Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Pharmacol ; 886: 173542, 2020 Nov 05.
Article in English | MEDLINE | ID: mdl-32910945

ABSTRACT

Cannabidiol (CBD) is a non-psychoactive component of Cannabis which has recently received regulatory consideration for the treatment of intractable forms of epilepsy such as the Dravet and the Lennox-Gastaut syndromes. The mechanisms of the antiepileptic effects of CBD are unclear, but several pre-clinical studies suggest the involvement of ion channels. Therefore, we have evaluated the effects of CBD on seven major cardiac currents shaping the human ventricular action potential and on Purkinje fibers isolated from rabbit hearts to assess the in vitro cardiac safety profile of CBD. We found that CBD inhibits with comparable micromolar potencies the peak and late components of the NaV1.5 sodium current, the CaV1.2 mediated L-type calcium current, as well as all the repolarizing potassium currents examined except Kir2.1. The most sensitive channels were KV7.1 and the least sensitive were KV11.1 (hERG), which underly the slow (IKs) and rapid (IKr) components, respectively, of the cardiac delayed-rectifier current. In the Purkinje fibers, CBD decreased the action potential (AP) duration more potently at half-maximal than at near complete repolarization, and slightly decreased the AP amplitude and its maximal upstroke velocity. CBD had no significant effects on the membrane resting potential except at the highest concentration tested under fast pacing rate. These data show that CBD impacts cardiac electrophysiology and suggest that caution should be exercised when prescribing CBD to carriers of cardiac channelopathies or in conjunction with other drugs known to affect heart rhythm or contractility.


Subject(s)
Action Potentials/drug effects , Cannabidiol/pharmacology , Heart Ventricles/drug effects , Heart/drug effects , Ion Channels/drug effects , Animals , Cannabidiol/toxicity , Channelopathies/complications , Humans , In Vitro Techniques , KCNQ1 Potassium Channel/drug effects , Membrane Potentials/drug effects , Myocytes, Cardiac/drug effects , NAV1.5 Voltage-Gated Sodium Channel/drug effects , Patch-Clamp Techniques , Purkinje Fibers/drug effects , Rabbits
3.
Sci Rep ; 10(1): 5627, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32221320

ABSTRACT

Automated patch clamp (APC) instruments enable efficient evaluation of electrophysiologic effects of drugs on human cardiac currents in heterologous expression systems. Differences in experimental protocols, instruments, and dissimilar site procedures affect the variability of IC50 values characterizing drug block potency. This impacts the utility of APC platforms for assessing a drug's cardiac safety margin. We determined variability of APC data from multiple sites that measured blocking potency of 12 blinded drugs (with different levels of proarrhythmic risk) against four human cardiac currents (hERG [IKr], hCav1.2 [L-Type ICa], peak hNav1.5, [Peak INa], late hNav1.5 [Late INa]) with recommended protocols (to minimize variance) using five APC platforms across 17 sites. IC50 variability (25/75 percentiles) differed for drugs and currents (e.g., 10.4-fold for dofetilide block of hERG current and 4-fold for mexiletine block of hNav1.5 current). Within-platform variance predominated for 4 of 12 hERG blocking drugs and 4 of 6 hNav1.5 blocking drugs. hERG and hNav1.5 block. Bland-Altman plots depicted varying agreement across APC platforms. A follow-up survey suggested multiple sources of experimental variability that could be further minimized by stricter adherence to standard protocols. Adoption of best practices would ensure less variable APC datasets and improved safety margins and proarrhythmic risk assessments.

4.
J Pharmacol Exp Ther ; 312(2): 619-26, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15356217

ABSTRACT

Heterologous expression and lesioning studies were conducted to identify possible subunit assembly partners in nicotinic acetylcholine receptors (nAChR) containing alpha6 subunits (alpha6(*) nAChR). SH-EP1 human epithelial cells were transfected with the requisite subunits to achieve stable expression of human alpha6beta2, alpha6beta4, alpha6beta2beta3, alpha6beta4beta3, or alpha6beta4beta3alpha5 nAChR. Cells expressing subunits needed to form alpha6beta4beta3alpha5 nAChR exhibited saturable [(3)H]epibatidine binding (K(d) = 95.9 +/- 8.3 pM and B(max) = 84.5 +/- 1.6 fmol/mg of protein). The rank order of binding competition potency (K(i)) for prototypical nicotinic compounds was alpha-conotoxin MII (6 nM) > nicotine (156 nM) approximately methyllycaconitine (200 nM) > alpha-bungarotoxin (>10 microM), similar to that for nAChR in dopamine neurons displaying a distinctive pharmacology. 6-Hydroxydopamine lesioning studies indicated that beta3 and alpha5 subunits are likely partners of the alpha6 subunits in nAChR expressed in dopaminergic cell bodies. Similar to findings in rodents, quantitative real-time reverse transcription-polymerase chain reactions of human brain indicated that alpha6 subunit mRNA expression was 13-fold higher in the substantia nigra than in the cortex or the rest of the brain. Thus, heterologous expression studies suggest that the human alpha5 subunit makes a critical contribution to alpha6beta4beta3alpha5 nAChR assembly into a ligand-binding form with native alpha6(*)-nAChR-like pharmacology and of potential physiological and pathophysiological relevance.


Subject(s)
Receptors, Nicotinic/biosynthesis , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Animals , Binding, Competitive/drug effects , Cell Line , Cell Membrane/metabolism , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Male , RNA/biosynthesis , RNA/isolation & purification , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Structure-Activity Relationship , Substantia Nigra/drug effects , Substantia Nigra/metabolism
5.
Hum Mol Genet ; 12(18): 2277-91, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12915482

ABSTRACT

Mutations of the parkin gene are the most frequent cause of early onset autosomal recessive parkinsonism (EO-AR). Here we show that inactivation of the parkin gene in mice results in motor and cognitive deficits, inhibition of amphetamine-induced dopamine release and inhibition of glutamate neurotransmission. The levels of dopamine are increased in the limbic brain areas of parkin mutant mice and there is a shift towards increased metabolism of dopamine by MAO. Although there was no evidence for a reduction of nigrostriatal dopamine neurons in the parkin mutant mice, the level of dopamine transporter protein was reduced in these animals, suggesting a decreased density of dopamine terminals, or adaptative changes in the nigrostriatal dopamine system. GSH levels were increased in the striatum and fetal mesencephalic neurons from parkin mutant mice, suggesting that a compensatory mechanism may protect dopamine neurons from neuronal death. These parkin mutant mice provide a valuable tool to better understand the preclinical deficits observed in patients with PD and to characterize the mechanisms leading to the degeneration of dopamine neurons that could provide new strategies for neuroprotection.


Subject(s)
Behavior, Animal/drug effects , Dopamine/metabolism , Gene Silencing , Neurotransmitter Uptake Inhibitors/metabolism , Ubiquitin-Protein Ligases/genetics , Alleles , Animals , Base Sequence , Body Temperature/genetics , Body Weight/genetics , Catecholamines/antagonists & inhibitors , Cells, Cultured , Dopamine/pharmacokinetics , Enzyme Inhibitors/pharmacology , Exons , Female , Homozygote , Introns , Male , Mice , Mice, Transgenic , Monoamine Oxidase/metabolism , Neurons/drug effects , Neurons/metabolism , Sequence Deletion , Ubiquitin-Protein Ligases/metabolism , alpha-Methyltyrosine/pharmacology
6.
Exp Neurol ; 176(1): 247-53, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12093102

ABSTRACT

The effects of a chronic treatment with the anti-glutamate and sodium channel modulating neuroprotective agent riluzole on the degeneration of dopamine-containing neurons were studied in the brain of weaver mutant mice. In these animals, as in Parkinson's disease, dopaminergic neurons of the nigro-striatal pathway undergo spontaneous and progressive cell death. Homozygous weaver mice were orally treated twice a day with either 8 mg/kg riluzole or placebo for 2 months. Quantification of tyrosine-hydroxylase and dopamine-transporter axonal immunostaining in the striatum revealed that riluzole significantly increased the density of striatal dopaminergic nerve terminals. These results suggest that riluzole protects dopaminergic processes in the weaver mice and/or promotes their neuroplasticity.


Subject(s)
Corpus Striatum/drug effects , Membrane Glycoproteins , Nerve Tissue Proteins , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Potassium Channels, Inwardly Rectifying , Riluzole/pharmacology , Substantia Nigra/drug effects , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins , Drug Administration Schedule , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Homozygote , Membrane Transport Proteins/biosynthesis , Mice , Mice, Neurologic Mutants , Neurons/metabolism , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Potassium Channels/genetics , Presynaptic Terminals/drug effects , Presynaptic Terminals/pathology , Substantia Nigra/metabolism , Substantia Nigra/pathology , Time , Tyrosine 3-Monooxygenase/biosynthesis
7.
Bioorg Med Chem ; 10(5): 1627-37, 2002 May.
Article in English | MEDLINE | ID: mdl-11886824

ABSTRACT

Excessive release of glutamate, a potent excitatory neurotransmitter, is thought to play an important role in a variety of acute and chronic neurological disorders, suggesting that excitatory amino acid antagonists may have broad therapeutic potential in neurology. Here, we describe the synthesis, pharmacological properties and neuroprotective activity of 9-carboxymethyl-imidazo-[1-2a]indeno[1-2e]pyrazin-4-one-2-carboxylic acid (RPR117824), an original selective AMPA antagonist. RPR117824 can be obtained through a six-step synthesis starting from (1-oxo-indan-4-yl) acetic acid, which has been validated on a gram-scale with an overall yield of 25%. Monosodium or disodium salts of the compound exhibit excellent solubility in saline (> or = 10 g/L), enabling intravenous administration. RPR117824 displays nanomolar affinity (IC(50)=18 nM) for AMPA receptors and competitive inhibition of electrophysiological responses mediated by AMPA receptors heterologously expressed in Xenopus oocytes (K(B)=5 nM) and native receptors in rat brain slices (IC(50)=0.36 microM). In in vivo testing, RPR117824 behaves as a powerful blocker of convulsions induced in mice or rats by supramaximal electroshock or chemoconvulsive agents such as pentylenetetrazole, bicuculline, isoniazide, strychnine, 4-aminopyridine and harmaline with half maximal effective doses ranging from 1.5 to 10 mg/kg following subcutaneous or intraperitoneal administration. In disease models in rats and gerbils, RPR117824 possesses significant neuroprotective activity in global and focal cerebral ischemia, and brain and spinal cord trauma.


Subject(s)
Anticonvulsants/chemical synthesis , Imidazoles/pharmacology , Neuroprotective Agents/chemical synthesis , Pyrazines/pharmacology , Receptors, AMPA/antagonists & inhibitors , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Brain Injuries/drug therapy , Brain Ischemia/drug therapy , Disease Models, Animal , Gerbillinae , Imidazoles/chemical synthesis , Imidazoles/chemistry , Male , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Oocytes , Protein Binding , Pyrazines/chemical synthesis , Pyrazines/chemistry , Radioligand Assay , Rats , Reperfusion Injury/drug therapy , Seizures/drug therapy , Spinal Cord Compression/drug therapy , Synapses/drug effects , Xenopus
8.
Eur J Neurosci ; 4(5): 420-424, 1992.
Article in English | MEDLINE | ID: mdl-12106350

ABSTRACT

Nitric oxide production in the cerebellum and induction of long-term potentiation (LTP) in the hippocampus have some characteristics in common: both phenomena are induced by activation of N-methyl-d-aspartate receptors and both are highly dependent on calcium-mediated processes. Here we provide evidence that endogenous nitric oxide production is necessary for synaptic plasticity in the CA1 hippocampus of the rat. LTP recorded in slices was blocked in a concentration-dependent manner by the nitric oxide synthase inhibitors l-NG-nitroarginine and l-NG-nitroarginine methyl ester, but l-NG-monomethylarginine was only marginally active. Bathing the slices with haemoglobin, a protein that scavenges nitric oxide, also resulted in a concentration-dependent blockade of LTP. Nitric oxide released locally from hydroxylamine produced a stable potentiation of synaptic transmission that was not additive with LTP induced by high-frequency stimulation. These results are fully consistent with the presumed retrograde messenger role of nitric oxide in LTP.

SELECTION OF CITATIONS
SEARCH DETAIL
...