Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 6(9): 1161-74, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25056913

ABSTRACT

Abnormal uterine activity in pregnancy causes a range of important clinical disorders, including preterm birth, dysfunctional labour and post-partum haemorrhage. Uterine contractile patterns are controlled by the generation of complex electrical signals at the myometrial smooth muscle plasma membrane. To identify novel targets to treat conditions associated with uterine dysfunction, we undertook a genome-wide screen of potassium channels that are enriched in myometrial smooth muscle. Computational modelling identified Kir7.1 as potentially important in regulating uterine excitability during pregnancy. We demonstrate Kir7.1 current hyper-polarizes uterine myocytes and promotes quiescence during gestation. Labour is associated with a decline, but not loss, of Kir7.1 expression. Knockdown of Kir7.1 by lentiviral expression of miRNA was sufficient to increase uterine contractile force and duration significantly. Conversely, overexpression of Kir7.1 inhibited uterine contractility. Finally, we demonstrate that the Kir7.1 inhibitor VU590 as well as novel derivative compounds induces profound, long-lasting contractions in mouse and human myometrium; the activity of these inhibitors exceeds that of other uterotonic drugs. We conclude Kir7.1 regulates the transition from quiescence to contractions in the pregnant uterus and may be a target for therapies to control uterine contractility.


Subject(s)
Potassium Channels, Inwardly Rectifying/physiology , Uterine Contraction/metabolism , Animals , Cell Line , Cricetinae , Cricetulus , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , In Vitro Techniques , Labor, Obstetric/metabolism , Membrane Potentials , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Pregnancy , Uterine Contraction/genetics
2.
Mol Pharmacol ; 86(2): 211-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24830940

ABSTRACT

Both human ether-à-go-go-related gene (hERG1) and the closely related human ether-à-go-go (hEAG1) channel are aberrantly expressed in a large proportion of human cancers. In the present study, we demonstrate that transfection of hERG1 into mouse fibroblasts is sufficient to induce many features characteristic of malignant transformation. An important finding of this work is that this transformation could be reversed by chronic incubation (for 2-3 weeks) with the hERG channel blocker dofetilide (100 nM), whereas more acute applications (for 1-2 days) were ineffective. The hERG1 expression resulted in a profound loss of cell contact inhibition, multiple layers of overgrowing cells, and high saturation densities. Cells also changed from fibroblast-like to a more spindle-shaped morphology, which was associated with a smaller cell size, a dramatic increase in cell polarization, a reduction in the number of actin stress fibers, and less punctate labeling of focal adhesions. Analysis of single-cell migration and scratch-wound closure clearly demonstrated that hERG1-expressing cells migrated more rapidly than vector-transfected control cells. In contrast to previous studies on hEAG1, there were no increases in rates of proliferation, or loss of growth factor dependency; however, hERG1-expressing cells were capable of substrate-independent growth. Allogeneic transplantation of hERG1-expressing cells into nude mice resulted in an increased incidence of tumors. In contrast to hEAG1, the mechanism of cellular transformation is dependent on ion conduction. Trafficking-deficient and conduction-deficient hERG1 mutants also prevented cellular transformation. These results provide evidence that hERG1 expression is sufficient to induce cellular transformation by a mechanism distinct from hEAG1. The most important conclusion of this study is that selective hERG1 channel blockers have therapeutic potential in the treatment of hERG1-expressing cancers.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Potassium Channel Blockers/pharmacology , Actins/metabolism , Animals , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Fibroblasts/drug effects , Focal Adhesions/metabolism , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Stress Fibers/metabolism , Transfection
3.
J Biomol Screen ; 14(7): 769-80, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19556611

ABSTRACT

Gamma-amino butyric acid (GABA)-activated Cl- channels are critical mediators of inhibitory postsynaptic potentials in the CNS. To date, rational design efforts to identify potent and selective GABA(A) subtype ligands have been hampered by the absence of suitable high-throughput screening approaches. The authors describe 384-well population patch-clamp (PPC) planar array electrophysiology methods for the study of GABA(A) receptor pharmacology. In HEK293 cells stably expressing human alpha1beta3gamma2 GABA(A) channels, GABA evoked outward currents at 0 mV of 1.05 +/- 0.08 nA, measured 8 s post GABA addition. The I(GABA) was linear and reversed close to the theoretical E(Cl) (-56 mV). Concentration-response curve analysis yielded a mean pEC(50) value of 5.4 and Hill slope of 1.5, and for a series of agonists, the rank order of potency was muscimol > GABA > isoguvacine. A range of known positive modulators, including diazepam and pentobarbital, produced concentration-dependent augmentation of the GABA EC( 20) response (1 microM). The competitive antagonists bicuculline and gabazine produced concentration-dependent, parallel, rightward displacement of GABA curves with pA(2) and slope values of 5.7 and 1.0 and 6.7 and 1.0, respectively. In contrast, picrotoxin (0.2-150 microM) depressed the maximal GABA response, implying a non-competitive antagonism. Overall, the pharmacology of human alpha1beta3gamma2 GABA(A) determined by PPC was highly similar to that obtained by conventional patch-clamp methods. In small-scale single-shot screens, Z' values of >0.5 were obtained in agonist, modulator, and antagonist formats with hit rates of 0% to 3%. The authors conclude that despite the inability of the method to resolve the peak agonist responses, PPC can rapidly and usefully quantify pharmacology for the alpha1beta3gamma2 GABA(A) isoform. These data suggest that PPC may be a valuable approach for a focused set and secondary screening of GABA(A) receptors and other slow ligand-gated ion channels.


Subject(s)
Electrophysiological Phenomena , Patch-Clamp Techniques/methods , Protein Subunits/metabolism , Receptors, GABA-A/metabolism , Recombinant Proteins/metabolism , Benzodiazepines/pharmacology , Cell Line , Drug Evaluation, Preclinical , Electrophysiological Phenomena/drug effects , Flumazenil/pharmacology , GABA-A Receptor Agonists , GABA-A Receptor Antagonists , Humans , Ion Channel Gating/drug effects
4.
Mol Pharmacol ; 76(3): 569-78, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19515965

ABSTRACT

A new small molecule, 4-(2-methoxy-phenylcarbamoyloxymethyl)-piperidine-1-carboxylic acid tert-butyl ester (GW542573X), is presented as an activator of small-conductance Ca(2+)-activated K(+) (SK, K(Ca)2) channels and distinguished from previously published positive modulators of SK channels, such as 1-ethyl-2-benzimidazolinone (1-EBIO) and cyclohexyl-[2-(3,5-dimethylpyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine (CyPPA), in several aspects. GW542573X is the first SK1-selective compound described: an EC(50) value of 8.2 +/- 0.8 microM (n = 6, [Ca(2+)](i) = 200 nM) was obtained from inside-out patches excised from hSK1-expressing HEK293 cells. Whole-cell experiments showed that hSK2 and hSK3 channels were more than 10 times, and hIK channels even more than 100 times, less sensitive to GW542573X. The Ca(2+)-response curve of hSK1 was left-shifted from an EC(50)(Ca(2+)) value of 410 +/- 20 nM (n = 9) to 240 +/- 10 nM (n = 5) in the presence of 10 microM GW542573X. In addition to this positive modulation, GW542573X activated SK1 in the absence of Ca(2+) and furthermore induced a 15% increase in the maximal current at saturating Ca(2+). Thus, GW542573X also acts as a genuine opener of the hSK1 channels, a mechanism of action (MOA) not previously obtained with SK channels. The differential potency on hSK1 and hSK3 enabled a chimera approach to elucidate site(s) important for this new MOA and selectivity property. A single amino acid (Ser293) located in S5 of hSK1 was essential, and substituting the corresponding Leu476 in hSK3 with serine conferred hSK1-like potency (EC(50) = 9.3 +/- 1.4 microM, n = 5). GW542573X may activate SK channels via interaction with "deep-pore" gating structures at the inner pore vestibule or the selectivity filter in contrast to 1-EBIO and CyPPA that exert positive modulation via the intracellular calmodulin binding domain.


Subject(s)
Carbamates/pharmacology , Piperidines/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/agonists , Small-Conductance Calcium-Activated Potassium Channels/genetics , Amino Acid Substitution , Carbamates/chemistry , Cell Line , Humans , Mutation , Piperidines/chemistry , Serine/genetics
5.
Comb Chem High Throughput Screen ; 12(1): 96-106, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19149495

ABSTRACT

The tractability of ion channels as drug targets has been significantly improved by the advent of planar array electrophysiology platforms which have dramatically increased the capacity for electrophysiological profiling of lead series compounds. However, the data quality and through-put obtained with these platforms is critically dependent on the robustness of the expression reagent being used. The generation of high quality, recombinant cell lines is therefore a key step in the early phase of ion channel drug discovery and this can present significant challenges due to the diversity and organisational complexity of many channel types. This article focuses on several complex and difficult to express ion channels and illustrates how improved stable cell lines can be obtained by integration of planar array electrophysiology systems into the cell line generation process per se. By embedding this approach at multiple stages (e.g., during development of the expression strategy, during screening and validation of clonal lines, and during characterisation of the final cell line), the cycle time and success rate in obtaining robust expression of complex multi-subunit channels can be significantly improved. We also review how recent advances in this technology (e.g., population patch clamp) have further widened the versatility and applicability of this approach.


Subject(s)
Cell Line/cytology , Drug Discovery/methods , Electrophysiology/methods , Ion Channels , Electrophysiology/instrumentation , Humans , Tissue Array Analysis
6.
BMC Biotechnol ; 7: 93, 2007 Dec 20.
Article in English | MEDLINE | ID: mdl-18096051

ABSTRACT

BACKGROUND: HERG potassium channel blockade is the major cause for drug-induced long QT syndrome, which sometimes cause cardiac disrhythmias and sudden death. There is a strong interest in the pharmaceutical industry to develop high quality medium to high-throughput assays for detecting compounds with potential cardiac liability at the earliest stages of drug development. Cultivation of cells at lower temperature has been used to improve the folding and membrane localization of trafficking defective hERG mutant proteins. The objective of this study was to investigate the effect of lower temperature maintenance on wild type hERG expression and assay performance. RESULTS: Wild type hERG was stably expressed in CHO-K1 cells, with the majority of channel protein being located in the cytoplasm, but relatively little on the cell surface. Expression at both locations was increased several-fold by cultivation at lower growth temperatures. Intracellular hERG protein levels were highest at 27 degrees C and this correlated with maximal 3H-dofetilide binding activity. In contrast, the expression of functionally active cell surface-associated hERG measured by patch clamp electrophysiology was optimal at 30 degrees C. The majority of the cytoplasmic hERG protein was associated with the membranes of cytoplasmic vesicles, which markedly increased in quantity and size at lower temperatures or in the presence of the Ca2+-ATPase inhibitor, thapsigargin. Incubation with the endocytic trafficking blocker, nocodazole, led to an increase in hERG activity at 37 degrees C, but not at 30 degrees C. CONCLUSION: Our results are consistent with the concept that maintenance of cells at reduced temperature can be used to boost the functional expression of difficult-to-express membrane proteins and improve the quality of assays for medium to high-throughput compound screening. In addition, these results shed some light on the trafficking of hERG protein under these growth conditions.


Subject(s)
Cell Culture Techniques/methods , Ether-A-Go-Go Potassium Channels/biosynthesis , Protein Transport , Animals , Biological Transport, Active/drug effects , CHO Cells , Ca(2+) Mg(2+)-ATPase/drug effects , Cricetinae , Cricetulus , Ether-A-Go-Go Potassium Channels/drug effects , Humans , Membrane Proteins , Nocodazole/pharmacology , Phenethylamines/pharmacology , Potassium Channel Blockers , Recombinant Proteins , Sulfonamides/pharmacology , Temperature , Thapsigargin/pharmacology , Transport Vesicles/drug effects , Up-Regulation
7.
Mol Biosyst ; 3(10): 714-22, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17882333

ABSTRACT

Population patch clamp (PPC) is a novel high throughput planar array electrophysiology technique that allows ionic currents to be recorded from populations of cells under voltage clamp. For the drug discovery pharmacologist, PPC promises greater speed and precision than existing methods for screening compounds at voltage-gated ion channel targets. Moreover, certain constitutively active or slow-ligand gated channels that have hitherto proved challenging to screen with planar array electrophysiology (e.g. SK/IK channels) are now more accessible. In this article we review early findings using PPC and provide a perspective on its likely impact on ion channel drug discovery. To support this, we include some new data on ion channel assay duplexing and on modulator assays, approaches that have thus far not been described.


Subject(s)
Ion Channels/metabolism , Patch-Clamp Techniques/methods , Animals , CHO Cells , Cricetinae , Cricetulus , Electrophysiology , Humans , Ion Channel Gating , Ion Channels/genetics , Patch-Clamp Techniques/instrumentation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
8.
J Biomol Screen ; 12(1): 50-60, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17092914

ABSTRACT

Planar array electrophysiology techniques were applied to assays for modulators of recombinant hIK and hSK3 Ca2+-activated K+ channels. In CHO-hIK-expressing cells, under asymmetric K+ gradients, small-molecule channel activators evoked time- and voltage-independent currents characteristic of those previously described by classical patch clamp electrophysiology methods. In single-hole (cell) experiments, the large cell-to-cell heterogeneity in channel expression rendered it difficult to generate activator concentration-response curves. However, in population patch clamp mode, in which signals are averaged from up to 64 cells, well-to-well variation was substantially reduced such that concentration-response curves could be easily constructed. The absolute EC50 values and rank order of potency for a range of activators, including 1-EBIO and DC-EBIO, corresponded well with conventional patch clamp data. Activator responses of hIK and hSK3 channels could be fully and specifically blocked by the selective inhibitors TRAM-34 and apamin, with IC50 values of 0.31 microM and 3 nM, respectively. To demonstrate assay precision and robustness, a test set of 704 compounds was screened in a 384-well format of the hIK assay. All plates had Z' values greater than 0.6, and the statistical cutoff for activity was 8%. Eleven hits (1.6%) were identified from this set, in addition to the randomly spiked wells with known activators. Overall, our findings demonstrate that population patch clamp is a powerful and enabling method for screening Ca2+-activated K+ channels and provides significant advantages over single-cell electrophysiology (IonWorks(HT)) and other previously published approaches. Moreover, this work demonstrates for the 1st time the utility of population patch clamp for ion channel activator assays and for non-voltage-gated ion channels.


Subject(s)
Electrophysiology/methods , Patch-Clamp Techniques/methods , Potassium Channels, Calcium-Activated/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Humans , Membrane Transport Modulators/pharmacology , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Reproducibility of Results
9.
Brain Res ; 1065(1-2): 37-46, 2005 Dec 14.
Article in English | MEDLINE | ID: mdl-16336949

ABSTRACT

The role of calcium-activated potassium channels in the regulation of neuronal hyperexcitability, as in epilepsy, is unclear. To examine this issue, we have used the acute hippocampal slice model of epileptiform activity to investigate the effects of an enhancer of SK channel activity, 1-ethyl-benzimidazolinone (EBIO). That EBIO is an SK channel modulator was confirmed by its potentiation of hSK1, hSK2, hSK3 and hIK currents (EC(50) values in the range of 130-870 microM) and its apamin (1 microM) sensitive reduction of the number of action potentials fired in CA3 pyramidal neurons in response to a depolarizing current step. In addition, while EBIO did not significantly affect electrically evoked glutamatergic synaptic transmission, it did inhibit epileptiform activity (IC(50) values in the range of 150-325 microM) induced by (1) modifying the extracellular ionic environment by removing extracellular Mg(2+) or elevating extracellular K(+) from 3.0 to 8.5 mM and (2) disinhibiting the slice using 3 mM pentylenetetrazol or combined application of 10 microM gabazine and 10 microM CGP55845. Furthermore, its inhibitory effect in the full disinhibition model of epileptiform activity (10 microM gabazine + 10 microM CGP55845) was occluded by the SK channel blocker apamin (300 nM-1 microM) which in its own right increased the duration and reduced the frequency of individual epileptiform bursts. In conclusion, compounds that enhance the activation of small conductance Ca(2+) -activated K(+) channels are effective inhibitors of epileptiform activity in vitro.


Subject(s)
Epilepsy/physiopathology , Hippocampus/physiopathology , Neurons/physiology , Potassium Channels, Calcium-Activated/physiology , Animals , Apamin/pharmacology , Benzimidazoles , CHO Cells , Calcium Channel Agonists , Cell Line , Cricetinae , Evoked Potentials/drug effects , Excitatory Postsynaptic Potentials/drug effects , Extracellular Space/drug effects , Extracellular Space/physiology , GABA Modulators/pharmacology , Humans , Magnesium Deficiency/physiopathology , Male , Membrane Potentials/drug effects , Models, Neurological , Patch-Clamp Techniques , Potassium/pharmacology , Rats , Recombinant Proteins
10.
J Neurosci ; 25(14): 3489-98, 2005 Apr 06.
Article in English | MEDLINE | ID: mdl-15814779

ABSTRACT

We investigated the role of small-conductance calcium-activated potassium (SK) and intermediate-conductance calcium-activated potassium channels in modulating sensory transmission from peripheral afferents into the rat spinal cord. Subunit-specific antibodies reveal high levels of SK3 immunoreactivity in laminas I, II, and III of the spinal cord. Among dorsal root ganglion neurons, both peripherin-positive (C-type) and peripherin-negative (A-type) cells show intense SK3 immunoreactivity. Furthermore, dorsal root-stimulated sensory responses recorded in vitro are inhibited when SK channel activity is increased with 1-ethyl-2-benzimidazolinone (1-EBIO). In vivo electrophysiological recordings show that neuronal responses to naturally evoked nociceptive and nonnociceptive stimuli increase after application of the selective SK channel blocker 8,14-diaza-1,7(1,4)-diquinolinacyclotetradecaphanedium di-trifluoroacetate (UCL 1848), indicating that SK channels are normally active in moderating afferent input. Conversely, neuronal responses evoked by mechanical stimuli are inhibited when SK channel activity is increased with 1-EBIO. These effects are reversed by the subsequent application of UCL 1848. Our data demonstrate that SK channels have an important role in controlling sensory input into the spinal cord.


Subject(s)
Neurons, Afferent/physiology , Pain/physiopathology , Potassium Channels, Calcium-Activated/physiology , Spinal Cord/cytology , Animals , Animals, Newborn , Benzimidazoles/pharmacology , Calcium Channel Agonists/pharmacology , Cell Line , Dose-Response Relationship, Drug , Electric Stimulation/methods , Ependyma/metabolism , Female , Ganglia, Spinal/cytology , Humans , Immunohistochemistry/methods , In Vitro Techniques , Intermediate Filament Proteins/metabolism , Male , Membrane Glycoproteins/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Nerve Fibers/drug effects , Nerve Fibers/physiology , Nerve Tissue Proteins/metabolism , Neurons, Afferent/drug effects , Patch-Clamp Techniques/methods , Peripherins , Physical Stimulation/methods , Potassium Channel Blockers/pharmacology , Potassium Channels, Calcium-Activated/drug effects , Quinolinium Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Synaptic Transmission/radiation effects , TRPV Cation Channels/metabolism , Time Factors , Transfection
11.
Naunyn Schmiedebergs Arch Pharmacol ; 369(6): 602-15, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15127180

ABSTRACT

The SK/IK family of small and intermediate conductance calcium-activated potassium channels contains four members, SK1, SK2, SK3 and IK1, and is important for the regulation of a variety of neuronal and non-neuronal functions. In this study we have analysed the distribution of these channels in human tissues and their cellular localisation in samples of colon and corpus cavernosum. SK1 mRNA was detected almost exclusively in neuronal tissues. SK2 mRNA distribution was restricted but more widespread than SK1, and was detected in adrenal gland, brain, prostate, bladder, liver and heart. SK3 mRNA was detected in almost every tissue examined. It was highly expressed in brain and in smooth muscle-rich tissues including the clitoris and the corpus cavernosum, and expression in the corpus cavernosum was upregulated up to 5-fold in patients undergoing sex-change operations. IK1 mRNA was present in surface-rich, secretory and inflammatory cell-rich tissues, highest in the trachea, prostate, placenta and salivary glands. In detailed immunohistochemical studies of the colon and the corpus cavernosum, SK1-like immunoreactivity was observed in the enteric neurons. SK3-like immunoreactivity was observed strongly in smooth muscle and vascular endothelium. IK1-like immunoreactivity was mainly observed in inflammatory cells and enteric neurons of the colon, but absent in corpus cavernosum. These distinctive patterns of distribution suggest that these channels are likely to have different biological functions and could be specifically targeted for a number of human diseases, such as irritable bowel syndrome, hypertension and erectile dysfunction.


Subject(s)
Colon/metabolism , Muscle, Smooth, Vascular/metabolism , Potassium Channels, Calcium-Activated/physiology , Adult , Aged , Blotting, Northern , Colon/blood supply , Electric Conductivity , Female , Gene Expression Regulation , Genitalia/blood supply , Genitalia/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Muscle, Smooth/blood supply , Muscle, Smooth/metabolism , Organ Specificity , Potassium Channels, Calcium-Activated/biosynthesis , Potassium Channels, Calcium-Activated/genetics , RNA, Messenger/analysis , Small-Conductance Calcium-Activated Potassium Channels
12.
Neuropharmacology ; 46(3): 425-38, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14975698

ABSTRACT

The voltage-gated sodium channel NaV1.8 (SNS, PN3) is thought to be a molecular correlate of the dorsal root ganglion (DRG) tetrodotoxin resistant (TTX-R) Na+ current. TTX-R/NaV1.8 is an attractive therapeutic drug target for inflammatory and neuropathic pain on the basis of its specific distribution in sensory neurones and its modulation by inflammatory mediators. However, detailed analysis of recombinant NaV1.8 has been hampered by difficulties in stably expressing the functional protein in mammalian cells. Here, we show stable expression and functional analysis of rat NaV1.8 (rNaV1.8) in the rat DRG/mouse N18Tg2 neuroblastoma hybridoma cell line ND7-23. Rat NaV1.8 Na+ currents were recorded (789 +/- 89 pA, n=62, over 20-cell passages) that qualitatively resembled DRG TTX-R in terms of gating kinetics and voltage-dependence of activation and inactivation. The local anaesthetic drug tetracaine produced tonic inhibition of rNaV1.8 (mean IC50 value 12.5 microM) and in repeated gating paradigms (2-10 Hz) also showed frequency-dependent block. There was a correlation between the ability of several analogues of the anticonvulsant/analgesic compound lamotrigine to inhibit TTX-R and rNaV1.8 (r=0.72, P<0.001). RT-PCR analysis of wild type ND7-23 cells revealed endogenous expression of the beta1 and beta3 accessory Na+ channel subunits-the possibility that the presence of these subunits assists and stabilises expression of rNaV1.8 is discussed. We conclude that the neuroblastoma ND7-23 cell line is a suitable heterologous expression system for rNaV1.8 Na+ channels in that it allows stable expression of a channel with biophysical properties that closely resemble the native TTX-R currents in DRG neurones. This reagent will prove useful in the search for pharmacological inhibitors of rNaV1.8 as novel analgesics.


Subject(s)
Ganglia, Spinal/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/physiology , Neuroblastoma/metabolism , Sodium Channels/biosynthesis , Sodium Channels/physiology , Animals , Cell Line , Cell Line, Tumor , Female , Ganglia, Spinal/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , NAV1.8 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Sodium Channels/genetics
13.
J Biomol Screen ; 8(1): 50-64, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12854998

ABSTRACT

To address the throughput restrictions of classical patch clamp electrophysiology, Essen Instruments has developed a plate-based electrophysiology measurement platform. The instrument is an integrated platform that consists of computer-controlled fluid handling, recording electronics, and processing tools capable of voltage clamp whole-cell recordings from thousands of individual cells per day. To establish a recording, the system uses a planar, multiwell substrate (a PatchPlate). The system effectively positions 1 cell into a hole separating 2 fluid compartments in each well of the substrate. Voltage control and current recordings from the cell membrane are made subsequent to gaining access to the cell interior by applying a permeabilizing agent to the intracellular side. Based on the multiwell design of the PatchPlate, voltage clamp recordings of up to 384 individual cells can be made in minutes and are comparable to measurements made using traditional electrophysiology techniques. An integrated pipetting system allows for up to 2 additions of modulation agents. Typical throughput, measurement fidelity, stability, and comparative pharmacology of a recombinant voltage-dependent sodium channel (hNav1.3) and a voltage-gated potassium channel (hKv1.5) exogenously expressed in CHO cells are presented. The IonWorks HT device can be used in biophysical and pharmacological profiling of ion channels in an environment compatible with high-capacity screening.


Subject(s)
Biological Assay/instrumentation , Electrophysiology/instrumentation , Animals , Biological Assay/methods , CHO Cells , Cricetinae , Electrophysiology/methods , Humans , Potassium Channels/metabolism , Sodium Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL