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










Publication year range
1.
J Magn Reson Imaging ; 41(6): 1622-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25168165

ABSTRACT

BACKGROUND: To characterize regional kidney sodium response by MRI following NKCC2 inhibition. METHODS: Regional renal sodium signals were monitored noninvasively using (23) Na-MRI at 9.4T with a temporal resolution of 1.5 min in anesthetized rats (N = 14). A mild NKCC2 inhibition was induced using a slow intravenous furosemide infusion. Time course of sodium signal was modeled as an exponential transient with a single characteristic time constant. RESULTS: Under normal physiological conditions, the renal sodium signals in medullary and cortical regions were stable and found to respond differently to furosemide challenge. Furosemide infusion at 1.2 mg/kg/h (N = 7) increased sodium signal in the cortex by 40 ± 6% (P < 7 × 10(-5) ) whereas decreased in the medulla by 29 ± 2% (P < 3 × 10(-6) ) with different temporal kinetics. The characteristic time constants of the change were determined to be: 8 ± 2 and 70 ± 10 min for medulla and cortex. Also, the medullary change occurred 9(±3) times faster than cortical independent of furosemide infusion rate up to 35 mg/kg/h. CONCLUSION: The pharmacological effects in terms of regional kidney sodium signal changes induced by NKCC2 inhibition are region-specific and highly predictable. Using noninvasive sodium MRI, we obtained regional renal sodium kinetics data sets in response to a low dose furosemide infusion in normal rats.


Subject(s)
Diuretics/pharmacology , Furosemide/pharmacology , Kidney Cortex/drug effects , Kidney Medulla/drug effects , Magnetic Resonance Imaging/methods , Sodium/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley
2.
Assay Drug Dev Technol ; 10(5): 417-31, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22881347

ABSTRACT

The renal outer medullary potassium (ROMK) channel is a member of the inwardly rectifying family of potassium (Kir) channels. ROMK (Kir1.1) is predominantly expressed in kidney where it plays a major role in the salt reabsorption process. Loss-of-function mutations in the human Kir1.1 channel are associated with antenatal Bartter's syndrome type II, a life-threatening salt and water balance disorder. Heterozygous carriers of Kir1.1 mutations associated with antenatal Bartter's syndrome have reduced blood pressure and a decreased risk of developing hypertension by age 60. These data suggest that Kir1.1 inhibitors could represent novel diuretics for the treatment of hypertension. Because little is known about the molecular pharmacology of Kir1.1 channels, assays that provide a robust, reliable readout of channel activity-while operating in high-capacity mode-are needed. In the present study, we describe high-capacity, 384- and 1,536-well plate, functional thallium flux, and IonWorks electrophysiology assays for the Kir1.1 channel that fulfill these criteria. In addition, 96-well (86)Rb(+) flux assays were established that can operate in the presence of 100% serum, and can provide an indication of the effect of a serum shift on compound potencies. The ability to grow Madin-Darby canine kidney cells expressing Kir1.1 in Transwell supports provides a polarized cell system that can be used to study the mechanism of Kir1.1 inhibition by different agents. All these functional Kir1.1 assays together can play an important role in supporting different aspects of drug development efforts during lead identification and/or optimization.


Subject(s)
Drug Discovery/methods , Potassium Channel Blockers/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Dogs , Humans , Madin Darby Canine Kidney Cells , Potassium Channel Blockers/blood , Potassium Channel Blockers/chemistry , Potassium Channels, Inwardly Rectifying/blood , Rats , Thallium/metabolism
3.
Mol Pharmacol ; 74(5): 1476-84, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18728100

ABSTRACT

Voltage-gated sodium (Na(V)1) channels play a critical role in modulating the excitability of sensory neurons, and human genetic evidence points to Na(V)1.7 as an essential contributor to pain signaling. Human loss-of-function mutations in SCN9A, the gene encoding Na(V)1.7, cause channelopathy-associated indifference to pain (CIP), whereas gain-of-function mutations are associated with two inherited painful neuropathies. Although the human genetic data make Na(V)1.7 an attractive target for the development of analgesics, pharmacological proof-of-concept in experimental pain models requires Na(V)1.7-selective channel blockers. Here, we show that the tarantula venom peptide ProTx-II selectively interacts with Na(V)1.7 channels, inhibiting Na(V)1.7 with an IC(50) value of 0.3 nM, compared with IC(50) values of 30 to 150 nM for other heterologously expressed Na(V)1 subtypes. This subtype selectivity was abolished by a point mutation in DIIS3. It is interesting that application of ProTx-II to desheathed cutaneous nerves completely blocked the C-fiber compound action potential at concentrations that had little effect on Abeta-fiber conduction. ProTx-II application had little effect on action potential propagation of the intact nerve, which may explain why ProTx-II was not efficacious in rodent models of acute and inflammatory pain. Mono-iodo-ProTx-II ((125)I-ProTx-II) binds with high affinity (K(d) = 0.3 nM) to recombinant hNa(V)1.7 channels. Binding of (125)I-ProTx-II is insensitive to the presence of other well characterized Na(V)1 channel modulators, suggesting that ProTx-II binds to a novel site, which may be more conducive to conferring subtype selectivity than the site occupied by traditional local anesthetics and anticonvulsants. Thus, the (125)I-ProTx-II binding assay, described here, offers a new tool in the search for novel Na(V)1.7-selective blockers.


Subject(s)
Action Potentials/drug effects , Nociceptors/drug effects , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Spider Venoms/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , DNA Primers , Humans , Ion Channel Gating , Male , Models, Animal , Molecular Sequence Data , Mutagenesis, Site-Directed , Radioligand Assay , Rats , Rats, Sprague-Dawley , Sequence Homology, Amino Acid , Sodium Channels/chemistry , Sodium Channels/genetics , Sodium Channels/physiology
4.
Mol Pharmacol ; 73(4): 1072-84, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18187582

ABSTRACT

Absorption of dietary cholesterol in the proximal region of the intestine is mediated by Niemann-Pick C1-like protein (NPC1L1) and is sensitive to the cholesterol absorption inhibitor ezetimibe (EZE). Although a correlation exists between EZE binding to NPC1L1 in vitro and efficacy in vivo, the precise nature of interaction(s) between NPC1L1, EZE, and cholesterol remain unclear. Here, we analyze the direct relationship between EZE analog binding to NPC1L1 and its influence on cholesterol influx in a novel in vitro system. Using the EZE analog [(3)H]AS, an assay that quantitatively measures the expression of NPC1L1 on the cell surface has been developed. It is noteworthy that whereas two cell lines (CaCo-2 and HepG2) commonly used for studying NPC1L1-dependent processes express almost undetectable levels of NPC1L1 at the cell surface, polarized Madin-Darby canine kidney (MDCKII) cells endogenously express 4 x 10(5) [(3)H]AS sites/cell under basal conditions. Depleting endogenous cholesterol with the HMG CoA reductase inhibitor lovastatin leads to a 2-fold increase in the surface expression of NPC1L1, supporting the contention that MDCKII cells respond to changes in cholesterol homeostasis by up-regulating a pathway for cholesterol influx. However, a significant increase in surface expression levels of NPC1L1 is necessary to characterize a pharmacologically sensitive, EZE-dependent pathway of cholesterol uptake in these cells. Remarkably, the affinity of EZE analogs for binding to NPC1L1 is almost identical to the IC(50) blocking cholesterol flux through NPC1L1 in MDCKII cells. From a mechanistic standpoint, these observations support the contention that EZE analogs and cholesterol share the same/overlapping binding site(s) or are tightly coupled through allosteric interactions.


Subject(s)
Azetidines/metabolism , Cholesterol/metabolism , Membrane Proteins/metabolism , Animals , Azetidines/chemistry , Caco-2 Cells , Cell Line , Cloning, Molecular , Dogs , Ezetimibe , Humans , Membrane Transport Proteins/metabolism , Reproducibility of Results , Sitosterols/metabolism , Sulfonamides/chemistry , Transfection , Tritium , beta-Lactams/metabolism
5.
Assay Drug Dev Technol ; 4(1): 37-48, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16506887

ABSTRACT

Clinical treatment of neuropathic pain can be achieved with a number of different drugs, some of which interact with all members of the voltage-gated sodium channel (NaV1) family. However, block of central nervous system and cardiac NaV1 channels can cause dose-limiting side effects, preventing many patients from achieving adequate pain relief. Expression of the tetrodotoxin-resistant NaV1.8 subtype is restricted to small-diameter sensory neurons, and several lines of evidence indicate a role for NaV1.8 in pain processing. Given these features, NaV1.8 subtype-selective blockers are predicted to be efficacious in the treatment of neuropathic pain and to be associated with fewer adverse effects than currently available therapies. To facilitate the identification of NaV1.8-specific inhibitors, we stably expressed the human NaV1.8 channel together with the auxiliary human beta1 subunit (NaV beta1) in human embryonic kidney 293 cells. Heterologously expressed human NaV1.8/NaV beta1 channels display biophysical properties that are similar to those of tetrodotoxin-resistant channels present in mouse dorsal root ganglion neurons. A membrane potential, fluorescence resonance energy transfer-based functional assay on a fluorometric imaging plate reader (FLIPR-Tetra, Molecular Devices, Sunnyvale, CA) platform has been established. This highcapacity assay is sensitive to known state-dependent NaV1 modulators and can be used to identify novel and selective NaV1.8 inhibitors.


Subject(s)
Membrane Potentials/physiology , Neurons, Afferent/physiology , Sodium Channels/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA Primers , Electrophysiology/methods , Fluorescence Resonance Energy Transfer/methods , Humans , Kidney , Models, Molecular , Molecular Sequence Data , NAV1.8 Voltage-Gated Sodium Channel , Peptide Fragments/immunology , Protein Conformation , Rabbits , Sodium Channels/genetics
6.
Diabetes ; 55(4): 1034-42, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16567526

ABSTRACT

Delayed-rectifier K+ currents (I(DR)) in pancreatic beta-cells are thought to contribute to action potential repolarization and thereby modulate insulin secretion. The voltage-gated K+ channel, K(V)2.1, is expressed in beta-cells, and the biophysical characteristics of heterologously expressed channels are similar to those of I(DR) in rodent beta-cells. A novel peptidyl inhibitor of K(V)2.1/K(V)2.2 channels, guangxitoxin (GxTX)-1 (half-maximal concentration approximately 1 nmol/l), has been purified, characterized, and used to probe the contribution of these channels to beta-cell physiology. In mouse beta-cells, GxTX-1 inhibits 90% of I(DR) and, as for K(V)2.1, shifts the voltage dependence of channel activation to more depolarized potentials, a characteristic of gating-modifier peptides. GxTX-1 broadens the beta-cell action potential, enhances glucose-stimulated intracellular calcium oscillations, and enhances insulin secretion from mouse pancreatic islets in a glucose-dependent manner. These data point to a mechanism for specific enhancement of glucose-dependent insulin secretion by applying blockers of the beta-cell I(DR), which may provide advantages over currently used therapies for the treatment of type 2 diabetes.


Subject(s)
Delayed Rectifier Potassium Channels/physiology , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/physiology , Potassium Channel Blockers/pharmacology , Amino Acid Sequence , Animals , Delayed Rectifier Potassium Channels/drug effects , Insulin Secretion , Islets of Langerhans/drug effects , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Molecular Sequence Data , Peptides/chemistry , Peptides/pharmacology , Potassium Channel Blockers/chemistry , Spider Venoms/chemistry , Spider Venoms/pharmacology
7.
Pain ; 117(1-2): 145-53, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16061326

ABSTRACT

A multitude of voltage-gated sodium channel subtypes (NaV1) are expressed in primary sensory neurons where they influence excitability via their role in the generation and propagation of action potentials. Peripheral nerve injury alters the expression of several NaV1subtypes, but among these only NaV1.3 is up-regulated in dorsal root ganglia (DRG) neurons. The increased expression of NaV1.3 implicates this subtype in the development and maintenance of neuropathic pain, but its contribution to neuropathic pain behavior has not been examined. Using the spared nerve injury (SNI) model, we found that peripheral nerve lesion increased NaV1.3-like immunoreactivity (-LI) in DRG neurons and that mechanical allodynia was partially alleviated following oral administration of two NaV1 blockers, mexiletine (30 and 100 mg/kg, p.o.) and lamotrigine (30 and 100 mg/kg, p.o.). Intrathecal administration of antisense oligonucleotides (4 days) selective for NaV1.3 decreased NaV1.3 immunostaining in the DRG by 50% in the SNI model, but did not attenuate mechanical or cold allodynia. Moreover, we found that only 18% of NaV1.3 positive neurons also expressed activated transcription factor-3 (ATF3), a marker of injured neurons. We then selectively axotomized a cutaneous nerve (sural) and a muscle nerve (gastrocnemius) in order to identify if NaV1.3 up-regulation is dependent on cutaneous and/or muscle afferent activation and found that the numbers of neurons expressing NaV1.3 was proportional to the magnitude of the injury, but independent of the nature of innervation. These results suggest that NaV1.3 increases in primary sensory neurons that are not directly damaged in response to injury. Thus, although NaV1.3 is up-regulated in a subpopulation of DRG neurons after injury, reduction in the expression of NaV1.3 subtype alone is not sufficient to influence the NaV1-dependent behavioral hypersensitivity associated with nerve injury.


Subject(s)
Nerve Tissue Proteins/metabolism , Neurons/metabolism , Pain/psychology , Peripheral Nervous System Diseases/psychology , Sodium Channels/metabolism , Up-Regulation/physiology , Analgesics/therapeutic use , Animals , Behavior, Animal , Disease Models, Animal , Dose-Response Relationship, Drug , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Immunohistochemistry/methods , Lamotrigine , Male , Mexiletine/therapeutic use , NAV1.3 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neurons/drug effects , Oligonucleotides/administration & dosage , Pain/drug therapy , Pain/etiology , Pain/metabolism , Pain Measurement/methods , Peripheral Nerves/metabolism , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/metabolism , Rats , Rats, Sprague-Dawley , Skin/metabolism , Sodium Channels/genetics , Sodium Channels/physiology , Time Factors , Triazines/therapeutic use , Up-Regulation/drug effects
8.
Proc Natl Acad Sci U S A ; 102(26): 9382-7, 2005 Jun 28.
Article in English | MEDLINE | ID: mdl-15964986

ABSTRACT

The transmission of pain signals after injury or inflammation depends in part on increased excitability of primary sensory neurons. Nociceptive neurons express multiple subtypes of voltage-gated sodium channels (NaV1s), each of which possesses unique features that may influence primary afferent excitability. Here, we examined the contribution of NaV1.9 to nociceptive signaling by studying the electrophysiological and behavioral phenotypes of mice with a disruption of the SCN11A gene, which encodes NaV1.9. Our results confirm that NaV1.9 underlies the persistent tetrodotoxin-resistant current in small-diameter dorsal root ganglion neurons but suggest that this current contributes little to mechanical thermal responsiveness in the absence of injury or to mechanical hypersensitivity after nerve injury or inflammation. However, the expression of NaV1.9 contributes to the persistent thermal hypersensitivity and spontaneous pain behavior after peripheral inflammation. These results suggest that inflammatory mediators modify the function of NaV1.9 to maintain inflammation-induced hyperalgesia.


Subject(s)
Anesthetics, Local/pharmacology , Neuropeptides/physiology , Nociceptors/metabolism , Pain , Sodium Channels/physiology , Tetrodotoxin/pharmacology , Animals , Behavior, Animal , DNA Primers/chemistry , DNA, Complementary/metabolism , Electrophysiology , Female , Ganglia, Spinal/metabolism , Gene Expression Regulation , Hyperalgesia/drug therapy , Inflammation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Genetic , NAV1.9 Voltage-Gated Sodium Channel , Neurons/metabolism , Neuropeptides/metabolism , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sodium/chemistry , Sodium Channels/metabolism
9.
J Physiol ; 567(Pt 1): 159-75, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15932888

ABSTRACT

Voltage-gated potassium (Kv) currents of human pancreatic islet cells were studied by whole-cell patch clamp recording. On average, 75% of the cells tested were identified as beta-cells by single cell, post-recording RT-PCR for insulin mRNA. In most cells, the dominant Kv current was a delayed rectifier. The delayed rectifier activated at potentials above -20 mV and had a V(1/2) for activation of -5.3 mV. Onset of inactivation was slow for a major component (tau = 3.2 s at +20 mV) observed in all cells; a smaller component (tau = 0.30 s) with an amplitude of approximately 25% was seen in some cells. Recovery from inactivation had a tau of 2.5 s at -80 mV and steady-state inactivation had a V(1/2) of -39 mV. In 12% of cells (21/182) a low-threshold, transient Kv current (A-current) was present. The A-current activated at membrane potentials above -40 mV, inactivated with a time constant of 18.5 ms at -20 mV, and had a V(1/2) for steady-state inactivation of -52 mV. TEA inhibited total Kv current with an IC50 = 0.54 mm and PAC, a disubstituted cyclohexyl Kv channel inhibitor, inhibited with an IC50 = 0.57 microm. The total Kv current was insensitive to margatoxin (100 nm), agitoxin-2 (50 nm), kaliotoxin (50 nm) and ShK (50 nm). Hanatoxin (100 nm) inhibited total Kv current by 65% at +20 mV. Taken together, these data provide evidence of at least two distinct types of Kv channels in human pancreatic beta-cells and suggest that more than one type of Kv channel may be involved in the regulation of glucose-dependent insulin secretion.


Subject(s)
Islets of Langerhans/physiology , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/physiology , Tetraethylammonium/pharmacology , Biophysical Phenomena , Biophysics , Cells, Cultured , Cyclohexanones/pharmacology , Delayed Rectifier Potassium Channels , Humans , Islets of Langerhans/cytology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurotoxins/pharmacology , Patch-Clamp Techniques , Peptides/pharmacology , Scorpion Venoms
10.
Brain Res ; 1029(1): 65-76, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15533317

ABSTRACT

One distinguishing feature of primary afferent neurons is their ability to bind the lectin IB(4). Previous work suggested that neurons in the inner part of lamina II (IIi), onto which IB(4)-positive sensory neurons project, facilitate nociceptive transmission following tissue or nerve injury. Using an IB(4)-saporin conjugate (IB(4)-SAP), we examined the contribution of IB(4)-positive neurons to nociceptive processing in rats with and without nerve injury. Intrasciatic injection of IB(4)-SAP (5 mug/5 mul) significantly decreased IB(4)-labeling and immunoreactive P(2)X(3) in the spinal cord and delayed the behavioral and neuroanatomical consequences of L5 spinal nerve ligation (SNL) injury. In the absence of injury, thermal and mechanical nociceptive thresholds increased 2 weeks post-treatment only in IB(4)-SAP-treated, but not control (saline or saporin only), rats. Acute NGF-induced hyperalgesia was also attenuated following IB(4)-SAP treatment. In the SNL model, mechanical allodynia failed to develop 1 and 2 weeks post-injury, but was fully established by 4 weeks. Moreover, neuropeptide Y immunoreactivity (NPY-ir), which increases in the spinal cord after nerve injury, was unchanged in IB(4)-SAP-treated animals whereas immunoreactive PKCgamma decreased 2, but not 4, weeks post-injury. Quantitative RT-PCR revealed a reduction in P(2)X(3) mRNA in L4 DRG of IB(4)-SAP-treated animals, but no change in TrkA expression. Our results suggest that IB(4)-positive neurons in L4 are required for the full expression of NGF-induced hyperalgesia and participate in the behavioral and anatomical consequences that follow injury to the L5 spinal nerve.


Subject(s)
Cytotoxins/metabolism , Lectins/metabolism , Nerve Degeneration/chemically induced , Neuralgia/physiopathology , Neurons, Afferent/pathology , Nociceptors/pathology , Pain/physiopathology , Peripheral Nervous System Diseases/physiopathology , Animals , Cytotoxins/pharmacology , Disease Models, Animal , Ganglia, Spinal/pathology , Ganglia, Spinal/physiopathology , Lectins/pharmacology , Ligation , Lumbosacral Region , Male , Neuralgia/pathology , Neurons, Afferent/drug effects , Nociceptors/drug effects , Pain/pathology , Pain Threshold , Peripheral Nervous System Diseases/pathology , Plant Lectins , Rats , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1 , Saporins , Spinal Nerves/injuries , Spinal Nerves/pathology , Spinal Nerves/physiopathology
11.
Biochemistry ; 43(30): 9866-76, 2004 Aug 03.
Article in English | MEDLINE | ID: mdl-15274641

ABSTRACT

Sodium channel blockers are used clinically to treat a number of neuropathic pain conditions, but more potent and selective agents should improve on the therapeutic index of currently used drugs. In a high-throughput functional assay, a novel sodium channel (Na(V)) blocker, N-[[2'-(aminosulfonyl)biphenyl-4-yl]methyl]-N'-(2,2'-bithien-5-ylmethyl)succinamide (BPBTS), was discovered. BPBTS is 2 orders of magnitude more potent than anticonvulsant and antiarrhythmic sodium channel blockers currently used to treat neuropathic pain. Resembling block by these agents, block of Na(V)1.2, Na(V)1.5, and Na(V)1.7 by BPBTS was found to be voltage- and use-dependent. BPBTS appeared to bind preferentially to open and inactivated states and caused a dose-dependent hyperpolarizing shift in the steady-state availability curves for all sodium channel subtypes tested. The affinity of BPBTS for the resting and inactivated states of Na(V)1.2 was 1.2 and 0.14 microM, respectively. BPBTS blocked Na(V)1.7 and Na(V)1.2 with similar potency, whereas block of Na(V)1.5 was slightly more potent. The slow tetrodotoxin-resistant Na(+) current in small-diameter DRG neurons was also potently blocked by BPBTS. [(3)H]BPBTS bound with high affinity to a single class of sites present in rat brain synaptosomal membranes (K(d) = 6.1 nM), and in membranes derived from HEK cells stably expressing Na(V)1.5 (K(d) = 0.9 nM). BPBTS dose-dependently attenuated nociceptive behavior in the formalin test, a rat model of tonic pain. On the basis of these findings, BPBTS represents a structurally novel and potent sodium channel blocker that may be used as a template for the development of analgesic agents.


Subject(s)
Amides/therapeutic use , Biphenyl Compounds/therapeutic use , Muscle Proteins/metabolism , Pain Measurement/drug effects , Sodium Channel Blockers/therapeutic use , Sodium Channels/metabolism , Amides/chemical synthesis , Amides/metabolism , Analgesics/chemical synthesis , Analgesics/metabolism , Analgesics/therapeutic use , Animals , Binding Sites , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/metabolism , Brain/metabolism , Cell Line , Disease Models, Animal , Formaldehyde/administration & dosage , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Humans , Mice , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , NAV1.2 Voltage-Gated Sodium Channel , NAV1.5 Voltage-Gated Sodium Channel , NAV1.7 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Sodium Channel Blockers/chemical synthesis , Sodium Channel Blockers/metabolism , Sodium Channels/biosynthesis , Sodium Channels/genetics , Succinates , Synaptosomes/metabolism , Tetrodotoxin/antagonists & inhibitors , Tetrodotoxin/chemistry
12.
Diabetes ; 53(3): 597-607, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14988243

ABSTRACT

Voltage-gated potassium channels (Kv channels) are involved in repolarization of excitable cells. In pancreatic beta-cells, prolongation of the action potential by block of delayed rectifier potassium channels would be expected to increase intracellular free calcium and to promote insulin release in a glucose-dependent manner. However, the specific Kv channel subtypes responsible for repolarization in beta-cells, most importantly in humans, are not completely resolved. In this study, we have investigated the expression of 26 subtypes from Kv subfamilies in human islet mRNA. The results of the RT-PCR analysis were extended by in situ hybridization and/or immunohistochemical analysis on sections from human or Rhesus pancreas. Cell-specific markers were used to show that Kv2.1, Kv3.2, Kv6.2, and Kv9.3 are expressed in beta-cells, that Kv3.1 and Kv6.1 are expressed in alpha-cells, and that Kv2.2 is expressed in delta-cells. This study suggests that more than one Kv channel subtype might contribute to the beta-cell delayed rectifier current and that this current could be formed by heterotetramers of active and silent subunits.


Subject(s)
Islets of Langerhans/physiology , Potassium Channels/genetics , Animals , Base Sequence , DNA Primers , Gene Expression Regulation , Humans , Islets of Langerhans/cytology , Macaca mulatta , Potassium Channels/classification , RNA, Messenger/genetics , Shab Potassium Channels , Species Specificity
13.
Biochemistry ; 41(50): 14734-47, 2002 Dec 17.
Article in English | MEDLINE | ID: mdl-12475222

ABSTRACT

Two peptides, ProTx-I and ProTx-II, from the venom of the tarantula Thrixopelma pruriens, have been isolated and characterized. These peptides were purified on the basis of their ability to reversibly inhibit the tetrodotoxin-resistant Na channel, Na(V) 1.8, and are shown to belong to the inhibitory cystine knot (ICK) family of peptide toxins interacting with voltage-gated ion channels. The family has several hallmarks: cystine bridge connectivity, mechanism of channel inhibition, and promiscuity across channels within and across channel families. The cystine bridge connectivity of ProTx-II is very similar to that of other members of this family, i.e., C(2) to C(16), C(9) to C(21), and C(15) to C(25). These peptides are the first high-affinity ligands for tetrodotoxin-resistant peripheral nerve Na(V) channels, but also inhibit other Na(V) channels (IC(50)'s < 100 nM). ProTx-I and ProTx-II shift the voltage dependence of activation of Na(V) 1.5 to more positive voltages, similar to other gating-modifier ICK family members. ProTx-I also shifts the voltage dependence of activation of Ca(V) 3.1 (alpha(1G), T-type, IC(50) = 50 nM) without affecting the voltage dependence of inactivation. To enable further structural and functional studies, synthetic ProTx-II was made; it adopts the same structure and has the same functional properties as the native peptide. Synthetic ProTx-I was also made and exhibits the same potency as the native peptide. Synthetic ProTx-I, but not ProTx-II, also inhibits K(V) 2.1 channels with 10-fold less potency than its potency on Na(V) channels. These peptides represent novel tools for exploring the gating mechanisms of several Na(V) and Ca(V) channels.


Subject(s)
Peptides/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism , Spider Venoms/pharmacology , Amino Acid Sequence , Animals , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/isolation & purification , Calcium Channel Blockers/pharmacology , Cell Line , Disulfides/chemistry , Electrophysiology , Humans , Ion Channel Gating/drug effects , Molecular Sequence Data , Peptides/chemistry , Peptides/isolation & purification , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/isolation & purification , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spider Venoms/chemistry , Spider Venoms/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...