Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 360
1.
Channels (Austin) ; 18(1): 2335469, 2024 Dec.
Article En | MEDLINE | ID: mdl-38564754

Studies in genetically modified animals and human genetics have recently provided new insight into the role of voltage-gated L-type Ca2+ channels in human disease. Therefore, the inhibition of L-type Ca2+ channels in vivo in wildtype and mutant mice by potent dihydropyridine (DHP) Ca2+ channel blockers serves as an important pharmacological tool. These drugs have a short plasma half-life in humans and especially in rodents and show high first-pass metabolism upon oral application. In the vast majority of in vivo studies, they have therefore been delivered through parenteral routes, mostly subcutaneously or intraperitoneally. High peak plasma concentrations of DHPs cause side effects, evident as DHP-induced aversive behaviors confounding the interpretation of behavioral readouts. Nevertheless, pharmacokinetic data measuring the exposure achieved with these applications are sparse. Moreover, parenteral injections require animal handling and can be associated with pain, discomfort and stress which could influence a variety of physiological processes, behavioral and other functional readouts. Here, we describe a noninvasive oral application of the DHP isradipine by training mice to quickly consume small volumes of flavored yogurt that can serve as drug vehicle. This procedure does not require animal handling, allows repeated drug application over several days and reproducibly achieves peak plasma concentrations over a wide range previously shown to be well-tolerated in humans. This protocol should facilitate ongoing nonclinical studies in mice exploring new indications for DHP Ca2+ channel blockers.


Calcium Channel Blockers , Calcium Channels, L-Type , Mice , Humans , Animals , Isradipine/pharmacology , Isradipine/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Administration, Oral
2.
Eur J Pharmacol ; 971: 176489, 2024 May 15.
Article En | MEDLINE | ID: mdl-38492875

Substance abuse disorder is a chronic condition for which pharmacological treatment options remain limited. L-type calcium channels (LTCC) have been implicated in drug-related plasticity and behavior. Specifically, dopaminergic neurons in the mesocorticolimbic pathway express Cav1.2 and Cav1.3 channels, which may regulate dopaminergic activity associated with reward behavior. Therefore, this study aimed to investigate the hypothesis that pre-administration of the LTCC blocker, isradipine can mitigate the effects of cocaine by modulating central glutamatergic transmission. For that, we administered isradipine at varying concentrations (1, 7.5, and 15 µg/µL) via intracerebroventricular injection in male Swiss mice. This pretreatment was carried out prior to subjecting animals to behavioral assessments to evaluate cocaine-induced locomotor sensitization and conditioned place preference (CPP). The results revealed that isradipine administered at a concentration of 1 µg/µL effectively attenuated both the sensitization and CPP induced by cocaine (15 mg/kg, via i. p.). Moreover, mice treated with 1 µg/µL of isradipine showed decreased presynaptic levels of glutamate and calcium in the cortex and hippocampus as compared to control mice following cocaine exposure. Notably, the gene expression of ionotropic glutamate receptors, AMPA, and NMDA, remained unchanged, as did the expression of Cav1.2 and Cav1.3 channels. Importantly, these findings suggest that LTCC blockage may inhibit behavioral responses to cocaine, most likely by decreasing glutamatergic input in areas related to addiction.


Calcium Channel Blockers , Cocaine , Mice , Male , Animals , Calcium Channel Blockers/pharmacology , Isradipine/pharmacology , Glutamic Acid , Cocaine/pharmacology , Dopamine/metabolism
3.
Eur J Neurosci ; 59(6): 1242-1259, 2024 Mar.
Article En | MEDLINE | ID: mdl-37941514

Ca2+ entry into nigrostriatal dopamine (DA) neurons and axons via L-type voltage-gated Ca2+ channels (LTCCs) contributes, respectively, to pacemaker activity and DA release and has long been thought to contribute to vulnerability to degeneration in Parkinson's disease. LTCC function is greater in DA axons and neurons from substantia nigra pars compacta than from ventral tegmental area, but this is not explained by channel expression level. We tested the hypothesis that LTCC control of DA release is governed rather by local mechanisms, focussing on candidate biological factors known to operate differently between types of DA neurons and/or be associated with their differing vulnerability to parkinsonism, including biological sex, α-synuclein, DA transporters (DATs) and calbindin-D28k (Calb1). We detected evoked DA release ex vivo in mouse striatal slices using fast-scan cyclic voltammetry and assessed LTCC support of DA release by detecting the inhibition of DA release by the LTCC inhibitors isradipine or CP8. Using genetic knockouts or pharmacological manipulations, we identified that striatal LTCC support of DA release depended on multiple intersecting factors, in a regionally and sexually divergent manner. LTCC function was promoted by factors associated with Parkinsonian risk, including male sex, α-synuclein, DAT and a dorsolateral co-ordinate, but limited by factors associated with protection, that is, female sex, glucocerebrosidase activity, Calb1 and ventromedial co-ordinate. Together, these data show that LTCC function in DA axons and isradipine effect are locally governed and suggest they vary in a manner that in turn might impact on, or reflect, the cellular stress that leads to parkinsonian degeneration.


Dopamine , Parkinson Disease , Female , Mice , Animals , Male , Isradipine/pharmacology , Isradipine/metabolism , Dopamine/metabolism , Calcium Channels, L-Type/metabolism , alpha-Synuclein/metabolism , Parkinson Disease/metabolism , Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , Substantia Nigra/metabolism , Risk Factors , Calcium/metabolism
4.
Brain Res ; 1818: 148528, 2023 11 01.
Article En | MEDLINE | ID: mdl-37567548

Methamphetamine (meth) is an addictive psychostimulant and there are no FDA-approved treatment options for patients suffering from meth use disorders. In addition to being addictive, meth is also neurotoxic and chronic administration results in degeneration of substantia nigra pars compacta (SNc) dopamine and locus coeruleus (LC) norepinephrine neurons in mice. Optimal treatment strategies for meth use disorders would attenuate maladaptive meth-seeking behavior as well as provide neuroprotection. The L-type calcium channel inhibitor isradipine and the monoamine oxidase (MAO) inhibitor rasagiline both prevent chronic meth-induced SNc and LC degeneration but effects on meth-seeking are unknown. To test whether these clinically available compounds can mitigate meth-seeking, mice were implanted with chronic indwelling jugular vein catheters and allowed to self-administer meth (0.1 mg/kg/infusion) for 10 consecutive days (2-hrs/day) on a fixed ratio (FR) 1 schedule of reinforcement with meth infusions paired to a cue light. One day after the last self-administration session mice were tested for cue-associated meth-seeking behavior wherein the meth-associated cue light was contingently presented but meth reinforcement withheld. Isradipine (3 mg/kg) attenuated cue-associated meth-seeking in both male and female mice. In contrast, rasagiline (1 mg/kg) had no effect on seeking in either sex. These results suggest that isradipine may have the potential to serve as a dual-purpose pharmacotherapy for meth use disorders by attenuating seeking behavior and providing neuroprotection.


Central Nervous System Stimulants , Methamphetamine , Mice , Male , Female , Animals , Methamphetamine/pharmacology , Isradipine/pharmacology , Calcium Channels, L-Type , Cues , Self Administration , Drug-Seeking Behavior/physiology
5.
Exp Clin Psychopharmacol ; 31(2): 507-522, 2023 Apr.
Article En | MEDLINE | ID: mdl-36595455

Given the personal and public health burden of addictive disorders, innovative approaches to treatment are sorely needed. This systematic review examined the use of the pharmacological agent isradipine in the context of potential applications for addiction treatment. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guided a comprehensive search of PubMed, Cochrane Library, and PsycINFO between the years 1985 to July 2022. Studies were included if isradipine was administered to adults with a current Diagnostic and Statistical Manual of Mental Disorders-5th edition diagnosis of a substance use disorder and/or to healthy volunteers alone and in conjunction with a substance (i.e, cocaine, methamphetamine, alcohol). A total of 16 studies with 252 participants were included in this review. Substantial variability was identified with study designs, isradipine dosages/dosing, and addictive substance of interest. Outcomes clustered in four categories: (a) cerebral blood flow (CBF), (b) hemodynamic effects, (c) subjective effects, and (d) cognitive effects. Isradipine was found to improve CBF in individuals with cocaine-induced hypoperfusion and in several studies was found to reduce parameters of blood pressure elevation after stimulant use. There were no significant findings on isradipine's effect on subjective reporting (i.e., craving, mood, drug affect) or cognition/attention. Given the limited number of studies identified in this review, there is insufficient data to draw clear conclusions. The direct effects of isradipine as a pharmacologic agent for addictive disorder treatment appear minimal, however, future work may benefit from examining the impact of isradipine as an augmentative agent within existing cue exposure paradigms for preventing cue-induced drug relapse. (PsycInfo Database Record (c) 2023 APA, all rights reserved).


Cocaine , Methamphetamine , Substance-Related Disorders , Adult , Humans , Isradipine/pharmacology , Isradipine/therapeutic use , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Substance-Related Disorders/drug therapy
6.
Neuron ; 108(6): 1091-1102.e5, 2020 12 23.
Article En | MEDLINE | ID: mdl-33080228

Corticostriatal synaptic integration is partitioned among striosome (patch) and matrix compartments of the dorsal striatum, allowing compartmentalized control of discrete aspects of behavior. Despite the significance of such organization, it's unclear how compartment-specific striatal output is dynamically achieved, particularly considering new evidence that overlap of afferents is substantial. We show that dopamine oppositely shapes responses to convergent excitatory inputs in mouse striosome and matrix striatal spiny projection neurons (SPNs). Activation of postsynaptic D1 dopamine receptors promoted the generation of long-lasting synaptically evoked "up-states" in matrix SPNs but opposed it in striosomes, which were more excitable under basal conditions. Differences in dopaminergic modulation were mediated, in part, by dendritic voltage-gated calcium channels (VGCCs): pharmacological manipulation of L-type VGCCs reversed compartment-specific responses to D1 receptor activation. These results support a novel mechanism for the selection of striatal circuit components, where fluctuating levels of dopamine shift the balance of compartment-specific striatal output.


Corpus Striatum/drug effects , Dendrites/drug effects , Neurons/drug effects , Receptors, Dopamine D1/antagonists & inhibitors , Animals , Benzazepines/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Corpus Striatum/metabolism , Dendrites/metabolism , Dopamine Antagonists/pharmacology , Isradipine/pharmacology , Mice , Mice, Transgenic , Neurons/metabolism , Receptors, Dopamine D1/metabolism , Synapses/drug effects , Synapses/metabolism
7.
Sci Signal ; 13(618)2020 02 11.
Article En | MEDLINE | ID: mdl-32047116

L-type voltage-gated Ca2+ channels (LTCCs) are implicated in neurodegenerative processes and cell death. Accordingly, LTCC antagonists have been proposed to be neuroprotective, although this view is disputed, because intentional LTCC activation can also have beneficial effects. LTCC-mediated Ca2+ influx influences mitochondrial function, which plays a crucial role in the regulation of cell viability. Hence, we investigated the effect of modulating LTCC-mediated Ca2+ influx on mitochondrial function in cultured hippocampal neurons. To activate LTCCs, neuronal activity was stimulated by increasing extracellular K+ or by application of the GABAA receptor antagonist bicuculline. The activity of LTCCs was altered by application of an agonistic (Bay K8644) or an antagonistic (isradipine) dihydropyridine. Our results demonstrated that activation of LTCC-mediated Ca2+ influx affected mitochondrial function in a bimodal manner. At moderate stimulation strength, ATP synthase activity was enhanced, an effect that involved Ca2+-induced Ca2+ release from intracellular stores. In contrast, high LTCC-mediated Ca2+ loads led to a switch in ATP synthase activity to reverse-mode operation. This effect, which required nitric oxide, helped to prevent mitochondrial depolarization and sustained increases in mitochondrial Ca2+ Our findings indicate a complex role of LTCC-mediated Ca2+ influx in the tuning and maintenance of mitochondrial function. Therefore, the use of LTCC inhibitors to protect neurons from neurodegeneration should be reconsidered carefully.


Calcium Channels, L-Type/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Mitochondria/metabolism , Neurons/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Biological Transport/drug effects , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Cell Survival/drug effects , Cells, Cultured , Hippocampus/cytology , Isradipine/pharmacology , Neurons/cytology , Neurons/drug effects , Rats, Sprague-Dawley
8.
Neurosci Bull ; 36(2): 165-173, 2020 Feb.
Article En | MEDLINE | ID: mdl-31482520

In the present study, we investigated the mechanisms underlying the mediation of iron transport by L-type Ca2+ channels (LTCCs) in primary cultured ventral mesencephalon (VM) neurons from rats. We found that co-treatment with 100 µmol/L FeSO4 and MPP+ (1-methyl-4-phenylpyridinium) significantly increased the production of intracellular reactive oxygen species, decreased the mitochondrial transmembrane potential and increased the caspase-3 activation compared to MPP+ treatment alone. Co-treatment with 500 µmol/L CaCl2 further aggravated the FeSO4-induced neurotoxicity in MPP+-treated VM neurons. Co-treatment with 10 µmol/L isradipine, an LTCC blocker, alleviated the neurotoxicity induced by co-application of FeSO4 and FeSO4/CaCl2. Further studies indicated that MPP+ treatment accelerated the iron influx into VM neurons. In addition, FeSO4 treatment significantly increased the intracellular Ca2+ concentration. These effects were blocked by isradipine. These results suggest that elevated extracellular Ca2+ aggravates iron-induced neurotoxicity. LTCCs mediate iron transport in dopaminergic neurons and this, in turn, results in elevated intracellular Ca2+ and further aggravates iron-induced neurotoxicity.


Calcium Channels, L-Type/metabolism , Calcium/metabolism , Dopaminergic Neurons/drug effects , Iron/toxicity , Mesencephalon/metabolism , 1-Methyl-4-phenylpyridinium/toxicity , Animals , Apoptosis/drug effects , Calcium Channel Blockers/pharmacology , Caspase 3/metabolism , Cells, Cultured , Female , Isradipine/pharmacology , Pregnancy , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
9.
Mol Psychiatry ; 25(10): 2373-2391, 2020 10.
Article En | MEDLINE | ID: mdl-31501511

Cocaine-associated memories are critical drivers of relapse in cocaine-dependent individuals that can be evoked by exposure to cocaine or stress. Whether these environmental stimuli recruit similar molecular and circuit-level mechanisms to promote relapse remains largely unknown. Here, using cocaine- and stress-primed reinstatement of cocaine conditioned place preference to model drug-associated memories, we find that cocaine drives reinstatement by increasing the duration that mice spend in the previously cocaine-paired context whereas stress increases the number of entries into this context. Importantly, both forms of reinstatement require Cav1.2 L-type Ca2+ channels (LTCCs) in cells of the prelimbic cortex that project to the nucleus accumbens core (PrL→NAcC). Utilizing fiber photometry to measure circuit activity in vivo in conjunction with the LTCC blocker, isradipine, we find that LTCCs drive differential recruitment of the PrL→ NAcC pathway during cocaine- and stress-primed reinstatement. While cocaine selectively activates PrL→NAcC cells prior to entry into the cocaine-paired chamber, a measure that is predictive of duration in that chamber, stress increases persistent activity of this projection, which correlates with entries into the cocaine-paired chamber. Using projection-specific chemogenetic manipulations, we show that PrL→NAcC activity is required for both cocaine- and stress-primed reinstatement, and that activation of this projection in Cav1.2-deficient mice restores reinstatement. These data indicate that LTCCs are a common mediator of cocaine- and stress-primed reinstatement. However, they engage different patterns of behavior and PrL→NAcC projection activity depending on the environmental stimuli. These findings establish a framework to further study how different environmental experiences can drive relapse, and supports further exploration of isradipine, an FDA-approved LTCC blocker, as a potential therapeutic for the prevention of relapse in cocaine-dependent individuals.


Calcium Channels, L-Type/metabolism , Cocaine/pharmacology , Corpus Striatum/drug effects , Frontal Lobe/drug effects , Memory/drug effects , Neural Pathways/drug effects , Stress, Psychological/psychology , Animals , Cocaine-Related Disorders/prevention & control , Corpus Striatum/cytology , Frontal Lobe/cytology , Isradipine/pharmacology , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects
10.
Pharmacol Biochem Behav ; 189: 172828, 2020 02.
Article En | MEDLINE | ID: mdl-31785245

Parkinson's disease (PD) is known as a movement disorder due to characteristic motor features. Existing therapies for PD are only symptomatic, and their efficacy decreases as disease progresses. Zebrafish, a vertebrate in which parkinsonism has been modelled, offers unique features for the identification of molecules with antiparkinsonian properties. Here, we developed a screening assay for the selection of neuroactive agents with antiparkinsonian potential. First, we performed a pharmacological validation of the phenotypes exhibited by the 6-hydroxydopamine zebrafish model, by testing the effects of known antiparkinsonian agents. These drugs were also tested for disease-modifying properties by whole mount immunohistochemistry to TH+ neurons and confocal microscopy in the dopaminergic diencephalic cluster of zebrafish. Next, we optimized a phenotypic screening using the 6-hydroxydopamine zebrafish model and tested 1600 FDA-approved bioactive drugs. We found that 6-hydroxydopamine-lesioned zebrafish larvae exhibit bradykinetic and dyskinetic-like behaviours that are rescued by the administration of levodopa, rasagiline, isradipine or amantadine. The rescue of dopaminergic cell loss by isradipine was also verified, through the observation of a higher number of TH+ neurons in 6-OHDA-lesioned zebrafish larvae treated with this compound as compared to untreated lesioned larvae. The phenotypic screening enabled us to identify several compounds previously positioned for PD, as well as, new molecules with potential antiparkinsonian properties. Among these, we selected stavudine, tapentadol and nabumetone as the most promising candidates. Our results demonstrate the functional similarities of the motor impairments exhibited by 6-hydroxydopamine-lesioned zebrafish with mammalian models of PD and with PD patients, and highlights novel molecules with antiparkinsonian potential.


Antiparkinson Agents/pharmacology , Antiparkinson Agents/therapeutic use , Larva/drug effects , Oxidopamine/pharmacology , Parkinson Disease/drug therapy , Zebrafish/growth & development , Amantadine/pharmacology , Amantadine/therapeutic use , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dopaminergic Neurons/drug effects , Drug Repositioning/methods , Indans/pharmacology , Indans/therapeutic use , Isradipine/pharmacology , Isradipine/therapeutic use , Levodopa/pharmacology , Levodopa/therapeutic use , Locomotion/drug effects , Motor Activity/drug effects , Phenotype
11.
Elife ; 82019 10 14.
Article En | MEDLINE | ID: mdl-31609202

Sensory hair cells in the ear utilize specialized ribbon synapses. These synapses are defined by electron-dense presynaptic structures called ribbons, composed primarily of the structural protein Ribeye. Previous work has shown that voltage-gated influx of Ca2+ through CaV1.3 channels is critical for hair-cell synapse function and can impede ribbon formation. We show that in mature zebrafish hair cells, evoked presynaptic-Ca2+ influx through CaV1.3 channels initiates mitochondrial-Ca2+ (mito-Ca2+) uptake adjacent to ribbons. Block of mito-Ca2+ uptake in mature cells depresses presynaptic-Ca2+ influx and impacts synapse integrity. In developing zebrafish hair cells, mito-Ca2+ uptake coincides with spontaneous rises in presynaptic-Ca2+ influx. Spontaneous mito-Ca2+ loading lowers cellular NAD+/NADH redox and downregulates ribbon size. Direct application of NAD+ or NADH increases or decreases ribbon size respectively, possibly acting through the NAD(H)-binding domain on Ribeye. Our results present a mechanism where presynaptic- and mito-Ca2+ couple to confer proper presynaptic function and formation.


Calcium Channels, L-Type/metabolism , Calcium/metabolism , Evoked Potentials, Auditory/physiology , Eye Proteins/metabolism , Hair Cells, Auditory/metabolism , Mitochondria/metabolism , Synapses/metabolism , Zebrafish Proteins/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Animals, Genetically Modified , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Calcium Signaling , Cell Size , Embryo, Nonmammalian , Eye Proteins/chemistry , Eye Proteins/genetics , Gene Expression , Hair Cells, Auditory/cytology , Hair Cells, Auditory/drug effects , Isradipine/pharmacology , Mitochondria/drug effects , Mitochondria/ultrastructure , NAD/metabolism , Oxidation-Reduction , Protein Binding , Protein Interaction Domains and Motifs , Ruthenium Compounds/pharmacology , Synapses/drug effects , Synapses/ultrastructure , Synaptic Transmission , Zebrafish , Zebrafish Proteins/agonists , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
12.
Autophagy ; 15(8): 1322-1332, 2019 08.
Article En | MEDLINE | ID: mdl-30755067

The pathophysiology of most neurodegenerative diseases includes aberrant accumulation of protein aggregates. Recent evidence highlights the role of protein degradation pathways in neurodegeneration. Concurrently, genetic tools have been generated to enable zebrafish, Danio rerio, to be used as an animal model to study neurodegenerative processes. In addition to optical clarity and fast ex utero development, the zebrafish brain is relatively small and has conserved structures with its mammalian counterparts. To take advantage of this model organism and to aid further studies on autophagy and neurodegeneration, we created a stable transgenic zebrafish line that expresses eGFP-Map1lc3b specifically in post-mitotic neurons under the elavl3 promoter. This line is useful for indirectly monitoring autophagic activity in neurons in vivo and screening for macroautophagy/autophagy-modulating compounds. We determined the applicability of this transgenic line by modulating and quantifying the number of autophagosomes via treatment with a known autophagy inducer (rapamycin) and inhibitors (3-methyladenine, protease inhibitors). Additionally, we proposed an in vivo method for quantifying rates of autophagosome accumulation, which can be used to infer occurrence of autophagic flux. Last, we tested two FDA-approved drugs currently undergoing clinical studies for Parkinson disease, isradipine and nilotinib, and found that isradipine did not modulate autophagy, whereas nilotinib induced both autophagosome number and autophagic flux. It is hoped that others will find this line useful as an in vivo vertebrate model to find or validate autophagy modulators that might be used to halt the progression of neurodegenerative diseases. Abbreviations: 3MA: 3-methyladenine; BafA: bafilomycin A1; dd: dorsal diencephalon; dpf: days post fertilization; e: eye; eGFP: enhanced green fluorescent protein; Elavl3: ELAV like neuron-specific RNA binding protein 3; FDA: Food and Drug Administration; hb: habenula; hpt, hours post treatment; Map1lc3b: microtubule-associated protein 1 light chain 3 beta; nt: neural tube; ot, optic tectum; P/E: pepstatin A and E64d; PD: Parkinson disease; PMTs: photomultiplier tubes; PTU: 1-phenyl-2-thiourea; Ta: annealing temperature; Tel, telencephalon.


Autophagy , Neurons/cytology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Cell Line, Tumor , Central Nervous System/metabolism , Green Fluorescent Proteins/metabolism , Humans , Isradipine/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Neurons/drug effects , Neurons/metabolism , Pyrimidines/pharmacology , Sirolimus/pharmacology , Zebrafish Proteins/metabolism
14.
Curr Drug Targets ; 20(2): 158-165, 2019.
Article En | MEDLINE | ID: mdl-28641520

Memories associated with substance use disorders, or substance-associated cues increase the likelihood of craving and relapse during abstinence. There is a growing consensus that manipulation of synaptic plasticity may reduce the strength of substance abuse-related memories. On the biological front, there are new insights that suggest memories associated with substance use disorder may follow unique neurobiological pathways that render them more accessible to pharmacological intervention. In parallel to this, research in neurochemistry has identified several potential candidate molecules that could influence the formation and maintenance of long-term memory. Drugs that target these molecules (blebbistatin, isradipine and zeta inhibitory peptide) have shown promise at the preclinical stage. In this review, we shall provide an overview of the evolving understanding on the biochemical mechanisms involved in memory formation and expound on the premise that substance use disorder is a learning disorder.


Memory, Long-Term/drug effects , Substance-Related Disorders/psychology , Cues , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Isradipine/pharmacology , Isradipine/therapeutic use , Molecular Targeted Therapy , Neuronal Plasticity/drug effects , Substance-Related Disorders/drug therapy
15.
Neuropsychopharmacology ; 43(12): 2361-2372, 2018 11.
Article En | MEDLINE | ID: mdl-29773910

Previous preclinical and clinical investigations have focused on the L-type calcium channel (LTCC) as a potential therapeutic target for substance abuse. While some clinical studies have examined the ability of LTCC blockers to alter cocaine's subjective effects, very few LTCC studies have examined cocaine relapse. Here, we examined whether ventral tegmental area (VTA)-specific or systemic administration of the LTCC inhibitor, isradipine, altered cocaine-seeking behavior in a rat model. Male Sprague-Dawley rats first received 10 days of cocaine self-administration training (2 h sessions), where active lever depression resulted in delivery of a ∼0.5 mg/kg cocaine infusion paired with a tone + light cue. Rats then underwent 10 days of forced abstinence, without access to cocaine or cocaine cues. Rats were then returned to the opertant chamber for the cue-induced cocaine-seeking test, where active lever depression in the original training context resulted in tone + light cue presentation. We found VTA specific or systemic isradipine administration robustly attenuated cocaine-seeking, without altering cocaine-taking nor natural reward seeking. Dopamine (DA) signaling in the nucleus accumbens (NAc) core is necessary and sufficient for cue-induced drug-seeking. Surprisingly in our study, isradipine enhanced tonic and phasic DA signaling in cocaine abstinent rats, with no change in sucrose abstinent nor naïve rats. Strikingly, isradipine's behavioral effects were dependent upon NAc core DA receptor activation. Together, our findings reveal a novel mechanism by which the FDA-approved drug, isradipine, could act to decrease cocaine relapse.


Calcium Channel Blockers/pharmacology , Cocaine/administration & dosage , Dopamine/metabolism , Drug-Seeking Behavior/drug effects , Isradipine/pharmacology , Nucleus Accumbens/metabolism , Ventral Tegmental Area/metabolism , Animals , Calcium Channels, L-Type/metabolism , Cues , Drug-Seeking Behavior/physiology , Male , Nucleus Accumbens/drug effects , Rats , Rats, Sprague-Dawley , Ventral Tegmental Area/drug effects
16.
J Clin Invest ; 128(6): 2266-2280, 2018 06 01.
Article En | MEDLINE | ID: mdl-29708514

The ability of the Cav1 channel inhibitor isradipine to slow the loss of substantia nigra pars compacta (SNc) dopaminergic (DA) neurons and the progression of Parkinson's disease (PD) is being tested in a phase 3 human clinical trial. But it is unclear whether and how chronic isradipine treatment will benefit SNc DA neurons in vivo. To pursue this question, isradipine was given systemically to mice at doses that achieved low nanomolar concentrations in plasma, near those achieved in patients. This treatment diminished cytosolic Ca2+ oscillations in SNc DA neurons without altering autonomous spiking or expression of Ca2+ channels, an effect mimicked by selectively knocking down expression of Cav1.3 channel subunits. Treatment also lowered mitochondrial oxidant stress, reduced a high basal rate of mitophagy, and normalized mitochondrial mass - demonstrating that Cav1 channels drive mitochondrial oxidant stress and turnover in vivo. Thus, chronic isradipine treatment remodeled SNc DA neurons in a way that should not only diminish their vulnerability to mitochondrial challenges, but to autophagic stress as well.


Calcium Signaling/drug effects , Dopaminergic Neurons/metabolism , Isradipine/pharmacology , Mitochondria/metabolism , Mitophagy/drug effects , Oxidative Stress/drug effects , Animals , Caveolin 1/metabolism , Dopaminergic Neurons/pathology , Humans , Male , Mice , Mitochondria/pathology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology
17.
Sci Rep ; 7(1): 7869, 2017 08 11.
Article En | MEDLINE | ID: mdl-28801600

The spontaneous activity of sinoatrial node (SAN) pacemaker cells is generated by a functional interplay between the activity of ionic currents of the plasma membrane and intracellular Ca2+ dynamics. The molecular correlate of a dihydropyridine (DHP)-sensitive sustained inward Na+ current (I st), a key player in SAN automaticity, is still unknown. Here we show that I st and the L-type Ca2+ current (I Ca,L) share CaV1.3 as a common molecular determinant. Patch-clamp recordings of mouse SAN cells showed that I st is activated in the diastolic depolarization range, and displays Na+ permeability and minimal inactivation and sensitivity to I Ca,L activators and blockers. Both CaV1.3-mediated I Ca,L and I st were abolished in CaV1.3-deficient (CaV1.3-/-) SAN cells but the CaV1.2-mediated I Ca,L current component was preserved. In SAN cells isolated from mice expressing DHP-insensitive CaV1.2 channels (CaV1.2DHP-/-), I st and CaV1.3-mediated I Ca,L displayed overlapping sensitivity and concentration-response relationships to the DHP blocker nifedipine. Consistent with the hypothesis that CaV1.3 rather than CaV1.2 underlies I st, a considerable fraction of I Ca,L was resistant to nifedipine inhibition in CaV1.2DHP-/- SAN cells. These findings identify CaV1.3 channels as essential molecular components of the voltage-dependent, DHP-sensitive I st Na+ current in the SAN.


Action Potentials/drug effects , Calcium Channels, L-Type/metabolism , Calcium/metabolism , Dihydropyridines/pharmacology , Heart Rate/drug effects , Action Potentials/genetics , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Cells, Cultured , Heart Rate/genetics , Isradipine/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Nifedipine/pharmacology , Patch-Clamp Techniques , Sinoatrial Node/cytology , Sinoatrial Node/drug effects , Sinoatrial Node/metabolism
18.
J Neurosci ; 37(28): 6761-6777, 2017 07 12.
Article En | MEDLINE | ID: mdl-28592699

Ca2+-influx through L-type Ca2+-channels (LTCCs) is associated with activity-related stressful oscillations of Ca2+ levels within dopaminergic (DA) neurons in the substantia nigra (SN), which may contribute to their selective degeneration in Parkinson's disease (PD). LTCC blockers were neuroprotective in mouse neurotoxin models of PD, and isradipine is currently undergoing testing in a phase III clinical trial in early PD. We report no evidence for neuroprotection by in vivo pretreatment with therapeutically relevant isradipine plasma levels, or Cav1.3 LTCC deficiency in 6-OHDA-treated male mice. To explain this finding, we investigated the pharmacological properties of human LTCCs during SN DA-like and arterial smooth muscle (aSM)-like activity patterns using whole-cell patch-clamp recordings in HEK293 cells (Cav1.2 α1-subunit, long and short Cav1.3 α1-subunit splice variants; ß3/α2δ1). During SN DA-like pacemaking, only Cav1.3 variants conducted Ca2+ current (ICa) at subthreshold potentials between action potentials. SN DA-like burst activity increased integrated ICa during (Cav1.2 plus Cav1.3) and after (Cav1.3) the burst. Isradipine inhibition was splice variant and isoform dependent, with a 5- to 11-fold lower sensitivity to Cav1.3 variants during SN DA-like pacemaking compared with Cav1.2 during aSM-like activity. Supratherapeutic isradipine concentrations reduced the pacemaker precision of adult mouse SN DA neurons but did not affect their somatic Ca2+ oscillations. Our data predict that Cav1.2 and Cav1.3 splice variants contribute differentially to Ca2+ load in SN DA neurons, with prominent Cav1.3-mediated ICa between action potentials and after bursts. The failure of therapeutically relevant isradipine levels to protect SN DA neurons can be explained by weaker state-dependent inhibition of SN DA LTCCs compared with aSM Cav1.2.SIGNIFICANCE STATEMENT The high vulnerability of dopamine (DA) neurons in the substantia nigra (SN) to neurodegenerative stressors causes Parkinson's disease (PD). Ca2+ influx through voltage-gated L-type Ca2+ channels (LTCCs), in particular Cav1.3, appears to contribute to this vulnerability, and the LTCC inhibitor isradipine is currently being tested as a neuroprotective agent for PD in a phase III clinical trial. However, in our study isradipine plasma concentrations approved for therapy were not neuroprotective in a PD mouse model. We provide an explanation for this observation by demonstrating that during SN DA-like neuronal activity LTCCs are less sensitive to isradipine than Cav1.2 LTCCs in resistance blood vessels (mediating dose-limiting vasodilating effects) and even at supratherapeutic concentrations isradipine fails to reduce somatic Ca2+ oscillations of SN DA neurons.


Calcium Channels, L-Type/metabolism , Calcium Signaling/physiology , Dopamine/metabolism , Dopaminergic Neurons/physiology , Isradipine/metabolism , Substantia Nigra/physiology , Animals , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Dopaminergic Neurons/drug effects , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Isradipine/pharmacology , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , Substantia Nigra/drug effects
19.
Oncotarget ; 8(29): 47284-47295, 2017 Jul 18.
Article En | MEDLINE | ID: mdl-28521299

The aim of this study is to investigate the effects of L-type calcium channels (LTCCs) on MPTP-induced dopamine (DA) neuron degeneration and iron accumulation in the substantia nigra (SN) of mice. By real-time PCR and western blots, we first quatified expressions of L-type Cav1.2 and Cav1.3 calcium channel α1 subunits in the SN of experimental mice treated with MPTP. We found that the expressions of Cav1.2 and Cav1.3 calcium channel α1 subunits markedly increased after MPTP treatment for 2 and 3 weeks. Secondly, we observed the effects of isradipine, a LTCC antagonist, on MPTP-induced DA neuron degeneration and iron accumulation in the SN. Our results showed that isradipine treatment prevented against MPTP-induced Cav1.2 and Cav1.3 calcium channel α1 subunits up-regulation in the SN. We also found that isradipine prevented against MPTP-induced DA neuron depletion in the SN and partly restored the DA content in the striatum. Moreover, we found that isradipine inhibited the increase of iron positive cells in the SN of the MPTP-treated mice. Finally, we investigated the effects of isradipine on cellular iron accumulation in the dopaminergic MES23.5 cell line. Our studies showed that MPP+ treatment accelerated iron influx in the MES23.5 cells. Treatment with Bayk8644 further aggravated iron accumulation. Treatment with isradipine prevented against MPP+-induced iron influx in the MES23.5 cells. These results suggest that up-regulation of LTCCs may be responsible for the DA neuron degeneration in the MPTP-treated mice, The LTCCs may directly contribute to iron influx into DA neurons, and isradipine may suppress cellular iron accumulation and prevents neurodegeneration.


Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Iron/metabolism , Isradipine/pharmacology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Biomarkers , Calcium Channel Blockers/pharmacology , Cell Line , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Disease Progression , Dopamine , Dopaminergic Neurons/pathology , Male , Mice , Neuroprotective Agents/pharmacology , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Substantia Nigra/pathology
20.
Artif Cells Nanomed Biotechnol ; 45(1): 139-145, 2017 Feb.
Article En | MEDLINE | ID: mdl-26829018

Context Isradipine is an effective calcium channel blocker used in the management of hypertension. It undergoes extensive first pass metabolism and has low oral bioavailability. Hence we attempted to develop isradipine-loaded invasomes. Objective The purpose of this work was to prepare and characterize invasomes carrier for isradipine, and to evaluate the optimized formulation obtained for pharmacodynamic study. Materials and methods Isradipine-loaded invasomes were prepared by conventional thin layer evaporation technique using Phospholipon® 90G, ß-citronellene (terpene) and ethanol. Prepared formulations were characterized in terms of size, size distribution, morphology, entrapment efficiency, and antihypertensive activity. Results and discussion It was observed that prepared isradipine-loaded invasomes delivers ameliorated flux, reasonable entrapment efficiency, and more effectiveness for transdermal delivery. The optimized formulation presented the particle size of 194 ± 18 nm, entrapment efficiency (88.46%), and attained mean transdermal flux of 22.80 ± 2.10 µg/cm2/h through rat skin. Confocal laser scanning microscopy revealed an enhanced permeation of Rhodamine-Red-loaded isradipine invasomes to the deeper layers of the rat skin. During antihypertensive study, the treatment group showed a substantial and constant decrease in blood pressure, for up to 24 h. The isradipine invasomes formulation was found to be effective, with a 20% reduction in blood pressure by virtue of better permeation through Wistar rat skin. Conclusion It was concluded that the developed isradipine invasomes accentuate the transdermal flux and the results obtained encouraged the use of the isradipine-loaded invasomes as the formulation for the potential management of hypertension.


Hypertension/drug therapy , Isradipine , Administration, Cutaneous , Animals , Hypertension/physiopathology , Isradipine/pharmacokinetics , Isradipine/pharmacology , Liposomes , Rats , Rats, Wistar
...