Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Mol Neurobiol ; 61(4): 2430-2445, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37889366

ABSTRACT

The ability of monkeys and rats to carry out spatial working memory tasks has been shown to depend on the persistent firing of pyramidal cells in the prefrontal cortex (PFC), arising from recurrent excitatory connections on dendritic spines. These spines express hyperpolarization-activated cyclic nucleotide-gated (HCN) channels whose open state is increased by cAMP signaling, and which markedly alter PFC network connectivity and neuronal firing. In traditional neural circuits, activation of these non-selective cation channels leads to neuronal depolarization and increased firing rate. Paradoxically, cAMP activation of HCN channels in PFC pyramidal cells reduces working memory-related neuronal firing. This suggests that activation of HCN channels may hyperpolarize rather than depolarize these neurons. The current study tested the hypothesis that Na+ influx through HCN channels activates Slack Na+-activated K+ (KNa) channels to hyperpolarize the membrane. We have found that HCN and Slack KNa channels co-immunoprecipitate in cortical extracts and that, by immunoelectron microscopy, they colocalize at postsynaptic spines of PFC pyramidal neurons. A specific blocker of HCN channels, ZD7288, reduces KNa current in pyramidal cells that express both HCN and Slack channels, but has no effect on KNa currents in an HEK cell line expressing Slack without HCN channels, indicating that blockade of HCN channels in neurons reduces K+ current indirectly by lowering Na+ influx. Activation of HCN channels by cAMP in a cell line expressing a Ca2+ reporter results in elevation of cytoplasmic Ca2+, but the effect of cAMP is reversed if the HCN channels are co-expressed with Slack channels. Finally, we used a novel pharmacological blocker of Slack channels to show that inhibition of Slack in rat PFC improves working memory performance, an effect previously demonstrated for blockers of HCN channels. Our results suggest that the regulation of working memory by HCN channels in PFC pyramidal neurons is mediated by an HCN-Slack channel complex that links activation HCN channels to suppression of neuronal excitability.


Subject(s)
Memory, Short-Term , Pyramidal Cells , Animals , Rats , Cyclic Nucleotide-Gated Cation Channels , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Memory, Short-Term/physiology , Neurons/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism
2.
Res Sq ; 2023 May 04.
Article in English | MEDLINE | ID: mdl-37205397

ABSTRACT

The ability of monkeys and rats to carry out spatial working memory tasks has been shown to depend on the persistent firing of pyramidal cells in the prefrontal cortex (PFC), arising from recurrent excitatory connections on dendritic spines. These spines express hyperpolarization-activated cyclic nucleotide-gated (HCN) channels whose open state is increased by cAMP signaling, and which markedly alter PFC network connectivity and neuronal firing. In traditional neural circuits, activation of these non-selective cation channels leads to neuronal depolarization and increased firing rate. Paradoxically, cAMP activation of HCN channels in PFC pyramidal cells reduces working memory-related neuronal firing. This suggests that activation of HCN channels may hyperpolarize rather than depolarize these neurons. The current study tested the hypothesis that Na+ influx through HCN channels activates Slack Na+-activated K+ (KNa) channels to hyperpolarize the membrane. We have found that HCN and Slack KNa channels coimmunoprecipitate in cortical extracts and that, by immunoelectron microscopy, they colocalize at postsynaptic spines of PFC pyramidal neurons. A specific blocker of HCN channels, ZD7288, reduces KNa current in pyramidal cells that express both HCN and Slack channels, but has no effect on KNa currents in an HEK cell line expressing Slack without HCN channels, indicating that blockade of HCN channels in neurons reduces K+ +current indirectly by lowering Na+ influx. Activation of HCN channels by cAMP in a cell line expressing a Ca2+ reporter results in elevation of cytoplasmic Ca2+, but the effect of cAMP is reversed if the HCN channels are co-expressed with Slack channels. Finally, we used a novel pharmacological blocker of Slack channels to show that inhibition of Slack in rat PFC improves working memory performance, an effect previously demonstrated for blockers of HCN channels. Our results suggest that the regulation of working memory by HCN channels in PFC pyramidal neurons is mediated by an HCN-Slack channel complex that links activation HCN channels to suppression of neuronal excitability.

3.
Adv Neurobiol ; 30: 255-285, 2023.
Article in English | MEDLINE | ID: mdl-36928854

ABSTRACT

CNS diseases, including psychiatric disorders, represent a significant opportunity for the discovery and development of new drugs and therapeutic treatments with the potential to have a significant impact on human health. CNS diseases, however, present particular challenges to therapeutic discovery efforts, and psychiatric diseases/disorders may be among the most difficult. With specific exceptions such as psychostimulants for ADHD, a large number of psychiatric patients are resistant to existing treatments. In addition, clinicians have no way of knowing which psychiatric patients will respond to which drugs. By definition, psychiatric diagnoses are syndromal in nature; determinations of efficacy are often self-reported, and drug discovery is largely model-based. While such models of psychiatric disease are amenable to screening for new drugs, whether cellular or whole-animal based, they have only modest face validity and, more importantly, predictive validity. Multiple academic, pharmaceutical industry, and government agencies are dedicated to the translation of new findings about the neurobiology of major psychiatric disorders into the discovery and advancement of novel therapies. The collaboration of these agencies provide a pathway for developing new therapeutics. These efforts will be greatly helped by recent advances in understanding the genetic bases of psychiatric disorders, the ongoing search for diagnostic and therapy-responsive biomarkers, and the validation of new animal models.


Subject(s)
Mental Disorders , Animals , Humans , Mental Disorders/drug therapy , Biomarkers
4.
Biochem Pharmacol ; 208: 115413, 2023 02.
Article in English | MEDLINE | ID: mdl-36646291

ABSTRACT

A number of mutations to members of several CNS potassium (K) channel families have been identified which result in rare forms of neonatal onset epilepsy, or syndromes of which one prominent characteristic is a form of epilepsy. Benign Familial Neonatal Convulsions or Seizures (BFNC or BFNS), also referred to as Self-Limited Familial Neonatal Epilepsy (SeLNE), results from mutations in 2 members of the KV7 family (KCNQ) of K channels; while generally self-resolving by about 15 weeks of age, these mutations significantly increase the probability of generalized seizure disorders in the adult, in some cases they result in more severe developmental syndromes. Epilepsy of Infancy with Migrating Focal Seizures (EIMSF), or Migrating Partial Seizures of Infancy (MMPSI), is a rare severe form of epilepsy linked primarily to gain of function mutations in a member of the sodium-dependent K channel family, KCNT1 or SLACK. Finally, KCNMA1 channelopathies, including Liang-Wang syndrome (LIWAS), are rare combinations of neurological symptoms including seizure, movement abnormalities, delayed development and intellectual disabilities, with Liang-Wang syndrome an extremely serious polymalformative syndrome with a number of neurological sequelae including epilepsy. These are caused by mutations in the pore-forming subunit of the large-conductance calcium-activated K channel (BK channel) KCNMA1. The identification of these rare but significant channelopathies has resulted in a resurgence of interest in their treatment by direct pharmacological or genetic modulation. We will briefly review the genetics, biophysics and pharmacology of these K channels, their linkage with the 3 syndromes described above, and efforts to more effectively target these syndromes.


Subject(s)
Channelopathies , Epilepsy, Benign Neonatal , Epilepsy , Infant, Newborn , Adult , Humans , Channelopathies/genetics , Channelopathies/therapy , Syndrome , Epilepsy/drug therapy , Epilepsy/genetics , Epilepsy, Benign Neonatal/genetics , Mutation , Calcium Channels/genetics , Potassium Channels, Sodium-Activated/genetics , Nerve Tissue Proteins/metabolism
5.
ACS Omega ; 7(43): 39097-39106, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36340178

ABSTRACT

Monitoring the anti-epileptic drug carbamazepine (CBZ) is crucial for proper dosing, optimizing a patient's clinical outcome, and managing their medication regimen. Due to its narrow therapeutic window and concentration-related toxicity, CBZ is prescribed and monitored in a highly personalized manner. We report an electrochemical conformation-changing aptasensor with two assay formats: a 30 min assay for routine monitoring and a 5 min assay for rapid emergency testing. To enable "sample-to-answer" testing, a de novo CBZ aptamer (K d < 12 nM) with conformational switching due to a G-quadruplex motif was labeled with methylene blue and immobilized on a gold electrode. The electrode fabrication and detection conditions were optimized using electrochemical techniques and visualized by atomic force microscopy (AFM). The aptasensor performance, including reproducibility, stability, and interference, was characterized using electrochemical impedance spectroscopy and voltammetry techniques. The aptasensor exhibited a wide dynamic range in buffer (10 nM to 100 µM) with limits of detection of 1.25 and 1.82 nM for the 5 and 30 min assays, respectively. The clinical applicability is demonstrated by detecting CBZ in finger prick blood samples (<50 µL). The proposed assays provide a promising method to enable point-of-care monitoring for timely personalized CBZ dosing.

6.
Adv Pharmacol ; 94: 213-254, 2022.
Article in English | MEDLINE | ID: mdl-35659373

ABSTRACT

Glucagon-like peptide-1 (GLP-1) receptor agonists are extensively used in type 2 diabetic patients for the effective control of hyperglycemia. It is now clear from outcomes trials that this class of drugs offers important additional benefits to these patients due to reducing the risk of developing major adverse cardiac events (MACE). This risk reduction is, in part, due to effective glycemic control in patients; however, the various outcomes trials, further validated by subsequent meta-analysis of the outcomes trials, suggest that the risk reduction in MACE is also dependent on glycemic-independent mechanisms operant in cardiovascular tissues. These glycemic-independent mechanisms are likely mediated by GLP-1 receptors found throughout the cardiovascular system and by the complex signaling cascades triggered by the binding of agonists to the G-protein coupled receptors. This heterogeneity of signaling pathways underlying different downstream effects of GLP-1 agonists, and the discovery of biased agonists favoring specific signaling pathways, may have import in the future treatment of MACE in these patients. We review the evidence supporting the glycemic-independent evidence for risk reduction of MACE by the GLP-1 receptor agonists and highlight the putative mechanisms underlying these benefits. We also comment on the different signaling pathways which appear important for mediating these effects.


Subject(s)
Cardiovascular Diseases , Cardiovascular System , Diabetes Mellitus, Type 2 , Cardiovascular Diseases/drug therapy , Cardiovascular System/metabolism , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/therapeutic use , Glucagon-Like Peptide-1 Receptor/agonists , Humans , Hypoglycemic Agents/therapeutic use
7.
Cell ; 182(5): 1170-1185.e9, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32795412

ABSTRACT

Loss of the gene (Fmr1) encoding Fragile X mental retardation protein (FMRP) causes increased mRNA translation and aberrant synaptic development. We find neurons of the Fmr1-/y mouse have a mitochondrial inner membrane leak contributing to a "leak metabolism." In human Fragile X syndrome (FXS) fibroblasts and in Fmr1-/y mouse neurons, closure of the ATP synthase leak channel by mild depletion of its c-subunit or pharmacological inhibition normalizes stimulus-induced and constitutive mRNA translation rate, decreases lactate and key glycolytic and tricarboxylic acid (TCA) cycle enzyme levels, and triggers synapse maturation. FMRP regulates leak closure in wild-type (WT), but not FX synapses, by stimulus-dependent ATP synthase ß subunit translation; this increases the ratio of ATP synthase enzyme to its c-subunit, enhancing ATP production efficiency and synaptic growth. In contrast, in FXS, inability to close developmental c-subunit leak prevents stimulus-dependent synaptic maturation. Therefore, ATP synthase c-subunit leak closure encourages development and attenuates autistic behaviors.


Subject(s)
Adenosine Triphosphate/metabolism , Fragile X Syndrome/metabolism , Protein Subunits/metabolism , Animals , Cell Line , Citric Acid Cycle/physiology , Fibroblasts/metabolism , Fragile X Mental Retardation Protein/metabolism , HEK293 Cells , Humans , Mice , Neurons/metabolism , RNA, Messenger , Synapses/metabolism
8.
Biochem Pharmacol ; 177: 113995, 2020 07.
Article in English | MEDLINE | ID: mdl-32339494

ABSTRACT

Few discoveries have influenced drug discovery programs more than the finding that mitochondrial membranes undergo swings in permeability in response to cellular perturbations. The conductor of these permeability changes is the aptly named mitochondrial permeability transition pore which, although not yet precisely defined, is comprised of several integral proteins that differentially act to regulate the flux of ions, proteins and metabolic byproducts during the course of cellular physiological functions but also pathophysiological insults. Pursuit of the pore's exact identity remains a topic of keen interest, but decades of research have unearthed provocative functions for the integral proteins leading to their evaluation to develop novel therapeutics for a wide range of clinical indications. Chief amongst these targeted, integral proteins have been the Voltage Dependent Anion Channel (VDAC) and the F1FO ATP synthase. Research associated with the roles and ligands of VDAC has been extensive and we will expand upon 3 examples of ligand:VDAC interactions for consideration of drug discovery projects: Tubulin:VDAC1, Hexokinase I/II:VDAC1 and olesoxime:VDAC1. The discoveries that cyclosporine blocks mitochondrial permeability transition via binding to cyclophilin D, and that cyclophilin D is an important component of F1FO ATP synthase, has heightened interest in the F1FO ATP synthase as a focal point for drug discovery, and we will discuss 2 plausible campaigns associated with disease indications. To date no drug has emerged from prospective targeting these integral proteins; however, continued exploration such as the approaches suggested in this Commentary will increase the likelihood of providing important therapeutics for severely unmet medical needs.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Cholestenones/therapeutic use , Cyclosporine/therapeutic use , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/genetics , Voltage-Dependent Anion Channel 1/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Cyclophilins/genetics , Cyclophilins/metabolism , Gene Expression Regulation , Hexokinase/genetics , Hexokinase/metabolism , Humans , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Permeability/drug effects , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tubulin/genetics , Tubulin/metabolism , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/metabolism
9.
Neuropharmacology ; 120: 11-19, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-26979921

ABSTRACT

An important goal of biomedical research is to translate basic research findings into useful medical advances. In the field of neuropharmacology this requires understanding disease mechanisms as well as the effects of drugs and other compounds on neuronal function. Our hope is that this information will result in new or improved treatment for CNS disease. Despite great progress in our understanding of the structure and functions of the CNS, the discovery of new drugs and their clinical development for many CNS disorders has been problematic. As a result, CNS drug discovery and development programs have been subjected to significant cutbacks and eliminations over the last decade. While there has been recent resurgence of interest in CNS targets, these past changes in priority of the pharmaceutical and biotech industries reflect several well-documented realities. CNS drugs in general have higher failure rates than non-CNS drugs, both preclinically and clinically, and in some areas, such as the major neurodegenerative diseases, the clinical failure rate for disease-modifying treatments has been 100%. The development times for CNS drugs are significantly longer for those drugs that are approved, and post-development regulatory review is longer. In this introduction we review some of the reasons for failure, delineating both scientific and technical realities, some unique to the CNS, that have contributed to this. We will focus on major neurodegenerative disorders, which affect millions, attract most of the headlines, and yet have witnessed the fewest successes. We will suggest some changes that, when coupled with the approaches discussed in the rest of this special volume, may improve outcomes in future CNS-targeted drug discovery and development efforts. This article is part of the Special Issue entitled "Beyond small molecules for neurological disorders".


Subject(s)
Biomedical Research , Central Nervous System Agents/therapeutic use , Central Nervous System Diseases/drug therapy , Drug Discovery , Animals , Central Nervous System Agents/pharmacology , Humans
10.
Mol Pharmacol ; 87(1): 1-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25332381

ABSTRACT

Inefficiency of oxidative phosphorylation can result from futile leak conductance through the inner mitochondrial membrane. Stress or injury may exacerbate this leak conductance, putting cells, and particularly neurons, at risk of dysfunction and even death when energy demand exceeds cellular energy production. Using a novel method, we have recently described an ion conductance consistent with mitochondrial permeability transition pore (mPTP) within the c-subunit of the ATP synthase. Excitotoxicity, reactive oxygen species-producing stimuli, or elevated mitochondrial matrix calcium opens the channel, which is inhibited by cyclosporine A and ATP/ADP. Here we show that ATP and the neuroprotective drug dexpramipexole (DEX) inhibited an ion conductance consistent with this c-subunit channel (mPTP) in brain-derived submitochondrial vesicles (SMVs) enriched for F1FO ATP synthase (complex V). Treatment of SMVs with urea denatured extramembrane components of complex V, eliminated DEX- but not ATP-mediated current inhibition, and reduced binding of [(14)C]DEX. Direct effects of DEX on the synthesis and hydrolysis of ATP by complex V suggest that interaction of the compound with its target results in functional conformational changes in the enzyme complex. [(14)C]DEX bound specifically to purified recombinant b and oligomycin sensitivity-conferring protein subunits of the mitochondrial F1FO ATP synthase. Previous data indicate that DEX increased the efficiency of energy production in cells, including neurons. Taken together, these studies suggest that modulation of a complex V-associated inner mitochondrial membrane current is metabolically important and may represent an avenue for the development of new therapeutics for neurodegenerative disorders.


Subject(s)
Benzothiazoles/pharmacology , Cyclosporine/pharmacology , Mitochondrial Membrane Transport Proteins/metabolism , Proton-Translocating ATPases/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Brain/cytology , Brain/enzymology , Humans , Mitochondrial Membranes/drug effects , Mitochondrial Permeability Transition Pore , Pramipexole
12.
Brain Res ; 1446: 1-11, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22364637

ABSTRACT

Cellular stress or injury can result in mitochondrial dysfunction, which has been linked to many chronic neurological disorders including amyotrophic lateral sclerosis (ALS) and Parkinson's disease (PD). Stressed and dysfunctional mitochondria exhibit an increase in large conductance mitochondrial membrane currents and a decrease in bioenergetic efficiency. Inefficient energy production puts cells, and particularly neurons, at risk of death when energy demands exceed cellular energy production. Here we show that the candidate ALS drug dexpramipexole (DEX; KNS-760704; ((6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine) and cyclosporine A (CSA) inhibited increases in ion conductance in whole rat brain-derived mitochondria induced by calcium or treatment with a proteasome inhibitor, although only CSA inhibited calcium-induced permeability transition in liver-derived mitochondria. In several cell lines, including cortical neurons in culture, DEX significantly decreased oxygen consumption while maintaining or increasing production of adenosine triphosphate (ATP). DEX also normalized the metabolic profile of injured cells and was protective against the cytotoxic effects of proteasome inhibition. These data indicate that DEX increases the efficiency of oxidative phosphorylation, possibly by inhibition of a CSA-sensitive mitochondrial conductance.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Energy Metabolism/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Neurons/ultrastructure , Propranolol/pharmacology , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Biophysical Phenomena/drug effects , Brain/cytology , Cell Survival/drug effects , Cells, Cultured , Cyclosporine/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Male , Mice , Mitochondrial Membranes/drug effects , Neuroblastoma/pathology , Neuroblastoma/ultrastructure , Oligopeptides/pharmacology , Oxygen Consumption/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
13.
Nat Med ; 17(12): 1652-6, 2011 Nov 20.
Article in English | MEDLINE | ID: mdl-22101764

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is characterized by upper and lower motor neuron dysfunction and loss, rapidly progressive muscle weakness, wasting and death. Many factors, including mitochondrial dysfunction, may contribute to ALS pathogenesis. Riluzole, which has shown only modest benefits in a measure of survival time without demonstrated effects on muscle strength or function, is the only approved treatment for ALS. We tested the putative mitochondrial modulator dexpramipexole (KNS-760704; (6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine) in subjects with ALS in a two-part, double-blind safety and tolerability study, with a preliminary assessment of its effects on functional decline and mortality. In part 1, the effects of dexpramipexole (50, 150 or 300 mg d(-1)) versus placebo were assessed over 12 weeks. In part 2, after a 4-week, single-blind placebo washout, continuing subjects were re-randomized to dexpramipexole at 50 mg d(-1) or 300 mg d(-1) as double-blind active treatment for 24 weeks. Dexpramipexole was safe and well tolerated. Trends showing a dose-dependent attenuation of the slope of decline of the ALS Functional Rating Scale-Revised (ALSFRS-R) in part 1 and a statistically significant (P = 0.046) difference between groups in a joint rank test of change from baseline in ALSFRS-R and mortality in part 2 strongly support further testing of dexpramipexole in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Benzothiazoles/therapeutic use , Muscles/physiopathology , Aged , Benzothiazoles/adverse effects , Double-Blind Method , Female , Humans , Male , Middle Aged , Mitochondria/drug effects , Muscle Strength/drug effects , Pramipexole , Riluzole/therapeutic use , Single-Blind Method
14.
J Clin Pharmacol ; 51(8): 1177-85, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20959524

ABSTRACT

Dexpramipexole (KNS-760704; [6R]-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine) is a novel synthetic amino-benzothiazole in development for the treatment of amyotrophic lateral sclerosis (ALS). Preclinical studies have shown that dexpramipexole is neuroprotective in vitro and in vivo, is highly orally bioavailable and water soluble, and rapidly achieves and maintains high central nervous system concentrations relative to plasma. Two phase 1 clinical studies were conducted to assess the safety, tolerability, and pharmacokinetics (PK) of single and multiple doses of dexpramipexole in 54 healthy male and female adults. The effect of food on the single-dose PK of dexpramipexole was also evaluated. Single doses (50 mg, 150 mg, or 300 mg) and multiple doses (50 mg twice daily, 100 mg twice daily, or 150 mg twice daily) of dexpramipexole over 4.5 days were safe and well tolerated. Dexpramipexole was rapidly absorbed, with time to maximum plasma concentration ranging from 1.8 to 2.6 hours and half-life ranging from 6.4 to 8.1 hours under fasted conditions, and was mostly eliminated in urine as unchanged parent drug (84%-90% of dose). Food had no effect on the single-dose PK of dexpramipexole. These findings support the ongoing development of dexpramipexole for the treatment of ALS and further evaluation of the compound's therapeutic potential in other neurodegenerative diseases.


Subject(s)
Benzothiazoles/adverse effects , Benzothiazoles/pharmacokinetics , Dopamine Agonists/adverse effects , Dopamine Agonists/pharmacokinetics , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacokinetics , Adult , Benzothiazoles/administration & dosage , Benzothiazoles/analysis , Dopamine Agonists/administration & dosage , Dopamine Agonists/analysis , Dose-Response Relationship, Drug , Double-Blind Method , Drugs, Investigational/administration & dosage , Drugs, Investigational/analysis , Female , Food-Drug Interactions , Half-Life , Humans , Intestinal Absorption , Male , Metabolic Clearance Rate , Middle Aged , Plasma/chemistry , Pramipexole , Urine/chemistry
15.
CNS Neurosci Ther ; 14(3): 215-26, 2008.
Article in English | MEDLINE | ID: mdl-18801114

ABSTRACT

Developing effective treatments for chronic neurodegenerative disorders such as amyotrophic lateral sclerosis (ALS) has proven extremely difficult. ALS is universally fatal, characterized by progressive weakness due to the degeneration of upper and lower motor neurons, and leads eventually to respiratory failure which is the usual cause of death. Only a single treatment has been approved, the modestly effective nonspecific neuroprotectant Rilutek (riluzole; 2-amino-6-(trifluoromethoxy)benzothiazole). KNS-760704 [(6R)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine dihydrochloride, RPPX], a synthetic amino-benzothiazole with demonstrated activity in maintaining mitochondrial function, is being developed as a treatment for ALS. It has proven to be effective in multiple in vitro and in vivo assays of neuroprotection, including the G93A-SOD1 mutant mouse model; however, its specific mechanism of action remains unknown. The potential of KNS-760604 as a treatment for ALS was first suggested by studies showing that its optical enantiomer, Mirapex[(6S)-4,5,6,7-tetrahydro-N6-propyl-2,6-benzothiazole-diamine; pramipexole dihydrochloride; PPX], a high-affinity agonist at dopamine D2, D3, and D4 receptors, exhibits important neuroprotective properties independent of its dopamine receptor agonism. In cell-based assays, both RPPX and PPX reduce the production of reactive oxygen species (ROS), attenuate the activation of apoptotic pathways, and increase cell survival in response to a variety of neurotoxins. However, PPX has limited utility as a clinical neuroprotective agent because the drug concentrations required for neuroprotection would likely produce unacceptable dopaminergic side effects. RPPX, on the other hand, while possessing the same neuroprotective potential as PPX, is a much lower-affinity dopamine receptor agonist and may therefore be more useful in the treatment of ALS. This review will examine the data supporting the hypothesis that the RPPX may have therapeutic potential for the treatment of neurodegenerative disorders including ALS. In addition, we will briefly review recent preclinical data in support of RPPX, and discuss the current status of its clinical development.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Benzothiazoles/pharmacology , Dopamine Agonists/therapeutic use , Neuroprotective Agents/therapeutic use , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Benzothiazoles/chemistry , Benzothiazoles/therapeutic use , Dopamine Agonists/chemistry , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotective Agents/chemistry , Pramipexole , Stereoisomerism
16.
Expert Opin Ther Targets ; 12(5): 565-81, 2008 May.
Article in English | MEDLINE | ID: mdl-18410240

ABSTRACT

BACKGROUND: Neuronal KCNQ channels (K(V)7.2-5) represent attractive targets for the development of therapeutics for chronic and neuropathic pain, migraine, epilepsy and other neuronal hyperexcitability disorders, although there has been only modest progress in translating this potential into useful therapeutics. OBJECTIVE: Compelling evidence of the importance of K(V)7 channels as neuronal regulatory elements, readily amenable to pharmacological modulation, has sustained widespread interest in these channels as drug targets. This review will update readers on key aspects of the characterization of these important ion channel targets, and will discuss possible current barriers to their exploitation for CNS therapeutics. METHODS: This article is based on a review of recent literature, with a focus on data pertaining to the roles of these channels in neurophysiology. In addition, I review some of the regulatory elements that influence the channels and how these may relate to channel pharmacology, and present a review of recent advances in neuronal K(V)7 channel pharmacology. CONCLUSIONS: These channels continue to be valid and approachable targets for CNS therapeutics. However, we may need to understand more about the roles of neuronal K(V)7 channels during the development of disease states, as well as to pay more attention to a detailed analysis of the molecular pharmacology of the different channel subfamily members and the modes of interaction of individual modulators, in order to successfully target these channels for therapeutic development.


Subject(s)
Central Nervous System Diseases/drug therapy , Drug Delivery Systems , KCNQ Potassium Channels/drug effects , Animals , Controlled Clinical Trials as Topic , Humans , KCNQ Potassium Channels/metabolism , KCNQ2 Potassium Channel/drug effects , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/drug effects , KCNQ3 Potassium Channel/metabolism , Neurons/metabolism , Potassium Channel Blockers/pharmacology
17.
J Med Chem ; 50(3): 528-42, 2007 Feb 08.
Article in English | MEDLINE | ID: mdl-17266205

ABSTRACT

Compound 8a (BMS-191011), an opener of the cloned large-conductance, Ca2+-activated potassium (maxi-K) channel, demonstrated efficacy in in vivo stroke models, which led to its nomination as a candidate for clinical evaluation. Its maxi-K channel opening properties were consistent with its structural topology, being derived by combining elements from other known maxi-K openers. However, 8a suffered from poor aqueous solubility, which complicated elucidation of SAR during in vitro evaluation. The activity of 8a in in vivo stroke models and studies directed toward improving its solubility are reported herein. Enhanced solubility was achieved by appending heterocycles to the 8a scaffold, and a notable observation was made that inclusion of a simple amino group (anilines 8k and 8l) yielded excellent in vitro maxi-K ion channel opening activity and enhanced brain-to-plasma partitioning compared to the appended heterocycles.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/physiology , Oxadiazoles/pharmacology , Animals , Brain/metabolism , Crystallography, X-Ray , Female , In Vitro Techniques , Ion Channel Gating , Molecular Structure , Oocytes/drug effects , Oocytes/physiology , Oxadiazoles/chemistry , Oxadiazoles/pharmacokinetics , Patch-Clamp Techniques , Plasma , Rats , Rats, Inbred SHR , Solubility , Stroke/drug therapy , Stroke/pathology , Structure-Activity Relationship , Xenopus laevis
18.
J Med Chem ; 50(5): 1050-7, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-17274609

ABSTRACT

The synthesis of a series of 3-beta-hydroxyethyl-4-arylquinolin-2-ones is described. These compounds contain hydrophilic and hydrophobic substituents ortho to the phenolic OH in the C ring of the quinolinone. Electrophysiological evaluation of the panel of compounds revealed that 11 and 16 with an unbranched ortho substituent retain activity as maxi-K ion channel openers. Members of this series of compounds can exist as stable atropisomers. Calculated estimates of the energy barrier for rotation around the aryl-aryl single bond in 3 is 31 kcal/mol. The atropisomers of (+/-)-3, (+/-)-4, and (+/-)-11 were separated by chiral HPLC and tested for their effect on maxi-K mediated outward current in hSlo injected X. laevis oocytes. The (-) isomer in each case was found to be more active than the corresponding (+) isomer, suggesting that the ion channel exhibits stereoselective activation. X-ray crystallographic structures of (+)-3 and (+)-11 were determined. Evaluation of the stability of (-)-3 at 80 degrees C in n-butanol indicated a 19.6% conversion to (+)-3 over 72 h. In human serum at 37 degrees C (-)-3 did not racemize over the course of the 30 h study.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/drug effects , Quinolines/chemical synthesis , Animals , Crystallography, X-Ray , Female , Humans , In Vitro Techniques , Ion Channel Gating , Large-Conductance Calcium-Activated Potassium Channels/physiology , Molecular Structure , Oocytes/drug effects , Oocytes/physiology , Patch-Clamp Techniques , Quinolines/chemistry , Quinolines/pharmacology , Stereoisomerism , Thermodynamics , Xenopus laevis
19.
Semin Cell Dev Biol ; 17(5): 555-64, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17141538

ABSTRACT

The voltage-gated calcium channels (VGCCs) are a large and functionally diverse group of ion channels found throughout the central nervous system (CNS) and the periphery. Neuronal functions include the control of neurotransmitter release and neuronal excitability in important pain pathways. In the current review we will give an overview of the data that has been generated in support of these channels performing a pivotal role in the pain pathway.


Subject(s)
Calcium Channels , Calcium/physiology , Ion Channel Gating/physiology , Nociceptors/physiology , Pain/metabolism , Animals , Calcium Channels/chemistry , Calcium Channels/physiology , Humans , Mice , Models, Biological , Pain/etiology
20.
Psychopharmacology (Berl) ; 188(4): 629-40, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17004085

ABSTRACT

RATIONALE: Alzheimer's dementia (AD) patients have profound deficits in cognitive and social functions, mediated in part by a decline in cholinergic function. Acetylcholinesterase inhibitors (AChEI) are the most commonly prescribed treatment for the cognitive deficits in AD patients, but their therapeutic effects are small, and it is still not clear if they primarily affect attention, memory, or some other cognitive/behavioral functions. OBJECTIVES: The objective of the present experiments was to explore the effects of donepezil (Aricepttrade mark), an AChEI, on behavioral deficits related exclusively to cholinergic dysfunction. MATERIALS AND METHODS: The effects of donepezil were assessed in Sprague-Dawley rats with scopolamine-induced deficits in a battery of cognitive/behavioral tests. RESULTS: Scopolamine produced deficits in contextual and cued fear conditioning, the 5-choice serial reaction time test, delayed nonmatching to position, the radial arm maze, and the Morris water maze. Analyses of the pattern and size of the effects revealed that donepezil produced very large effects on scopolamine-induced deficits in psychomotor function (approximately 20-50% of the variance), moderate-sized effects on scopolamine-induced deficits in simple conditioning and attention (approximately 3-10% of the variance), but only small effects on scopolamine-induced deficits in higher cognitive functions of working memory and spatial mapping (approximately 1% of the variance). CONCLUSIONS: These results are consistent with the limited efficacy of donepezil on higher cognitive function in AD patients, and suggest that preclinical behavioral models could be used not only to determine if novel treatments have some therapeutic potential, but also to predict more precisely what the pattern and size of the effects might be.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Cognition Disorders/drug therapy , Indans/pharmacology , Piperidines/pharmacology , Psychomotor Disorders/drug therapy , Animals , Attention/drug effects , Cognition Disorders/chemically induced , Conditioning, Psychological/drug effects , Donepezil , Male , Maze Learning/drug effects , Memory/drug effects , Muscarinic Antagonists , Psychomotor Disorders/chemically induced , Rats , Rats, Sprague-Dawley , Scopolamine
SELECTION OF CITATIONS
SEARCH DETAIL
...