Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 114(39): 10491-10496, 2017 09 26.
Article in English | MEDLINE | ID: mdl-28874544

ABSTRACT

Poison-dart Phyllobates terribilis frogs sequester lethal amounts of steroidal alkaloid batrachotoxin (BTX) in their skin as a defense mechanism against predators. BTX targets voltage-gated Na+ channels and enables them to open persistently. How BTX autoresistance arises in such frogs remains a mystery. The BTX receptor has been delineated along the Na+ channel inner cavity, which is formed jointly by four S6 transmembrane segments from domains D1 to D4. Within the P. terribilis muscle Na+ channel, five amino acid (AA) substitutions have been identified at D1/S6 and D4/S6. We therefore investigated the role of these naturally occurring substitutions in BTX autoresistance by introducing them into rat Nav1.4 muscle Na+ channel, both individually and in combination. Our results showed that combination mutants containing an N1584T substitution all conferred a complete BTX-resistant phenotype when expressed in mammalian HEK293t cells. The single N1584T mutant also retained its functional integrity and became exceptionally resistant to 5 µM BTX, aside from a small residual BTX effect. Single and combination mutants with the other four S6 residues (S429A, I433V, A445D, and V1583I) all remained highly BTX sensitive. These findings, along with diverse BTX phenotypes of N1584K/A/D/T mutant channels, led us to conclude that the conserved N1584 residue is indispensable for BTX actions, probably functioning as an integral part of the BTX receptor. Thus, complete BTX autoresistance found in P. terribilis muscle Na+ channels could emerge primarily from a single AA substitution (asparagine→threonine) via a single nucleotide mutation (AAC→ACC).


Subject(s)
Amino Acid Substitution/genetics , Batrachotoxins/toxicity , Drug Resistance/genetics , NAV1.4 Voltage-Gated Sodium Channel/genetics , Animals , Anura/metabolism , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutagenesis, Site-Directed , NAV1.4 Voltage-Gated Sodium Channel/metabolism , Protein Domains/genetics , Protein Domains/physiology , Rats , Sodium Channels/genetics
2.
Anesth Analg ; 122(3): 719-729, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26646348

ABSTRACT

BACKGROUND: Antidepressant S-duloxetine alleviates intractable pain associated with diabetic peripheral neuropathy and fibromyalgia. It also reduces both acute and persistent pain in various animal models. This study addresses whether the enantiomer, R-duloxetine, and the homolog, N-methyl duloxetine, could act as analgesics and whether they block neuronal Na⁺ channels. METHODS: The rat incision plus extension model on the dorsothoracic skin was applied to evoke postoperative mechanoallodynia and hyperalgesia, measured for 5 days postoperatively by local responses to von Frey filaments. R-Duloxetine and N-methyl duloxetine were administered systemically (intraperitoneal) or locally (subcutaneous [SC]) 1 hour before the surgery. The block of Na currents in rat neuronal GH3 cells was determined under the whole-cell configuration. RESULTS: Ipsilateral SC injections (2 mg/0.4 mL) of R-duloxetine and N-methyl duloxetine reduced both postoperative allodynia and hyperalgesia by approximately 89% to 99% in the area under the curve of skin responses next to incision over 5 days. Systemic intraperitoneal injections at a higher dosage (10 mg) had smaller analgesic effects (reduced by approximately 53%-69%), whereas contralateral SC injections (10 mg) were ineffective. Both R-duloxetine and N-methyl duloxetine blocked neuronal Na⁺ currents, with a higher affinity for the inactivated than the resting states. In addition, both drugs elicited significant use-dependent block of Na currents when stimulated at 5 Hz. CONCLUSIONS: R-Duloxetine and N-methyl duloxetine are highly effective against postoperative pain using the skin incision model, and they elicit both tonic and use-dependent block of neuronal Na⁺ channels. Our results suggest that R-duloxetine and N-methyl duloxetine are applicable as novel analgesics.


Subject(s)
Analgesics/pharmacology , Antidepressive Agents, Second-Generation/pharmacology , Duloxetine Hydrochloride/analogs & derivatives , Duloxetine Hydrochloride/pharmacology , Pain, Postoperative/drug therapy , Animals , Cell Line , Hyperalgesia/drug therapy , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Neurons/drug effects , Patch-Clamp Techniques , Physical Stimulation , Pituitary Gland/cytology , Pituitary Gland/drug effects , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology
3.
Anesth Analg ; 121(2): 532-44, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26049779

ABSTRACT

BACKGROUND: Duloxetine is an antidepressant effective for major depressive disorder and also the alleviation of pain for patients with diabetic peripheral neuropathy, chronic musculoskeletal pain, and fibromyalgia. How duloxetine works in pain relief remains unknown. In this study, we address whether duloxetine could act as an analgesic via systemic and local applications. METHODS: Efficacies of bupivacaine and duloxetine applied subcutaneously at the incision site against acute postoperative pain were compared after rat skin incision. Contralateral and intraperitoneal injections were used to assess systemic efficacy of duloxetine. Local anesthetic actions were assayed through functional block of the rat sciatic nerve. Inhibition by duloxetine of neuronal Na channels was characterized in rat GH3 cells. RESULTS: Our studies showed that subcutaneous duloxetine (2 mg) reduced hyperalgesia and allodynia for several days after skin incision, whereas subcutaneous bupivacaine (2 mg) did not. Contralaterally injected duloxetine (10 mg) had minimal effects on postoperative pain. Intraperitoneal duloxetine also reduced both allodynia and hyperalgesia, albeit at higher doses (10-20 mg). Duloxetine (2 mg) inhibited motor and nociceptive functions via sciatic nerve block for approximately 24 hours. It also reduced Na currents with 50% inhibitory concentrations of 30.4 ± 1.2 µM and 4.26 ± 0.19 µM (n = 8) for resting and fast-inactivated channels, respectively. Furthermore, duloxetine (10 µM) elicited additional use-dependent block of peak Na currents by approximately 70% when stimulated at 5 Hz. CONCLUSIONS: Our results demonstrate that duloxetine can act as a local anesthetic and an analgesic drug via both local and systemic applications. Because duloxetine inhibits neuronal Na currents with high potency, it may exert its antihyperalgesic effects through inhibition of the spontaneous nerve impulses that result from peripheral injury, encompassing its actions on multiple central nervous system and peripheral targets.


Subject(s)
Analgesics/pharmacology , Dermatologic Surgical Procedures , Hyperalgesia/prevention & control , Pain, Postoperative/prevention & control , Thiophenes/pharmacology , Administration, Cutaneous , Analgesics/administration & dosage , Anesthetics, Local/pharmacology , Animals , Bupivacaine/pharmacology , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Duloxetine Hydrochloride , Hyperalgesia/diagnosis , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Injections, Intraperitoneal , Male , Pain Measurement , Pain, Postoperative/diagnosis , Pain, Postoperative/metabolism , Pain, Postoperative/physiopathology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Sodium Channels/metabolism , Thiophenes/administration & dosage , Time Factors
4.
Mol Pharmacol ; 85(5): 692-702, 2014 May.
Article in English | MEDLINE | ID: mdl-24563546

ABSTRACT

Lacosamide is an anticonvulsant hypothesized to enhance slow inactivation of neuronal Na(+) channels for its therapeutic action. Cardiac Na(+) channels display less and incomplete slow inactivation, but their sensitivity toward lacosamide remains unknown. We therefore investigated the action of lacosamide in human cardiac Nav1.5 and Nav1.5-CW inactivation-deficient Na(+) channels. Lacosamide showed little effect on hNav1.5 Na(+) currents at 300 µM when cells were held at -140 mV. With 30-second conditioning pulses from -90 to -50 mV; however, hNav1.5 Na(+) channels became sensitive to lacosamide with IC50 (50% inhibitory concentration) around 70-80 µM. Higher IC50 values were found at -110 and -30 mV. The development of lacosamide block at -70 mV was slow in wild-type Na(+) channels (τ; 8.04 ± 0.39 seconds, n = 8). This time constant was significantly accelerated in hNav1.5-CW inactivation-deficient counterparts. The recovery from lacosamide block at -70 mV for 10 seconds was relatively rapid in wild-type Na(+) channels (τ; 639 ± 90 milliseconds, n = 8). This recovery was accelerated further in hNav1.5-CW counterparts. Unexpectedly, lacosamide elicited a time-dependent block of persistent hNav1.5-CW Na(+) currents with an IC50 of 242 ± 19 µM (n = 5). Furthermore, both hNav1.5-CW/F1760K mutant and batrachotoxin-activated hNav1.5 Na(+) channels became completely lacosamide resistant, indicating that the lacosamide receptor overlaps receptors for local anesthetics and batrachotoxin. Our results together suggest that lacosamide targets the intermediate preopen and open states of hNav1.5 Na(+) channels. Lacosamide may thus track closely the conformational changes at the hNav1.5-F1760 region along the activation pathway.


Subject(s)
Acetamides/metabolism , Acetamides/pharmacology , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Signal Transduction/physiology , Sodium Channel Blockers/metabolism , Sodium Channel Blockers/pharmacology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Lacosamide , Membrane Potentials/drug effects , Membrane Potentials/physiology , Protein Binding/physiology , Signal Transduction/drug effects , Time Factors
5.
Channels (Austin) ; 7(4): 263-74, 2013.
Article in English | MEDLINE | ID: mdl-23695971

ABSTRACT

Miniature persistent late Na(+) currents in cardiomyocytes have been linked to arrhythmias and sudden death. The goals of this study are to establish a stable cell line expressing robust persistent cardiac Na(+) currents and to test Class 1 antiarrhythmic drugs for selective action against resting and open states. After transient transfection of an inactivation-deficient human cardiac Na(+) channel clone (hNav1.5-CW with L409C/A410W double mutations), transfected mammalian HEK293 cells were treated with 1 mg/ml G-418. Individual G-418-resistant colonies were isolated using glass cylinders. One colony with high expression of persistent Na(+) currents was subjected to a second colony selection. Cells from this colony remained stable in expressing robust peak Na(+) currents of 996 ± 173 pA/pF at +50 mV (n = 20). Persistent late Na(+) currents in these cells were clearly visible during a 4-second depolarizing pulse albeit decayed slowly. This slow decay is likely due to slow inactivation of Na(+) channels and could be largely eliminated by 5 µM batrachotoxin. Peak cardiac hNav1.5-CW Na(+) currents were blocked by tetrodotoxin with an IC(50) value of 2.27 ± 0.08 µM (n = 6). At clinic relevant concentrations, Class 1 antiarrhythmics are much more selective in blocking persistent late Na(+) currents than their peak counterparts, with a selectivity ratio ranging from 80.6 (flecainide) to 3 (disopyramide). We conclude that (1) Class 1 antiarrhythmics differ widely in their resting- vs. open-channel selectivity, and (2) stably transfected HEK293 cells expressing large persistent hNav1.5-CW Na(+) currents are suitable for studying as well as screening potent open-channel blockers.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Electrophysiological Phenomena/drug effects , Ion Channel Gating/drug effects , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Transfection , Batrachotoxins/pharmacology , HEK293 Cells , Humans , Mutation , Myocytes, Cardiac/drug effects , Time Factors
6.
Reg Anesth Pain Med ; 34(4): 333-9, 2009.
Article in English | MEDLINE | ID: mdl-19574866

ABSTRACT

BACKGROUND AND OBJECTIVES: Elevated extracellular calcium ion has been shown to shift the voltage dependence of Na+- and K+-ion channels rightward, making the nerve less excitable. We hypothesized that calcium chloride (CaCl2) when used as an adjuvant prolongs and intensifies the block by local anesthetics (LAs). We investigated the effects of LAs combined with calcium in rat sciatic nerve blockade and in cultured rat GH3 cells expressing Na+ channels. Furthermore, we tested for histologic changes due to CaCl2. METHODS: We anesthetized rats with sevoflurane, exposed the sciatic nerves, and injected 0.2 mL of 1% lidocaine or 0.1% bupivacaine, alone or coadministered with 0.625%, 1.25%, 2.5%, or 5% CaCl2 (n = 8-10 per group). We assessed the complete-block time and complete-recovery time of proprioception, motor function, and nocifensive reaction. To elucidate the mechanism of nerve block, we performed electrophysiology experiments in cultured rat GH3 cells. Sciatic nerves were harvested at day 7 and stained with hemotoxylin/eosin. RESULTS: The addition of CaCl2 overall prolonged the duration of blockade by lidocaine or bupivacaine. Adding 10 mM CaCl2 to 300 microM lidocaine caused a right shift of the steady-state Na+-channel inactivation curve, indicating that the CaCl2 reduced the potency of lidocaine. Rat sciatic nerves treated with 1% lidocaine coadministered with 5% CaCl2 showed microscopic signs of neurotoxicity. CONCLUSIONS: The mechanism of prolonged nerve block of CaCl2 coadministered with LAs seems to be a raised threshold for nerve excitation. Major histopathologic changes at higher concentrations of CaCl2 are evident, and therefore, clinical application as an adjuvant to LAs seems unlikely.


Subject(s)
Anesthetics, Local , Bupivacaine , Calcium Chloride/pharmacology , Lidocaine , Nerve Block , Sciatic Nerve/drug effects , Animals , Drug Interactions , Electrophysiology , Male , Motor Activity/drug effects , Motor Activity/physiology , Proprioception/physiology , Rats , Rats, Sprague-Dawley , Sciatic Nerve/pathology , Sciatic Nerve/physiology , Sodium Channels/drug effects , Time Factors
7.
Anesthesiology ; 111(1): 127-37, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19512868

ABSTRACT

BACKGROUND: Nociceptive-selective local anesthesia is produced by entry of the permanently charged lidocaine-derivative QX-314 into nociceptors when coadministered with capsaicin, a transient receptor potential vanilloid 1 (TRPV1) channel agonist. However, the pain evoked by capsaicin before establishment of the QX-314-mediated block would limit clinical utility. Because TRPV1 channels are also activated by lidocaine, the authors tested whether lidocaine can substitute for capsaicin to introduce QX-314 into nociceptors through TRPV1 channels and produce selective analgesia. METHODS: Lidocaine (0.5% [17.5 mM], 1% [35 mM], and 2% [70 mM]) alone, QX-314 (0.2% [5.8 mM]) alone, and a combination of the two were injected subcutaneously and adjacent to the sciatic nerve in rats and mice. Mechanical and thermal responsiveness were measured, as was motor block. RESULTS: Coapplication of 0.2% QX-314 with lidocaine prolonged the nociceptive block relative to lidocaine alone, an effect attenuated in TRPV1 knockout mice. The 0.2% QX-314 alone had no effect when injected intraplantary or perineurally, and it produced only weak short-lasting inhibition of the cutaneous trunci muscle reflex. Perisciatic nerve injection of lidocaine with QX-314 produced a differential nociceptive block much longer than the transient motor block, lasting 2 h (for 1% lidocaine) to 9 h (2% lidocaine). Triple application of lidocaine, QX-314, and capsaicin further increased the duration of the differential block. CONCLUSIONS: Coapplication of lidocaine and its quaternary derivative QX-314 produces a long-lasting, predominantly nociceptor-selective block, likely by facilitating QX-314 entry through TRPV1 channels. Delivery of QX-314 into nociceptors by using lidocaine instead of capsaicin produces sustained regional analgesia without nocifensive behavior.


Subject(s)
Lidocaine/analogs & derivatives , Lidocaine/administration & dosage , Pain Measurement/drug effects , Sodium Channel Blockers/administration & dosage , Animals , Cells, Cultured , Drug Therapy, Combination , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Time Factors
8.
J Membr Biol ; 229(1): 1-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19418088

ABSTRACT

Articaine is widely used as a local anesthetic (LA) in dentistry, but little is known regarding its blocking actions on Na+ channels. We therefore examined the state-dependent block of articaine first in rat skeletal muscle rNav1.4 Na+ channels expressed in Hek293t cells. Articaine exhibited a weak block of resting rNav1.4 Na+ channels at -140 mV with a 50% inhibitory concentration (IC(50)) of 378 +/- 26 microM (n = 5). The affinity was higher for inactivated Na+ channels measured at -70 mV with an IC50 value of 40.6 +/- 2.7 microM (n = 5). The open-channel block by articaine was measured using inactivation-deficient rNav1.4 Na+ channels with an IC50 value of 15.8 +/- 1.5 microM (n = 5). Receptor mapping demonstrated that articaine interacted strongly with a D4S6 phenylalanine residue, which is known to form a part of the LA receptor. Thus the block of rNav1.4 Na+ channels by articaine is via the conserved LA receptor in a highly state-dependent manner, with a ranking order of open (23.9x) > inactivated (9.3x) > resting (1x) state. Finally, the open-channel block by articaine was likewise measured in inactivation-deficient hNav1.7 and rNav1.8 Na+ channels, with IC(50) values of 8.8 +/- 0.1 and 22.0 +/- 0.5 microM, respectively (n = 5), indicating that the high-affinity open-channel block by articaine is indeed preserved in neuronal Na+ channel isoforms.


Subject(s)
Carticaine/administration & dosage , Ion Channel Gating/physiology , Membrane Potentials/physiology , Muscle Proteins/physiology , Sodium Channels/physiology , Anesthetics, Local/administration & dosage , Cell Line , Dose-Response Relationship, Drug , Humans , Membrane Potentials/drug effects , NAV1.4 Voltage-Gated Sodium Channel
9.
Anesthesiology ; 109(5): 872-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18946300

ABSTRACT

BACKGROUND: Transient receptor potential vanilloid 1 channels integrate nociceptive stimuli and are predominantly expressed by unmyelinated C-fiber nociceptors, but not low-threshold mechanoreceptive sensory or motor fibers. A recent report showed that the transient receptor potential vanilloid 1 channel agonist capsaicin allows a hydrophilic quaternary ammonium derivative of lidocaine, QX-314, to selectively block C fibers without motor block. The authors tested whether a similar differential block would be produced using amphipathic N-methyl amitriptyline, amitriptyline, bupivacaine, or lidocaine, either alone or together with 0.05% capsaicin, in a rat sciatic nerve block model. METHODS: Rats (n = 8/group) were anesthetized with sevoflurane, and 0.2 ml of drug was injected either alone or with capsaicin (simultaneously or 10 min later) next to the sciatic nerve in the sciatic notch. Motor function was assessed by the extensor postural thrust. Nociception was evaluated by the nocifensive withdrawal reflex and vocalization evoked by pinch of a skin fold over the lateral metatarsus (cutaneous pain) with a serrated forceps. RESULTS: N-Methyl amitriptyline, amitriptyline, bupivacaine, or lidocaine, followed by injection of capsaicin 10 min later, each elicited a predominantly nociceptive-specific blockade. In comparison, simultaneous application of each local anesthetic with capsaicin did not elicit a clinically significant differential block, with the exception of N-methyl amitriptyline. CONCLUSIONS: Both tertiary amine local anesthetics and their quaternary ammonium derivatives can elicit a predominantly sensory/nociceptor selective block when followed by injection of capsaicin. The combined application of transient receptor potential vanilloid 1 channel agonists and various local anesthetics or their quaternary ammonium derivatives is an appealing strategy to achieve a long-lasting differential block in regional analgesia.


Subject(s)
Anesthetics, Local/administration & dosage , Capsaicin/administration & dosage , Nerve Block/methods , Pain Measurement/drug effects , Sciatic Nerve/drug effects , Animals , Drug Therapy, Combination , Male , Pain/physiopathology , Pain/prevention & control , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Sciatic Nerve/physiology
10.
Anesth Analg ; 107(4): 1397-405, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18806059

ABSTRACT

BACKGROUND: Bulleyaconitine A (BLA) is an analgesic and antiinflammatory drug isolated from Aconitum plants. BLA has several potential targets, including voltage-gated Na+ channels. We tested whether BLA elicited long-lasting cutaneous analgesia, when co-injected with lidocaine and epinephrine, as a model for prolonged infiltration anesthesia. METHODS: The local anesthetic properties of BLA were assessed by the patch-clamp technique in HEK293t cells expressing Nav1.7 and Nav1.8 neuronal Na+ channels, both crucial for nociception. Drug solutions (0.6 mL) were injected subcutaneously via rat shaved dorsal skin. Inhibition of the cutaneous trunci muscle reflex was evaluated by pinpricks. Skin cross-sections were stained with hematoxylin and eosin or with antibodies against PGP9.5. RESULTS: BLA at 10 microM interacted minimally with resting or inactivated Nav1.7 and Nav1.8 Na+ channels when infrequently stimulated to +50 mV for 3 ms. However, when stimulated at 2 Hz for 1000 pulses, their peak Na+ currents were >90% reduced by BLA. This use-dependent inhibition was not significantly reversed after 15-min washing. Complete nociceptive blockade after injection of lidocaine (0.5%)/epinephrine (1:200,000) lasted for approximately 1 h in rats; full recovery occurred after approximately 6 h. Co-injection of 0.125 mM BLA with lidocaine/epinephrine increased the duration of complete nociceptive blockade to 24 h. Full recovery occurred after approximately 6 days. Skin histology including peripheral nerve fibers appeared unaffected by BLA. CONCLUSIONS: BLA inhibits Nav1.7 and Nav1.8 Na+ currents in a use-dependent manner. Co-injection of BLA at

Subject(s)
Aconitine/analogs & derivatives , Adjuvants, Anesthesia/administration & dosage , Analgesia , Anesthetics, Local/administration & dosage , Skin/innervation , Aconitine/administration & dosage , Aconitine/pharmacology , Aconitine/toxicity , Adjuvants, Anesthesia/pharmacology , Adjuvants, Anesthesia/toxicity , Anesthetics, Combined , Anesthetics, Local/pharmacology , Anesthetics, Local/toxicity , Animals , Dose-Response Relationship, Drug , Epinephrine/administration & dosage , Hepatocytes , Humans , Injections, Subcutaneous , Lidocaine/administration & dosage , NAV1.7 Voltage-Gated Sodium Channel , NAV1.8 Voltage-Gated Sodium Channel , Patch-Clamp Techniques , Rats , Skin/pathology , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism , Vasoconstrictor Agents/administration & dosage , Vasoconstrictor Agents/pharmacology
11.
J Membr Biol ; 222(2): 79-90, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18418539

ABSTRACT

Antidepressants, such as traditional tricyclic antidepressants (TCAs), are the first-line treatment for various pain syndromes. Available evidence indicates that TCAs may target Na+ channels for their analgesic action. In this report, we examined the effects of contemporary antidepressants sertraline and paroxetine on (1) neuronal Na+ channels expressed in GH3 cells and (2) muscle rNav1.4 Na+ channels heterologously expressed in Hek293t cells. Our results showed that both antidepressants blocked Na+ channels in a highly state-dependent manner. The 50% inhibitory concentrations (IC50) for sertraline and paroxetine ranged approximately 18-28 microM: for resting block and approximately 2-8 microM: for inactivated block of neuronal and rNav1.4 Na+ channels. Surprisingly, the IC50 values for both drugs were about 0.6-0.7 microM: for the open channel block of persistent late Na+ currents generated through inactivation-deficient rNav1.4 mutant Na+ channels. For comparison, the open channel block in neuronal hNav1.7 counterparts yielded IC50 values around 0.3-0.4 microM: for both drugs. Receptor mapping using fast inactivation-deficient rNav1.4-F1579A/K mutants with reduced affinities toward local anesthetics (LAs) and TCAs indicated that the F1579 residue is not involved in the binding of sertraline and paroxetine. Thus, sertraline and paroxetine are potent open channel blockers that target persistent late Na+ currents preferentially, but their block is not mediated via the phenylalanine residue at the known LA/TCA receptor site.


Subject(s)
Antidepressive Agents/pharmacology , Paroxetine/pharmacology , Sertraline/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Amino Acid Substitution , Anesthetics, Local/pharmacology , Animals , Binding Sites/genetics , Cell Line , Humans , Kinetics , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutagenesis, Site-Directed , NAV1.7 Voltage-Gated Sodium Channel , Phenylalanine/chemistry , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium Channels/chemistry , Sodium Channels/genetics , Sodium Channels/metabolism
12.
Mol Pharmacol ; 73(3): 940-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18079277

ABSTRACT

Ranolazine is an antianginal agent that targets a number of ion channels in the heart, including cardiac voltage-gated Na(+) channels. However, ranolazine block of muscle and neuronal Na(+) channel isoforms has not been examined. We compared the state- and use-dependent ranolazine block of Na(+) currents carried by muscle Nav1.4, cardiac Nav1.5, and neuronal Nav1.7 isoforms expressed in human embryonic kidney 293T cells. Resting and inactivated block of Na(+) channels by ranolazine were generally weak, with a 50% inhibitory concentration (IC(50)) >/= 60 microM. Use-dependent block of Na(+) channel isoforms by ranolazine during repetitive pulses (+50 mV/10 ms at 5 Hz) was strong at 100 microM, up to 77% peak current reduction for Nav1.4, 67% for Nav1.5, and 83% for Nav1.7. In addition, we found conspicuous time-dependent block of inactivation-deficient Nav1.4, Nav1.5, and Nav1.7 Na(+) currents by ranolazine with estimated IC(50) values of 2.4, 6.2, and 1.7 microM, respectively. On- and off-rates of ranolazine were 8.2 microM(-1) s(-1) and 22 s(-1), respectively, for Nav1.4 open channels and 7.1 microM(-1) s(-1) and 14 s(-1), respectively, for Nav1.7 counterparts. A F1579K mutation at the local anesthetic receptor of inactivation-deficient Nav1.4 Na(+) channels reduced the potency of ranolazine approximately 17-fold. We conclude that: 1) both muscle and neuronal Na(+) channels are as sensitive to ranolazine block as their cardiac counterparts; 2) at its therapeutic plasma concentrations, ranolazine interacts predominantly with the open but not resting or inactivated Na(+) channels; and 3) ranolazine block of open Na(+) channels is via the conserved local anesthetic receptor albeit with a relatively slow on-rate.


Subject(s)
Acetanilides/pharmacology , Ion Channel Gating/physiology , Muscle Proteins/drug effects , Muscles/physiology , Neurons/physiology , Piperazines/pharmacology , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Sodium Channels/physiology , Acetanilides/chemistry , Cell Line , Dose-Response Relationship, Drug , Electrophysiology , Humans , Inhibitory Concentration 50 , Ion Channel Gating/drug effects , Kidney/cytology , Kinetics , Molecular Structure , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle Proteins/physiology , Muscles/drug effects , Mutation , NAV1.4 Voltage-Gated Sodium Channel , NAV1.7 Voltage-Gated Sodium Channel , Neurons/drug effects , Patch-Clamp Techniques , Piperazines/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Ranolazine , Recombinant Proteins/metabolism , Sodium Channels/genetics , Sodium Channels/metabolism , Transfection
13.
Reg Anesth Pain Med ; 32(4): 288-95, 2007.
Article in English | MEDLINE | ID: mdl-17720112

ABSTRACT

BACKGROUND AND OBJECTIVES: Magnesium sulfate (MgSO(4)) is well known as an antagonist of N-methyl-d-aspartate receptors and was used for intrathecal analgesia a century ago. However, the effects of MgSO(4) combined with local anesthetics (LAs) on peripheral nerves are unclear. We tested the hypothesis that MgSO(4) could be used as an adjuvant to prolong and intensify conduction block by amide-type LAs in a rat sciatic-nerve block model. Further, the mechanism of possible synergy between LAs and MgSO(4) was investigated in whole-cell mode patch-clamp experiments. METHODS: Sciatic nerves were exposed to 2%/73.9 mM lidocaine, 0.25%/7.7 mM bupivacaine, and 0.5%/15.4 mM ropivacaine, with or without addition of 1.25%, 2.5%, or 5% MgSO(4)/50.7 mM, and nerve block characteristics were assessed. To elucidate the LA-MgSO(4) interaction, voltage-dependent inactivation curves were determined in cultured rat GH(3) cells that expressed neuronal Na(+) channels. RESULTS: Unexpectedly, the addition of MgSO(4) overall significantly shortened the duration of block by lidocaine, bupivacaine, and ropivacaine. The steady-state inactivation of Na(+) channels in the presence of 300 muM lidocaine was almost unchanged by the addition of 10 mM MgSO(4), indicating that MgSO(4) does not affect the potency of lidocaine toward the inactivated Na(+) channel. CONCLUSIONS: MgSO(4) coadministered with amide-type LAs shortened the duration of sciatic-nerve block in rats. Therefore, it does not seem to be useful as an adjuvant for peripheral-nerve block. The mechanism of this observed antagonism is unclear but appears to be independent of the action of LAs and MgSO(4) at the LA receptor within the Na(+) channel.


Subject(s)
Anesthetics, Local/pharmacology , Magnesium Sulfate/pharmacology , Sciatic Nerve/metabolism , Sodium Channels/drug effects , Amides , Anesthetics, Local/administration & dosage , Animals , Bupivacaine , Dose-Response Relationship, Drug , Lidocaine , Magnesium Sulfate/administration & dosage , Motor Activity/drug effects , Nerve Block/methods , Pain Measurement/drug effects , Proprioception/drug effects , Rats , Rats, Sprague-Dawley , Ropivacaine , Sciatic Nerve/drug effects
14.
Anesthesiology ; 107(1): 82-90, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17585219

ABSTRACT

BACKGROUND: Bulleyaconitine A (BLA) is an active ingredient of Aconitum bulleyanum plants. BLA has been approved for the treatment of chronic pain and rheumatoid arthritis in China, but its underlying mechanism remains unclear. METHODS: The authors examined (1) the effects of BLA on neuronal voltage-gated Na channels in vitro under the whole cell patch clamp configuration and (2) the sensory and motor functions of rat sciatic nerve after single BLA injections in vivo. RESULTS: BLA at 10 microm did not affect neuronal Na currents in clonal GH3 cells when stimulated infrequently to +50 mV. When stimulated at 2 Hz for 1,000 pulses (+50 mV for 4 ms), BLA reduced the peak Na currents by more than 90%. This use-dependent reduction of Na currents by BLA reversed little after washing. Single injections of BLA (0.2 ml at 0.375 mm) into the rat sciatic notch not only blocked sensory and motor functions of the sciatic nerve but also induced hyperexcitability, followed by sedation, arrhythmia, and respiratory distress. When BLA at 0.375 mm was coinjected with 2% lidocaine (approximately 80 mm) or epinephrine (1:100,000) to reduce drug absorption by the bloodstream, the sensory and motor functions of the sciatic nerve remained fully blocked for approximately 4 h and regressed completely after approximately 7 h, with minimal systemic effects. CONCLUSIONS: BLA reduces neuronal Na currents strongly at +50 mV in a use-dependent manner. When coinjected with lidocaine or epinephrine, BLA elicits prolonged block of both motor and sensory functions in rats with minimal adverse effects.


Subject(s)
Aconitine/analogs & derivatives , Aconitum/chemistry , Analgesics/pharmacology , Anesthetics, Local , Aconitine/pharmacology , Alkaloids/pharmacology , Animals , Behavior, Animal/drug effects , Cell Line , Electrophysiology , Epinephrine/pharmacology , Lidocaine/pharmacology , Male , Muscle, Skeletal/drug effects , Pain Measurement/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Reflex/drug effects , Sodium Channels/drug effects , Vasoconstrictor Agents/pharmacology
15.
Pflugers Arch ; 454(2): 277-87, 2007 May.
Article in English | MEDLINE | ID: mdl-17205354

ABSTRACT

Sequence alignment of four S6 segments in the human cardiac Na+ channel suggests that serine-401 (hNav1.5-S401) at D1S6 along with asparagine-927 (N927) at D2S6, serine-1458 (S1458) at D3S6, and phenylalanine-1760 (F1760) at D4S6 may jointly form a pore-facing S(401)N(927)S(1458)F(1760) ring. Importantly, this pore-facing structure is adjacent to the putative gating-hinge (G(400)G(926)G(1457)S(1759)) and close to the selectivity filter. Within this SNSF ring, only S401 has not yet been identified as a batrachotoxin (BTX) sensing residue. We therefore created S401 mutants with 12 substitutions (S401C,W,P,A,K,F,R,E,L,N,D,G) and assayed their BTX sensitivity. All S401 mutants expressed Na+ currents but often with altered gating characteristics. Ten mutants were found sensitive to 5 muM BTX, which eliminated Na+ channel fast inactivation after repetitive pulses. However, S401K and S401R became BTX resistant. In addition, the block of open and inactivated hNav1.5-S401K Na+ channels by local anesthetic bupivacaine was reduced by approximately 8-10-fold, but not the block of resting Na+ channels. Qualitatively, these ligand-sensing phenotypes of hNav1.5-S401K channels resemble those of S1458K and F1760K channels reported earlier. Together, our results support that residue hNav1.5-S401 at D1S6 is facing the inner cavity and is in close proximity to the receptor sites for BTX and for local anesthetics.


Subject(s)
Anesthetics, Local/metabolism , Batrachotoxins/metabolism , Muscle Proteins/physiology , Serine/metabolism , Sodium Channels/physiology , Amino Acid Sequence , Amino Acid Substitution , Anesthetics, Local/pharmacology , Batrachotoxins/pharmacology , Binding Sites/genetics , Bupivacaine/pharmacology , Cell Line , Computer Simulation , Electric Stimulation , Electrophysiology , Gene Expression/genetics , Humans , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Models, Molecular , Molecular Sequence Data , Muscle Proteins/genetics , Muscle Proteins/metabolism , Mutagenesis, Site-Directed , NAV1.5 Voltage-Gated Sodium Channel , Protein Binding , Recombinant Proteins/metabolism , Serine/genetics , Sodium/metabolism , Sodium Channels/genetics , Sodium Channels/metabolism , Transfection
16.
Pain ; 127(1-2): 73-83, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16962240

ABSTRACT

Capsaicin elicits burning pain via the activation of the vanilloid receptor (TRPV1). Intriguingly, several reports showed that capsaicin also inhibits Na+ currents but the mechanisms remain unclear. To explore this non-TRPV1 action we applied capsaicin to HEK293 cells stably expressing inactivation-deficient rat skeletal muscle Na+ mutant channels (rNav1.4-WCW). Capsaicin elicited a conspicuous time-dependent block of inactivation-deficient Na+ currents. The 50% inhibitory concentration (IC50) of capsaicin for open Na+ channels at +30 mV was measured 6.8+/-0.6 microM (n=5), a value that is 10-30 times lower than those for resting (218 microM) and inactivated (74 microM) wild-type Na+ channels. On-rate and off-rate constants for capsaicin open-channel block at +30 mV were estimated to be 6.37 microM(-1) s(-1) and 34.4 s(-1), respectively, with a calculated dissociation constant (KD) of 5.4 microM. Capsaicin at 30 microM produced approximately 70% additional use-dependent block of remaining rNav1.4-WCW Na+ currents during repetitive pulses at 1 Hz. Site-directed mutagenesis showed that the local anesthetic receptor was not responsible for the capsaicin block of the inactivation-deficient Na+ channel. Interestingly, capsaicin elicited little time-dependent block of batrachotoxin-modified rNav1.4-WCW Na+ currents, indicating that batrachotoxin prevents capsaicin binding. Finally, neuronal open Na+ channels endogenously expressed in GH3 cells were as sensitive to capsaicin block as rNav1.4 counterparts. We conclude that capsaicin preferentially blocks persistent late Na+ currents, probably via a receptor that overlaps the batrachotoxin receptor but not the local anesthetic receptor. Drugs that target such a non-TRPV1 receptor could be beneficial for patients with neuropathic pain.


Subject(s)
Capsaicin/administration & dosage , Ion Channel Gating/physiology , Kidney/physiology , Membrane Potentials/physiology , Muscle Fibers, Skeletal/physiology , Sodium Channels/physiology , TRPV Cation Channels/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Ion Channel Gating/drug effects , Kidney/drug effects , Membrane Potentials/drug effects , Muscle Fibers, Skeletal/drug effects , Rats , Sodium Channel Blockers/administration & dosage , Sodium Channels/drug effects , TRPV Cation Channels/chemistry
17.
Channels (Austin) ; 1(3): 179-88, 2007.
Article in English | MEDLINE | ID: mdl-18690024

ABSTRACT

Batrachotoxin (BTX) not only keeps the voltage-gated Na(+) channel open persistently but also reduces its single-channel conductance. Although a BTX receptor has been delimited within the inner cavity of Na(+) channels, how Na(+) ions flow through the BTX-bound permeation pathway remains unclear. In this report we tested a hypothesis that Na(+) ions traverse a narrow gap between bound BTX and residue N927 at D2S6 of cardiac hNa(v)1.5 Na(+) channels. We found that BTX at 5 microM indeed elicited a strong block of hNa(v)1.5-N927K currents (approximately 70%) after 1000 repetitive pulses (+50 mV/20 ms at 2 Hz) without any effects on Na(+) channel gating. Once occurred, this unique use-dependent block of hNa(v)1.5-N927K Na(+) channels recovered little at holding potential (-140 mV), demonstrating that BTX block is irreversible under our experimental conditions. Such an irreversible effect likewise developed in fast inactivation-deficient hNa(v)1.5-N927K Na(+) channels albeit with a faster on-rate; approximately 90% of peak Na(+) currents were abolished by BTX after 200 repetitive pulses (+50 mV/20 ms). This use-dependent block of fast inactivation-deficient hNa(v)1.5-N927K Na(+) channels by BTX was duration dependent. The longer the pulse duration the larger the block developed. Among N927K/W/R/H/D/S/Q/G/E substitutions in fast inactivation-deficient hNa(v)1.5 Na(+) channels, only N927K/R Na(+) currents were highly sensitive to BTX block. We conclude that (a) BTX binds within the inner cavity and partly occludes the permeation pathway and (b) residue hNa(v)1.5-N927 is critical for ion permeation between bound BTX and D2S6, probably because the side-chain of N927 helps coordinate permeating Na(+) ions.


Subject(s)
Batrachotoxins/pharmacology , Muscle Proteins/antagonists & inhibitors , Mutation , Myocytes, Cardiac/drug effects , Sodium Channel Blockers/pharmacology , Sodium/metabolism , Animals , Asparagine , Batrachotoxins/chemistry , Batrachotoxins/metabolism , Binding Sites , Cell Line , Cell Membrane Permeability , Humans , Ion Channel Gating/drug effects , Membrane Potentials , Models, Molecular , Molecular Structure , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel , Protein Binding , Protein Conformation , Rats , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/metabolism , Sodium Channels/chemistry , Sodium Channels/genetics , Sodium Channels/metabolism , Time Factors , Transfection
18.
J Neurophysiol ; 96(4): 1848-59, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16807347

ABSTRACT

The calmodulin (CaM) inhibitor trifluoperazine (TFP) can produce analgesia when given intrathecally to rats; however, the mechanism is not known. We asked whether TFP could modulate the Na(v)1.7 sodium channel, which is highly expressed in the peripheral nervous system and plays an important role in nociception. We show that 500 nM and 2 muM TFP induce major decreases in Na(v)1.7 and Na(v)1.4 current amplitudes and that 2 muM TFP causes hyperpolarizing shifts in the steady-state inactivation of Na(v)1.7 and Na(v)1.4. CaM can bind to the C-termini of voltage-gated sodium channels and modulate their functional properties; therefore we investigated if TFP modulation of sodium channels was due to CaM inhibition. However, the TFP inhibition was not replicated by whole cell dialysis of a calmodulin inhibitory peptide, indicating that major effects of TFP do not involve a disruption of CaM-channel interactions. Rather, our data show that TFP inhibition is state dependent and that the majority of the TFP inhibition depends on specific amino-acid residues in the local anesthetic receptor site in sodium channels. TFP was also effective in vivo in causing motor and sensory blockade after subfascial injection to the rat sciatic nerve. The state-dependent block of Na(v)1.7 channels with nanomolar concentrations of TFP raises the possibility that TFP, or TFP analogues, might be useful for regional anesthesia and pain management and could be more potent than traditional local anesthetics.


Subject(s)
Analgesics/pharmacology , Muscle Proteins/drug effects , Receptors, Drug/physiology , Sodium Channels/drug effects , Trifluoperazine/pharmacology , Action Potentials/physiology , Anesthetics, Local/pharmacology , Calmodulin/antagonists & inhibitors , Calmodulin/physiology , Cell Line , Dose-Response Relationship, Drug , Humans , Muscle Proteins/genetics , Muscle Proteins/physiology , Mutation , NAV1.4 Voltage-Gated Sodium Channel , NAV1.7 Voltage-Gated Sodium Channel , Patch-Clamp Techniques , Receptors, Drug/drug effects , Sciatic Nerve/drug effects , Sodium Channels/genetics , Sodium Channels/physiology
19.
J Gen Physiol ; 127(3): 277-89, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16505148

ABSTRACT

Resurgent tail Na(+) currents were first discovered in cerebellar Purkinje neurons. A recent study showed that a 14-mer fragment of a mouse beta4 subunit, beta4(154-167), acts as an intracellular open-channel blocker and elicits resurgent currents in Purkinje neurons (Grieco, T.M., J.D. Malhotra, C. Chen, L.L. Isom, and I.M. Raman. 2005. Neuron. 45:233-244). To explore these phenotypes in vitro, we characterized beta4(154-167) actions in inactivation-deficient cardiac hNav1.5 Na(+) channels expressed in human embryonic kidney 293t cells. Intracellular beta4(154-167) from 25-250 microM elicited a conspicuous time-dependent block of inactivation-deficient Na(+) currents at 50 mV in a concentration-dependent manner. On and off rates for beta4(154-167) binding were estimated at 10.1 microM(-1)s(-1) and 49.1 s(-1), respectively. Upon repolarization, large tail currents emerged with a slight delay at -140 mV, probably as a result of the rapid unblocking of beta4(154-167). Near the activation threshold (approximately -70 mV), resurgent tail currents were robust and long lasting. Likewise, beta4(154-167) induces resurgent currents in wild-type hNav1.5 Na(+) channels, although to a lesser extent. The inactivation peptide acetyl-KIFMK-amide not only restored the fast inactivation phenotype in hNav1.5 inactivation-deficient Na(+) channels but also elicited robust resurgent currents. When modified by batrachotoxin (BTX), wild-type hNav1.5 Na(+) channels opened persistently but became resistant to beta4(154-167) and acetyl-KIFMK-amide block. Finally, a lysine substitution of a phenylalanine residue at D4S6, F1760, which forms a part of receptors for local anesthetics and BTX, rendered cardiac Na(+) channels resistant to beta4(154-167). Together, our in vitro studies identify a putative S6-binding site for beta4(154-167) within the inner cavity of hNav1.5 Na(+) channels. Such an S6 receptor readily explains (1) why beta4(154-167) gains access to its receptor as an open-channel blocker, (2), why bound beta4(154-167) briefly prevents the activation gate from closing by a "foot-in-the-door" mechanism during deactivation, (3) why BTX inhibits beta4(154-167) binding by physical exclusion, and (4) why a lysine substitution of residue F1760 eliminates beta4(154-167) binding.


Subject(s)
Ion Channel Gating/physiology , Kidney/metabolism , Membrane Potentials/physiology , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/metabolism , Peptides/administration & dosage , Sodium Channels/metabolism , Amino Acid Substitution , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , Ion Channel Gating/drug effects , Kidney/drug effects , Kinetics , Membrane Potentials/drug effects , Mice , Muscle Proteins/genetics , Mutagenesis, Site-Directed , NAV1.5 Voltage-Gated Sodium Channel , Sodium Channels/genetics , Structure-Activity Relationship , Time Factors
20.
Reg Anesth Pain Med ; 31(1): 14-8, 2006.
Article in English | MEDLINE | ID: mdl-16418019

ABSTRACT

BACKGROUND AND OBJECTIVES: The adrenergic drug ephedrine inhibits Na(+) current in cultured cells expressing Na(+) channels and provides dose-dependent reversible rat sciatic nerve blockade. The dosage required for peripheral nerve blockade in humans would cause unacceptable cardiovascular side effects. We therefore hypothesized that either lidocaine or bupivacaine would show synergy with ephedrine in a rat sciatic nerve block model, allowing a dose reduction and limiting side effects while improving efficacy. METHODS: Sciatic nerves of anesthetized rats were exposed by lateral incision of the thighs; 0.2 mL of different concentrations of the following drugs was injected sub-fascially: bupivacaine, lidocaine, and ephedrine alone and bupivacaine or lidocaine combined with ephedrine (n=8 per group). After animals recovered from anesthesia, a blinded investigator evaluated motor blockade (push against a balance) and sensory/nociceptive blockade (to pinch of the fifth toe) at predefined time intervals. RESULTS: Ephedrine combined with bupivacaine interacted additively for both motor and sensory blockade, and ephedrine combined with lidocaine interacted antagonistically (sub-additive) for sensory blockade and additively for motor blockade. CONCLUSIONS: A combination of ephedrine with either lidocaine or bupivacaine is unlikely to allow a significant dose reduction because of a lack of synergy. Furthermore, the cardiovascular side effects will limit the maximal tolerable dosage of ephedrine.


Subject(s)
Adrenergic Agents/administration & dosage , Anesthetics, Local/administration & dosage , Bupivacaine/administration & dosage , Ephedrine/administration & dosage , Lidocaine/administration & dosage , Nerve Block , Sciatic Nerve/drug effects , Animals , Dose-Response Relationship, Drug , Drug Combinations , Evoked Potentials, Motor/drug effects , Evoked Potentials, Somatosensory/drug effects , Male , Models, Animal , Pain Measurement , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...