Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Biol Pharm Bull ; 43(8): 1241-1247, 2020.
Article in English | MEDLINE | ID: mdl-32741944

ABSTRACT

Retinal pigment epithelial (RPE) cells form the outer blood-retinal barrier (BRB) and regulate drug/compound exchange between the neural retina and blood in the fenestrated blood vessels of retinal choroid via membrane transporters. Recent studies have elucidated that RPE cells express hemichannels, which are opened by extracellular Ca2+ depletion and accept several drugs/compounds as a transporting substrate. The objective of this study was to elucidate the hemichannel-mediated compound transport properties of the outer BRB. In human RPE cells, namely ARPE-19 cells, time-dependent uptake of fluorescent hemichannel substrates, such as Lucifer Yellow, sulforhodamine-101 (SR-101), and propidium iodide (PI) was promoted under Ca2+-depleted conditions. The uptake of these substrates under Ca2+-depleted conditions exhibited saturable kinetics with a Michaelis-Menten constant (Km) of 87-109 µM. In addition, SR-101 and PI uptake by ARPE-19 cells was dependent of extracellular Ca2+ concentration, and that under Ca2+-depleted conditions was significantly decreased by typical substrates and/or inhibitors for hemichannels. Moreover, Ca2+-depleted conditions promoted the efflux transport of calcein from ARPE-19 cells, and the promoted calcein efflux transport was significantly inhibited by a typical hemichannel inhibitor. These results suggested that hemichannels at the outer BRB were involved in the influx and efflux transport of drugs/compounds.


Subject(s)
Blood-Retinal Barrier/physiology , Calcium/physiology , Retinal Pigment Epithelium/metabolism , Cells, Cultured , Epithelial Cells/metabolism , Humans , Isoquinolines/pharmacokinetics , Propidium/pharmacokinetics , Retinal Pigment Epithelium/cytology , Rhodamines/pharmacokinetics
2.
Biochim Biophys Acta ; 1848(10 Pt A): 2118-25, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26112464

ABSTRACT

Ca2+ activation and membrane electroporation by 10-ns and 4-ms electric pulses (nsEP and msEP) were compared in rat embryonic cardiomyocytes. The lowest electric field which triggered Ca2+ transients was expectedly higher for nsEP (36 kV/cm) than for msEP (0.09 kV/cm) but the respective doses were similar (190 and 460 mJ/g). At higher intensities, both stimuli triggered prolonged firing in quiescent cells. An increase of basal Ca2+ level by >10 nM in cells with blocked voltage-gated Ca2+ channels and depleted Ca2+ depot occurred at 63 kV/cm (nsEP) or 0.14 kV/cm (msEP) and was regarded as electroporation threshold. These electric field values were at 150-230% of stimulation thresholds for both msEP and nsEP, notwithstanding a 400,000-fold difference in pulse duration. For comparable levels of electroporative Ca2+ uptake, msEP caused at least 10-fold greater uptake of propidium than nsEP, suggesting increased yield of larger pores. Electroporation by msEP started Ca2+ entry abruptly and locally at the electrode-facing poles of cell, followed by a slow diffusion to the center. In a stark contrast, nsEP evoked a "supra-electroporation" pattern of slower but spatially uniform Ca2+ entry. Thus nsEP and msEP had comparable dose efficiency, but differed profoundly in the size and localization of electropores.


Subject(s)
Cell Membrane Permeability/physiology , Electroporation/methods , Myocytes, Cardiac/physiology , Myocytes, Cardiac/radiation effects , Propidium/pharmacokinetics , Animals , Cell Membrane Permeability/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Metabolic Clearance Rate/radiation effects , Radiation Dosage , Rats , Static Electricity
3.
Circ Res ; 110(10): 1311-21, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22492531

ABSTRACT

RATIONALE: Electrical conduction through gap junction channels between endothelial cells of resistance vessels is integral to blood flow control. Small and intermediate-conductance Ca(2+)-activated K(+) channels (SK(Ca)/IK(Ca)) initiate electrical signals in endothelial cells, but it is unknown whether SK(Ca)/IK(Ca) activation alters signal transmission along the endothelium. OBJECTIVE: We tested the hypothesis that SK(Ca)/IK(Ca) activity regulates electrical conduction along the endothelium of resistance vessels. METHODS AND RESULTS: Freshly isolated endothelial cell tubes (60 µm wide; 1-3 mm long; cell length, ≈35 µm) from mouse skeletal muscle feed (superior epigastric) arteries were studied using dual intracellular microelectrodes. Current was injected (±0.1-3 nA) at site 1 while recording membrane potential (V(m)) at site 2 (separation distance=50-2000 µm). SK(Ca)/IK(Ca) activation (NS309, 1 µmol/L) reduced the change in V(m) along endothelial cell tubes by ≥50% and shortened the electrical length constant (λ) from 1380 to 850 µm (P<0.05) while intercellular dye transfer (propidium iodide) was maintained. Activating SK(Ca)/IK(Ca) with acetylcholine or SKA-31 also reduced electrical conduction. These effects of SK(Ca)/IK(Ca) activation persisted when hyperpolarization (>30 mV) was prevented with 60 mmol/L [K(+)](o). Conversely, blocking SK(Ca)/IK(Ca) (apamin+charybdotoxin) depolarized cells by ≈10 mV and enhanced electrical conduction (ie, changes in V(m)) by ≈30% (P<0.05). CONCLUSIONS: These findings illustrate a novel role for SK(Ca)/IK(Ca) activity in tuning electrical conduction along the endothelium of resistance vessels by governing signal dissipation through changes in membrane resistance. Voltage-insensitive ion channels can thereby tune intercellular electrical signaling independent from gap junction channels.


Subject(s)
Endothelium, Vascular/physiology , Epigastric Arteries/physiology , Gap Junctions/physiology , Potassium Channels, Calcium-Activated/physiology , Vascular Resistance/physiology , Acetylcholine/pharmacology , Animals , Benzothiazoles/pharmacology , Electric Conductivity , Epigastric Arteries/drug effects , Indicators and Reagents/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Microelectrodes , Nitric Oxide/metabolism , Potassium Channels, Calcium-Activated/agonists , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Propidium/pharmacokinetics , Regional Blood Flow/physiology , Signal Transduction/physiology , Vascular Resistance/drug effects , Vasodilator Agents/pharmacology
4.
J Theor Biol ; 360: 83-94, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25010659

ABSTRACT

The aim of this paper is to present a new model of in vitro cell electropermeabilization, which describes separately the conducting state and the permeable state of the membrane submitted to high voltage pulses. We first derive the model based on the experimental observations and we present the numerical methods to solve the non-linear partial differential equations. We then present numerical simulations that corroborate qualitatively the experimental data dealing with the uptake of propidium iodide (PI) after millipulses. This tends to justify the validity of our modeling. Forthcoming work will be to calibrate the parameters of the model for quantitative description of the uptake.


Subject(s)
Cell Membrane/metabolism , Electric Conductivity , Models, Biological , Computer Simulation , In Vitro Techniques , Permeability , Propidium/pharmacokinetics
5.
Biol Pharm Bull ; 37(12): 1958-62, 2014.
Article in English | MEDLINE | ID: mdl-25451844

ABSTRACT

We previously demonstrated that the P2X7 receptor (P2X7R), a purinergic receptor, expressed by mouse cultured cortical astrocytes is constitutively activated without any exogenous stimulus, differing from the case of neurons. It is well known that astrocytic morphology differs between in vitro and in vivo situations, implying different functionalities. Brain acute slices are widely accepted as an in vitro experimental system that reflects in vivo cell conditions better than in vitro cell culture ones. We examined whether astrocytic P2X7Rs exhibited constitutive activation in mouse cortical slices. In acute cortical slices, P2X7R-immunoreactivity was detected in both glial fibrillary acidic protein-immunopositive astrocytes and microtubule-associated protein 2-immunopositive neurons. Astrocytic, but not neuronal, spontaneous uptake of propidium iodide, an indicator of P2X7R channel/pore activity, was inhibited by representative antagonists of P2X7R, but they had no effect on the uptake by astrocytes in membrane-permeabilized fixed slices. These findings indicate that astrocytes, but not neurons, in acute cortical slices exhibit constitutive activation of P2X7Rs under non-stimulated resting conditions as in the case of cell culture systems.


Subject(s)
Astrocytes/metabolism , Brain/cytology , Neurons/metabolism , Receptors, Purinergic P2X7/metabolism , Animals , Biological Transport , Cells, Cultured , Female , Gene Expression Regulation/physiology , Mice , Propidium/pharmacokinetics , Receptors, Purinergic P2X7/genetics , Staining and Labeling
6.
J Cell Physiol ; 227(4): 1428-37, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21618533

ABSTRACT

Purinergic receptors in lens epithelium suggest lens function can be altered by chemical signals from aqueous humor or the lens itself. Here we show release of ATP by intact porcine lenses exposed to hyposmotic solution (200 mOsm). 18α-glycyrrhetinic acid (AGA) added together with probenecid eliminated the ATP increase. N-ethylmaleimide (200 µM), an exocytotic inhibitor, had no significant effect on ATP increase. Lenses exposed to hyposmotic solution displayed a ~400% increase of propidium iodide (PI) entry into the epithelium. The increased ability of PI (MW 668) to enter the epithelium suggests possible opening of connexin and/or pannexin hemichannels. This is consistent with detection of connexin 43, connexin 50, and pannexin 1 in the epithelium and the ability of AGA + probenecid to prevent ATP release. Na,K-ATPase activity doubled in the epithelium of lenses exposed to hyposmotic solution. The increase of Na,K-ATPase activity did not occur when apyrase was used to prevent extracellular ATP accumulation or when AGA + probenecid prevented ATP release. The increase of Na,K-ATPase activity was inhibited by the purinergic P2 antagonist reactive blue-2 and pertussis toxin, a G-protein inhibitor, but not by the P2X antagonist PPADS. Hyposmotic solution activated Src family kinase (SFK) in the epithelium, judged by Western blot. The SFK inhibitor PP2 abolished both SFK activation and the Na,K-ATPase activity increase. In summary, hyposmotic shock-induced ATP release is sufficient to activate a purinergic receptor- and SFK-dependent mechanism that stimulates Na,K-ATPase activity. The responses might signify an autoregulatory loop initiated by mechanical stress or osmotic swelling.


Subject(s)
Adenosine Triphosphate/metabolism , Lens, Crystalline/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Connexins/metabolism , Epithelium/metabolism , Homeostasis , Hypotonic Solutions , In Vitro Techniques , Osmotic Pressure , Propidium/pharmacokinetics , Receptors, Purinergic/metabolism , Sus scrofa , src-Family Kinases/metabolism
7.
Biol Pharm Bull ; 34(6): 921-4, 2011.
Article in English | MEDLINE | ID: mdl-21628896

ABSTRACT

In our previous study, we reported that urechistachykinin I (U I) and II (U II) exerted antimicrobial effects. To find out how the tachykinin consensus sequence of the urechistachykinin peptide family affects its antimicrobial activity, analogues substituting the amino acid residues phenylalanine (Phe-6; Anal 1), glycine (Gly-8; Anal 2), and arginine (Arg-10; Anal 3) of U II to alanine (Ala) were designed. Subsequently, the antimicrobial activity was shown on the order of Anal 3>U II=Anal 2>Anal 1, and this activity pattern was correlated with membrane studies such as propidium iodide (PI) influx and fluorescein isothiocyanate dextran (FD) leakage assay. These results suggest that the antimicrobial activity is related to the hydrophobicity values of the peptides. In regards to the activity of U II, it is determined that the hydrophobic Phe-6 plays a more critical role than Gly-8 or Arg-10.


Subject(s)
Anti-Infective Agents/pharmacology , Conserved Sequence , Neuropeptides/chemistry , Peptide Fragments/pharmacology , Tachykinins/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Annelida/metabolism , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/chemistry , Biological Transport/drug effects , Cell Membrane Permeability/drug effects , Circular Dichroism , Consensus Sequence , Dextrans/pharmacokinetics , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacokinetics , Fungi/drug effects , Fungi/metabolism , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Hydrophobic and Hydrophilic Interactions , Indicators and Reagents/pharmacokinetics , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Propidium/pharmacokinetics , Protein Structure, Secondary
8.
Biotechniques ; 69(2): 88-98, 2020 08.
Article in English | MEDLINE | ID: mdl-32393121

ABSTRACT

Fluorophores SYTO 9 and propidium iodide (PI) are extensively applied in medicine, food industry and environmental monitoring to assess the viability of bacteria. However, the actual performance of these dyes remains largely unknown. In addition, their effects on the physiology of cells have not been elucidated. Here we characterized the effects of these two dyes on Brevibacillus brevis under optimized staining. We found that SYTO 9 entered cells continuously while PI tended to adhere to the cell wall before entering the cell. In addition, results showed that a high amount of the dyes altered the physicochemical properties of membranes, improving their breakthrough. These results provide new perspectives and ideas for improving the characterization of bacterial viability using flow cytometry.


Subject(s)
Brevibacillus , Fluorescent Dyes , Organic Chemicals , Propidium , Brevibacillus/cytology , Brevibacillus/drug effects , Brevibacillus/metabolism , Cell Wall/chemistry , Cell Wall/metabolism , Flow Cytometry , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Kinetics , Microbial Viability/drug effects , Organic Chemicals/chemistry , Organic Chemicals/pharmacokinetics , Propidium/chemistry , Propidium/pharmacokinetics
9.
Food Microbiol ; 26(5): 542-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19465252

ABSTRACT

The effect of thermal and pressure treatments on Lactobacillus rhamnosus ATCC 53103 was evaluated by flow cytometric analysis in conjunction to standard cultivation techniques. A double staining technique with fluorochromes carboxyfluorescein diacetate (cFDA) and propidium iodide (PI) revealed that depending on temperature regime used heat-killed cells had different fluorescence behaviors. Cells killed at 60 degrees C were not stained at all whereas heat treatment at 75 degrees C resulted in a single population entirely labelled by PI. These findings indicated that thermal-induced cell death was achievable with or without membrane degradation. Hydrostatic pressures beyond 400 MPa inactivated L. rhamnosus ATCC 53103 in a different way. It was observed that the irreversible damage of the membrane-bound transport systems could be largely accounted for the cause of high pressure-induced cell death.


Subject(s)
Flow Cytometry , Fluorescent Dyes/pharmacokinetics , Food Preservation/methods , Hot Temperature , Hydrostatic Pressure , Lacticaseibacillus rhamnosus/growth & development , Cell Separation , Colony Count, Microbial , Fluoresceins/pharmacokinetics , Food Microbiology , Kinetics , Probiotics , Propidium/pharmacokinetics
10.
Bioelectrochemistry ; 122: 123-133, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29627664

ABSTRACT

Bipolar cancellation refers to a phenomenon when applying a second electric pulse reduces ("cancels") cell membrane damage by a preceding electric pulse of the opposite polarity. Bipolar cancellation is a reason why bipolar nanosecond electric pulses (nsEP) cause weaker electroporation than just a single unipolar phase of the same pulse. This study was undertaken to explore the dependence of bipolar cancellation on nsEP parameters, with emphasis on the amplitude ratio of two opposite polarity phases of a bipolar pulse. Individual cells (CHO, U937, or adult mouse ventricular cardiomyocytes (VCM)) were exposed to either uni- or bipolar trapezoidal nsEP, or to nanosecond electric field oscillations (NEFO). The membrane injury was evaluated by time-lapse confocal imaging of the uptake of propidium (Pr) or YO-PRO-1 (YP) dyes and by phosphatidylserine (PS) externalization. Within studied limits, bipolar cancellation showed little or no dependence on the electric field intensity, pulse repetition rate, chosen endpoint, or cell type. However, cancellation could increase for larger pulse numbers and/or for longer pulses. The sole most critical parameter which determines bipolar cancellation was the phase ratio: maximum cancellation was observed with the 2nd phase of about 50% of the first one, whereas a larger 2nd phase could add a damaging effect of its own. "Swapping" the two phases, i.e., delivering the smaller phase before the larger one, reduced or eliminated cancellation. These findings are discussed in the context of hypothetical mechanisms of bipolar cancellation and electroporation by nsEP.


Subject(s)
Cell Membrane Permeability , Electroporation/methods , Propidium/pharmacokinetics , Quinolinium Compounds/pharmacokinetics , Animals , Benzoxazoles/administration & dosage , Benzoxazoles/pharmacokinetics , CHO Cells , Cell Membrane/metabolism , Cells, Cultured , Cricetulus , Electricity , Female , Mice , Myocytes, Cardiac/metabolism , Phosphatidylserines/metabolism , Propidium/administration & dosage , Quinolinium Compounds/administration & dosage
11.
IEEE Trans Nanobioscience ; 17(1): 62-69, 2018 03.
Article in English | MEDLINE | ID: mdl-29570076

ABSTRACT

Electroporation, a physical transfection method to introduce genomic molecules in selective living cells, could be implemented by microelectrode devices. A local electric field generated by a finer electrode can induces cytomembrane poration in the electrode vicinity. To employ fine, high-speed scanning electrodes, we developed a fine virtual cathode pattern, which was generated on a cell adhesive surface of 100-nm-thick SiN membrane by inverted-electron beam lithography. The SiN membrane works as both a vacuum barrier and the display screen of the virtual cathode. The kinetic energy of the incident primary electrons to the SiN membrane was completely blocked, whereas negative charges and leaking electric current appeared on the surface of the dielectric SiN membrane within a region of 100 nm. Locally controlled transmembrane molecular delivery was demonstrated on adhered C2C12 myoblast cells in a culturing medium with fluorescent dye propidium iodide (PI). Increasing fluorescence of pre-diluted PI indicated local poration and transmembrane inflow at the virtual cathode position, as well as intracellular diffusion. The transmembrane inflows depended on beam duration time and acceleration voltage. At the post-molecular delivery, a slight decrease in intracellular PI fluorescence intensity indicates membrane recovery from the poration. Cell viability was confirmed by time-lapse cell imaging of post-exposure cell migration.


Subject(s)
Drug Delivery Systems/instrumentation , Electroporation/instrumentation , Electroporation/methods , Animals , Cell Line , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane Permeability , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Mice , Microelectrodes , Propidium/chemistry , Propidium/pharmacokinetics
12.
PLoS One ; 13(1): e0191522, 2018.
Article in English | MEDLINE | ID: mdl-29342216

ABSTRACT

The incidence of multidrug-resistant (MDR) organisms, including methicillin-resistant Staphylococcus aureus (MRSA), is a serious threat to public health. Progress in developing new therapeutics is being outpaced by antibiotic resistance development, and alternative agents that rapidly permeabilize bacteria hold tremendous potential for treating MDR infections. A new class of glycopolymers includes polycationic poly-N (acetyl, arginyl) glucosamine (PAAG) is under development as an alternative to traditional antibiotic strategies to treat MRSA infections. This study demonstrates the antibacterial activity of PAAG against clinical isolates of methicillin and mupirocin-resistant Staphylococcus aureus. Multidrug-resistant S. aureus was rapidly killed by PAAG, which completely eradicated 88% (15/17) of all tested strains (6-log reduction in CFU) in ≤ 12-hours at doses that are non-toxic to mammalian cells. PAAG also sensitized all the clinical MRSA strains (17/17) to oxacillin as demonstrated by the observed reduction in the oxacillin MIC to below the antibiotic resistance breakpoint. The effect of PAAG and standard antibiotics including vancomycin, oxacillin, mupirocin and bacitracin on MRSA permeability was studied by measuring propidium iodide (PI) uptake by bacterial cells. Antimicrobial resistance studies showed that S. aureus developed resistance to PAAG at a rate slower than to mupirocin but similar to bacitracin. PAAG was observed to resensitize drug-resistant S. aureus strains sampled from passage 13 and 20 of the multi-passage resistance study, reducing MICs of mupirocin and bacitracin below their clinical sensitivity breakpoints. This class of bacterial permeabilizing glycopolymers may provide a new tool in the battle against multidrug-resistant bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Glucosamine/analogs & derivatives , Methicillin-Resistant Staphylococcus aureus/drug effects , Polymers/pharmacology , Polysaccharides/pharmacology , Anti-Bacterial Agents/chemistry , Drug Resistance, Multiple, Bacterial , Glucosamine/chemistry , Glucosamine/pharmacology , Glycosides , Humans , In Vitro Techniques , Methicillin Resistance , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/metabolism , Microbial Sensitivity Tests , Mupirocin/pharmacology , Permeability/drug effects , Polymers/chemistry , Polysaccharides/chemistry , Propidium/pharmacokinetics , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology
13.
Brain Res ; 1147: 39-50, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17368578

ABSTRACT

The blood-brain barrier (BBB) is a structural and functional barrier that regulates the passage of molecules into and out of the brain to maintain the neural microenvironment. We have previously developed the in vitro BBB model with human brain microvascular endothelial cells (HBMEC). However, in vivo HBMEC are shown to interact with astrocytes and also exposed to shear stress through blood flow. In an attempt to develop the BBB model to mimic the in vivo condition we constructed the flow-based in vitro BBB model using HBMEC and human fetal astrocytes (HFA). We also examined the effect of astrocyte-conditioned medium (ACM) in lieu of HFA to study the role of secreted factor(s) on the BBB properties. The tightness of HBMEC monolayer was assessed by the permeability of dextran and propidium iodide as well as by measuring the transendothelial electrical resistance (TEER). We showed that the HBMEC permeability was reduced and TEER was increased by non-contact, co-cultivation with HFA and ACM. The exposure of HBMEC to shear stress also exhibited decreased permeability. Moreover, HFA/ACM and shear flow exhibited additive effect of decreasing the permeability of HBMEC monolayer. In addition, we showed that the HBMEC expression of ZO-1 (tight junction protein) was increased by co-cultivation with ACM and in response to shear stress. These findings suggest that the non-contact co-cultivation with HFA helps maintain the barrier properties of HBMEC by secreting factor(s) into the medium. Our in vitro flow model system with the cells of human origin should be useful for studying the interactions between endothelial cells, glial cells, and secreted factor(s) as well as the role of shear stress in the barrier property of HBMEC.


Subject(s)
Astrocytes/physiology , Blood-Brain Barrier/physiology , Brain/physiology , Capillary Permeability/physiology , Endothelial Cells/physiology , Indicators and Reagents/pharmacokinetics , Biological Factors/metabolism , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Brain/blood supply , Brain/cytology , Capillary Permeability/drug effects , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Dextrans/pharmacokinetics , Endothelial Cells/drug effects , Humans , Propidium/pharmacokinetics , Shear Strength , Stress, Mechanical
14.
Toxicology ; 238(1): 60-9, 2007 Aug 16.
Article in English | MEDLINE | ID: mdl-17618031

ABSTRACT

Recombinant human P2X(7) receptors, C-terminally labelled with enhanced green fluorescent protein (P2X(7)-EGFP), were transiently expressed in HEK293 cells. Activation of these receptors by their preferential agonist 2',3'-O-(4-benzoylbenzoyl)-ATP (BzATP) induced inward currents and propidium ion uptake indicating the opening of cationic channels and of large pores permeable for dye molecules, respectively. Two mutants of P2X(7) receptors (P2X(7)-EGFP-I568N, -E496A) representing polymorphisms in the P2X(7) gene known to interfere with normal receptor-trafficking and with optimal assembly of its subunits, responded with much lower current amplitudes to BzATP than their wild-type counterpart. Similarly, the normal propidium ion uptake induced by BzATP at the wild-type P2X(7) receptor was abolished by the two mutants. Confocal laser scanning microscopy indicated that in vitro ischemia of 12h duration increased the integration of P2X(7)-EGFP, but not of its two mutants, into the plasma membrane of HEK293 cells. Further, this ischemic stimulus facilitated the current response to BzATP in HEK293 cells permanently transfected with P2X(7) receptors. Finally, the fluorescence intensity per cell measured by flow cytometry and P2X(7) antibodies directed against an extracellular, but not an intracellular epitope of the receptor, were also increased. In conclusion, P2X(7) receptors may alter their trafficking properties during ischemia and thereby contribute to the ATP-induced damage of various cell-types including neurons.


Subject(s)
Cell Membrane/metabolism , Glucose/pharmacology , Oxygen/pharmacology , Receptors, Purinergic P2/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Apoptosis/drug effects , Cell Hypoxia , Cell Line , Cell Membrane/physiology , Dose-Response Relationship, Drug , Flow Cytometry , Glucose/deficiency , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Membrane Potentials/drug effects , Microscopy, Confocal , Mutation , Patch-Clamp Techniques , Propidium/metabolism , Propidium/pharmacokinetics , Protein Transport/drug effects , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/physiology , Receptors, Purinergic P2X7 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology , Time Factors , Transfection
15.
Clin Cancer Res ; 12(4): 1355-64, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16489094

ABSTRACT

PURPOSE: Heterogeneous sensitivity of melanoma cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis may lead to outgrowth of TRAIL-resistant cells and limit successful treatment by TRAIL. The present study aims to better understand the biological characteristics of melanoma cells resistant to TRAIL-induced apoptosis. EXPERIMENTAL DESIGN: We generated TRAIL-resistant melanoma cells by prolonged exposure to TRAIL and characterized the cells in terms of their sensitivity to killing induced by a panel of cytotoxic agents using biological and biochemical methods. RESULTS: TRAIL-resistant melanoma cells are cross-resistant to apoptosis induced by another death ligand FasL, the DNA-damaging agent cisplatin, the histone deacetylase inhibitor suberic bishydroxamate, and the antimicrotubule Vinca alkaloid, vincristine. The apoptotic signaling seemed to be inhibited upstream of mitochondrial apoptotic events and was associated with decreased expression of multiple apoptotic mediators, including pro-caspase-8, Fas-associated death domain, Bid, Bim, p53, and the products of its proapoptotic target genes. Despite being resistant to apoptosis, TRAIL-resistant melanoma cells were more vulnerable to cisplatin-induced nonapoptotic cell death. This was characterized by lack of DNA fragmentation, delayed externalization of phosphatidylserine, caspase and p53 independence, and severe mitochondrial disruption, and was preceded by poly(ADP)ribose polymerase (PARP) activation and depletion of intracellular ATP, indicative of necrotic cell death. Inhibition of PARP activity partially converted the mode of cell death from necrosis to apoptosis. CONCLUSIONS: TRAIL-resistant melanoma cells are cross-resistant to apoptosis induced by various apoptotic stimuli but are more sensitive to nonapoptotic cell death induced by cisplatin. Exploration of chemotherapy-induced nonapoptotic cell death may provide an alternative strategy in overcoming resistance of melanoma cells to TRAIL-induced apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Fas Ligand Protein , Flow Cytometry , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Melanoma/metabolism , Melanoma/pathology , Membrane Potentials/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Necrosis/chemically induced , Poly(ADP-ribose) Polymerases , Propidium/pharmacokinetics , TNF-Related Apoptosis-Inducing Ligand , Time Factors , Tumor Necrosis Factors/pharmacology , Tumor Suppressor Protein p53/metabolism , Vincristine/pharmacology
16.
Int J Nanomedicine ; 12: 5659-5672, 2017.
Article in English | MEDLINE | ID: mdl-28848345

ABSTRACT

PURPOSE: Pulsed-laser irradiation of light-absorbing gold nanoparticles (AuNPs) attached to cells transiently increases cell membrane permeability for targeted molecule delivery. Here, we targeted EGFR on the ovarian carcinoma cell line OVCAR-3 with AuNPs. In order to optimize membrane permeability and to demonstrate molecule delivery into adherent OVCAR-3 cells, we systematically investigated different experimental conditions. MATERIALS AND METHODS: AuNPs (30 nm) were functionalized by conjugation of the antibody cetuximab against EGFR. Selective binding of the particles was demonstrated by silver staining, multiphoton imaging, and fluorescence-lifetime imaging. After laser irradiation, membrane permeability of OVCAR-3 cells was studied under different conditions of AuNP concentration, cell-incubation medium, and cell-AuNP incubation time. Membrane permeability and cell viability were evaluated by flow cytometry, measuring propidium iodide and fluorescein isothiocyanate-dextran uptake. RESULTS: Adherently growing OVCAR-3 cells can be effectively targeted with EGFR-AuNP. Laser irradiation led to successful permeabilization, and 150 kDa dextran was successfully delivered into cells with about 70% efficiency. CONCLUSION: Antibody-targeted and laser-irradiated AuNPs can be used to deliver molecules into adherent cells. Efficacy depends not only on laser parameters but also on AuNP:cell ratio, cell-incubation medium, and cell-AuNP incubation time.


Subject(s)
Cell Membrane Permeability/drug effects , Drug Delivery Systems/methods , Gold/chemistry , Lasers , Metal Nanoparticles/chemistry , Cell Line, Tumor , Cell Membrane Permeability/radiation effects , Cell Survival/drug effects , Cetuximab/administration & dosage , Cetuximab/chemistry , Dextrans/pharmacokinetics , ErbB Receptors/immunology , ErbB Receptors/metabolism , Female , Flow Cytometry/methods , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacokinetics , Humans , Metal Nanoparticles/administration & dosage , Molecular Targeted Therapy , Propidium/pharmacokinetics
17.
J Neurosci Methods ; 269: 1-5, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27179931

ABSTRACT

BACKGROUND: Organotypic hippocampal slice cultures (OHSCs) are an attractive in vitro model to examine mechanisms of neuronal injury, because the normal hippocampal architecture, function and cellular diversity are mostly preserved. The effects of exposure to excitotoxins such as N-methyl-d-aspartate (NMDA) on cell viability can be determined by propidium iodide (PI) staining. NEW METHOD: We describe a simple method to objectively quantify cell death in NMDA exposed slice cultures using PI that provides a standardized means of quantifying cell death in hippocampal subfields without the need to induce maximal cell death in each slice. The method employs separation of subfields using simple landmarks and densitometric quantification of PI intensity in 10 template-oriented counting fields. RESULTS: We show that exposure to increasing concentrations of NMDA results in a dose-dependent increase in PI uptake. Additionally, our method facilitates the comparison of cell death in different hippocampal subfields, such as dentate gyrus, CA1 and CA3. Our results show marked differences of PI uptake in the hippocampal regions with the CA1 area being most sensitive to NMDA-induced injury. COMPARISON WITH EXISTING METHOD(S): The method provides a standardized format for quantifying PI exclusion in OHSCs that can be applied to cultures of differing shapes and sizes, permits comparisons between hippocampal subfields and does not require induction of maximal cell death. CONCLUSION: The method of quantifying PI uptake described herein allows for an objective, quantitative and reproducible analysis and comparison of cell death in distinct regions of OHSCs.


Subject(s)
Cell Death/drug effects , Hippocampus/drug effects , N-Methylaspartate/toxicity , Propidium/pharmacokinetics , Staining and Labeling , Tissue Culture Techniques , Animals , Cell Count , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Densitometry , Dose-Response Relationship, Drug , Female , Hippocampus/metabolism , Hippocampus/pathology , Image Processing, Computer-Assisted , Male , Microscopy, Fluorescence , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Rats, Sprague-Dawley
18.
Cell Death Differ ; 8(1): 83-99, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11313706

ABSTRACT

Biological activities of oxysterols seem tightly regulated. Therefore, the ability to induce cell death of structurally related oxysterols, such as those oxidized at C7(7alpha-, 7beta-hydroxycholesterol, and 7-ketocholesterol), was investigated on U937 cells at different times of treatment in a concentration range of 5-80 microg/ml. Whereas all oxysterols accumulate inside the cells, strong inhibition of cell growth and increased permeability to propidium iodide were observed only with 7beta-hydroxycholesterol and 7-ketocholesterol, which trigger an apoptotic process characterized by the occurrence of cells with fragmented and/or condensed nuclei, and by various cellular dysfunctions: loss of mitochondrial transmembrane potential, cytosolic release of cytochrome c, activation of caspase-9 and -3 with subsequent enhanced activity of caspase-3, degradation of poly(ADP-ribose) polymerase, and increased accumulation of cellular C16 : 0 and C24 : 1 ceramide species. This ceramide generation is not attributed to caspase activation since inhibition of 7beta-hydroxycholesterol- and 7-ketocholesterol-induced apoptosis by Z-VAD-fmk (100 microM), a broad spectrum caspase inhibitor, did not reduce C16 : 0 and C24 : 1 ceramide species accumulation. Conversely, when U937 cells were treated with 7beta-hydroxycholesterol and 7-ketocholesterol in the presence of fumonisin B1 (100 microM), a specific inhibitor of ceramide synthase, C16 : 0 and C24 : 1 ceramide species production was completely abrogated whereas apoptosis was not prevented. Noteworthy, 7alpha-hydroxycholesterol induced only a slight inhibition of cell growth. Collectively, these results are consistent with the notion that the alpha or beta hydroxyl radical position of oxysterols oxidized at C7 plays a key role in the induction of the apoptotic process. In addition, our findings demonstrate that 7beta-hydroxycholesterol- and 7-ketocholesterol-induced apoptosis involve the mitochondrial signal transduction pathway and they suggest that C16 : 0 and C24 : 1 ceramide species generated through ceramide synthase play a minor role in the commitment of 7beta-hydroxycholesterol- and 7-ketocholesterol-induced cell death.


Subject(s)
Apoptosis , Caspases/metabolism , Ceramides/biosynthesis , Fumonisins , Hydroxycholesterols/pharmacology , Ketocholesterols/pharmacology , U937 Cells/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Carboxylic Acids/pharmacology , Caspase 3 , Caspase 9 , Caspase Inhibitors , Cell Death/drug effects , Cell Division/drug effects , Cell Membrane Permeability/drug effects , Cytochrome c Group/metabolism , Cytosol/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Hydroxycholesterols/pharmacokinetics , Ketocholesterols/pharmacokinetics , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Propidium/pharmacokinetics , U937 Cells/cytology , U937 Cells/metabolism
19.
Neuroscience ; 136(1): 259-67, 2005.
Article in English | MEDLINE | ID: mdl-16182452

ABSTRACT

Many patients display elevated levels of serum cortisol following acute ischemic stroke. Given that glucocorticoids may potentiate some forms of insult, these studies examined the effects of corticosterone or dexamethasone exposure on cytotoxicity following oxygen-glucose deprivation in the cerebellum, a brain region susceptible to stroke. In organotypic cerebellar slice cultures prepared from neonatal rat pups, 90-min of oxygen-glucose deprivation at 15 days in vitro resulted in significant cytotoxicity at 24-, 48-, and 72-h post-oxygen-glucose deprivation, as measured by uptake of propidium iodide. Exposure of cultures following oxygen-glucose deprivation to the antioxidant trolox (500 microM), but not to the glucocorticoid receptor antagonist RU486 (10 microM), completely blocked oxygen-glucose deprivation-induced cytotoxicity. Corticosterone (1 microM) or dexamethasone (10 microM) exposure alone did not significantly increase propidium iodide uptake above levels observed in control cultures. However, corticosterone or dexamethasone exposure after oxygen-glucose deprivation potentiated oxygen-glucose deprivation-mediated propidium iodide uptake at each time point. Trolox, as well as RU486, co-exposure of cultures to corticosterone or dexamethasone after oxygen-glucose deprivation abolished all cytotoxicity. In conclusion, these data demonstrated that glucocorticoid exposure modulated oxygen-glucose deprivation-mediated propidium iodide uptake, which likely involved glucocorticoid receptor activation and pro-oxidant effects.


Subject(s)
Cerebellum/drug effects , Cerebellum/physiopathology , Corticosterone/pharmacology , Dexamethasone/pharmacology , Glucose/deficiency , Hypoxia/physiopathology , Animals , Antioxidants/pharmacology , Cell Death/drug effects , Cerebellum/metabolism , Chromans/pharmacology , Drug Synergism , Female , In Vitro Techniques , Male , Mifepristone/pharmacology , Propidium/pharmacokinetics , Rats , Rats, Sprague-Dawley
20.
Neurotox Res ; 28(4): 352-60, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26286504

ABSTRACT

Several neurotransmitter and neuromodulatory systems can control physiological glutamatergic activity. For example, opioid receptor ligands were shown to partially inhibit N-methyl-D-aspartic acid (NMDA) receptor-dependent glutamatergic excitotoxicity. Also, the endogenous tridecapeptide neurotensin (NT) was found to modulate excessive glutamate release and glutamate receptor activity in neurons. Alternatively to the one target-one drug approach, it has been well documented that hybrid compounds encompassing two pharmacophores in one molecular scaffold can represent more potent drugs. Moreover, such structures with dual activity can potentially enable a reduction of undesirable side effects and/or improved bioavailability. Herein, we describe the neuroprotective potential of an opioid-NT hybrid peptide (PK20), which was recently designed and synthesized within our group. The protective properties of PK20, assessed in an in vitro model of excitotoxic injury in organotypic hippocampal slice cultures subjected to NMDA, were compared to the effects caused by NT. Our results indicate that PK20 is a potent anti-neurodegenerative agent. Moreover, co-administered with NMDA, PK20 (25-100 ng/ml) dose-dependently reduced hippocampal cell death, determined by a decrease in the propidium iodide signal. We also report for the first time the significant NT-induced neuroprotective effect, as its application (50-100 ng/ml) to hippocampal slice cultures protected CA1 damage against neurotoxicity caused by NMDA.


Subject(s)
Hippocampus/drug effects , Neuroprotective Agents/pharmacology , Neurotensin/pharmacology , Oligopeptides/pharmacology , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Male , N-Methylaspartate/toxicity , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Neurotensin/administration & dosage , Neurotensin/chemistry , Oligopeptides/administration & dosage , Oligopeptides/chemistry , Organ Culture Techniques , Propidium/pharmacokinetics , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL