Colistin ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Bacterial outer membrane ,Polymyxin B ,medicine.drug - Abstract
Polymyxins are last-line antibiotics against life-threatening multidrug-resistant Gram-negative bacteria. Unfortunately, polymyxin resistance is increasingly reported, leaving a total lack of therapies. Using lipidomics and transcriptomics, we discovered that polymyxin B induced lipid A deacylation via pagL in both polymyxin-resistant and -susceptible Pseudomonas aeruginosa. Our results demonstrated that the deacylation of lipid A is an "innate immunity" response to polymyxins and a key compensatory mechanism to the aminoarabinose modification to confer high-level polymyxin resistance in P. aeruginosa. Furthermore, cutting-edge neutron reflectometry studies revealed that an assembled outer membrane (OM) with the less hydrophobic penta-acylated lipid A decreased polymyxin B penetration, compared to the hexa-acylated form. Polymyxin analogues with enhanced hydrophobicity displayed superior penetration into the tail regions of the penta-acylated lipid A OM. Our findings reveal a previously undiscovered mechanism of polymyxin resistance, wherein polymyxin-induced lipid A remodeling affects the OM packing and hydrophobicity, perturbs polymyxin penetration, and thereby confers high-level resistance.
- Published
- 2017
37. Plasma Protein Binding Structure–Activity Relationships Related to the N-Terminus of Daptomycin
- Author
-
Tony Velkov, Mark E. Cooper, Johnson Mak, Mei-Ling Han, Elena K. Schneider, Sue C Nang, Johnny X. Huang, Vincenzo Carbone, Yan Zhu, Jian Li, and Keith K. Khoo
- Subjects
Models, Molecular ,0301 basic medicine ,030106 microbiology ,Serum albumin ,Serum Albumin, Human ,Plasma protein binding ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Daptomycin ,polycyclic compounds ,medicine ,Humans ,chemistry.chemical_classification ,Binding Sites ,biology ,Chemistry ,Lipopeptide ,Fatty acid ,Hemopexin ,Blood Proteins ,Surface Plasmon Resonance ,Human serum albumin ,Blood proteins ,Molecular Docking Simulation ,030104 developmental biology ,Infectious Diseases ,Biochemistry ,alpha 1-Antitrypsin ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Protein Binding ,medicine.drug - Abstract
Daptomycin is a lipopeptide antibiotic that is highly bound to plasma proteins. To date, the plasma components and structure-activity relationships responsible for the plasma protein binding profile of daptomycin remain uncharacterized. In the present study we have employed a surface plasmon resonance assay together with molecular docking techniques to investigate the plasma protein binding structure-activity relationships related to the N-terminal fatty acyl of daptomycin. Three compounds were investigated: (1) native daptomycin, which displays an N-terminal n-decanoyl fatty acid side chain, and two analogues with modifications to the N-terminal fatty acyl chain; (2) des-acyl daptomycin; and (3) acetyl-daptomycin. The surface plasmon resonance (SPR) data showed that the binding profile of native daptomycin was in the rank order human serum albumin (HSA) ≫ α-1-antitrypsin > low-density lipoprotein ≥ hemoglobin > sex hormone binding globulin > α-1-acid-glycoprotein (AGP) > hemopexin > fibrinogen > α-2-macroglobulin > β2-microglobulin > high-density lipoprotein > fibronectin > haptoglobulin > transferrin > immunoglobulin G. Notably, binding to fatty acid free HSA was greater than binding to nondelipidated HSA. SPR and ultrafiltration studies also indicated that physiological concentrations of calcium increase binding of daptomycin and acetyl-daptomycin to HSA and AGP. A molecular model of the daptomycin-human serum albumin A complex is presented that illustrates the pivotal role of the N-terminal fatty acyl chain of daptomycin for binding to drug site 1 of HSA. In proof-of-concept, the capacity of physiological cocktails of the identified plasma proteins to inhibit the antibacterial activity of daptomycin was assessed with in vitro microbiological assays. We show that HSA, α-1-antitrypsin, low-density lipoprotein, sex hormone binding globulin, α-1-acid-glycoprotein, and hemopexin are responsible for the majority of the sequestering activity in human plasma. The findings are relevant to medicinal chemistry programs focused on the development of next-generation daptomycin lipopeptides. Tailored modifications to the N-terminal fatty acyl domain of the daptomycin molecule should yield novel daptomycin lipopeptides with more ideal plasma protein binding profiles to increase the levels of active (free) drug in plasma and improved in vivo activity.
- Published
- 2017
38. Curcumin Attenuates Colistin-Induced Neurotoxicity in N2a Cells via Anti-inflammatory Activity, Suppression of Oxidative Stress, and Apoptosis
- Author
-
Chongshan Dai, Daowen Li, Roberto Cappai, Tony Velkov, Giuseppe D. Ciccotosto, Sanlei Xie, Shusheng Tang, and Xilong Xiao
- Subjects
0301 basic medicine ,Curcumin ,Cell Survival ,Neuroscience (miscellaneous) ,Apoptosis ,Biology ,Pharmacology ,medicine.disease_cause ,Cell Line ,Superoxide dismutase ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,Kinase activity ,Colistin ,Anti-Inflammatory Agents, Non-Steroidal ,Neurotoxicity ,Glutathione ,medicine.disease ,Oxidative Stress ,030104 developmental biology ,Neurology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Oxidative stress ,Intracellular - Abstract
Neurotoxicity is an unwanted side-effect seen in patients receiving therapy with the "last-line" polymyxin antibiotics. This is the first study to show that colistin-induced neurotoxicity in neuroblastoma-2a (N2a) cells gives rise to an inflammatory response involving the IL-1β/p-IκB-α/NF-κB pathway. Pretreatment with curcumin at 5, 10, and 20 μM for 2 h prior to colistin (200 μM) exposure for 24 h, produced an anti-inflammatory effect by significantly down-regulating the expression of the pro-inflammatory mediators cyclooxygenase-2 (COX-2), phosphorylation of the inhibitor of nuclear factor-kappa B (NF-κB) (p-IκB)-α, and concomitantly NF-κB levels. Moreover, curcumin significantly decreased intracellular reactive oxygen species (ROS) production and increased the activities of the anti-ROS enzymes superoxide dismutase, catalase, and the intracellular levels of glutathione. Curcumin pretreatment also protected the cells from colistin-induced mitochondrial dysfunction, caspase activation, and subsequent apoptosis. Overall, our findings demonstrate for the first time, a potential role for curcumin for treating polymyxin-induced neurotoxicity through the modulation of NF-κB signaling and its potent anti-oxidative and anti-apoptotic effects.
- Published
- 2016
39. Development of HPLC and LC–MS/MS methods for the analysis of ivacaftor, its major metabolites and lumacaftor in plasma and sputum of cystic fibrosis patients treated with ORKAMBI or KALYDECO
- Author
-
Elena K. Schneider, Dominic Keating, Felisa Reyes-Ortega, Tony Velkov, John W Wilson, Jian Li, and Tom Kotsimbos
- Subjects
0301 basic medicine ,Cystic Fibrosis ,Clinical Biochemistry ,Luma ,Aminopyridines ,Quinolones ,Aminophenols ,Biochemistry ,Cystic fibrosis ,Article ,Analytical Chemistry ,Ivacaftor ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,Limit of Detection ,Tandem Mass Spectrometry ,medicine ,Humans ,Benzodioxoles ,Chromatography, High Pressure Liquid ,Chromatography ,medicine.diagnostic_test ,Chemistry ,Lumacaftor ,Sputum ,Cell Biology ,General Medicine ,medicine.disease ,030104 developmental biology ,030228 respiratory system ,Therapeutic drug monitoring ,Pharmacodynamics ,sense organs ,medicine.symptom ,medicine.drug - Abstract
ORKAMBI (ivacaftor-lumacaftor [LUMA]) and KALYDECO (ivacaftor; IVA) are two new breakthrough cystic fibrosis (CF) drugs that directly modulate the activity and trafficking of the defective CFTR underlying the CF disease state. Currently, no therapeutic drug monitoring assays exist for these very expensive, albeit, important drugs. In this study, for the first time HPLC and LC-MS methods were developed and validated for rapid detection and quantification of IVA and its major metabolites hydroxymethyl-IVA M1 (active) and IVA-carboxylate M6 (inactive); and LUMA in the plasma and sputum of CF patients. With a mobile phase consisting of acetonitrile/water:0.1% formic acid (60:40 v/v) at a flow rate of 1 mL/min, a linear correlation was observed over a concentration range from 0.01 to 10 μg/mL in human plasma (IVA R2>0.999, IVA M1 R2> 0.9961, IVA M6 R2>0.9898, LUMA R2>0.9954). The assay was successfully utilized to quantify the concentration of LUMA, IVA, M1 and M6 in the plasma and sputum of CF patients undergoing therapy with KALYDECO (IVA 150 mg/q12 h) or ORKAMBI (200 mg/q12 h LUMA-125 mg/q12 h IVA). The KALYDECO patient exhibited an IVA plasma concentration of 0.97 μg/mL at 2.5 h post dosage. M1 and M6 plasma concentrations were 0.50 μg/mL and 0.16 μg/mL, respectively. Surprisingly, the ORKAMBI patient displayed very low plasma concentrations of IVA (0.06 μg/mL) and M1 (0.07 μg/mL). The M6 concentrations (0.15 μg/mL) were comparable to those of the KALYDECO patient. However, we observed a relatively high plasma concentration of LUMA (4.42 μg/mL). This reliable and novel method offers a simple and sensitive approach for therapeutic drug monitoring of KALYDECO and ORKAMBI in plasma and sputum. The introduction of the assay into the clinical setting will facilitate pharmacokinetics/pharmacodynamic analysis and assist clinicians to develop more cost effective and efficacious dosage regimens for these breakthrough CF drugs.
- Published
- 2016
40. Nuclear heterogeneity is prevalent in high-quality fractionated human sperm cells typically used for assisted conception
- Author
-
E Schneider, N Cole, Jacob Netherton, Ana Izabel Silva Balbin Villaverde, H Zhang, Mark Baker, Louise Hetherington, Rachel A Ogle, and Tony Velkov
- Subjects
Male ,Population ,030232 urology & nephrology ,Semen ,Biology ,Andrology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Humans ,education ,Child ,Infertility, Male ,education.field_of_study ,030219 obstetrics & reproductive medicine ,Rehabilitation ,Obstetrics and Gynecology ,Embryo ,Chromomycin A3 ,Sperm ,Spermatozoa ,Chromatin ,Staining ,Reproductive Medicine ,chemistry ,Fertilization ,Percoll - Abstract
STUDY QUESTION What is the nuclear heterogeneity of high-density purified human spermatozoa typically used for IVF purposes. SUMMARY ANSWER The data show that while density gradient separation has improved the overall sperm population, there is still a large degree of nuclear heterogeneity within these cells. WHAT IS KNOWN ALREADY Chromomycin A3 (CMA3) is an important DNA binding fluorochrome for the assessment of male-factor fertility. It is typically used to predict IVF outcomes on entire sperm ejaculates with very high receiver operating characteristic. Here we used CMA3 to characterise typical populations of human spermatozoa that would be used for IVF purposes after density gradient separation. STUDY DESIGN, SIZE, DURATION We compared the intensity of CMA3 binding within high-dense sperm populations obtained from men. Binding heterogeneity was confirmed through fluorescence microscopy and FACS analysis independently. We also looked at CMA3 staining directly with head morphology in this sperm population. Finally, we looked at electron micrographs of nuclear heterogeneity (vacuoles, chromatin compaction) of spermatozoa following density gradient sorting of CMA3-stained cells. PARTICIPANTS/MATERIALS, SETTING, METHODS We used sperm donors who had fathered one or more children. Semen was collected after 2 days abstinence and purified over Percoll gradients. Only the high-quality spermatozoa, the same used for assisted conception, were then used. Cells were stained with CMA3 and sorted using FACS. Following this, electron micrographs were used to assess nuclear heterogeneity of CMA3-dependent sorted spermatozoa. MAIN RESULTS AND THE ROLE OF CHANCE CMA3 staining occurs within morphologically normal as well as abnormal spermatozoa. High-intensity CMA3-stained sperm possessed large vacuoles that were not seen in the low-CMA3 population. In addition, the high-CMA3 stained cells possess higher amounts of nuclear granulation. LIMITATIONS, REASONS FOR CAUTION The present study only describes the issues within the chromatin of these cells and does not suggest an alternate selection technique. WIDER IMPLICATIONS OF THE FINDINGS CMA3 is one of the better reported prognostic assays in predicting pregnancy outcomes, especially in cases where the male is at fault. However, it is clear that even in fractionated populations of human spermatozoa, there are sperm cells that are morphologically normal yet possess high levels of CMA3 staining and chromatin granulation. The implication of this is that the embryologist, whom selects on the basis of sperm morphology, may choose a cell with poor chromatin, which may lead to poor embryo outcomes. STUDY FUNDING/COMPETING INTEREST(S) The project was funded by the National Health and Medical Research council, APP1118943. The authors have no conflict of interest to declare. TRIAL REGISTRATION NUMBER N/A.
- Published
- 2019
41. Synergistic Combination of Polymyxin B and Enrofloxacin Induced Metabolic Perturbations in Extensive Drug-Resistant Pseudomonas aeruginosa
- Author
-
Yu-Wei Lin, Mei-Ling Han, Jinxin Zhao, Yan Zhu, Gauri Rao, Alan Forrest, Jiangning Song, Keith S. Kaye, Paul Hertzog, Anthony Purcell, Darren Creek, Qi Tony Zhou, Tony Velkov, and Jian Li
- Subjects
0301 basic medicine ,medicine.drug_class ,Polymyxin ,Antibiotics ,Drug resistance ,medicine.disease_cause ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Metabolome ,Enrofloxacin ,Pharmacology (medical) ,extensive drug-resistant ,Pharmacology ,Pseudomonas aeruginosa ,Chemistry ,polymyxin ,lcsh:RM1-950 ,metabolomics ,3. Good health ,030104 developmental biology ,lcsh:Therapeutics. Pharmacology ,030220 oncology & carcinogenesis ,Colistin ,enrofloxacin ,Polymyxin B ,medicine.drug - Abstract
Polymyxins are used as a last-resort class of antibiotics against multidrug-resistant (MDR) Gram-negative Pseudomonas aeruginosa. As polymyxin monotherapy is associated with potential development of resistance, combination therapy is highly recommended. This study investigated the mechanism underlying the synergistic killing of polymyxin B and enrofloxacin against extensive drug-resistant (XDR) P. aeruginosa. An XDR isolate P. aeruginosa 12196 was treated with clinically relevant concentrations of polymyxin B (2 mg/L) and enrofloxacin (1 mg/L) alone or in combination. Metabolome profiles were investigated from bacterial samples collected at 1-and 4-h posttreatment using liquid chromatography with tandem mass spectrometry (LC-MS/MS), and data were analyzed using univariate and multivariate statistics. Significantly perturbed metabolites (q < 0.05, fold change ≥ 2) were subjected to pathway analysis. The synergistic killing by polymyxin B–enrofloxacin combination was initially driven by polymyxin B as indicated by the perturbation of lipid metabolites at 1 h in particular. The killing was subsequently driven by enrofloxacin via the inhibition of DNA replication, resulting in the accumulation of nucleotides at 4 h. Furthermore, the combination uniquely altered levels of metabolites in energy metabolism and cell envelope biogenesis. Most importantly, the combination significantly minimized polymyxin resistance via the inhibition of lipid A modification pathway, which was most evident at 4 h. This is the first study to elucidate the synergistic mechanism of polymyxin B–enrofloxacin combination against XDR P. aeruginosa. The metabolomics approach taken in this study highlights its power to elucidate the mechanism of synergistic killing by antibiotic combinations at the systems level.
- Published
- 2019
42. Inhalable liposomal powder formulations for co-delivery of synergistic ciprofloxacin and colistin against multi-drug resistant gram-negative lung infections
- Author
-
Shaoning Wang, Guihong Chai, Jian Li, Yu-Wei Lin, Shihui Yu, Weisan Pan, Qi Tony Zhou, Peizhi Zou, and Tony Velkov
- Subjects
medicine.drug_class ,Antibiotics ,Pharmaceutical Science ,02 engineering and technology ,Pharmacology ,030226 pharmacology & pharmacy ,Article ,03 medical and health sciences ,0302 clinical medicine ,Ciprofloxacin ,Administration, Inhalation ,Drug Resistance, Bacterial ,medicine ,Humans ,Respiratory Tract Infections ,Aerosolization ,Liposome ,Chemistry ,Colistin ,Drug Synergism ,021001 nanoscience & nanotechnology ,Drug Resistance, Multiple ,Anti-Bacterial Agents ,Drug Combinations ,A549 Cells ,Drug delivery ,Liposomes ,Pseudomonas aeruginosa ,Mannitol ,Powders ,0210 nano-technology ,Antibacterial activity ,Gram-Negative Bacterial Infections ,medicine.drug - Abstract
The aim of this study was to design and characterize dry powder inhaler formulations of ciprofloxacin and colistin co-loaded liposomes prepared by the ultrasonic spray-freeze-drying (USFD) technique. Liposomal formulations and powder production parameters were optimized to achieve optimal characteristics and in-vitro performance such as encapsulation efficiency (EE), particle size, particle distribution index (PDI), fine particle fraction (FPF), emitted dose (ED) and in vitro antibacterial activity. The formulation (F6) with the mannitol (5% w/v) as the internal lyoprotectant and sucrose (5%, w/v), mannitol (10%, w/v) and leucine (5%, w/w) as the external lyoprotectants / aerosolization enhancers showed an optimal rehydrated EE values of ciprofloxacin and colistin (44.9 ± 0.9% and 47.0 ± 0.6%, respectively) as well as satisfactory aerosol performance (FPF: 45.8 ± 2.2% and 43.6 ± 1.6%, respectively; ED: 97.0 ± 0.5% and 95.0 ± 0.6%, respectively). For the blank liposomes, there was almost no inhibitory effect on the cell proliferation in human lung epithelial A549 cells, showing that the lipid materials used in the liposome formulation is safe for use in pulmonary drug delivery. The cytotoxicity study demonstrated that the optimized liposomal formulation (F6) was not cytotoxic at least at the drug concentrations of colistin 5 μg/mL and ciprofloxacin 20 μg/mL. Colistin (2 mg/L) monotherapy showed no antibacterial effect against P. aeruginosa H131300444 and H133880624. Ciprofloxacin (8 mg/L) monotherapy showed moderate bacterial killing for both clinical isolates; however, regrowth was observed in 6 h for P. aeruginosa H133880624. The liposomal formulation displayed superior antibacterial activity against clinical isolates of Pseudomonas aeruginosa H131300444 and P. aeruginosa H133880624 compared to each antibiotic per se. These results demonstrate that the liposomal powder formulation prepared by USFD could potentially be a pulmonary delivery system for antibiotic combination to treat multi-drug resistant Gram-negative lung infections.
- Published
- 2019
43. The impact of backbone N ‐methylation on the structure‐activity relationship of Leu 10 ‐teixobactin
- Author
-
John A. Karas, Tony Velkov, Jian Li, James D. Swarbrick, Elena K Schneider-Futschik, Daniel Hoyer, and Maytham Hussein
- Subjects
Pharmacology ,Depsipeptide ,Natural product ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Teixobactin ,General Medicine ,N methylation ,010402 general chemistry ,Antimicrobial ,01 natural sciences ,Biochemistry ,Combinatorial chemistry ,0104 chemical sciences ,chemistry.chemical_compound ,Mechanism of action ,Structural Biology ,Drug Discovery ,medicine ,Rapid access ,Molecular Medicine ,Structure–activity relationship ,medicine.symptom ,Molecular Biology - Abstract
Antimicrobial resistance is a serious threat to global human health; therefore, new anti-infective therapeutics are required. The cyclic depsi-peptide teixobactin exhibits potent antimicrobial activity against several Gram-positive pathogens. To study the natural product's mechanism of action and improve its pharmacological properties, efficient chemical methods for preparing teixobactin analogues are required to expedite structure-activity relationship studies. Described herein is a synthetic route that enables rapid access to analogues. Furthermore, our new N-methylated analogues highlight that hydrogen bonding along the N-terminal tail is likely to be important for antimicrobial activity.
- Published
- 2019
44. Comparative Metabolomics Reveals Key Pathways Associated With the Synergistic Killing of Colistin and Sulbactam Combination Against Multidrug-Resistant Acinetobacter baumannii
- Author
-
Mei-Ling Han, Xiaofen Liu, Tony Velkov, Yu-Wei Lin, Yan Zhu, Darren J. Creek, Christopher K. Barlow, Heidi H. Yu, Zhihui Zhou, Jing Zhang, and Jian Li
- Subjects
0301 basic medicine ,Combination therapy ,medicine.drug_class ,β-lactam ,Polymyxin ,Antibiotics ,Pharmacology ,peptidoglycan ,combination therapy ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,polycyclic compounds ,Pharmacology (medical) ,biology ,polymyxin ,lipopolysaccharide ,lcsh:RM1-950 ,Sulbactam ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,bacterial infections and mycoses ,metabolomics ,Uridine ,Acinetobacter baumannii ,030104 developmental biology ,lcsh:Therapeutics. Pharmacology ,chemistry ,030220 oncology & carcinogenesis ,Colistin ,bacteria ,Peptidoglycan ,medicine.drug - Abstract
Background: Polymyxins are a last-line class of antibiotics against multidrug-resistant Acinetobacter baumannii. However, polymyxin resistance can emerge with monotherapy, highlighting the need for synergistic combination therapies. Polymyxins in combination with β-lactams have shown remarkable synergy against multidrug-resistant A. baumannii. Methods: Liquid chromatography-mass spectrometry-based metabolomics was conducted to investigate the metabolic perturbations in an A. baumannii clinical isolate, AB090342, in response to colistin (1 mg/L), sulbactam (128 mg/L), and their combination at 1, 4, and 24 h. Metabolomics data were analyzed using univariate and multivariate statistics, and metabolites showing ≥2-fold changes were subjected to pathway analysis. Results: The synergistic activity of colistin-sulbactam combination was initially driven by colistin through perturbation of fatty acid and phospholipid levels at 1 h. Cell wall biosynthesis was perturbed by sulbactam alone and the combination over 24 h; this was demonstrated by the decreased levels of two important precursors, uridine diphosphate-N-acetylglucosamine and uridine diphosphate-N-acetylmuramate, together with perturbed lysine and amino sugar metabolism. Moreover, sulbactam alone and the combination significantly depleted nucleotide metabolism and the associated arginine biosynthesis, glutamate metabolism, and pentose phosphate pathway. Notably, the colistin-sulbactam combination decreased amino acid and nucleotide levels more dramatically at 4 h compared with both monotherapies. Conclusions: This is the first metabolomics study revealing the time-dependent synergistic activity of colistin and sulbactam against A. baumannii, which was largely driven by sulbactam through the inhibition of cell wall biosynthesis. Our mechanistic findings may help optimizing synergistic colistin combinations in patients.
- Published
- 2019
45. Synthesis and structure-activity relationships of teixobactin
- Author
-
James D. Swarbrick, Jian Li, John A. Karas, Andrew M. Giltrap, Zhisen Kang, Tony Velkov, Fan Chen, Maytham Hussein, Daniel Hoyer, Elena K Schneider-Futschik, and Richard J. Payne
- Subjects
Modern medicine ,medicine.drug_class ,Antibiotics ,Antimicrobial peptides ,Teixobactin ,Computational biology ,Microbial Sensitivity Tests ,Biology ,010402 general chemistry ,01 natural sciences ,General Biochemistry, Genetics and Molecular Biology ,chemistry.chemical_compound ,Structure-Activity Relationship ,Antibiotic resistance ,History and Philosophy of Science ,Depsipeptides ,Drug Resistance, Multiple, Bacterial ,medicine ,Animals ,Humans ,Enduracididine ,Natural product ,Lipid II ,010405 organic chemistry ,General Neuroscience ,Bacterial Infections ,0104 chemical sciences ,chemistry - Abstract
The discovery of antibiotics has led to the effective treatment of bacterial infections that were otherwise fatal and has had a transformative effect on modern medicine. Teixobactin is an unusual depsipeptide natural product that was recently discovered from a previously unculturable soil bacterium and found to possess potent antibacterial activity against several Gram positive pathogens, including methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococci. One of the key features of teixobactin as an antibiotic lead is that resistance could not be generated in a laboratory setting. This is proposed to be a result of a mechanism of action that involves binding to essential cell wall synthesis building blocks, lipid II and lipid III. Since the initial isolation report in 2015, significant efforts have been made to understand its unique mechanism of action, develop efficient synthetic routes for its production, and thus enable the generation of analogues for structure-activity relationship studies and optimization of its pharmacological properties. Our review provides a comprehensive treatise on the progress in understanding teixobactin chemistry, structure-activity relationships, and mechanisms of antibacterial activity. Teixobactin represents an exciting starting point for the development of new antibiotics that can be used to combat multidrug-resistant bacterial ("superbug") infections.
- Published
- 2019
46. Polymyxin resistance in Klebsiella pneumoniae: multifaceted mechanisms utilized in the presence and absence of the plasmid-encoded phosphoethanolamine transferase gene mcr-1
- Author
-
Chongshan Dai, Mei-Ling Han, Jiping Wang, Sue C Nang, Marina Harper, Heidi H. Yu, Tony Velkov, Von Vergel L. Torres, and Jian Li
- Subjects
Microbiology (medical) ,Klebsiella pneumoniae ,medicine.drug_class ,Polymyxin ,Population ,Drug resistance ,Microbial Sensitivity Tests ,Lipid A ,Mice ,Plasmid ,Bacterial Proteins ,Drug Resistance, Bacterial ,medicine ,Animals ,Humans ,Pharmacology (medical) ,education ,Polymyxin B ,Original Research ,Pharmacology ,education.field_of_study ,biology ,Chemistry ,biology.organism_classification ,Ethanolaminephosphotransferase ,Molecular biology ,Anti-Bacterial Agents ,Klebsiella Infections ,Infectious Diseases ,Mutation ,MCR-1 ,Female ,Gene Deletion ,medicine.drug - Abstract
ObjectivesUntil plasmid-mediated mcr-1 was discovered, it was believed that polymyxin resistance in Gram-negative bacteria was mainly mediated by the chromosomally-encoded EptA and ArnT, which modify lipid A with phosphoethanolamine (pEtN) and 4-amino-4-deoxy-l-arabinose (l-Ara4N), respectively. This study aimed to construct a markerless mcr-1 deletion mutant in Klebsiella pneumoniae, validate a reliable reference gene for reverse transcription quantitative PCR (RT–qPCR) and investigate the interactions among mcr-1, arnT and eptA, in response to polymyxin treatments using pharmacokinetics/pharmacodynamics (PK/PD).MethodsAn isogenic markerless mcr-1 deletion mutant (II-503Δmcr-1) was generated from a clinical K. pneumoniae II-503 isolate. The efficacy of different polymyxin B dosage regimens was examined using an in vitro one-compartment PK/PD model and polymyxin resistance was assessed using population analysis profiles. The expression of mcr-1, eptA and arnT was examined using RT–qPCR with a reference gene pepQ, and lipid A was profiled using LC-MS. In vivo polymyxin B efficacy was investigated in a mouse thigh infection model.ResultsIn K. pneumoniae II-503, mcr-1 was constitutively expressed, irrespective of polymyxin exposure. Against II-503Δmcr-1, an initial bactericidal effect was observed within 4 h with polymyxin B at average steady-state concentrations of 1 and 3 mg/L, mimicking patient PK. However, substantial regrowth and concomitantly increased expression of eptA and arnT were detected. Predominant l-Ara4N-modified lipid A species were detected in II-503Δmcr-1 following polymyxin B treatment.ConclusionsThis is the first study demonstrating a unique markerless deletion of mcr-1 in a clinical polymyxin-resistant K. pneumoniae. The current polymyxin B dosage regimens are suboptimal against K. pneumoniae, regardless of mcr, and can lead to the emergence of resistance.
- Published
- 2019
47. Effects of antibiotic component on in-vitro bacterial killing, physico-chemical properties, aerosolization and dissolution of a ternary-combinational inhalation powder formulation of antibiotics for pan-drug resistant Gram-negative lung infections
- Author
-
Sharad Mangal, Dmitry Zemlyanov, Jiayang Huang, Nivedita Shetty, Tony Velkov, Hee Jun Park, Qi Tony Zhou, Yu-Wei Lin, Jian Li, and Heidi H. Yu
- Subjects
Chemical Phenomena ,Surface Properties ,Drug Compounding ,Pharmaceutical Science ,02 engineering and technology ,Microbial Sensitivity Tests ,030226 pharmacology & pharmacy ,Article ,03 medical and health sciences ,0302 clinical medicine ,Drug Stability ,medicine ,polycyclic compounds ,Solubility ,Desiccation ,Particle Size ,Dissolution ,Aerosolization ,Aerosols ,Chemistry ,Colistin ,Meropenem ,021001 nanoscience & nanotechnology ,bacterial infections and mycoses ,Anti-Bacterial Agents ,Drug Combinations ,Drug Liberation ,Spray drying ,Particle ,Particle size ,Powders ,Rifampin ,0210 nano-technology ,Ternary operation ,Nuclear chemistry ,medicine.drug - Abstract
Combinational antibiotic formulations have emerged as an important strategy to combat antibiotic resistance. The main objective of this study was to examine effects of individual components on the antimicrobial activity, physico-chemical properties, aerosolization and dissolution of powder aerosol formulations when three synergistic drugs were co-spray dried. A ternary dry powder formulation consisting of meropenem (75.5 %w/w), colistin (15.1 %w/w) and rifampicin (9.4 %w/w) at the selected ratio was produced by spray drying. The ternary formulation was characterized for in-vitro antibacterial activity, physico-chemical properties, surface composition, aerosol performance and dissolution. All of the formulations demonstrated excellent aerosolization behavior achieving a fine particle fraction of >70%, which was substantially higher than those for the Meropenem-SD and Colistin-Meropenem formulations. The results indicated that rifampicin controlled the surface morphology of the ternary and binary combination formulations resulting in the formation of highly corrugated particles. Advanced characterization of surface composition by XPS supported the hypothesis that rifampicin was enriched on the surface of the combination powder formulations. All spray-dried formulations were amorphous and absorbed substantial amount of water at the elevated humidity. Storage at the elevated humidity caused a substantial decline in aerosolization performance for the Meropenem-SD and Colistin-Meropenem, which was attributed to increased inter-particulate capillary forces or particle fusion. In contrast, the ternary combination and binary Meropenem-Rifampicin formulations showed no change in aerosol performance at the elevated storage humidity conditions; attributable to the enriched hydrophobicity of rifampicin on the particle surface that acted as a barrier against moisture condensation and particle fusion. Interestingly, in the ternary formulation rifampicin enrichment on the surface did not interfere with the dissolution of other two components (i.e. meropenem and colistin). Our study provides an insight on the impact of each component on the performance of co-spray dried combinational formulations.
- Published
- 2019
48. Comparative Metabolomics and Transcriptomics Reveal Multiple Pathways Associated with Polymyxin Killing in Pseudomonas aeruginosa
- Author
-
Tony Purcell, Darren J. Creek, Hsin-Hui Shen, Samuel M. Moskowitz, Tony Velkov, Mei-Ling Han, Yan Zhu, Yu-Wei Lin, Paul J. Hertzog, Alina D. Gutu, and Jian Li
- Subjects
Physiology ,medicine.drug_class ,Polymyxin ,lcsh:QR1-502 ,medicine.disease_cause ,polymyxins ,Biochemistry ,Microbiology ,lcsh:Microbiology ,Lipid A ,03 medical and health sciences ,chemistry.chemical_compound ,transcriptomics ,Genetics ,medicine ,lipid A modification ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,glycerophospholipids ,0303 health sciences ,030306 microbiology ,Pseudomonas aeruginosa ,Chemistry ,lipopolysaccharide ,Therapeutics and Prevention ,metabolomics ,QR1-502 ,3. Good health ,Computer Science Applications ,Multiple drug resistance ,Modeling and Simulation ,Colistin ,lipids (amino acids, peptides, and proteins) ,Peptidoglycan ,Bacterial outer membrane ,Polymyxin B ,medicine.drug ,Research Article - Abstract
Pseudomonas aeruginosa has been highlighted by the recent WHO Global Priority Pathogen List due to multidrug resistance. Without new antibiotics, polymyxins remain a last-line therapeutic option for this difficult-to-treat pathogen. The emergence of polymyxin resistance highlights the growing threat to our already very limited antibiotic armamentarium and the urgency to understand the exact mechanisms of polymyxin activity and resistance. Integration of the correlative metabolomics and transcriptomics results in the present study discovered that polymyxin treatment caused significant perturbations in the biosynthesis of lipids, lipopolysaccharide, and peptidoglycan, central carbon metabolism, and oxidative stress. Importantly, lipid A modifications were surprisingly rapid in response to polymyxin treatment at clinically relevant concentrations. This is the first study to reveal the dynamics of polymyxin-induced cellular responses at the systems level, which highlights that combination therapy should be considered to minimize resistance to the last-line polymyxins. The results also provide much-needed mechanistic information which potentially benefits the discovery of new-generation polymyxins., Polymyxins are a last-line therapy against multidrug-resistant Pseudomonas aeruginosa; however, resistance to polymyxins has been increasingly reported. Therefore, understanding the mechanisms of polymyxin activity and resistance is crucial for preserving their clinical usefulness. This study employed comparative metabolomics and transcriptomics to investigate the responses of polymyxin-susceptible P. aeruginosa PAK (polymyxin B MIC, 1 mg/liter) and its polymyxin-resistant pmrB mutant PAKpmrB6 (MIC, 16 mg/liter) to polymyxin B (4, 8, and 128 mg/liter) at 1, 4, and 24 h, respectively. Our results revealed that polymyxin B at 4 mg/liter induced different metabolic and transcriptomic responses between polymyxin-susceptible and -resistant P. aeruginosa. In strain PAK, polymyxin B significantly activated PmrAB and the mediated arn operon, leading to increased 4-amino-4-deoxy-L-arabinose (L-Ara4N) synthesis and the addition to lipid A. In contrast, polymyxin B did not increase lipid A modification in strain PAKpmrB6. Moreover, the syntheses of lipopolysaccharide and peptidoglycan were significantly decreased in strain PAK but increased in strain PAKpmrB6 due to polymyxin B treatment. In addition, 4 mg/liter polymyxin B significantly perturbed phospholipid and fatty acid levels and induced oxidative stress in strain PAK, but not in PAKpmrB6. Notably, the increased trehalose-6-phosphate levels indicate that polymyxin B potentially caused osmotic imbalance in both strains. Furthermore, 8 and 128 mg/liter polymyxin B significantly elevated lipoamino acid levels and decreased phospholipid levels but without dramatic changes in lipid A modification in wild-type and mutant strains, respectively. Overall, this systems study is the first to elucidate the complex and dynamic interactions of multiple cellular pathways associated with the polymyxin mode of action against P. aeruginosa. IMPORTANCE Pseudomonas aeruginosa has been highlighted by the recent WHO Global Priority Pathogen List due to multidrug resistance. Without new antibiotics, polymyxins remain a last-line therapeutic option for this difficult-to-treat pathogen. The emergence of polymyxin resistance highlights the growing threat to our already very limited antibiotic armamentarium and the urgency to understand the exact mechanisms of polymyxin activity and resistance. Integration of the correlative metabolomics and transcriptomics results in the present study discovered that polymyxin treatment caused significant perturbations in the biosynthesis of lipids, lipopolysaccharide, and peptidoglycan, central carbon metabolism, and oxidative stress. Importantly, lipid A modifications were surprisingly rapid in response to polymyxin treatment at clinically relevant concentrations. This is the first study to reveal the dynamics of polymyxin-induced cellular responses at the systems level, which highlights that combination therapy should be considered to minimize resistance to the last-line polymyxins. The results also provide much-needed mechanistic information which potentially benefits the discovery of new-generation polymyxins.
- Published
- 2019
49. Polymyxins: Mode of Action
- Author
-
Tony Velkov and Zhifeng Li
- Subjects
Computer science ,medicine.drug_class ,Polymyxin ,Respiratory chain ,Lipopeptide ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Antibiotic resistance ,chemistry ,Action (philosophy) ,NADH quinone oxidoreductase ,medicine ,030212 general & internal medicine ,Biochemical engineering ,Mode of action - Abstract
The dry antibiotic development pipeline coupled with the emergence of multi-drug resistant Gram-negative 'superbugs' has driven the revival of the polymyxin lipopeptide antibiotics. Understanding the mode of action of antibiotics is an important precursor for optimizing their use and development. This chapter provides a concise treatise of the current knowledge-based on the primary mode of action of polymyxins as well as recent developments in understanding of bacterial cell responses and secondary modes of action.
- Published
- 2019
50. An 'Unlikely' Pair: The Antimicrobial Synergy of Polymyxin B in Combination with the Cystic Fibrosis Transmembrane Conductance Regulator Drugs KALYDECO and ORKAMBI
- Author
-
Qi Tony Zhou, Mei-Ling Han, Mark T. Muller, Jian Li, Tony Velkov, Elena K. Schneider, Yohei Doi, Mohammad Abul Kalam Azad, Mark Baker, Johnny X. Huang, Mark E. Cooper, Jiping Wang, and Phillip J. Bergen
- Subjects
0301 basic medicine ,Cystic Fibrosis ,medicine.drug_class ,Polymyxin ,Aminopyridines ,Cystic Fibrosis Transmembrane Conductance Regulator ,Quinolones ,Pharmacology ,Aminophenols ,medicine.disease_cause ,Cystic fibrosis ,Article ,Microbiology ,Ivacaftor ,03 medical and health sciences ,chemistry.chemical_compound ,medicine ,Humans ,Benzodioxoles ,Polymyxin B ,biology ,Pseudomonas aeruginosa ,business.industry ,Lumacaftor ,Drug Synergism ,Antimicrobial ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,Anti-Bacterial Agents ,030104 developmental biology ,Infectious Diseases ,chemistry ,biology.protein ,Drug Therapy, Combination ,lipids (amino acids, peptides, and proteins) ,business ,medicine.drug - Abstract
Novel combination therapies are desperately needed for combating lung infections caused by bacterial "superbugs". This study aimed to investigate the synergistic antibacterial activity of polymyxin B in combination with the cystic fibrosis (CF) drugs KALYDECO (ivacaftor) and ORKAMBI (ivacaftor + lumacaftor) against Gram-negative pathogens that commonly colonize the CF lung, in particular, the problematic Pseudomonas aeruginosa. The in vitro synergistic activity of polymyxin B combined with ivacaftor or lumacaftor was assessed using checkerboard and static time-kill assays against a panel of polymyxin-susceptible and polymyxin-resistant P. aeruginosa isolates from the lungs of CF patients. Polymyxin B, ivacaftor, and lumacaftor were ineffective when used individually against polymyxin-resistant (MIC ≥ 4 mg/L) isolates. However, when used together, the combination of clinically relevant concentrations of polymyxin B (2 mg/L) combined with ivacaftor (8 mg/L) or ivacaftor (8 mg/L) + lumacaftor (8 mg/L) displayed synergistic killing activity against polymyxin-resistant P. aeruginosa isolates as demonstrated by a 100-fold decrease in the bacterial count (CFU/mL) even after 24 h. The combinations also displayed excellent antibacterial activity against P. aeruginosa under CF relevant conditions in a sputum medium assay. The combination of lumacaftor (alone) with polymyxin B showed additivity against P. aeruginosa. The potential antimicrobial mode of action of the combinations against P. aeruginosa was investigated using different methods. Treatment with the combinations induced cytosolic GFP release from P. aeruginosa cells and showed permeabilizing activity in the nitrocefin assay, indicating damage to both the outer and inner Gram-negative cell membranes. Moreover, scanning and transmission electron micrographs revealed that the combinations produce outer membrane damage to P. aeruginosa cells that is distinct from the effect of each compound per se. Ivacaftor was also shown to be a weak inhibitor of the bacterial DNA gyrase and topoisomerase IV with no effect on either human type I or type IIα topoisomerases. Lumacaftor displayed the ability to increase the cellular production of damaging reactive oxygen species. In summary, the combination of polymyxin B with KALYDECO or ORKAMBI exhibited synergistic activity against highly polymyxin-resistant P. aeruginosa CF isolates and can be potentially useful for otherwise untreatable CF lung infections.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.