Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Br J Pharmacol ; 181(12): 1734-1756, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38157867

ABSTRACT

BACKGROUND AND PURPOSE: Neuropathic pain affects up to 10% of the global population and is caused by an injury or a disease affecting the somatosensory, peripheral, or central nervous system. NP is characterized by chronic, severe and opioid-resistant properties. Therefore, its clinical management remains very challenging. The N-type voltage-gated calcium channel, Cav2.2, is a validated target for therapeutic intervention in chronic and neuropathic pain. The conotoxin ziconotide (Prialt®) is an FDA-approved drug that blocks Cav2.2 channel but needs to be administered intrathecally. Thus, although being principally efficient, the required application route is very much in disfavour. EXPERIMENTAL APPROACH AND KEY RESULTS: Here, we describe an orally available drug candidate, RD2, which competes with ziconotide binding to Cav2.2 at nanomolar concentrations and inhibits Cav2.2 almost completely reversible. Other voltage-gated calcium channel subtypes, like Cav1.2 and Cav3.2, were affected by RD2 only at concentrations higher than 10 µM. Data from sciatic inflammatory neuritis rat model demonstrated the in vivo proof of concept, as low-dose RD2 (5 mg·kg-1) administered orally alleviated neuropathic pain compared with vehicle controls. High-dose RD2 (50 mg·kg-1) was necessary to reduce pain sensation in acute thermal response assessed by the tail flick test. CONCLUSIONS AND IMPLICATIONS: Taken together, these results demonstrate that RD2 has antiallodynic properties. RD2 is orally available, which is the most convenient application form for patients and caregivers. The surprising and novel result from standard receptor screens opens the room for further optimization into new promising drug candidates, which address an unmet medical need.


Subject(s)
Calcium Channel Blockers , Calcium Channels, N-Type , Neuralgia , Animals , Humans , Male , Mice , Rats , Administration, Oral , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Calcium Channels, N-Type/metabolism , Calcium Channels, N-Type/drug effects , Dose-Response Relationship, Drug , Mice, Inbred C57BL , Neuralgia/drug therapy , omega-Conotoxins/administration & dosage , omega-Conotoxins/pharmacology , omega-Conotoxins/therapeutic use , Rats, Inbred Lew
2.
Naunyn Schmiedebergs Arch Pharmacol ; 393(9): 1715-1728, 2020 09.
Article in English | MEDLINE | ID: mdl-32388600

ABSTRACT

The present research work was designed to examine the neuroprotective effect of ethanolic extract of Solanum virginianum Linn. (SV) in chronic construction injury (CCI) of sciatic nerve-induced neuropathic pain in rats. The extract was initially standardized by high-performance thin-layer chromatography using solasodine as a biomarker and was then subjected to assess the degree of mechanical allodynia, thermal allodynia, mechanical hyperalgesia, thermal hyperalgesia and biochemical evaluations. Administration of SV (100 and 200 mg/kg; p.o.) and pregabalin (10 mg/kg; p.o.) as a reference standard significantly debilitated hyperalgesia and allodynia and notably restored the altered antioxidant level and pro-inflammatory cytokine (IL-1ß and TNF-α) expression in a dose-dependent manner. Further, to appraise the mechanistic approach of solasodine, docking simulation studies were done on the 3D structure of the voltage-gated N-type calcium channel (Cav 2.2), R-type calcium channel (Cav 2.3) and sodium channel (Nav 1.7), and the results revealed that solasodine properly positioned into Phe 19, Leu 32, Met 51 and Met 71 (FLMM pocket) of Cav 2.2 and Cav 2.3 and being a competitor of Ca2+/N-lobe it may inactivate these calcium channels but did not bind into the desired binding pocket of Nav 1.7. Thus, the study confirmed the role of solasodine as a major biomarker for the observed neuroprotective nature of Solanum virginianum.


Subject(s)
Analgesics/pharmacology , Hyperalgesia/prevention & control , Molecular Docking Simulation , Neuralgia/prevention & control , Pain Threshold/drug effects , Plant Extracts/pharmacology , Sciatic Neuropathy/drug therapy , Solanaceous Alkaloids/pharmacology , Solanum , Analgesics/isolation & purification , Analgesics/metabolism , Animals , Behavior, Animal/drug effects , Binding Sites , Binding, Competitive , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/metabolism , Disease Models, Animal , Ethanol/chemistry , Female , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Male , Neuralgia/metabolism , Neuralgia/physiopathology , Plant Extracts/isolation & purification , Plant Extracts/metabolism , Protein Binding , Rats, Wistar , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/physiopathology , Solanaceous Alkaloids/isolation & purification , Solanaceous Alkaloids/metabolism , Solanum/chemistry , Solvents/chemistry
3.
Sci Rep ; 8(1): 11328, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054511

ABSTRACT

Sophorae Flavescentis Radix (SFR) is a medicinal herb with many functions that are involved in anti-inflammation, antinociception, and anticancer. SFR is also used to treat a variety of itching diseases. Matrine (MT) is one of the main constituents in SFR and also has the effect of relieving itching, but the antipruritic mechanism is still unclear. Here, we investigated the effect of MT on anti-pruritus. In acute and chronic itch models, MT significantly inhibited the scratching behavior not only in acute itching induced by histamine (His), chloroquine (CQ) and compound 48/80 with a dose-depended manner, but also in the chronic pruritus models of atopic dermatitis (AD) and acetone-ether-water (AEW) in mice. Furthermore, MT could be detected in the blood after intraperitoneal injection (i.p.) and subcutaneous injection (s.c.). Finally, electrophysiological and calcium imaging results showed that MT inhibited the excitatory synaptic transmission from dorsal root ganglion (DRG) to the dorsal horn of the spinal cord by suppressing the presynaptic N-type calcium channel. Taken together, we believe that MT is a novel drug candidate in treating pruritus diseases, especially for histamine-independent and chronic pruritus, which might be attributed to inhibition of the presynaptic N-type calcium channel.


Subject(s)
Alkaloids/administration & dosage , Antipruritics/administration & dosage , Calcium Channel Blockers/administration & dosage , Pruritus/drug therapy , Quinolizines/administration & dosage , Alkaloids/chemistry , Animals , Antipruritics/chemistry , Calcium/metabolism , Calcium Channel Blockers/chemistry , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/genetics , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Male , Mice , Pruritus/genetics , Pruritus/pathology , Quinolizines/chemistry , Sophora/chemistry , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Matrines
4.
J Neurosci ; 33(49): 19314-25, 2013 Dec 04.
Article in English | MEDLINE | ID: mdl-24305827

ABSTRACT

Free fatty acids receptor 3 (FFA3, GPR41) and 2 (FFA2, GPR43), for which the short-chain fatty acids (SCFAs) acetate and propionate are agonist, have emerged as important G-protein-coupled receptors influenced by diet and gut flora composition. A recent study (Kimura et al., 2011) demonstrated functional expression of FFA3 in the rodent sympathetic nervous system (SNS) providing a potential link between nutritional status and autonomic function. However, little is known of the source of endogenous ligands, signaling pathways, or effectors in sympathetic neurons. In this study, we found that FFA3 and FFA2 are unevenly expressed in the rat SNS with higher transcript levels in prevertebral (e.g., celiac-superior mesenteric and major pelvic) versus paravertebral (e.g., superior cervical and stellate) ganglia. FFA3, whether heterologously or natively expressed, coupled via PTX-sensitive G-proteins to produce voltage-dependent inhibition of N-type Ca(2+) channels (Cav2.2) in sympathetic neurons. In addition to acetate and propionate, we show that ß-hydroxybutyrate (BHB), a metabolite produced during ketogenic conditions, is also an FFA3 agonist. This contrasts with previous interpretations of BHB as an antagonist at FFA3. Together, these results indicate that endogenous BHB levels, especially when elevated under certain conditions, such as starvation, diabetic ketoacidosis, and ketogenic diets, play a potentially important role in regulating the activity of the SNS through FFA3.


Subject(s)
3-Hydroxybutyric Acid/pharmacology , Calcium Channels, N-Type/drug effects , Neurons/physiology , Receptors, G-Protein-Coupled/agonists , Sympathetic Nervous System/physiology , Animals , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Electrophysiological Phenomena/physiology , Fluorescence Resonance Energy Transfer , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/physiology , HeLa Cells , Humans , In Situ Hybridization , Ketone Bodies/pharmacology , Ligands , Male , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Sympathetic Nervous System/cytology , Transfection
5.
Assay Drug Dev Technol ; 10(6): 542-50, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22428804

ABSTRACT

Ca(V)2.2 (N-type) calcium channels are key regulators of neurotransmission. Evidence from knockout animals and localization studies suggest that Ca(V)2.2 channels play a critical role in nociceptive transmission. Additionally, ziconotide, a selective peptide inhibitor of Ca(V)2.2 channels, is clinically used to treat refractory pain. However, the use of ziconotide is limited by its low therapeutic index, which is believed, at least in part, to be a consequence of ziconotide inhibiting Ca(V)2.2 channels regardless of the channel state. Subsequent efforts have focused on the discovery of state-dependent inhibitors that preferentially bind to the inactivated state of Ca(V)2.2 channels in order to achieve an improved safety profile relative to ziconotide. Much less attention has been paid to understanding the binding kinetics of these state-dependent inhibitors. Here, we describe a novel electrophysiology-based assay on an automated patch platform designed to differentiate Ca(V)2.2 inhibitors based on their combined state dependence and kinetics. More specifically, this assay assesses inactivated state block, closed state block, and monitors the kinetics of recovery from block when channels move between states. Additionally, a use-dependent assay is described that uses a train of depolarizing pulses to drive channels to a similar level of inactivation for comparison. This use-dependent protocol also provides information on the kinetics of block development. Data are provided to show how these assays can be utilized to screen for kinetic diversity within and across chemical classes.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Electrophysiology/methods , Animals , Automation , Biological Assay , Cell Line , Data Interpretation, Statistical , Drug Evaluation, Preclinical , Indoles/pharmacology , Kinetics , Patch-Clamp Techniques , Pyrimidines/pharmacology , Rats , Structure-Activity Relationship , Triazines/pharmacology , Triazoles/pharmacology , omega-Conotoxins/pharmacology
6.
Heart Vessels ; 27(4): 419-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21989861

ABSTRACT

The L/N-type calcium channel blocker cilnidipine has been shown to suppress aldosterone production induced by angiotensin II (Ang II) in vitro. In addition, cilnidipine also suppresses the reflex tachycardia induced by its antihypertensive action in vivo. We investigated the effects of cilnidipine on the reflex aldosterone production induced by its antihypertensive action, to identify the differences in the effects of cilnidipine from those of the L-type calcium channel blocker nifedipine. Male SHR/Izm rats were anesthetized by intraperitoneal injection of pentobarbital sodium, and administered an intravenous infusion of saline supplemented or not with Ang II for 30 min. Blood pressure was monitored continuously in the femoral artery. Each of the calcium channel blockers under study was administered intravenously as a bolus through the femoral vein 1 min after the start of the Ang II infusion, and blood samples were collected 30 min after the start of the Ang II infusion. Following administration at nonhypotensive doses, all calcium channel blockers tended to decrease the plasma aldosterone. In particular, cilnidipine significantly suppressed the plasma aldosterone levels. On the other hand, under the condition of Ang II-induced hypertension, administration of a hypotensive dosage of cilnidipine showed no effect on the plasma aldosterone levels, whereas a hypotensive dosage of nifedipine significantly increased the plasma aldosterone levels. Our results suggest that the L/N-type calcium channel blocker cilnidipine reduces the plasma aldosterone level by suppressing the aldosterone production induced by reflex upregulation of the renin-angiotensin-aldosterone system associated with reduction of the blood pressure.


Subject(s)
Aldosterone/blood , Antihypertensive Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Channels, N-Type/drug effects , Dihydropyridines/pharmacology , Hypertension/drug therapy , Reflex/drug effects , Angiotensin II/administration & dosage , Animals , Antihypertensive Agents/administration & dosage , Blood Pressure/drug effects , Calcium Channel Blockers/administration & dosage , Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Dihydropyridines/administration & dosage , Disease Models, Animal , Down-Regulation , Hypertension/blood , Hypertension/physiopathology , Injections, Intravenous , Male , Nifedipine/pharmacology , Rats , Rats, Inbred SHR , Renin-Angiotensin System/drug effects
7.
Exp Physiol ; 95(10): 982-93, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20696781

ABSTRACT

Presynaptic imidazoline receptors (R(i-pre)) are found in the sympathetic axon terminals of animal and human cardiovascular systems, and they regulate blood pressure by modulating the release of peripheral noradrenaline (NA). The cellular mechanism of R(i-pre)-induced inhibition of NA release is unknown. We, therefore, investigated the effect of R(i-pre) activation on voltage-dependent Ca(2+) channels in rat superior cervical ganglion (SCG) neurons, using the conventional whole-cell patch-clamp method. Cirazoline (30 µM), an R(i-pre) agonist as well as an α-adrenoceptor (R(α)) agonist, decreased Ca(2+) currents (I(Ca)) by about 50% in a voltage-dependent manner with prepulse facilitation. In the presence of low-dose rauwolscine (3 µM), which blocks the α(2)-adrenoceptor (R(α2)), cirazoline still inhibited I(Ca) by about 30%, but prepulse facilitation was significantly attenuated. This inhibitory action of cirazoline was almost completely prevented by high-dose rauwolscine (30 µM), which blocks R(i-pre) as well as R(α2). In addition, pretreatment with LY320135 (10 µM), another R(i-pre) antagonist, in combination with low-dose rauwolscine (3 µM), also blocked the R(α2)-resistant effect of cirazoline. Addition of guanosine-5-O-(2-thiodiphosphate) (2 mm) to the internal solutions significantly attenuated the action of cirazoline. However, pertussis toxin (500 ng ml(1)) did not significantly influence the inhibitory effect of cirazoline. Moreover, cirazoline (30 µM) suppressed M current in SCG neurons cultured overnight. Finally, omega-conotoxin (omega-CgTx) GVIA (1 µM) obstructed cirazoline-induced current inhibition, and cirazoline (30 µM) significantly decreased the frequency of action potential firing in a partly reversible manner. This cirazoline-induced inhibition of action potential firing was almost completely occluded in the presence of omega-CgTx. Taken together, our results suggest that activation of R(i-pre) in SCG neurons reduced N-type I(Ca) in a pertussis toxin- and voltage-insensitive pathway, and this inhibition attenuated repetitive action potential firing in SCG neurons.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Calcium Signaling/drug effects , Imidazoles/pharmacology , Imidazoline Receptors/agonists , Neurons/drug effects , Presynaptic Terminals/drug effects , Receptors, Presynaptic/agonists , Superior Cervical Ganglion/drug effects , Action Potentials , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Benzofurans/pharmacology , Calcium Channels, N-Type/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/pharmacology , Imidazoline Receptors/metabolism , Kinetics , Male , Neurons/metabolism , Norepinephrine/metabolism , Patch-Clamp Techniques , Pertussis Toxin/pharmacology , Potassium Channels/drug effects , Potassium Channels/metabolism , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Presynaptic/metabolism , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/metabolism , Thionucleotides/pharmacology , Yohimbine/pharmacology , omega-Conotoxin GVIA/pharmacology
8.
Biol Pharm Bull ; 32(10): 1695-700, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19801830

ABSTRACT

Cilnidipine is a 1,4-dihydropyridine-derived voltage-dependent calcium channel (VDCC) blocker and suppresses N-type VDCC currents in addition to L-type VDCC currents. An earlier investigation has suggested that intrathecally injected cilnidipine produces antinociception by blocking N-type VDCCs in mice. The present study using the rat formalin model examined antinociceptive effects of intrathecally and orally administered cilnidipine to elucidate a putative site of antinociception of cilnidipine, assess the efficacy of oral cilnidipine for pain relief, and clarify the mechanism(s) responsible for the antinociceptive effect of oral cilnidipine. Cilnidipine (whether intrathecal or oral) suppressed nociception in phases 1 and 2 of the formalin model. In addition, the potency of oral cilnidipine to suppress formalin-induced nociception in phase 2 was greater than that of oral gabapentin, a clinically available drug for treatment of neuropathic pain. Cilnidipine elicited antinociceptive effects without neurological side-effects including serpentine-like tail movement, whole body shaking, and allodynia. Such side-effects can be induced by higher doses of intrathecal ziconotide, a clinically available N-type VDCC blocker. In contrast, orally administered nifedipine, an L-type VDCC blocker, had no effect on either phase of formalin-induced nociception. These results suggest that cilnidipine acts on the spinal cord to produce antinociception and is efficacious for pain relief after oral administration with better safety profile than that of ziconotide. Furthermore, the failure of orally administered nifedipine to affect formalin-induced nociception raises the possibility that oral cilnidipine produces antinociception through, at least in part, spinal N-type VDCC blockade.


Subject(s)
Analgesics/therapeutic use , Calcium Channel Blockers/therapeutic use , Calcium Channels, N-Type/drug effects , Dihydropyridines/therapeutic use , Pain/drug therapy , Spinal Cord/drug effects , Administration, Oral , Amines/pharmacology , Amines/therapeutic use , Analgesics/adverse effects , Analgesics/pharmacology , Animals , Calcium Channel Blockers/adverse effects , Calcium Channel Blockers/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Cyclohexanecarboxylic Acids/therapeutic use , Dihydropyridines/adverse effects , Dihydropyridines/pharmacology , Formaldehyde , Gabapentin , Male , Models, Animal , Nifedipine/pharmacology , Nifedipine/therapeutic use , Pain/chemically induced , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/pharmacology , gamma-Aminobutyric Acid/therapeutic use , omega-Conotoxins/adverse effects , omega-Conotoxins/therapeutic use
9.
Eur J Pharmacol ; 587(1-3): 42-7, 2008 Jun 10.
Article in English | MEDLINE | ID: mdl-18468596

ABSTRACT

N-type Ca2+ channels located on presynaptic nerve terminals regulate neurotransmitter release, including that from the spinal terminations of primary afferent nociceptors. Pharmacological and ion-channel gene knockdown approaches in animals have revealed N-type Ca2+ channels to be particularly attractive molecular targets for the discovery and development of new analgesic drugs. In recent years, some non-peptide small molecular N-type Ca2+ channel blockers have been reported. However, low selectivity and some side effects limit their further development. To overcome these disadvantages, some new compounds were designed and synthesized in our institute by optimizing the 4-amino-piperidine template. C101, one of these compounds, was demonstrated to block N-type Ca2+ channels with higher selectivity. It was found that C101 produced concentration-dependent inhibition on N-type Ca2+ channels expressed in Xenopus oocytes with an IC50 is 2.2+/-0.6 microM. The current-voltage relationship was not altered after 2-min exposure to C101. However, the steady-state inactivation relationship curve was shifted to more negative potentials for channels. Therefore, it seemed that C101 blocks the inactivated channel. C101 did not present any remarkable effects on voltage-gated potassium, sodium channels in cultured rat hippocampal neurons, and L-, P/Q-, R-type calcium channels and HERG channels expressed in Xenopus oocytes. The results suggested that C101 was a high selective blocker targeting N-type Ca2+ channels, and may have a potential to be developed as a novel analgesic agent.


Subject(s)
Calcium Channel Blockers , Calcium Channels, N-Type/drug effects , Piperidines/pharmacology , Animals , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Cells, Cultured , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Electrophysiology , Ether-A-Go-Go Potassium Channels/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , Humans , Oocytes/metabolism , Rats , Sodium Channels/drug effects , Xenopus
10.
J Physiol ; 580(Pt.3): 883-94, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17303639

ABSTRACT

N-type calcium channels contribute to the release of glutamate from primary afferent terminals synapsing onto nocisponsive neurons in the dorsal horn of the spinal cord, but little is known of functional adaptations to these channels in persistent pain states. Subtype-selective conotoxins and other drugs were used to determine the role of different calcium channel types in a rat model of inflammatory pain. Electrically evoked primary afferent synapses onto lumber dorsal horn neurons were examined three days after induction of inflammation with intraplantar complete Freund's adjuvant. The maximal inhibitory effect of the N-type calcium channel blockers, omega-conotoxins CVID and MVIIA, were attenuated in NK1 receptor-positive lamina I neurons after inflammation, but the potency of CVID was unchanged. This was associated with reduced inhibition of the frequency of asynchronous-evoked synaptic events by CVID studied in the presence of extracellular strontium, suggesting reduced N-type channel contribution to primary afferent synapses after inflammation. After application of CVID, the relative contributions of P/Q and L channels to primary afferent transmission and the residual current were unchanged by inflammation, suggesting the adaptation was specific to N-type channels. Blocking T-type channels did not affect synaptic amplitude under control or inflamed conditions. Reduction of N-type channel contribution to primary afferent transmission was selective for NK1 receptor-positive neurons identified by post hoc immunohistochemistry and did not occur at synapses in laminae II(o) or II(i), or inhibitory synapses. These results suggest that inflammation selectively downregulates N-type channels in the terminals of primary afferents synapsing onto (presumed) nociceptive lamina I NK1 receptor-positive neurons.


Subject(s)
Calcium Channels, N-Type/metabolism , Inflammation/physiopathology , Posterior Horn Cells/metabolism , Receptors, Neurokinin-1/metabolism , Synaptic Transmission , Afferent Pathways/physiopathology , Animals , Calcium Channels, N-Type/drug effects , Chronic Disease , Down-Regulation , Electric Stimulation , Evoked Potentials/drug effects , Female , Freund's Adjuvant , Hindlimb , Inflammation/chemically induced , Inflammation/metabolism , Male , Nociceptors , Pain/metabolism , Pain/physiopathology , Rats , Rats, Sprague-Dawley , Spinal Cord/physiopathology , Synapses , Venoms/pharmacology , omega-Conotoxins
11.
Bioorg Med Chem ; 15(2): 1091-105, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17074493

ABSTRACT

Virtual screening of the commercial databases was done by using a three dimensional pharmacophore previously developed for T-type calcium channel blockers using CATALYSTtrade mark program. Biological evaluation of 25 selected virtual hits resulted in the discovery of a highly potent compound VH04 with IC(50) value of 0.10 microM, eight times as potent as the known selective T-type calcium channel blocker, mibefradil. Search for similar compounds yielded several hits with micro-molar IC(50) values and high T-type calcium channel selectivity. Based on the structure of the virtual hits, small molecule libraries with novel scaffolds were designed, synthesis and biological evaluation of which are currently in progress. This result shows a successful example of ligand based drug discovery of potent T-type calcium channel blockers.


Subject(s)
Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/drug effects , Animals , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/genetics , Calcium Channels, T-Type/genetics , Catalysis , Cell Line , Computer Simulation , Databases, Factual , Electrophysiology , Humans , Mibefradil/pharmacology , Models, Molecular , Oocytes/metabolism , Patch-Clamp Techniques , RNA, Complementary/biosynthesis , RNA, Complementary/genetics , Xenopus
12.
J Neurophysiol ; 97(1): 5-14, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17021021

ABSTRACT

Previous studies have shown that corticosterone enhances whole cell calcium currents in CA1 pyramidal neurons, through a pathway involving binding of glucocorticoid receptor homodimers to the DNA. We examined whether glucocorticoids show selectivity for L- over N-type of calcium currents. Moreover, we addressed the putative gene targets that eventually lead to the enhanced calcium currents. Electrophysiological recordings were performed in nucleated patches that allow excellent voltage control. Calcium currents in these patches almost exclusively involve N- and L-type channels. We found that L- but not N-type calcium currents were largely enhanced after treatment with a high dose of corticosterone sufficient to activate glucocorticoid receptors. Voltage dependency and kinetic properties of the currents were unaffected by the hormone. Nonstationary noise analysis suggests that the increased current is not caused by a larger unitary conductance, but rather to a doubling of the number of functional channels. Quantitative real-time PCR revealed that transcripts of the Ca(v)1 subunits encoding for the N- or L-type calcium channels are not upregulated in the mouse CA1 area; instead, a strong, direct, and consistent upregulation of the beta4 subunit was observed. This indicates that the corticosteroid-induced increase in number of L-type calcium channels is not caused by a simple transcriptional regulation of the pore-forming subunit of the channels.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling/genetics , Glucocorticoids/metabolism , Hippocampus/metabolism , Animals , Calcium/metabolism , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/genetics , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , Glucocorticoids/pharmacology , Hippocampus/drug effects , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Protein Subunits/drug effects , Protein Subunits/genetics , Protein Subunits/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
13.
Bioorg Med Chem Lett ; 17(3): 662-7, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17134896

ABSTRACT

A series of new N-type (Ca(v)2.2) calcium channel blockers derived from the 'hit' structures 2-(3-bromo-4-fluorophenyl)-3-(2-pyridin-2-ylethyl)thiazolidin-4-one 9 and its 2-[4-(4-bromophenyl)pyridin-3-yl]-3-isobutyl analogue 10 is described. Extensive SAR studies using a range of synthetic approaches resulted in novel, patented compounds with IC50 values of up to 0.2 microM in an in vitro IMR32 assay, and selectivities for N/L of up to 30-fold. The new compounds described have potential in treatment of neuropathic pain.


Subject(s)
Analgesics/chemical synthesis , Analgesics/pharmacology , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Thiazolidinediones/chemical synthesis , Thiazolidinediones/pharmacology , Calcium Channels, L-Type/drug effects , Cell Line, Tumor , Chromatography, High Pressure Liquid , Drug Evaluation, Preclinical , Humans , Indicators and Reagents , Spectrophotometry, Ultraviolet , Structure-Activity Relationship
14.
J Biol Chem ; 281(29): 20011-7, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16704976

ABSTRACT

Voltage-gated calcium channels (Ca(v)) 2.2 currents are potentiated by phorbol-12-myristate, 13-acetate (PMA), whereas Ca(v) 2.3 currents are increased by both PMA and acetyl-beta-methylcholine (MCh). MCh-selective sites were identified in the alpha(1) 2.3 subunit, whereas the identified PMA sites responded to both PMA and MCh (Kamatchi, G. L., Franke, R., Lynch, C., III, and Sando, J. J. (2004) J. Biol. Chem. 279, 4102-4109; Fang, H., Franke, R., Patanavanich, S., Lalvani, A., Powell, N. K., Sando, J. J., and Kamatchi, G. L. (2005) J. Biol. Chem. 280, 23559-23565). The hypothesis that PMA sites in the alpha(1) 2.2 subunit are homologous to the PMA-responsive sites in alpha(1) 2.3 subunit was tested with Ser/Thr --> Ala mutations in the alpha(1) 2.2 subunit. WT alpha(1) 2.2 or mutants were expressed in Xenopus oocytes in combination with beta1b and alpha2/delta subunits. Inward current (I(Ba)) was recorded using Ba(2+) as the charge carrier. T422A, S1757A, S2108A, or S2132A decreased the PMA response. In contrast, S425A increased the response to PMA, and thus, it was considered an inhibitory site. Replacement of each of the identified stimulatory Ser/Thr sites with Asp increased the basal current and decreased the PMA-induced enhancement, consistent with regulation by phosphorylation at these sites. Multiple mutant combinations showed (i) greater inhibition than that caused by the single Ala mutations; (ii) that enhancement observed when Thr-422 and Ser-2108 are available may be inhibited by the presence of Ser-425; and (iii) that the combination of Thr-422, Ser-2108, and either Ser-1757 or Ser-2132 can provide a greater response to PMA when Ser-425 is replaced with Ala. The homologous sites in alpha(1) 2.2 and alpha(1) 2.3 subunits seem to be functionally different. The existence of an inhibitory phosphorylation site in the I-II linker seems to be unique to the alpha(1) 2.2 subunit.


Subject(s)
Calcium Channels, N-Type/physiology , Serine , Tetradecanoylphorbol Acetate/pharmacology , Alternative Splicing , Amino Acid Substitution , Animals , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/genetics , DNA, Complementary/genetics , Female , Genetic Variation , Kinetics , Methacholine Chloride/pharmacology , Oocytes/physiology , Phosphorylation , Protein Kinase C/metabolism , Protein Subunits/genetics , Protein Subunits/physiology , Rats , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Superior Cervical Ganglion/physiology , Xenopus laevis
15.
Bioorg Med Chem ; 14(15): 5333-9, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16616501

ABSTRACT

Antiallergic drug cyproheptadine (Cyp) is known to have inhibitory activities for L-type calcium channels in addition to histamine and serotonin receptors. Since we found that Cyp had an inhibitory activity against N-type calcium channel, Cyp was optimized to obtain more selective N-type calcium channel blocker with analgesic action. As a consequence of the optimization, we found 13 with potent N-type calcium channel inhibitory activity which had lower inhibitory activities against L-type calcium channel, histamine (H1), and serotonin (5-HT2A) receptors than those of Cyp. 13 showed an oral analgesic activity in rat formalin-induced pain model.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Cyproheptadine/analogs & derivatives , Cyproheptadine/pharmacology , Drug Design , Administration, Oral , Animals , Behavior, Animal/drug effects , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/chemistry , Cell Line, Tumor , Cyproheptadine/chemistry , Drug Evaluation, Preclinical , Formaldehyde/chemistry , Guinea Pigs , Humans , In Vitro Techniques , Male , Molecular Structure , Pain/chemically induced , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Histamine/drug effects , Receptors, Serotonin/drug effects , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
16.
J Pharmacol Sci ; 100(4): 243-61, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16565579

ABSTRACT

Benidipine is a dihydropyridine-derived calcium channel blocker developed in Japan, with several unique mechanisms of action, that is, triple calcium channels (L, N, and T) blocking action with a membrane approach. Benidipine has relatively high vascular selectivity and is expected to show protective effects on vascular endothelial cells. Renal protective effects of benidipine also have been shown in several basic and clinical studies. Moreover, anti-oxidative action and enhancing nitric oxide production have been noted with this drug, following its cardio-protective effects in patients with ischemic heart diseases. In fact, benidipine exerted a better prognostic effect than other calcium channel blockers in the therapy for patients with vasospastic angina. In addition, benidipine showed reliable antihypertensive, renoprotective effects if used in combination with angiotensin II type 1 receptor blockers (ARBs) when adequate anti-hypertensive effects are not achieved by ARBs alone, indicating that benidipine is an useful calcium channel blocker in combination therapy for hypertension. Benidipine was launched on the Japanese market 14 years ago, but few severe side effects have been reported, suggesting that this is a drug with established safety and long-acting pharmacological effects.


Subject(s)
Blood Pressure/drug effects , Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Vasodilator Agents/pharmacology , Angina Pectoris/drug therapy , Animals , Calcium Channel Blockers/pharmacokinetics , Calcium Channel Blockers/therapeutic use , Calcium Channels, L-Type/drug effects , Calcium Channels, N-Type/drug effects , Calcium Channels, T-Type/drug effects , Clinical Trials as Topic , Diabetic Nephropathies/drug therapy , Dihydropyridines/pharmacokinetics , Dihydropyridines/therapeutic use , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Heart/drug effects , Humans , Hypertension/drug therapy , Kidney/blood supply , Kidney/drug effects , Renal Circulation , Vasodilator Agents/pharmacokinetics , Vasodilator Agents/therapeutic use
17.
Glia ; 49(2): 259-74, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15472989

ABSTRACT

Muller cells mediate retinal function by stabilizing the ionic environment and signal glial network activity via calcium waves. Using whole-cell patch clamp recording, we describe a high-voltage-activated, slowly inactivating Ca channel current in isolated salamander Muller cells that has unusual pharmacological properties. The Ca channel current has an activation midpoint of approximately -8 mV and an inactivation midpoint of approximately -26 mV in 10 mM Ba2+. The time constant for inactivation is approximately 380 ms at potentials positive to zero. The current is blocked by Cd2+ with an EC50 of <100 nM. nisoldipine (10 microM) blocks approximately 50%, while nifedipine (1 microM), diltiazem (20 microM), and verapamil (50 microM) each block one-third of the current. In contrast to its typical actions, BayK 8644 blocks the current by approximately 25%. Blockers of other Ca channel subtypes were also tested: omega-agatoxin IVA (200 nM) blocked only 13% of the Ca channel current, while omega-conotoxin GVIA (1 microM) blocked 84% of the current. Immnohistochemistry supported the presence of alpha1A, alpha1B, alpha1C, and alpha1D Ca channel subunits. Mapping of dihydropyridine-binding sites with DM-BODIPY revealed a distribution of channels over the entire membrane of the Muller cell with a higher density at the apical region. Overall, these observations suggest either the presence of a mix of L- and N-type Ca channels or a single, unconventional HVA Ca channel subtype sharing L- and N-type Ca channel characteristics.


Subject(s)
Calcium Channels/physiology , Neuroglia/physiology , Retina/physiology , Ambystoma , Animals , Binding Sites/drug effects , Binding Sites/physiology , Boron Compounds , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/physiology , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/physiology , Cell Membrane/metabolism , In Vitro Techniques , Larva , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neuroglia/drug effects , Neurotoxins/pharmacology , Patch-Clamp Techniques , Protein Subunits/drug effects , Protein Subunits/metabolism , Receptor Aggregation/physiology , Retina/drug effects
18.
Neurochem Int ; 45(6): 885-93, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15312983

ABSTRACT

The unconventional gaseous transmitter nitric oxide (NO) markedly influences most of mechanisms involved in the regulation of intracellular Ca2+ homeostasis. In excitable cells, Ca2+ signaling mainly depends on the activity of voltage-gated Ca2+ channels (VGCCs). In the present paper, we will review data from our laboratory and others characterizing NO-induced modulation of Ca(v)1 (L-type) and Ca(v)2.2 (N-type) channels. In particular, we will explore experimental evidence indicating that NO's inhibition of channel gating is produced via cGMP-dependent protein kinase and examine some of the numerous cell functions that are potentially influenced by the action of NO on Ca2+ channels.


Subject(s)
Calcium Channels, L-Type/drug effects , Calcium Channels, N-Type/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Nitric Oxide/physiology , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Humans
19.
J Pharmacol Exp Ther ; 305(2): 460-6, 2003 May.
Article in English | MEDLINE | ID: mdl-12606627

ABSTRACT

We used fluorescence resonance energy transfer imaging of enhanced cyan fluorescent protein (CFP)-tagged and enhanced yellow fluorescent protein (YFP)-tagged protein pairs to examine the hypothesis that G protein gamma subunit-like (GGL) domain-containing regulators of G protein signaling (RGS) can directly bind to the Gbeta5 subunit of heterotrimeric G proteins in vivo. We observed that Gbeta5 could interact with Ggamma2 and Ggamma13, after their expression in human embryonic kidney 293 cells. Interestingly, although untagged Ggamma3 did not interact with Gbeta5, CFP-tagged Ggamma3 strongly interacted with YFP-tagged Gbeta5 in FRET studies. Moreover, CFP-Ggamma3 supported Ca(2+) channel inhibition when paired with Gbeta5 or YFP-Gbeta5, indicating a "gain of function" for CFP-Ggamma3. Gbeta5 could also interact with RGS11 and its N-terminal, but not its C-terminal domain. On the other hand, RGS11 did not interact with Gbeta1. These studies demonstrate that the GGL domain-containing N terminus of RGS 11 can directly interact with Gbeta5 in vivo and supports the hypothesis that this interaction may contribute to the specificity of Gbeta5 interactions with cellular effector molecules.


Subject(s)
GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , Heterotrimeric GTP-Binding Proteins/chemistry , RGS Proteins/chemistry , RGS Proteins/genetics , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/drug effects , Cells, Cultured , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Electrophysiology , Energy Transfer , Fluorescent Dyes , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/pharmacology , Humans , Patch-Clamp Techniques , RGS Proteins/pharmacology , Transfection
20.
Bioorg Med Chem Lett ; 12(17): 2267-9, 2002 Sep 02.
Article in English | MEDLINE | ID: mdl-12161113

ABSTRACT

This study was performed to determine the structure-activity relationships (SAR) of L-cysteine based N-type calcium channel blockers. Basic nitrogen was introduced into the C-terminal lipophilic moiety of L-cysteine with a view toward improvement of its physicochemical properties. L-Cysteine derivative 9 was found to be a potent and selective N-type calcium channel blocker with IC(50) of 0.33 microM in calcium influx assay using IMR-32 cells and was 15-fold selective for N-type calcium channels over L-type channels. Compound 9 showed improved oral analgesic efficacy in the rat formalin induced pain model and the rat chronic constriction injury (CCI) model, which is one of the most reliable models of chronic neuropathic pain, without any significant effect on blood pressure or neurological behavior.


Subject(s)
Calcium Channel Blockers/chemistry , Calcium Channels, N-Type/drug effects , Cysteine/analogs & derivatives , Pain/drug therapy , Administration, Oral , Analgesics/administration & dosage , Analgesics/chemistry , Analgesics/pharmacology , Animals , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/pharmacology , Constriction, Pathologic , Drug Evaluation, Preclinical , Hydrophobic and Hydrophilic Interactions , Inhibitory Concentration 50 , Pain/chemically induced , Rats , Structure-Activity Relationship , Therapeutic Equivalency
SELECTION OF CITATIONS
SEARCH DETAIL