Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Nature ; 567(7748): 341-346, 2019 03.
Article in English | MEDLINE | ID: mdl-30842654

ABSTRACT

Cancer-specific inhibitors that reflect the unique metabolic needs of cancer cells are rare. Here we describe Gboxin, a small molecule that specifically inhibits the growth of primary mouse and human glioblastoma cells but not that of mouse embryonic fibroblasts or neonatal astrocytes. Gboxin rapidly and irreversibly compromises oxygen consumption in glioblastoma cells. Gboxin relies on its positive charge to associate with mitochondrial oxidative phosphorylation complexes in a manner that is dependent on the proton gradient of the inner mitochondrial membrane, and it inhibits the activity of F0F1 ATP synthase. Gboxin-resistant cells require a functional mitochondrial permeability transition pore that regulates pH and thus impedes the accumulation of Gboxin in the mitochondrial matrix. Administration of a metabolically stable Gboxin analogue inhibits glioblastoma allografts and patient-derived xenografts. Gboxin toxicity extends to established human cancer cell lines of diverse organ origin, and shows that the increased proton gradient and pH in cancer cell mitochondria is a mode of action that can be targeted in the development of antitumour reagents.


Subject(s)
Glioblastoma/drug therapy , Glioblastoma/metabolism , Oxidative Phosphorylation/drug effects , Allografts , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cell Line, Tumor , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Hydrogen-Ion Concentration , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/enzymology , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore , Neoplasm Transplantation , Organ Specificity , Proton-Motive Force/drug effects , Proton-Translocating ATPases/antagonists & inhibitors , Proton-Translocating ATPases/metabolism , Xenograft Model Antitumor Assays
2.
J Appl Microbiol ; 135(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38678002

ABSTRACT

AIMS: This study aimed to develop an editable structural scaffold for improving drug development, including pharmacokinetics and pharmacodynamics of antibiotics by using synthetic compounds derived from a (hetero)aryl-quinoline hybrid scaffold. METHODS AND RESULTS: In this study, 18 CF3-substituted (hetero)aryl-quinoline hybrid molecules were examined for their potential antibacterial activity against Staphylococcus aureus by determining minimal inhibitory concentrations. These 18 synthetic compounds represent modifications to key regions of the quinoline N-oxide scaffold, enabling us to conduct a structure-activity relationship analysis for antibacterial potency. Among the compounds, 3 m exhibited potency against with both methicillin resistant S. aureus strains, as well as other Gram-positive bacteria, including Enterococcus faecalis and Bacillus subtilis. We demonstrated that 3 m disrupted the bacterial proton motive force (PMF) through monitoring the PMF and conducting the molecular dynamics simulations. Furthermore, we show that this mechanism of action, disrupting PMF, is challenging for S. aureus to overcome. We also validated this PMF inhibition mechanism of 3 m in an Acinetobacter baumannii strain with weaken lipopolysaccharides. Additionally, in Gram-negative bacteria, we demonstrated that 3 m exhibited a synergistic effect with colistin that disrupts the outer membrane of Gram-negative bacteria. CONCLUSIONS: Our approach to developing editable synthetic novel antibacterials underscores the utility of CF3-substituted (hetero)aryl-quinoline scaffold for designing compounds targeting the bacterial proton motive force, and for further drug development, including pharmacokinetics and pharmacodynamics.


Subject(s)
Anti-Bacterial Agents , Indoles , Microbial Sensitivity Tests , Proton-Motive Force , Quinolines , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Quinolines/pharmacology , Quinolines/chemistry , Proton-Motive Force/drug effects , Indoles/pharmacology , Indoles/chemistry , Structure-Activity Relationship , Methicillin-Resistant Staphylococcus aureus/drug effects , Molecular Dynamics Simulation , Acinetobacter baumannii/drug effects , Enterococcus faecalis/drug effects , Staphylococcus aureus/drug effects , Bacillus subtilis/drug effects
3.
Pharmacol Res ; 175: 105978, 2022 01.
Article in English | MEDLINE | ID: mdl-34813930

ABSTRACT

The widespread dissemination of antibiotic resistance genes (ARGs) is a serious problem and constitutes a threat for public health. Plasmid-mediated conjugative transfer of ARGs is recognized as one of the most important pathways accounting for this global crisis. Inhibiting the conjugative transfer of resistant gene-bearing plasmids provides a feasible strategy to prevent the spread of antibiotic resistance. Here we found that melatonin, a neurohormone secreted from pineal gland, substantially inhibited the horizontal transfer of RP4-7 plasmid in a dose-dependent manner. Furthermore, melatonin could also suppress the conjugal frequency of different types of clinical plasmids that carrying colistin resistance gene mcr-1 rather than blaNDM or tet(X) genes. Next, we investigated the mechanisms underlying the inhibitory effect of melatonin on conjugation. As a result, we showed that the addition of melatonin markedly reduced bacterial membrane permeability and inhibited the oxidative stress. In line with these observations, the conjugative transfer-related genes were regulated accordingly. Most importantly, we uncovered that melatonin disrupted bacterial proton motive force (PMF), which is an essential bacterial energy metabolism substance and is important for conjugative process. Collectively, these results provide implications that some non-antibiotics such as melatonin are effective inhibitors of transmission of ARGs and raise a promising strategy to confront the increasing resistant infections.


Subject(s)
Drug Resistance, Microbial/genetics , Melatonin/pharmacology , Proton-Motive Force/drug effects , Adenosine Triphosphate/metabolism , Ampicillin , Animals , Anti-Bacterial Agents , Cell Membrane Permeability/drug effects , Chloramphenicol , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli Infections , Escherichia coli Proteins/genetics , Female , Genes, Bacterial , Mice, Inbred ICR , Plasmids , Reactive Oxygen Species/metabolism
4.
PLoS Pathog ; 15(4): e1007697, 2019 04.
Article in English | MEDLINE | ID: mdl-31034512

ABSTRACT

Antibiotic susceptibility of bacterial pathogens is typically evaluated using in vitro assays that do not consider the complex host microenvironment. This may help explaining a significant discrepancy between antibiotic efficacy in vitro and in vivo, with some antibiotics being effective in vitro but not in vivo or vice versa. Nevertheless, it is well-known that antibiotic susceptibility of bacteria is driven by environmental factors. Lung epithelial cells enhance the activity of aminoglycoside antibiotics against the opportunistic pathogen Pseudomonas aeruginosa, yet the mechanism behind is unknown. The present study addresses this gap and provides mechanistic understanding on how lung epithelial cells stimulate aminoglycoside activity. To investigate the influence of the local host microenvironment on antibiotic activity, an in vivo-like three-dimensional (3-D) lung epithelial cell model was used. We report that conditioned medium of 3-D lung cells, containing secreted but not cellular components, potentiated the bactericidal activity of aminoglycosides against P. aeruginosa, including resistant clinical isolates, and several other pathogens. In contrast, conditioned medium obtained from the same cell type, but grown as conventional (2-D) monolayers did not influence antibiotic efficacy. We found that 3-D lung cells secreted endogenous metabolites (including succinate and glutamate) that enhanced aminoglycoside activity, and provide evidence that bacterial pyruvate metabolism is linked to the observed potentiation of antimicrobial activity. Biochemical and phenotypic assays indicated that 3-D cell conditioned medium stimulated the proton motive force (PMF), resulting in increased bacterial intracellular pH. The latter stimulated antibiotic uptake, as determined using fluorescently labelled tobramycin in combination with flow cytometry analysis. Our findings reveal a cross-talk between host and bacterial metabolic pathways, that influence downstream activity of antibiotics. Understanding the underlying basis of the discrepancy between the activity of antibiotics in vitro and in vivo may lead to improved diagnostic approaches and pave the way towards novel means to stimulate antibiotic activity.


Subject(s)
Culture Media, Conditioned/pharmacology , Lung/metabolism , Metabolome , Proton-Motive Force/drug effects , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Tobramycin/pharmacology , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Humans , Lung/drug effects , Lung/microbiology , Microbial Sensitivity Tests , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology
5.
Soft Matter ; 16(11): 2725-2735, 2020 Mar 18.
Article in English | MEDLINE | ID: mdl-32115597

ABSTRACT

Transmembrane pH gradient poly(isoprene)-block-poly(ethylene glycol) (PI-b-PEG) polymersomes were investigated for their potential use in the detoxification of ammonia, a metabolite that is excessively present in patients suffering from urea cycle disorders and advanced liver diseases, and which causes neurotoxic effects (e.g., hepatic encephalopathy). Polymers varying in PI and PEG block length were synthesized via nitroxide-mediated polymerization and screened for their ability to self-assemble into polymersomes in aqueous media. Ammonia sequestration by the polymersomes was investigated in vitro. While most vesicular systems were able to capture ammonia in simulated intestinal fluids, uptake was lost in partially dehydrated medium mimicking conditions in the colon. Polymeric crosslinking of residual olefinic bonds in the PI block increased polymersome stability, partially preserving the ammonia capture capacity in the simulated colon environment. These more stable vesicular systems hold promise for the chronic oral treatment of hyperammonemia.


Subject(s)
Ammonia/chemistry , Drug Carriers/chemistry , Hepatic Encephalopathy/drug therapy , Inactivation, Metabolic/genetics , Ammonia/metabolism , Butadienes/chemistry , Butadienes/pharmacology , Drug Carriers/pharmacology , Fluorescein-5-isothiocyanate/chemistry , Hemiterpenes/chemistry , Hemiterpenes/pharmacology , Hepatic Encephalopathy/etiology , Hepatic Encephalopathy/metabolism , Humans , Hydrogen-Ion Concentration , Liver Diseases/complications , Liver Diseases/drug therapy , Liver Diseases/metabolism , Methacrylates/chemistry , Particle Size , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polymerization , Polymers/chemistry , Polymers/pharmacology , Proton-Motive Force/drug effects , Urea Cycle Disorders, Inborn/complications , Urea Cycle Disorders, Inborn/drug therapy , Urea Cycle Disorders, Inborn/metabolism , Water/metabolism
6.
Biochem Biophys Res Commun ; 507(1-4): 407-413, 2018 12 09.
Article in English | MEDLINE | ID: mdl-30449596

ABSTRACT

Bacterial persisters comprise a small fraction of phenotypically heterogeneous variants with transient capability for survival when exposed to high concentrations of antibiotic. In aquatic pathogenic bacteria Aeromonas veronii, Small Protein B (SmpB), the core factor of trans-translation system, was identified as a new persistence-related gene. The SmpB deletion exhibited a higher susceptibility and lower persister cell formation under aminoglycosides antibiotics pressure compared with wild type. The transcriptional and translational activities of smpB gene were significantly enhanced by the gentamicin challenge in exponential phase, but not changed in stationary phase. The transcriptomic analysis revealed that the smpB deletion stimulated the production of proton-motive force (PMF). The cell survival induced by carbonyl cyanide m-chlorophenyl hydrazone (CCCP) further verified that SmpB variation affected the quantities of PMF. Taken together, these results uncovered a novel mechanism of persister formation mediated by SmpB under aminoglycosides treatments.


Subject(s)
Aeromonas veronii/metabolism , Aminoglycosides/pharmacology , Down-Regulation/drug effects , Proton-Motive Force/drug effects , RNA-Binding Proteins/metabolism , Aeromonas veronii/drug effects , Anti-Bacterial Agents/pharmacology , Electron Transport/drug effects , Gene Deletion , Gentamicins/pharmacology , Microbial Sensitivity Tests , Protein Biosynthesis/drug effects
7.
Proc Natl Acad Sci U S A ; 112(51): E7073-82, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26644565

ABSTRACT

There is a growing need for new antibiotics. Compounds that target the proton motive force (PMF), uncouplers, represent one possible class of compounds that might be developed because they are already used to treat parasitic infections, and there is interest in their use for the treatment of other diseases, such as diabetes. Here, we tested a series of compounds, most with known antiinfective activity, for uncoupler activity. Many cationic amphiphiles tested positive, and some targeted isoprenoid biosynthesis or affected lipid bilayer structure. As an example, we found that clomiphene, a recently discovered undecaprenyl diphosphate synthase inhibitor active against Staphylococcus aureus, is an uncoupler. Using in silico screening, we then found that the anti-glioblastoma multiforme drug lead vacquinol is an inhibitor of Mycobacterium tuberculosis tuberculosinyl adenosine synthase, as well as being an uncoupler. Because vacquinol is also an inhibitor of M. tuberculosis cell growth, we used similarity searches based on the vacquinol structure, finding analogs with potent (∼0.5-2 µg/mL) activity against M. tuberculosis and S. aureus. Our results give a logical explanation of the observation that most new tuberculosis drug leads discovered by phenotypic screens and genome sequencing are highly lipophilic (logP ∼5.7) bases with membrane targets because such species are expected to partition into hydrophobic membranes, inhibiting membrane proteins, in addition to collapsing the PMF. This multiple targeting is expected to be of importance in overcoming the development of drug resistance because targeting membrane physical properties is expected to be less susceptible to the development of resistance.


Subject(s)
Anti-Infective Agents/pharmacology , Proton-Motive Force/drug effects , Uncoupling Agents/pharmacology , Alkyl and Aryl Transferases/antagonists & inhibitors , Anti-Infective Agents/chemistry , Biophysical Phenomena , Clomiphene/pharmacology , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular , Molecular Dynamics Simulation , Molecular Structure , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/growth & development , Piperidines/pharmacology , Quinolines/pharmacology , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Uncoupling Agents/chemistry
8.
AAPS PharmSciTech ; 19(7): 2898-2907, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30209787

ABSTRACT

This study described a pH-gradient dissolution method combined with flux measurements as an in vitro tool for assessing the risk of bioavailability reduction due to drug-drug interactions (DDI) caused by acid reducing agents (ARAs). The device incorporates absorption chambers into USP II dissolution vessels, with fiber optic UV-probes monitoring concentration in situ. Dosage forms of Genentech BCS class II drugs, GDC-0810, GDC-0941, and compound A, were tested by starting the dissolution in either pH 1.6 or pH 4.0 media then converting to FaSSIF after 30 min. GDC-0810 showed no significant difference in flux between the two conversion experiments. A supersaturation phase was observed for GDC-0941 in the pH 1.6 experiments after media conversion to FaSSIF; however, it did not appear to occur in the pH 4.0 experiment due to low drug solubility at pH 4.0, resulting in a 95% decrease in flux compared to pH 1.6 experiment. The extent of flux reduction and the total accumulated API mass in the absorption chamber agreed well with the 89% reduction in mean Cmax and the 82% reduction in mean AUC from dog PK study between animals treated with pentagastrin and famotidine. Testing of the compound A optimized formulation tablets showed a 25% reduction in flux and in vitro absorbed amount by changing pH 1.6 to 4.0, correlating well with the AUC decrease in clinical studies. Good correlation between in vitro data and in vivo PK data demonstrated the applicability of the method for formulators to develop drug products mitigating DDI from ARAs.


Subject(s)
Cinnamates/chemistry , Cinnamates/pharmacokinetics , Indazoles/chemistry , Indazoles/pharmacokinetics , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Dogs , Drug Interactions/physiology , Humans , Hydrogen-Ion Concentration , Proton-Motive Force/drug effects , Proton-Motive Force/physiology , Solubility , Tablets
9.
Biochim Biophys Acta ; 1858(2): 274-80, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26615918

ABSTRACT

Biologically active, artificially synthesized two-peptide bacteriocin PlnEF was used to study its mode of action on sensitive bacteria Lactobacillus plantarum pl2. The data obtained showed that PlnEF induced membrane permeabilization, allowing for the efflux of electrolytes, which was evidenced by the increased extracellular conductivity, the dissipation of transmembrane electrical potential and pH gradient, and rapid intracellular ATP depletion after L. plantarum pl2 cells were treated with PlnEF for minutes. Laser confocal microscopy showed that PlnEF accumulated very quickly in L. plantarum pl2 cells and the accumulation of PlnEF caused damage to cell membrane. Scanning electron microscopy and transmission electron microscopy further showed that PlnEF induced morphological changes and structure disruption to L. plantarum pl2 cells, such as the formation of blebs, microspheres, membrane deformation and cell lysis. In summary, the data obtained show that PlnEF caused cell membrane damage to L. plantarum pl2 cells. Our study reveals the antimicrobial mechanism of two-peptide bacteriocin PlnEF against L. plantarum.


Subject(s)
Bacteriocins/pharmacology , Cell Membrane/metabolism , Lactobacillus plantarum/metabolism , Membrane Potentials/drug effects , Proton-Motive Force/drug effects , Bacteriocins/chemistry , Cell Membrane/chemistry
10.
Nature ; 473(7346): 216-20, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21562562

ABSTRACT

Bacterial persistence is a state in which a sub-population of dormant cells, or 'persisters', tolerates antibiotic treatment. Bacterial persisters have been implicated in biofilms and in chronic and recurrent infections. Despite this clinical relevance, there are currently no viable means for eradicating persisters. Here we show that specific metabolic stimuli enable the killing of both Gram-negative (Escherichia coli) and Gram-positive (Staphylococcus aureus) persisters with aminoglycosides. This potentiation is aminoglycoside-specific, it does not rely on growth resumption and it is effective in both aerobic and anaerobic conditions. It proceeds by the generation of a proton-motive force which facilitates aminoglycoside uptake. Our results demonstrate that persisters, although dormant, are primed for metabolite uptake, central metabolism and respiration. We show that aminoglycosides can be used in combination with specific metabolites to treat E. coli and S. aureus biofilms. Furthermore, we demonstrate that this approach can improve the treatment of chronic infections in a mouse urinary tract infection model. This work establishes a strategy for eradicating bacterial persisters that is based on metabolism, and highlights the importance of the metabolic environment to antibiotic treatment.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Aerobiosis , Anaerobiosis , Animals , Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Disease Models, Animal , Drug Synergism , Escherichia coli Infections/drug therapy , Female , Mice , Proton-Motive Force/drug effects , Staphylococcal Infections/drug therapy , Urinary Tract Infections/drug therapy
11.
Proc Natl Acad Sci U S A ; 111(13): 4844-9, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24550475

ABSTRACT

The SecY/61 complex forms the protein-channel component of the ubiquitous protein secretion and membrane protein insertion apparatus. The bacterial version SecYEG interacts with the highly conserved YidC and SecDF-YajC subcomplex, which facilitates translocation into and across the membrane. Together, they form the holo-translocon (HTL), which we have successfully overexpressed and purified. In contrast to the homo-dimeric SecYEG, the HTL is a hetero-dimer composed of single copies of SecYEG and SecDF-YajC-YidC. The activities of the HTL differ from the archetypal SecYEG complex. It is more effective in cotranslational insertion of membrane proteins and the posttranslational secretion of a ß-barreled outer-membrane protein driven by SecA and ATP becomes much more dependent on the proton-motive force. The activity of the translocating copy of SecYEG may therefore be modulated by association with different accessory subcomplexes: SecYEG (forming SecYEG dimers) or SecDF-YajC-YidC (forming the HTL). This versatility may provide a means to refine the secretion and insertion capabilities according to the substrate. A similar modularity may also be exploited for the translocation or insertion of a wide range of substrates across and into the endoplasmic reticular and mitochondrial membranes of eukaryotes.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Proton-Motive Force , Adenosine Triphosphate/pharmacology , Cross-Linking Reagents/metabolism , Escherichia coli/drug effects , Escherichia coli Proteins/isolation & purification , Membrane Proteins/isolation & purification , Models, Biological , Protein Binding/drug effects , Protein Stability/drug effects , Protein Subunits/metabolism , Protein Transport/drug effects , Proton-Motive Force/drug effects , Ribosomes/drug effects , Ribosomes/metabolism
12.
Proc Natl Acad Sci U S A ; 110(40): 16169-74, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24046367

ABSTRACT

Identifying the mechanism of action for antibacterial compounds is essential for understanding how bacteria interact with one another and with other cell types and for antibiotic discovery efforts, but determining a compound's mechanism of action remains a serious challenge that limits both basic research and antibacterial discovery programs. Here, we show that bacterial cytological profiling (BCP) is a rapid and powerful approach for identifying the cellular pathway affected by antibacterial molecules. BCP can distinguish between inhibitors that affect different cellular pathways as well as different targets within the same pathway. We use BCP to demonstrate that spirohexenolide A, a spirotetronate that is active against methicillin-resistant Staphylococcus aureus, rapidly collapses the proton motive force. BCP offers a simple, one-step assay that can be broadly applied, solving the longstanding problem of how to rapidly determine the cellular target of thousands of compounds.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cytological Techniques/methods , Drug Evaluation, Preclinical/methods , Drug Resistance, Multiple, Bacterial/genetics , Staphylococcus aureus/drug effects , Flow Cytometry , Heterocyclic Compounds, 4 or More Rings/pharmacology , Microbial Sensitivity Tests , Microscopy, Fluorescence , Principal Component Analysis , Proton-Motive Force/drug effects
13.
Can J Microbiol ; 61(4): 263-71, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25728340

ABSTRACT

The antibacterial mechanism of a Cinnamomum cassia essential oil from Vietnam and of its main component (trans-cinnamaldehyde, 90% (m/m) of C. cassia essential oil) against a Listeria innocua strain was investigated to estimate their potential for food preservation. In the presence of C. cassia essential oil or trans-cinnamaldehyde at their minimal bactericidal concentration (2700 µg·mL(-1)), L. innocua cells fluoresced green after staining with Syto9® and propidium iodide, as observed by epifluorescence microscopy, suggesting that the perturbation of membrane did not cause large pore formation and cell lysis but may have introduced the presence of viable but nonculturable bacteria. Moreover, the fluidity, potential, and intracellular pH of the cytoplasmic membrane were perturbed in the presence of the essential oil or trans-cinnamaldehyde. However, these membrane perturbations were less severe in the presence of trans-cinnamaldehyde than in the presence of multicomponent C. cassia essential oil. This indicates that in addition to trans-cinnamaldehyde, other minor C. cassia essential oil components play a major role in its antibacterial activity against L. innocua cells.


Subject(s)
Acrolein/analogs & derivatives , Anti-Bacterial Agents/pharmacology , Cinnamomum/chemistry , Listeria/drug effects , Oils, Volatile/pharmacology , Plant Extracts/pharmacology , Acrolein/chemistry , Acrolein/pharmacology , Anti-Bacterial Agents/chemistry , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Listeria/chemistry , Listeria/growth & development , Listeria/metabolism , Membrane Fluidity/drug effects , Microbial Viability/drug effects , Oils, Volatile/chemistry , Plant Extracts/chemistry , Proton-Motive Force/drug effects
14.
Biol Res ; 48: 17, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25889629

ABSTRACT

BACKGROUND: Karwinskia humboldtiana (Kh) is a poisonous plant of the rhamnacea family. To elucidate some of the subcellular effects of Kh toxicity, membrane fluidity and ATPase activities as hydrolytic and as proton-pumping activity were assessed in rat liver submitochondrial particles. Rats were randomly assigned into control non-treated group and groups that received 1, 1.5 and 2 g/Kg body weight of dry powder of Kh fruit, respectively. Rats were euthanized at day 1 and 7 after treatment. RESULTS: Rats under Kh treatment at all dose levels tested, does not developed any neurologic symptoms. However, we detected alterations in membrane fluidity and ATPase activity. Lower dose of Kh on day 1 after treatment induced higher mitochondrial membrane fluidity than control group. This change was strongly correlated with increased ATPase activity and pH gradient driven by ATP hydrolysis. On the other hand, membrane fluidity was hardly affected on day 7 after treatment with Kh. Surprisingly, the pH gradient driven by ATPase activity was significantly higher than controls despite an diminution of the hydrolytic activity of ATPase. CONCLUSIONS: The changes in ATPase activity and pH gradient driven by ATPase activity suggest an adaptive condition whereby the fluidity of the membrane is altered.


Subject(s)
Adenosine Triphosphatases/metabolism , Karwinskia/toxicity , Membrane Fluidity/drug effects , Mitochondria, Liver/drug effects , Animals , Fruit/toxicity , Male , Mitochondria, Liver/enzymology , Proton-Motive Force/drug effects , Random Allocation , Rats, Sprague-Dawley , Subcellular Fractions/drug effects , Submitochondrial Particles/drug effects
15.
J Biol Chem ; 288(15): 10567-77, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23457298

ABSTRACT

Superoxide flashes are transient bursts of superoxide production within the mitochondrial matrix that are detected using the superoxide-sensitive biosensor, mitochondria-targeted circularly permuted YFP (mt-cpYFP). However, due to the pH sensitivity of mt-cpYFP, flashes were suggested to reflect transient events of mitochondrial alkalinization. Here, we simultaneously monitored flashes with mt-cpYFP and mitochondrial pH with carboxy-SNARF-1. In intact cardiac myocytes and purified skeletal muscle mitochondria, robust mt-cpYFP flashes were accompanied by only a modest increase in SNARF-1 ratio (corresponding to a pH increase of <0.1), indicating that matrix alkalinization is minimal during an mt-cpYFP flash. Individual flashes were also accompanied by stepwise increases of MitoSOX signal and decreases of NADH autofluorescence, supporting the superoxide origin of mt-cpYFP flashes. Transient matrix alkalinization induced by NH4Cl only minimally influenced flash frequency and failed to alter flash amplitude. However, matrix acidification modulated superoxide flash frequency in a bimodal manner. Low concentrations of nigericin (< 100 nM) that resulted in a mild dissipation of the mitochondrial pH gradient increased flash frequency, whereas a maximal concentration of nigericin (5 µm) collapsed the pH gradient and abolished flash activity. These results indicate that mt-cpYFP flash events reflect a burst in electron transport chain-dependent superoxide production that is coincident with a modest increase in matrix pH. Furthermore, flash activity depends strongly on a combination of mitochondrial oxidation and pH gradient.


Subject(s)
Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Superoxides/metabolism , Ammonium Chloride/pharmacology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Benzopyrans/pharmacology , Cells, Cultured , Electron Transport Chain Complex Proteins/metabolism , Fluorescent Dyes/pharmacology , Hydrogen-Ion Concentration , Ionophores/pharmacology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Naphthols/pharmacology , Nigericin/pharmacology , Oxidation-Reduction/drug effects , Proton-Motive Force/drug effects , Proton-Motive Force/physiology , Rats , Rats, Sprague-Dawley , Rhodamines/pharmacology
16.
Antimicrob Agents Chemother ; 58(9): 5519-27, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25001307

ABSTRACT

Bis-thiazolium salts constitute a new class of antihematozoan drugs that inhibit parasite phosphatidylcholine biosynthesis. They specifically accumulate in Plasmodium- and Babesia-infected red blood cells (IRBC). Here, we provide new insight into the choline analogue albitiazolium, which is currently being clinically tested against severe malaria. Concentration-dependent accumulation in P. falciparum-infected erythrocytes reached steady state after 90 to 120 min and was massive throughout the blood cycle, with cellular accumulation ratios of up to 1,000. This could not occur through a lysosomotropic effect, and the extent did not depend on the food vacuole pH, which was the case for the weak base chloroquine. Analysis of albitiazolium accumulation in P. falciparum IRBC revealed a high-affinity component that was restricted to mature stages and suppressed by pepstatin A treatment, and thus likely related to drug accumulation in the parasite food vacuole. Albitiazolium also accumulated in a second high-capacity component present throughout the blood cycle that was likely not related to the food vacuole and also observed with Babesia divergens-infected erythrocytes. Accumulation was strictly glucose dependent, drastically inhibited by H+/K+ and Na+ ionophores upon collapse of ionic gradients, and appeared to be energized by the proton-motive force across the erythrocyte plasma membrane, indicating the importance of transport steps for this permanently charged new type of antimalarial agent. This specific, massive, and irreversible accumulation allows albitiazolium to restrict its toxicity to hematozoa-infected erythrocytes. The intraparasitic compartmentation of albitiazolium corroborates a dual mechanism of action, which could make this new type of antimalarial agent resistant to parasite resistance.


Subject(s)
Antimalarials/metabolism , Erythrocytes/metabolism , Thiazoles/metabolism , Antimalarials/pharmacology , Babesia/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Drug Resistance/drug effects , Erythrocytes/drug effects , Humans , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Proton-Motive Force/drug effects , Thiazoles/pharmacology
17.
Antimicrob Agents Chemother ; 58(2): 923-30, 2014.
Article in English | MEDLINE | ID: mdl-24277026

ABSTRACT

Bacterial resistance to antibiotics and biocides is an increasing public health problem. Genes encoding integral membrane proteins belonging to the DedA family are present in most bacterial genomes, including Escherichia coli. An E. coli strain lacking partially redundant DedA family genes yqjA and yghB (strain BC202) displays temperature sensitivity and cell division defects. These phenotypes can be corrected by overexpression of mdfA, an Na(+)-K(+)/H(+) antiporter of the major facilitator superfamily. We show that BC202 is hypersensitive to several biocides and cationic compounds that are known substrates of several multidrug resistance transporters, including MdfA, EmrE, and AcrB. The introduction of deletions of genes encoding these drug transporters into BC202 results in additional sensitivity. Expression of wild-type yghB or yqjA can restore drug resistance, but this is eliminated upon mutation of two membrane-embedded acidic amino acids (E39 or D51 in either protein). This dependence upon membrane-embedded acidic amino acids is a hallmark of proton-dependent antiporters. Overexpression of mdfA in BC202 or artificially restoring proton motive force (PMF) restores wild-type resistance to substrates of MdfA as well as other drug resistance transporters such as EmrE and AcrAB. These results suggest that YqjA and YghB may be membrane transporters required for PMF-dependent drug efflux in E. coli.


Subject(s)
Conserved Sequence , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Proton-Motive Force/genetics , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Antiporters/genetics , Antiporters/metabolism , Cell Division , Drug Resistance, Bacterial/drug effects , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Molecular Sequence Data , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Mutation , Proton-Motive Force/drug effects , Sequence Alignment
18.
FASEB J ; 27(9): 3818-26, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23733749

ABSTRACT

Toward generating new tools for fighting multidrug-resistant (MDR) bacteria, we assessed the ability of a membrane-active peptide to sensitize gram-negative bacteria to various antibiotics. The mechanism for affecting inner and/or outer membrane functions was assessed by complementary biophysical methods (SPR, DSC, ITC). The implication of efflux pumps was examined using Acr-AB mutants, as tested with representative antibiotics, host defense peptides, and synthetic mimics. The ability to affect disease course systemically was compared for a single therapy and combination therapy, using the mouse thigh-infection model. The data show that potent antibiotic action can be provoked in vitro and in vivo, by a treatment combining two antibacterial compounds whose individual inefficiency against gram-negative bacteria stems from their efflux. Thus, at subminimal inhibitory concentrations, the lipopeptide-like sequence, N(α)(ω7)dodecenoyl-lysyl-[lysyl-aminododecanoyl-lysyl]-amide (designated C12(ω7)K-ß12), has, nonetheless, rapidly achieved a transient membrane depolarization, which deprived bacteria of the proton-motive force required for active efflux. Consequently, bacteria became significantly sensitive to intracellular targeting antibiotics. Collectively, these findings suggest a potentially useful approach for expanding the antibiotics sensitivity spectrum of MDR gram-negative bacteria to include efflux substrates.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Lipopeptides/pharmacology , Membrane Potentials/drug effects , Animals , Anti-Bacterial Agents/chemistry , Calorimetry, Differential Scanning , Lipopeptides/chemistry , Magnetic Resonance Spectroscopy , Male , Mice , Microbial Sensitivity Tests , Peptidomimetics , Proton-Motive Force/drug effects , Surface Plasmon Resonance , Thigh/microbiology
19.
FASEB J ; 27(7): 2862-72, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23515444

ABSTRACT

The type III secretion apparatus (T3SA) participates in the secretion of bacterial proteins called effectors, although the detailed mechanism of effector secretion remains unclear. T3SA and flagellum were shown to branch from a common ancestor and also show structural similarity. In addition, both T3SA-dependent effector secretion and flagellar rotation were reported to require proton-motive force (PMF) for activity. From these reports, we hypothesized that T3SA, like the flagellum, would rotate via PMF and that this rotation is responsible for effector secretion. To observe T3SA rotation, we constructed a novel observation system by modifying the tip of T3SA on bacterial cell membranes with an observation probe, which allowed documentation of T3SA rotation for the first time. T3SA rotation was stopped by the addition of a protonophore that decreases PMF. Moreover, increased viscosity of the observation medium inhibited both rotation of T3SA associated with beads and effector secretion. These results suggested that effector secretion would follow the PMF-dependent rotation of T3SA and could be inhibited by preventing T3SA rotation. Moreover, the motion-track analysis of bead rotation suggested that the T3SA needle might be flexible. Consequently, we propose a "rotational secretion model" as a novel effector secretion mechanism of T3SA.


Subject(s)
Bacterial Proteins/metabolism , Cell Membrane/physiology , Proton-Motive Force/physiology , Pseudomonas aeruginosa/physiology , ADP Ribose Transferases/metabolism , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Blotting, Western , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Flagella/metabolism , Flagella/physiology , GTPase-Activating Proteins/metabolism , Microscopy, Atomic Force , Models, Biological , Models, Molecular , Mutation , Oligopeptides/genetics , Oligopeptides/metabolism , Polyethylene Glycols/pharmacology , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Proton Ionophores/pharmacology , Proton-Motive Force/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Rotation
20.
Nature ; 451(7177): 489-92, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-18216859

ABSTRACT

Bacterial flagella contain a specialized secretion apparatus that functions to deliver the protein subunits that form the filament and other structures to outside the membrane. This apparatus is related to the injectisome used by many gram-negative pathogens and symbionts to transfer effector proteins into host cells; in both systems this export mechanism is termed 'type III' secretion. The flagellar secretion apparatus comprises a membrane-embedded complex of about five proteins, and soluble factors, which include export-dedicated chaperones and an ATPase, FliI, that was thought to provide the energy for export. Here we show that flagellar secretion in Salmonella enterica requires the proton motive force (PMF) and does not require ATP hydrolysis by FliI. The export of several flagellar export substrates was prevented by treatment with the protonophore CCCP, with no accompanying decrease in cellular ATP levels. Weak swarming motility and rare flagella were observed in a mutant deleted for FliI and for the non-flagellar type-III secretion ATPases InvJ and SsaN. These findings show that the flagellar secretion apparatus functions as a proton-driven protein exporter and that ATP hydrolysis is not essential for type III secretion.


Subject(s)
Energy Metabolism , Flagella/metabolism , Proton-Motive Force/physiology , Salmonella enterica/metabolism , Adenosine Triphosphate/metabolism , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Energy Metabolism/drug effects , Flagella/chemistry , Hydrogen-Ion Concentration , Mutation/genetics , Protein Transport/drug effects , Protein Transport/genetics , Proton-Motive Force/drug effects , Proton-Translocating ATPases/metabolism , Salmonella enterica/enzymology , Salmonella enterica/genetics
SELECTION OF CITATIONS
SEARCH DETAIL