Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 52
1.
Eur J Pharmacol ; 939: 175437, 2023 Jan 15.
Article En | MEDLINE | ID: mdl-36502961

Pimavanserin is a highly selective 5-HT2A inverse agonist in current medical use. Prior studies suggest that 5-HT2A serotonin receptors may play a role in anxiety and emotional memory. Therefore, pimavanserin was tested in a rat model of PTSD to determine whether it might ameliorate PTSD-like symptoms. The lifetime prevalence of PTSD is estimated to be 125% higher in women than men. Consequently, in an effort to create a robust model of PTSD that was more representative of human PTSD prevalence, 20-week old female rats of the emotionally hyperreactive Lewis strain were used for these studies. The rats were single-housed and exposed twice to restraint stress coupled with predator odor or to a sham-stressed condition. Twenty days after the second stress or sham-stress exposure, rats were injected with saline alone or with 0.3 or 1.0 mg/kg pimavanserin, doses that were confirmed to substantially block 5-HT2A receptor activity in this study without causing any non-specific behavioral or adverse effects. One hour later, rats were tested for anxiety through acoustic startle response, the elevated plus-maze and three parameters of open field behavior. Five days later, blood was sampled for plasma corticosterone. The stressed/saline-injected rats had higher anxiety scores and corticosterone levels than sham-stressed/saline-injected rats. Pimavanserin significantly and generally dose-dependently reversed these persistent stress effects, but had no significant effect on the behavioral measures in normal, non-stressed rats. These results, consistent with a role for the 5-HT2A receptor, suggest that pimavanserin might have potential to reduce some consequences of traumatic stress.


Stress Disorders, Post-Traumatic , Animals , Female , Humans , Male , Rats , Anxiety/drug therapy , Anxiety/etiology , Corticosterone/pharmacology , Disease Models, Animal , Drug Inverse Agonism , Rats, Inbred Lew , Receptor, Serotonin, 5-HT2A , Reflex, Startle , Serotonin/pharmacology , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/etiology , Stress, Psychological/complications , Stress, Psychological/drug therapy
2.
Alzheimers Dement (N Y) ; 8(1): e12247, 2022.
Article En | MEDLINE | ID: mdl-35128032

INTRODUCTION: Psychosis in Alzheimer's disease (AD) is associated with grave clinical consequences including a precipitous cognitive decline and a hastened demise. These outcomes are aggravated by use of existing antipsychotic medications, which are also associated with cognitive decline and increased mortality; preclinical models that would develop new therapeutic approaches are desperately needed. The current report evaluates the ability of the neoteric antipsychotic, pimavanserin, to normalize hyperkinesis and sensorimotor gating in the novel catechol-O-methyltransferase (COMT) deleted P301L/COMT- and rTg(P301L)4510 models of psychotic AD, and the impact of pimavanserin on tau pathology. METHODS: Female P301L/COMT- mice were behaviorally characterized for abnormalities of locomotion and sensorimotor gating, and biochemically characterized for patterns of tau phosphorylation relative to relevant controls utilizing high-sensitivity tau enzyme-linked immunosorbent assay (ELISA). Female P301L/COMT- and rTg(P301L)4510 mice were randomized to pimavanserin or vehicle treatment to study the ability of pimavanserin to normalize locomotion and rescue sensorimotor gating. Additionally, high-sensitivity tau ELISA was used to investigate the impact of treatment on tau phosphorylation. RESULTS: P301L/COMT- mice evidenced a hyperlocomotive phenotype and deficits of sensorimotor gating relative to wild-type mice on the same background, and increased tau phosphorylation relative to COMT-competent P301L mice. Pimavanserin normalized the hyperkinetic phenotype in both the P301L/COMT- and rTg(P301L)4510 mice but had no impact on sensorimotor gating in either model. Pimavanserin treatment had little impact on tau phosphorylation patterns. DISCUSSION: These data suggest that pimavanserin ameliorates tau-driven excessive locomotion. Given the morbidity associated with aberrant motor behaviors such as pacing in AD and lack of effective treatments, future studies of the impact of pimavanserin on actigraphy in patients with this syndrome may be warranted.

3.
CNS Drugs ; 35(7): 727-741, 2021 07.
Article En | MEDLINE | ID: mdl-34224112

Psychosis occurs across a wide variety of dementias with differing etiologies, including Alzheimer's dementia, Parkinson's dementia, Lewy body dementia, frontotemporal dementia, and vascular dementia. Pimavanserin, a selective serotonin 5-HT2A receptor (5-HT2AR) inverse agonist, has shown promising results in clinical trials by reducing the frequency and/or severity of hallucinations and delusions and the risk of relapse of these symptoms in patients with dementia-related psychosis. A literature review was conducted to identify mechanisms that explain the role of 5-HT2ARs in both the etiology and treatment of dementia-related psychosis. This review revealed that most pathological changes commonly associated with neurodegenerative diseases cause one or more of the following events to occur: reduced synaptic contact of gamma aminobutyric acid (GABA)-ergic interneurons with glutamatergic pyramidal cells, reduced cortical innervation from subcortical structures, and altered 5-HT2AR expression levels. Each of these events promotes increased pyramidal cell hyperexcitability and disruption of excitatory/inhibitory balance, facilitating emergence of psychotic behaviors. The brain regions affected by these pathological changes largely coincide with areas expressing high levels of 5-HT2ARs. At the cellular level, 5-HT2ARs are most highly expressed on cortical glutamatergic pyramidal cells, where they regulate pyramidal cell excitability. The common effects of different neurodegenerative diseases on pyramidal cell excitability together with the close anatomical and functional connection of 5-HT2ARs to pyramidal cell excitability may explain why suppressing 5-HT2AR activity could be an effective strategy to treat dementia-related psychosis.


Dementia , Psychotic Disorders , Pyramidal Cells , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Antipsychotic Agents/pharmacology , Cortical Excitability/drug effects , Cortical Excitability/physiology , Dementia/complications , Dementia/pathology , Dementia/physiopathology , Psychotic Disorders/drug therapy , Psychotic Disorders/etiology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Receptor, Serotonin, 5-HT2A/metabolism
4.
ACS Chem Neurosci ; 12(12): 2088-2098, 2021 06 16.
Article En | MEDLINE | ID: mdl-34032411

Neurodegeneration and impaired neural development are a common feature of many neuropsychiatric disorders. Second-generation antipsychotics (SGAs) and certain atypical antidepressants display neuroprotective effects. Though these drugs interact with many molecular targets, a common shared attribute is high antagonist potency at 5-HT2A receptors. Pimavanserin is a selective 5-HT2A inverse agonist/antagonist that was recently FDA approved for treating hallucinations and delusions associated with Parkinson's disease. Unlike SGAs, pimavanserin lacks activity at other targets like dopamine, histamine, muscarinic, and adrenergic receptors. To investigate whether selective 5-HT2A inverse agonists have neuroprotective properties, pimavanserin and another selective 5-HT2A inverse agonist, M100907, were applied to primary cultures of dopaminergic neurons treated with 1-methyl-4-phenylpyridinium (MPP+). Both pimavanserin and M100907 protected dopaminergic neurons against MPP+-induced cell death. The neuroprotective effects of pimavanserin required signaling through the extracellular signal-regulated kinase 1/2 pathway, restored mitochondrial function, and reduced oxidative stress. Further investigation showed that pimavanserin promotes the release of brain-derived neurotrophic factor and glial-derived neurotrophic factor (GDNF) and that the neuroprotective effects of pimavanserin were blocked by antibodies to GDNF but not by anti-tyrosine receptor kinase B receptor antibodies. Thus, pimavanserin induces release of neurotrophic factors and protects dopaminergic neurons against MPP+ toxicity in a GDNF-dependent manner.


1-Methyl-4-phenylpyridinium , Neuroprotective Agents , 1-Methyl-4-phenylpyridinium/toxicity , Dopaminergic Neurons , Glial Cell Line-Derived Neurotrophic Factor , Neuroprotective Agents/pharmacology , Piperidines , Urea/analogs & derivatives
5.
J Neurochem ; 156(5): 658-673, 2021 03.
Article En | MEDLINE | ID: mdl-33278025

Amyloid-ß (Aß) peptide aggregation into soluble oligomers and insoluble plaques is a precipitating event in the pathogenesis of Alzheimer's disease (AD). Given that synaptic activity can regulate Aß generation, we postulated that 5HT2A -Rs may regulate Aß as well. We treated APP/PS1 transgenic mice with the selective 5HT2A inverse agonists M100907 or Pimavanserin systemically and measured brain interstitial fluid (ISF) Aß levels in real-time using in vivo microdialysis. Both compounds reduced ISF Aß levels by almost 50% within hours, but had no effect on Aß levels in 5HT2A -R knock-out mice. The Aß-lowering effects of Pimavanserin were blocked by extracellular-regulated kinase (ERK) and NMDA receptor inhibitors. Chronic administration of Pimavanserin by subcutaneous osmotic pump to aged APP/PS1 mice significantly reduced CSF Aß levels and Aß pathology and improved cognitive function in these mice. Pimavanserin is FDA-approved to treat Parkinson's disease psychosis, and also has been shown to reduce psychosis in a variety of other dementia subtypes including Alzheimer's disease. These data demonstrate that Pimavanserin may have disease-modifying benefits in addition to its efficacy against neuropsychiatric symptoms of Alzheimer's disease. Read the Editorial Highlight for this article on page 560.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Drug Inverse Agonism , Piperidines/therapeutic use , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin 5-HT2 Receptor Agonists/therapeutic use , Urea/analogs & derivatives , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/biosynthesis , Animals , Female , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C3H , Mice, Transgenic , Piperidines/pharmacology , Serotonin 5-HT2 Receptor Agonists/pharmacology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Serotonin 5-HT2 Receptor Antagonists/therapeutic use , Urea/pharmacology , Urea/therapeutic use
6.
Neurosci Lett ; 713: 134524, 2019 11 20.
Article En | MEDLINE | ID: mdl-31585211

Previous work has shown that chronic nicotine administration causes adaptive changes in 5-HT2A receptor expression. Based on this relationship, it was hypothesized that inactivating 5-HT2A receptors with the inverse agonists pimavanserin and volinanserin (MDL100907), would reduce the symptoms of nicotine withdrawal syndrome. Sprague-Dawley rats were rendered nicotine-dependent by subcutaneous infusion of nicotine bitartrate, 9 mg/kg/day for seven days. The infusions were then terminated, and 22 h later, rats were observed under "blind" conditions for somatically expressed behavioral nicotine withdrawal signs. One hour before observations, the nicotine dependent rats were injected i.p. with saline alone, or either 0.3 or 1.0 mg/kg pimavanserin in saline. Total withdrawal signs were reduced in a dose-dependent manner. A one-way ANOVA (total withdrawal signs as a function of dose) was highly significant, as was the descending linear trend of withdrawal signs as a function of dose. The 1.0 mg/kg dose reduced withdrawal signs nearly to the level exhibited by a comparison group of non-dependent rats injected with saline. A second experiment was conducted in a similar manner, which showed that volinanserin at 1.0 mg/kg but not 0.25 mg/kg also reduced nicotine withdrawal signs to nearly the level of non-dependent rats. In vitro experiments demonstrated that both pimavanserin and volinanserin potently antagonize 5-HT2A receptors, with approximately 25-fold, and 300-fold selectivity over 5-HT2C receptors, respectively. The results suggest that the 5-HT2A receptor contributes to mediating nicotine withdrawal syndrome, and thus represents a potential target for interventions to aid smoking cessation.


Fluorobenzenes/pharmacology , Nicotine/adverse effects , Piperidines/pharmacology , Receptor, Serotonin, 5-HT2A/drug effects , Substance Withdrawal Syndrome/prevention & control , Urea/analogs & derivatives , Animals , Dose-Response Relationship, Drug , Drug Inverse Agonism , Male , Rats , Receptor, Serotonin, 5-HT2C/drug effects , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Urea/pharmacology
8.
Neurosci Lett ; 684: 98-103, 2018 09 25.
Article En | MEDLINE | ID: mdl-29981879

Considerable evidence suggests the Neuropeptide FF (NPFF) and related peptides exert pro-nociceptive and anti-opiate actions, particularly at the supra-spinal level, which may contribute to opiate dependence. The FF1 receptor subtype appears to be primarily responsible for anti-opiate effects. In contrast, stimulation of the FF2 receptor primarily induces pro-opiate effects. AC-262620 is a small molecule, systemically active, selective FF1 receptor antagonist. An initial experiment showed that 10 mg/kg i.p. AC-262620 significantly reduced subsequent naloxone-precipitated somatically expressed withdrawal signs in rats infused s.c. for seven days with 0.3 mg/kg/hr morphine sulfate. A second experiment showed that the same dose of AC-262620 significantly reduced subsequent spontaneous withdrawal signs 23.75 h after termination of seven days s.c. infusion of 0.6 mg/kg/hr morphine sulfate. Chronic nicotine intake may contribute to dependence by overstimulating opiate receptors through release of opiate peptides. By analogy to opiate dependence, it was hypothesized that FF1 receptor activation contributes to nicotine dependence and withdrawal syndrome. AC-262620 significantly reduced somatically expressed withdrawal signs precipitated by the nicotinic antagonist mecamylamine in rats infused for seven days with nicotine bitartrate. Taken together, these findings suggest that NPFF or related neuropeptides contribute to opiate, as well as nicotine, dependence and withdrawal syndrome through the FF1 receptor.


Opioid-Related Disorders/drug therapy , Receptors, Neuropeptide/antagonists & inhibitors , Substance Withdrawal Syndrome/drug therapy , Tobacco Use Disorder/drug therapy , Analgesics, Opioid/adverse effects , Animals , Male , Morphine/adverse effects , Nicotine/adverse effects , Opioid-Related Disorders/psychology , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide/physiology , Substance Withdrawal Syndrome/psychology , Tobacco Use Disorder/psychology
9.
ACS Chem Neurosci ; 8(9): 2065-2077, 2017 09 20.
Article En | MEDLINE | ID: mdl-28691794

Retinoid X receptors (RXRs) play key roles in many physiological processes in both the periphery and central nervous system. In addition, RXRs form heterodimers with other nuclear receptors to exert their physiological effects. The nuclear receptor related 1 protein (NURR1) is particularly interesting because of its role in promoting differentiation and survival of dopamine neurons. However, only a small number of RXR-heterodimer selective modulators are available, with limited chemical diversity. This work describes the synthesis, biochemical evaluation, and structural elucidation of a novel series of RXR ligands with strongly biased interactions with RXRα-NURR1 heterodimers. Targeted modifications to the small molecule biaryl scaffold caused local RXRα side-chain disturbances and displacement of secondary structural elements upon ligand binding. This resulted in the repositioning of protein helices in the heterodimer interface of RXRα, alterations in homo- versus heterodimer formation, and modulation of activation function 2 (AF2). The data provide a rationale for the design of RXR ligands consisting of a highly conserved hydrophilic region, strongly contributing to the ligand affinity, and a variable hydrophobic region, which efficiently probes the effects of structural changes at the level of the ligand on co-regulator recruitment or the RXRα-NURR1 dimerization interface.


Nuclear Receptor Subfamily 4, Group A, Member 2/chemistry , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Retinoid X Receptor alpha/chemistry , Retinoid X Receptor alpha/metabolism , Drug Design , Escherichia coli , Esters/chemistry , Ethers/chemistry , Humans , Hydrogen Bonding , Ligands , Models, Molecular , Molecular Structure , Nuclear Receptor Subfamily 4, Group A, Member 2/agonists , Nuclear Receptor Subfamily 4, Group A, Member 2/antagonists & inhibitors , Protein Binding , Protein Multimerization , Retinoid X Receptor alpha/agonists , Retinoid X Receptor alpha/antagonists & inhibitors , Two-Hybrid System Techniques
10.
Pharmacology ; 99(3-4): 128-138, 2017.
Article En | MEDLINE | ID: mdl-27992867

We have previously reported that amiodarone interacts with a novel allosteric site on muscarinic receptors. Amiodarone's most striking effect is to enhance the maximal response elicited by muscarinic agonists at the M1, M3, and M5 receptors. Furthermore, the quaternary analog N-ethylamiodarone (NEA) is inhibitory at these receptors and appears to compete with amiodarone at that allosteric site. In the present studies, we show that dronedarone also modulates Gq-mediated responses at M1 and M3, although in a more discriminating manner. For example, dronedarone markedly enhances pilocarpine-stimulated release of arachidonic acid from CHO cells, via the M3 receptor subtype, but does not affect the acetylcholine-stimulated response. Such probe-dependent effects are diagnostic of an allosteric interaction. In comparison to these effects at M3, dronedarone is strongly inhibitory toward both pilocarpine and acetylcholine at the M1 subtype. The effects of dronedarone are consistent with an interaction at the amiodarone site: dronedarone inhibits the enhancement of acetylcholine's response produced by amiodarone at the M3 subtype; also, NEA reverses the enhancement of pilocarpine's response at M3 produced by either dronedarone or amiodarone. In studies with the M1-selective allosteric agonist 4-[3-(4-butylpiperidin-1-yl)-propyl]-7-fluoro-4H-benzo[1,4]oxazin-3-one (AC-260584), amiodarone enhanced the maximal response observed, whereas dronedarone was inhibitory. On the other hand, benzyl quinolone carboxylic acid, the well-known allosteric ligand that dramatically enhances the potency of acetylcholine at the M1 subtype, had no effect on the response profile of AC-260584. In summary, dronedarone acts at M1 and M3 muscarinic receptors in a manner that complements amiodarone and provides an additional tool with which to investigate this novel allosteric site.


Amiodarone/analogs & derivatives , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M3/agonists , Receptor, Muscarinic M3/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Amiodarone/chemistry , Amiodarone/metabolism , Amiodarone/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Dronedarone
11.
Mol Imaging ; 152016.
Article En | MEDLINE | ID: mdl-27553293

Activation of retinoid X receptors (RXRs) has been proposed as a therapeutic mechanism for the treatment of neurodegeneration, including Alzheimer's and Parkinson's diseases. We previously reported radiolabeling of a Food and Drug Administration-approved RXR agonist, bexarotene, by copper-mediated [(11)C]CO2 fixation and preliminary positron emission tomography (PET) neuroimaging that demonstrated brain permeability in nonhuman primate with regional binding distribution consistent with RXRs. In this study, the brain uptake and saturability of [(11)C]bexarotene were studied in rats and nonhuman primates by PET imaging under baseline and greater target occupancy conditions. [(11)C]Bexarotene displays a high proportion of nonsaturable uptake in the brain and is unsuitable for RXR occupancy measurements in the central nervous system.


Brain/diagnostic imaging , Neuroimaging/methods , Tetrahydronaphthalenes/pharmacology , Animals , Bexarotene , Brain/metabolism , Carbon Radioisotopes/chemistry , Male , Positron-Emission Tomography , Primates , Radiopharmaceuticals/chemistry , Rats , Retinoid X Receptors/agonists , Retinoid X Receptors/metabolism , Tetrahydronaphthalenes/chemistry
12.
ACS Chem Neurosci ; 7(9): 1180-7, 2016 09 21.
Article En | MEDLINE | ID: mdl-27456785

Chemotherapy-induced neuropathic pain (CINP) remains a major unmet medical need. Estrogen receptor beta (ERß)-selective agonists represent a novel strategy for treating CINP because they are neuroprotective and may also have anticancer activity. We confirmed that ERß-selective agonists have antiallodynic effects in the spinal nerve ligation model of neuropathic pain. We then showed that structurally diverse ERß-selective agonists also relieved allodynia in CINP caused by taxol, oxaliplatin, and vincristine. These effects were receptor subtype specific and mediated by ERß receptors as ERα-selective and nonselective estrogen agonists were inactive, a mixture of an ERß and ERα agonist was inactive, and ERß-selective antagonists blocked the effects of the ERß-selective agonists. The efficacy and potency of ERß-agonists was greater in male rats than females. To address the possibility that AC-186 might stimulate proliferation of cancers, rendering it unsuitable for treating CINP, we evaluated proliferative effects of AC-186 on prostate cancer cells and found it inhibited growth (LNCaP cells) or had no effect (PC3 cells) on these cells. Thus, ERß-selective agonists exhibit potential for treating CINP.


Antineoplastic Agents/adverse effects , Estrogen Receptor beta/metabolism , Estrogens/therapeutic use , Neuralgia/chemically induced , Neuralgia/drug therapy , Animals , Cell Line, Tumor , Disease Models, Animal , Estrogens/chemistry , Female , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Male , Neuralgia/etiology , Pain Measurement , Pain Threshold/drug effects , Prostatic Neoplasms/drug therapy , Rats , Rats, Sprague-Dawley
13.
J Med Chem ; 59(3): 1232-8, 2016 Feb 11.
Article En | MEDLINE | ID: mdl-26820900

The nuclear receptor Nurr1 can be activated by RXR via heterodimerization (RXR-Nurr1) and is a promising target for treating neurodegenerative diseases. We herein report the enantioselective synthesis and SAR of sterically constricted benzofurans at RXR. The established SAR, using whole cell functional assays, lead to the full agonist 9a at RXR (pEC50 of 8.2) and RXR-Nurr1. The X-ray structure shows enantiomeric discrimination where 9a optimally addresses the ligand binding pocket of RXR.


Benzofurans/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Protein Multimerization/drug effects , Retinoid X Receptors/metabolism , Benzofurans/chemical synthesis , Benzofurans/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Nuclear Receptor Subfamily 4, Group A, Member 2/agonists , Retinoid X Receptors/agonists , Structure-Activity Relationship
14.
Neurosci Lett ; 584: 141-5, 2015 Jan 01.
Article En | MEDLINE | ID: mdl-25459291

Neuropeptide FF (NPFF) modulates opiate actions. It has pro-nociceptive effects, primarily through the NPFF receptor 1 subtype, and anti-nociceptive effects, primarily through the NPFFR2 subtype. AC-263093 is a small l, organic, systemically active molecule that was previously shown to functionally activate NPFFR2, but not NPFFR1. It was hypothesized that AC-263093 would attenuate morphine tolerance. Rats were tested for radiant heat tail-flick latency before and after 5 mg/kg morphine sulfate s.c. They were then rendered morphine-tolerant by continuous subcutaneous infusion of 17.52 mg/kg/day morphine sulfate. On the seventh day of infusion, they were retested for analgesia 10 and 20 min after 5mg/kg morphine sulfate s.c. Tolerance was indicated by reduction of morphine analgesia from the pre-infusion test. Fifty minutes prior to morphine challenge, rats received either 10 mg/kg i.p. AC-263093 or injection vehicle alone. AC-2623093-treated rats had far smaller tolerance scores than control rats. This drug effect was significant, p = 0.015. The same dose of AC-263093 had almost no analgesic effect in non-tolerant, saline-infused rats. In vitro experiments revealed that AC-263093 had equal affinity for NPFFR1 and NPFFR2, and functionally inactivated NPFFR1, in addition to its previously shown ability to activate NPFFR2. Thus, altering the balance between activation of NPFF receptor subtypes may provide one approach to reversing opiate tolerance.


Analgesics, Opioid/pharmacology , Morphine/pharmacology , Receptors, Neuropeptide/metabolism , Animals , Drug Interactions , Drug Tolerance , Hydrazines/pharmacology , Male , Oligopeptides/pharmacology , Radioligand Assay , Rats, Sprague-Dawley , Receptors, Neuropeptide/antagonists & inhibitors
15.
Neurochem Res ; 39(10): 2008-17, 2014 Oct.
Article En | MEDLINE | ID: mdl-24682754

Parkinson's disease psychosis (PDP) is a condition that may develop in up to 60 % of Parkinson's patients, and is a major reason for nursing home placement for those affected. There are no FDA approved drugs for PDP but low doses of atypical anti-psychotic drugs (APDs) are commonly prescribed off-label. Only low-dose clozapine has shown efficacy in randomized controlled trials, but all APDs have black box warnings related to the increased mortality and morbidity when used in elderly demented patients. Using molecular pharmacological profiling of a large collection of marketed drugs, we discovered that potent inverse agonist activity against 5-HT2A serotonin receptors was a common feature of atypical APDs, especially the atypical APDs used to treat PDP. Since low-dose clozapine therapy selectively blocks this receptor, it was hypothesized that a highly selective 5-HT2A receptor inverse agonist might provide good symptom control in patients suffering from PDP, with a greatly improved safety and tolerability profile. A high throughput screening and subsequent chemical lead optimization campaign to develop potent, selective 5-HT2A receptor inverse agonists was launched, eventually resulting in the discovery of pimavanserin. Pimavanserin displays nanomolar potency as a 5-HT2A receptor inverse agonist, selectivity for 5-HT2A over 5-HT2C receptors, and no meaningful activity at any other G-protein coupled receptor. It demonstrated robust activity in preclinical models of schizophrenia and PDP, and did not worsen motoric symptoms, in contrast to the APDs tested. In a Phase III clinical trial, pimavanserin showed highly significant benefits in the primary endpoint, the scale for assessment of positive symptoms-PD, a scale adapted for use in PDP. In addition, improvements in all other efficacy endpoints, including physician's clinical global impression, caregiver burden, night-time sleep quality and daytime wakefulness, were seen. Pimavanserin demonstrated good safety and tolerability and did not worsen motoric symptoms as assessed by the unified Parkinson's disease rating scale parts II and III. An open-label extension study has further demonstrated that pimavanserin is safe and well-tolerated with long-term use. Pimavanserin may therefore offer a viable treatment option for patients suffering from PDP.


Parkinson Disease/drug therapy , Piperidines/therapeutic use , Psychotic Disorders/drug therapy , Urea/analogs & derivatives , Drug Discovery , Humans , Parkinson Disease/physiopathology , Piperidines/chemistry , Urea/chemistry , Urea/therapeutic use
16.
Bioorg Med Chem Lett ; 24(8): 1944-7, 2014 Apr 15.
Article En | MEDLINE | ID: mdl-24666648

Bexarotene, a retinoid X receptor (RXR) agonist, is being tested as a potential disease modifying treatment for neurodegenerative conditions. To limit the peripheral exposure of bexarotene and release it only in the affected areas of the brain, we designed a prodrug strategy based on the enzyme NAD(P)H/quinone oxidoreductase (NQO1) that is elevated in neurodegenerative diseases. A series of indolequinones (known substrates of NQO1) was synthesized and coupled to bexarotene. Bexarotene-3-(hydroxymethyl)-5-methoxy-1,2-dimethyl-1H-indole-4,7-dione ester 7a was cleaved best by NQO1. The prodrugs are not cleaved by esterase.


Drug Delivery Systems , Indoles/chemical synthesis , NAD(P)H Dehydrogenase (Quinone)/chemistry , Prodrugs/chemical synthesis , Tetrahydronaphthalenes/chemistry , Tetrahydronaphthalenes/chemical synthesis , Bexarotene , Indolequinones/chemical synthesis , Indolequinones/chemistry , Indolequinones/pharmacology , Indoles/chemistry , Indoles/pharmacology , Molecular Structure , Prodrugs/chemistry , Prodrugs/pharmacology , Retinoid X Receptors/agonists , Tetrahydronaphthalenes/pharmacology
17.
J Pharmacol Exp Ther ; 348(1): 116-24, 2014 Jan.
Article En | MEDLINE | ID: mdl-24204014

Use of molecular pharmacology to reprofile older drugs discovered before the advent of recombinant technologies is a fruitful method to elucidate mechanisms of drug action, expand understanding of structure-activity relationships between drugs and receptors, and in some cases, repurpose approved drugs. The H3 histamine receptor is a G-protein-coupled receptor (GPCR) primarily expressed in the central nervous system where among many things it modulates cognitive processes, nociception, feeding and drinking behavior, and sleep/wakefulness. In binding assays and functional screens of the H3 histamine receptor, the antiarrhythmic drugs lorcainide and amiodarone were identified as potent, selective antagonists/inverse agonists of human and rat H3 histamine receptors, with relatively little or no activity at over 20 other monoamine GPCRs, including H1, H2, and H4 receptors. Potent antagonism of H3 receptors was unique to amiodarone and lorcainide of 20 antiarrhythmic drugs tested, representing six pharmacological classes. These results expand the pharmacophore of H3 histamine receptor antagonist/inverse agonists and may explain, in part, the effects of lorcainide on sleep in humans.


Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Benzeneacetamides/pharmacology , Drug Inverse Agonism , Histamine Agonists/pharmacology , Piperidines/pharmacology , Receptors, Histamine H3 , Amiodarone/chemistry , Amiodarone/metabolism , Animals , Anti-Arrhythmia Agents/chemistry , Anti-Arrhythmia Agents/metabolism , Benzeneacetamides/chemistry , Benzeneacetamides/metabolism , Cell Proliferation/drug effects , HEK293 Cells , Histamine Agonists/chemistry , Histamine Agonists/metabolism , Humans , Mice , NIH 3T3 Cells , Piperidines/chemistry , Piperidines/metabolism , Rats , Receptors, Histamine H3/metabolism
18.
ACS Chem Neurosci ; 4(11): 1430-8, 2013 Nov 20.
Article En | MEDLINE | ID: mdl-24117438

Nurr1 is a nuclear hormone receptor (NucHR) strongly implicated in the growth, maintenance, and survival of dopaminergic neurons. Nurr1 may be unable to bind ligands directly, but it forms heterodimers with other NucHRs that do. Using bioluminescence resonance energy transfer (BRET) assays to directly monitor interactions of Nurr1 with other NucHRs, we found the cancer drug bexarotene (Targretin, also LGD1069) displayed biased interactions with Nurr1-RXR heterodimers compared with RXR-RXR homodimers. Remarkably, at doses up to 100-fold lower than those effective in rodent cancer models, bexarotene rescued dopamine neurons and reversed behavioral deficits in 6-hydroxydopamine (6-OHDA) lesioned rats. Compared to the high doses used in cancer therapy, low doses of bexarotene have significantly milder side effects including a reduced increase in plasma triglycerides and less suppression of thyroid function. On the basis of extrapolations from rat to human doses, we hypothesize that low oral doses of bexarotene may provide an effective and tolerated therapy for Parkinson's disease (PD).


Behavior, Animal/drug effects , Bioluminescence Resonance Energy Transfer Techniques/methods , Dopaminergic Neurons/drug effects , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Tetrahydronaphthalenes/administration & dosage , Administration, Oral , Animals , Behavior, Animal/physiology , Bexarotene , Cell Survival/drug effects , Cell Survival/physiology , Disease Models, Animal , Dopaminergic Neurons/pathology , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Infusions, Subcutaneous , Injections, Intraventricular , Male , Nuclear Receptor Subfamily 4, Group A, Member 2/chemistry , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , Parkinson Disease/physiopathology , Primary Cell Culture , Protein Multimerization , Rats , Rats, Sprague-Dawley , Tetrahydronaphthalenes/therapeutic use
19.
J Pharmacol Exp Ther ; 347(3): 697-704, 2013 Dec.
Article En | MEDLINE | ID: mdl-24078870

AC-55541 [N-[[1-(3-bromo-phenyl)-eth-(E)-ylidene-hydrazinocarbonyl]-(4-oxo-3,4-dihydro-phthalazin-1-yl)-methyl]-benzamide] and AC-264613 [2-oxo-4-phenylpyrrolidine-3-carboxylic acid [1-(3-bromo-phenyl)-(E/Z)-ethylidene]-hydrazide] are the first two small-molecule agonists described for the G protein-coupled receptor protease-activated receptor 2 (PAR2), but whether they activate PAR2 through a similar mechanism as its tethered peptide ligand or soluble peptide mimetics of its tethered peptide ligand is unclear. Extracellular loop 2 (ECL2) has been shown to play a critical role in the activation mechanism of PAR2. Therefore, we constructed a series of PAR2 receptors mutated in ECL2, including a previously described polymorphic variant of PAR2 (F240S), and compared AC-55541 and AC-264613 to SLIGRL and a potent analog of SLIGRL called 2-furoyl LIGRLO in a series of functional assays, including cellular proliferation, phosphatidylinositol hydrolysis, and ß-arrestin recruitment assays. Surprisingly, receptors with the F240S mutation were constitutively active in all functional assays tested. Furthermore, AC-55541 and AC-264613 were potentiated over 30-fold at the receptors with the F240S mutation, whereas SLIGRL and 2-furoyl LIGRLO were much less affected. In contrast, mutagenesis of charged residues in ECL2 confirmed their important role in the actions of peptide agonists of PAR2, whereas these mutations did not significantly affect activation of PAR2 by AC-55541 or AC-264613. These results suggest that F240S PAR2 receptors may be useful in screens to detect novel small-molecule PAR2 modulators and that further work on the biological importance of the F240S PAR2 variant is warranted.


Receptor, PAR-2/agonists , Receptor, PAR-2/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Arrestins/metabolism , Bioluminescence Resonance Energy Transfer Techniques , Cell Proliferation/drug effects , Hydrolysis , Mice , Molecular Sequence Data , Mutation/physiology , Oligopeptides/pharmacology , Phosphatidylinositols/metabolism , Receptor, PAR-2/genetics , beta-Arrestins
20.
ACS Chem Neurosci ; 4(9): 1249-55, 2013 Sep 18.
Article En | MEDLINE | ID: mdl-23898966

Drugs that selectively activate estrogen receptor ß (ERß) are potentially safer than the nonselective estrogens currently used in hormonal replacement treatments that activate both ERß and ERα. The selective ERß agonist AC-186 was evaluated in a rat model of Parkinson's disease induced through bilateral 6-hydroxydopamine lesions of the substantia nigra. In this model, AC-186 prevented motor, cognitive, and sensorimotor gating deficits and mitigated the loss of dopamine neurons in the substantia nigra, in males, but not in females. Furthermore, in male rats, 17ß-estradiol, which activates ERß and ERα with equal potency, did not show the same neuroprotective benefits as AC-186. Hence, in addition to a beneficial safety profile for use in both males and females, a selective ERß agonist has a differentiated pharmacological profile compared to 17ß-estradiol in males.


Cyclohexanes/therapeutic use , Estrogen Receptor beta/agonists , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/drug therapy , Phenols/therapeutic use , Animals , Corpus Striatum/chemistry , Corpus Striatum/drug effects , Corpus Striatum/pathology , Cyclohexanes/chemistry , Cyclohexanes/pharmacology , Cytokines/analysis , Drug Evaluation, Preclinical , Estradiol/therapeutic use , Exploratory Behavior/drug effects , Female , Male , Molecular Structure , Nerve Tissue Proteins/analysis , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Ovariectomy , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Phenols/chemistry , Phenols/pharmacology , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects , Rotarod Performance Test , Sex Factors , Substantia Nigra/chemistry , Substantia Nigra/drug effects , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/analysis
...