Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 5176, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890279

ABSTRACT

The longevity of grafts remains a major challenge in allogeneic transplantation due to immune rejection. Systemic immunosuppression can impair graft function and can also cause severe adverse effects. Here, we report a local immuno-protective strategy to enhance post-transplant persistence of allografts using a mesenchymal stem cell membrane-derived vesicle (MMV)-crosslinked hydrogel (MMV-Gel). MMVs are engineered to upregulate expression of Fas ligand (FasL) and programmed death ligand 1 (PD-L1). The MMVs are retained within the hydrogel by crosslinking. The immuno-protective microenvironment of the hydrogel protects allografts by presenting FasL and PD-L1. The binding of these ligands to T effector cells, the dominant contributors to graft destruction and rejection, results in apoptosis of T effector cells and generation of regulatory T cells. We demonstrate that implantation with MMV-Gel prolongs the survival and function of grafts in mouse models of allogeneic pancreatic islet cells and skin transplantation.


Subject(s)
Fas Ligand Protein , Hydrogels , Islets of Langerhans Transplantation , Mice, Inbred C57BL , Skin Transplantation , T-Lymphocytes, Regulatory , Transplantation, Homologous , Animals , Hydrogels/chemistry , Mice , Fas Ligand Protein/metabolism , Fas Ligand Protein/immunology , T-Lymphocytes, Regulatory/immunology , Islets of Langerhans Transplantation/methods , Skin Transplantation/methods , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mice, Inbred BALB C , Graft Survival/drug effects , Graft Survival/immunology , Graft Rejection/prevention & control , Graft Rejection/immunology , Humans , Male , Apoptosis/drug effects
2.
Sci Adv ; 10(11): eadk2444, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38478602

ABSTRACT

Cancer vaccines show huge potential for cancer prevention and treatment. However, their efficacy remains limited due to weak immunogenicity regarding inefficient stimulation of cytotoxic T lymphocyte (CTL) responses. Inspired by the unique characteristic and biological function of high-density lipoprotein (HDL), we here develop an HDL-mimicking nanovaccine with the commendable lymph-targeted capacity to potently elicit antitumor immunity using lipid nanoparticle that is co-loaded with specific cancer cytomembrane harboring a collection of tumor-associated antigens and an immune adjuvant. The nanoparticulate impact is explored on the efficiency of lymphatic targeting and dendritic cell uptake. The optimized nanovaccine promotes the co-delivery of antigens and adjuvants to lymph nodes and maintains antigen presentation of dendritic cells, resulting in long-term immune surveillance as the elevated frequency of CTLs within lymphoid organs and tumor tissue. Immunization of nanovaccine suppresses tumor formation and growth and augments the therapeutic efficacy of checkpoint inhibitors notably on the high-stemness melanoma in the mouse models.


Subject(s)
Melanoma , Nanoparticles , Neoplasms , Animals , Mice , Nanovaccines , T-Lymphocytes, Cytotoxic , Melanoma/pathology , Antigens, Neoplasm , Adjuvants, Immunologic/pharmacology , Immunotherapy/methods , Mice, Inbred C57BL
3.
Article in English | MEDLINE | ID: mdl-32194202

ABSTRACT

To determine whether (+)-catharanthine induces sedative- or anxiolytic/anxiogenic-like activity in male mice, proper animal paradigms were used. The results showed that (+)-catharanthine induces sedative-like activity in the 63-72 mg/Kg dose range in a flumazenil-insensitive manner, but neither this effect nor anxiolytic/anxiogenic-like activity was observed at lower doses. To determine the underlying molecular mechanism of the sedative-like activity, electrophysiological and radioligand binding experiments were performed with (+)-catharanthine and (±)-18-methoxycoronaridine [(±)-18-MC] on GABAA (GABAARs) and glycine receptors (GlyRs). Coronaridine congeners both activated and potentiated a variety of human (h) GABAARs, except hρ1. (+)-Catharanthine-induced potentiation followed this receptor selectivity (EC50's in µM): hα1ß2 (4.6 ± 0.8) > hα2ß2γ2 (12.6 ± 3.8) ~ hα1ß2γ2 (14.4 ± 4.6) indicating that both α1 and α2 are equally important, whereas γ2 is not necessary. (+)-Catharanthine was >2-fold more potent and efficient than (±)-18-MC at hα1ß2γ2. (+)-Catharanthine also potentiated, whereas (±)-18-MC inhibited, hα1 GlyRs with very low potency. Additional [3H]-flunitrazepam competition binding experiments using rat cerebellum membranes clearly demonstrated that these ligands do not bind to the benzodiazepine site. This is supported by the observed activity at hα1ß2 (lacking the BDZ site) and similar effects between α1- and α2-containing GABAARs. Our study shows, for the first time, that (+)-catharanthine induced sedative-like effects in mice, and coronaridine congeners potentiated human α1ß2γ2, α1ß2, and hα2ß2γ2, but not ρ1, GABAARs, both in a benzodiazepine-insensitive fashion, whereas only (+)-catharanthine slightly potentiated GlyRs.


Subject(s)
Benzodiazepines/metabolism , Hypnotics and Sedatives/metabolism , Ibogaine/analogs & derivatives , Ibogaine/metabolism , Receptors, GABA-A/metabolism , Animals , Benzodiazepines/pharmacology , Dose-Response Relationship, Drug , GABA-A Receptor Agonists/metabolism , GABA-A Receptor Agonists/pharmacology , HEK293 Cells , Humans , Hypnotics and Sedatives/pharmacology , Ibogaine/pharmacology , Locomotion/drug effects , Locomotion/physiology , Male , Maze Learning/physiology , Mice
4.
Behav Brain Res ; 378: 112278, 2020 01 27.
Article in English | MEDLINE | ID: mdl-31629836

ABSTRACT

Stroke leads to devastating outcomes including impairments of sensorimotor and cognitive function that may be long lasting. New intervention strategies are needed to overcome the long-lasting effects of ischemic injury. Previous studies determined that treatment with 5-methoxyindole-2-carboxylic acid (MICA) conferred chemical preconditioning and neuroprotection against stroke. The purpose of the current study was to determine whether the preconditioning can lead to functional improvements after stroke (done by transient middle cerebral artery occlusion). After 4 weeks of MICA feeding, half the rats underwent ischemic injury, while the other half remained intact. After one week recovery, all the rats were tested for motor and cognitive function (rotorod and water maze). At the time of euthanasia, measurements of long-term potentiation (LTP) were performed. While stroke injury led to motor and cognitive dysfunction, MICA supplementation did not reverse these impairments. However, MICA supplementation did improve stroke-related impairments in hippocampal LTP. The dichotomy of the outcomes suggest that more studies are needed to determine optimum duration and dosage for MICA to lead to substantial motor and cognitive improvements, along with LTP change and neuroprotection.


Subject(s)
Hippocampus/drug effects , Indoles/pharmacology , Ischemic Stroke/drug therapy , Ischemic Stroke/prevention & control , Ischemic Stroke/physiopathology , Long-Term Potentiation/drug effects , Maze Learning/drug effects , Neuroprotective Agents/pharmacology , Psychomotor Performance/drug effects , Recovery of Function/drug effects , Animals , Behavior, Animal/drug effects , Dietary Supplements , Dihydrolipoamide Dehydrogenase/drug effects , Disease Models, Animal , Indoles/administration & dosage , Infarction, Middle Cerebral Artery/complications , Ischemic Stroke/etiology , Male , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley
5.
Nat Commun ; 9(1): 2082, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29802295

ABSTRACT

Acid-sensing ion channels (ASICs) evolved to sense changes in extracellular acidity with the divalent cation calcium (Ca2+) as an allosteric modulator and channel blocker. The channel-blocking activity is most apparent in ASIC3, as removing Ca2+ results in channel opening, with the site's location remaining unresolved. Here we show that a ring of rat ASIC3 (rASIC3) glutamates (Glu435), located above the channel gate, modulates proton sensitivity and contributes to the formation of the elusive Ca2+ block site. Mutation of this residue to glycine, the equivalent residue in chicken ASIC1, diminished the rASIC3 Ca2+ block effect. Atomistic molecular dynamic simulations corroborate the involvement of this acidic residue in forming a high-affinity Ca2+ site atop the channel pore. Furthermore, the reported observations provide clarity for past controversies regarding ASIC channel gating. Our findings enhance understanding of ASIC gating mechanisms and provide structural and energetic insights into this unique calcium-binding site.


Subject(s)
Acid Sensing Ion Channels/chemistry , Binding Sites/physiology , Calcium/metabolism , Ion Channel Gating/physiology , Acid Sensing Ion Channels/genetics , Acid Sensing Ion Channels/metabolism , Animals , CHO Cells , Cations, Divalent/metabolism , Cricetulus , Glutamic Acid/genetics , Glutamic Acid/metabolism , Glycine/genetics , Glycine/metabolism , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Domains/physiology , Structure-Activity Relationship
6.
Neuropharmacology ; 133: 171-180, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29378213

ABSTRACT

This study aimed to address the mechanisms and reinforcing effects of three synthetic cathinone analogs of MDMA commonly reported in "Ecstasy" formulations: methylone, butylone, and pentylone. Whole-cell patch clamp techniques were used to assess the mechanism of each compound at the dopamine and serotonin transporters. Separate groups of rats were trained to discriminate methamphetamine, DOM, or MDMA from vehicle. Substitution studies were performed in each group and antagonism studies with SCH23390 were performed against each compound that produced substitution. Self-administration of each compound was evaluated under a progressive ratio schedule of reinforcement. Each compound produced an inward current at the serotonin transporter, but little or no current at the dopamine transporter. Each of the test compounds substituted fully for the discriminative stimulus effects of methamphetamine, methylone and butylone substituted partially for DOM and fully for MDMA, whereas pentylone failed to substitute for DOM and substituted only partially for MDMA. SCH23390 fully and dose-dependently attenuated methamphetamine-appropriate responding produced by each test compound, but was least potent against pentylone. MDMA-appropriate responding was minimally affected by SCH23390. Each test compound was robustly self-administered with pentylone producing the greatest self-administration at the doses tested. Given the prevalence of synthetic cathinones in "Ecstasy" formulations, these data indicate that adulterated "Ecstasy" formulations may drive more compulsive drug use than those containing only MDMA.


Subject(s)
Hallucinogens/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Reinforcement, Psychology , Substance-Related Disorders/physiopathology , Animals , Benzazepines/pharmacology , Central Nervous System Stimulants/pharmacology , Conditioning, Operant/drug effects , Disease Models, Animal , Dopamine Antagonists/pharmacology , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Drug Combinations , HEK293 Cells , Humans , Male , Methamphetamine/analogs & derivatives , Methamphetamine/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/analogs & derivatives , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Self Administration , Serotonin/pharmacology , Substance-Related Disorders/etiology
7.
Neuropharmacology ; 121: 167-178, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28456686

ABSTRACT

Efavirenz is a widely prescribed medicine used to treat type 1 human immunodeficiency virus (HIV-1), the most prevalent pathogenic strain of the virus responsible for the acquired immune deficiency syndrome (AIDS) pandemic. Under prescribed dosing conditions, either alone or in combination therapy, efavirenz-induced CNS disturbances are frequently reported. Efavirenz was recently reported to interact in a similar concentration range with a number of receptors, transporters and ion channels including recombinant rat α1ß2γ2 GABAA receptors whose actions were potentiated (Gatch et al., 2013; Dalwadi et al., 2016). Now we report on the molecular mechanism of efavirenz on GABAA receptors as a function of concentration and subunit composition via whole-cell recordings of GABA-activated currents from HEK293 cells expressing varying subunit configurations of GABAA receptors. Efavirenz elicited dual effects on the GABA response; it allosterically potentiated currents at low concentrations, whereas it inhibited currents at higher concentrations. The allosteric potentiating action on GABAA receptors was pronounced in the α1ß2γ2, α2ß2γ2 and α4ß2γ2 configurations, greatly diminished in the α6ß2γ2 configuration, and completely absent in the α3ß2γ2 or α5ß2γ2 configuration. In stark contrast, the inhibitory modulation of efavirenz at higher concentrations was evident in all subunit configurations examined. Moreover, efavirenz-induced modulatory effects were dependent on GABA concentration ([GABA]), with a pronounced impact on currents activated by low [GABA] but little effect at saturating [GABA]. Mutation of a highly-conserved threonine to phenylalanine in transmembrane domain 2 of the α1 subunit abolished the inhibitory effect of efavirenz in α1ß2 receptors. Finally, mutations of any of the three conserved extracellular residues in α1/2/4 subunits to the conserved residues at the corresponding positions in α3/5 subunits (i.e., R84P, M89L or I120L) completely eliminated the potentiating effect of efavirenz in α1ß2γ2 configuration. These findings demonstrate that efavirenz's positive allosteric modulation of the GABAA receptor is mediated via a novel allosteric site associated with the extracellular domain of the receptor.


Subject(s)
Benzoxazines/pharmacology , Receptors, GABA-A/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Alkynes , Allosteric Regulation , Animals , Cyclopropanes , Diazepam/pharmacology , Dose-Response Relationship, Drug , Flumazenil/pharmacology , GABA Modulators/pharmacology , HEK293 Cells , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mutagenesis/genetics , Patch-Clamp Techniques , Protein Domains/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Receptors, GABA-A/genetics , Transfection , gamma-Aminobutyric Acid/pharmacology
8.
Neuropharmacology ; 119: 100-110, 2017 06.
Article in English | MEDLINE | ID: mdl-28390894

ABSTRACT

Methylene blue (MB) is commonly used in diagnostic procedures and is also used to treat various medical conditions. Neurological effects of MB have been reported in clinical observations and experimental studies. Thus the modulation of GABAA receptor function by MB was investigated. Whole-cell GABA-activated currents were recorded from HEK293 cells expressing various GABAA receptor subunit configurations. MB inhibition of GABA currents was apparent at 3 µM, and it had an IC50 of 31 µM in human α1ß2γ2 receptors. The MB action was rapid and reversible. MB inhibition was not mediated via the picrotoxin site, as a mutation (T6'F of the ß2 subunit) known to confer resistance to picrotoxin had no effect on MB-induced inhibition. Blockade of GABAA receptors by MB was demonstrated across a range of receptors expressing varying subunits, including those expressed at extrasynaptic sites. The sensitivity of α1ß2 receptors to MB was similar to that observed in α1ß2γ2 receptors, indicating that MB's action via the benzodiazepine or Zn2+ site is unlikely. MB-induced inhibition of GABA response was competitive with respect to GABA. Furthermore, mutation of α1 F64 to A and ß2 Y205 to F in the extracellular N-terminus, both residues which are known to comprise GABA binding pocket, remarkably diminished MB inhibition of GABA currents. These data suggest that MB inhibits GABAA receptor function by direct or allosteric interaction with the GABA binding site. Finally, in mouse hippocampal CA1 pyramidal neurons, MB inhibited GABA-activated currents as well as GABAergic IPSCs. We demonstrate that MB directly inhibits GABAA receptor function, which may underlie some of the effects of MB on the CNS.


Subject(s)
Binding Sites/drug effects , Enzyme Inhibitors/pharmacology , Methylene Blue/pharmacology , Neurons/drug effects , Receptors, GABA-A/metabolism , Animals , Animals, Newborn , Binding Sites/genetics , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , HEK293 Cells , Hippocampus/cytology , Humans , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Protein Binding/drug effects , Rats , Receptors, GABA-A/genetics , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
9.
Pharmacol Res ; 110: 10-24, 2016 08.
Article in English | MEDLINE | ID: mdl-27157251

ABSTRACT

Efavirenz is highly effective at suppressing HIV-1, and the WHO guidelines list it as a component of the first-line antiretroviral (ARV) therapies for treatment-naïve patients. Though the pharmacological basis is unclear, efavirenz is commonly associated with a risk for neuropsychiatric adverse events (NPAEs) when taken at the prescribed dose. In many patients these NPAEs appear to subside after several weeks of treatment, though long-term studies show that in some patients the NPAEs persist. In a recent study focusing on the abuse potential of efavirenz, its receptor psychopharmacology was reported to include interactions with a number of established molecular targets for known drugs of abuse, and it displayed a prevailing behavioral profile in rodents resembling an LSD-like activity. In this report, we discovered interactions with additional serotonergic targets that may be associated with efavirenz-induced NPAEs. The most robust interactions were with 5-HT3A and 5-HT6 receptors, with more modest interactions noted for the 5-HT2B receptor and monoamine oxidase A. From a molecular mechanistic perspective, efavirenz acts as a 5-HT6 receptor inverse agonist of Gs-signaling, 5-HT2A and 5-HT2C antagonist of Gq-signaling, and a blocker of the 5-HT3A receptor currents. Efavirenz also completely or partially blocks agonist stimulation of the M1 and M3 muscarinic receptors, respectively. Schild analysis suggests that efavirenz competes for the same site on the 5-HT2A receptor as two known hallucinogenic partial agonists (±)-DOI and LSD. Prolonged exposure to efavirenz reduces 5-HT2A receptor density and responsiveness to 5-HT. Other ARVs such as zidovudine, nevirapine and emtricitabine did not share the same complex pharmacological profile as efavirenz, though some of them weakly interact with the 5-HT6 receptor or modestly block GABAA currents.


Subject(s)
Anti-HIV Agents/toxicity , Benzoxazines/toxicity , Brain/drug effects , HIV Infections/drug therapy , HIV-1/drug effects , Receptors, Serotonin/drug effects , Reverse Transcriptase Inhibitors/toxicity , Serotonin Antagonists/toxicity , Alkynes , Animals , Anti-HIV Agents/metabolism , Benzoxazines/metabolism , Binding, Competitive , Brain/metabolism , CHO Cells , Calcium Signaling/drug effects , Cricetulus , Cyclopropanes , Dose-Response Relationship, Drug , Drug Partial Agonism , Guinea Pigs , HEK293 Cells , HIV Infections/diagnosis , HIV Infections/virology , HIV-1/pathogenicity , HeLa Cells , Humans , Membrane Potentials , Monoamine Oxidase Inhibitors/toxicity , Protein Binding , Radioligand Assay , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism , Receptors, Serotonin/genetics , Receptors, Serotonin/metabolism , Reverse Transcriptase Inhibitors/metabolism , Time Factors , Transfection
10.
Endocrinology ; 157(5): 2067-79, 2016 05.
Article in English | MEDLINE | ID: mdl-26990062

ABSTRACT

Progesterone (P4) exerts robust cytoprotection in brain slice cultures (containing both neurons and glia), yet such protection is not as evident in neuron-enriched cultures, suggesting that glia may play an indispensable role in P4's neuroprotection. We previously reported that a membrane-associated P4 receptor, P4 receptor membrane component 1, mediates P4-induced brain-derived neurotrophic factor (BDNF) release from glia. Here, we sought to determine whether glia are required for P4's neuroprotection and whether glia's roles are mediated, at least partially, via releasing soluble factors to act on neighboring neurons. Our data demonstrate that P4 increased the level of mature BDNF (neuroprotective) while decreasing pro-BDNF (potentially neurotoxic) in the conditioned media (CMs) of cultured C6 astrocytes. We examined the effects of CMs derived from P4-treated astrocytes (P4-CMs) on 2 neuronal models: 1) all-trans retinoid acid-differentiated SH-SY5Y cells and 2) mouse primary hippocampal neurons. P4-CM increased synaptic marker expression and promoted neuronal survival against H2O2. These effects were attenuated by Y1036 (an inhibitor of neurotrophin receptor [tropomysin-related kinase] signaling), as well as tropomysin-related kinase B-IgG (a more specific inhibitor to block BDNF signaling), which pointed to BDNF as the key protective component within P4-CM. These findings suggest that P4 may exert its maximal protection by triggering a glia-neuron cross talk, in which P4 promotes mature BDNF release from glia to enhance synaptogenesis as well as survival of neurons. This recognition of the importance of glia in mediating P4's neuroprotection may also inform the design of effective therapeutic methods for treating diseases wherein neuronal death and/or synaptic deficits are noted.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Membrane Proteins/metabolism , Neuroglia/metabolism , Neurons/metabolism , Progesterone/pharmacology , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Mice , Neuroglia/cytology , Neuroglia/drug effects , Neurons/cytology , Neurons/drug effects , Propionates/pharmacology , Thiazolidines/pharmacology
11.
Br J Pharmacol ; 172(10): 2519-31, 2015 May.
Article in English | MEDLINE | ID: mdl-25573298

ABSTRACT

BACKGROUND AND PURPOSE: Cognitive deficits in patients with Alzheimer's disease, Parkinson's disease, traumatic brain injury and stroke often involve alterations in cholinergic signalling. Currently available therapeutic drugs provide only symptomatic relief. Therefore, novel therapeutic strategies are needed to retard and/or arrest the progressive loss of memory. EXPERIMENTAL APPROACH: Scopolamine-induced memory impairment provides a rapid and reversible phenotypic screening paradigm for cognition enhancement drug discovery. Male C57BL/6J mice given scopolamine (1 mg·kg(-1) ) were used to evaluate the ability of LS-1-137, a novel sigma (σ1) receptor-selective agonist, to improve the cognitive deficits associated with muscarinic antagonist administration. KEY RESULTS: LS-1-137 is a high-affinity (Ki = 3.2 nM) σ1 receptor agonist that is 80-fold selective for σ1, compared with σ2 receptors. LS-1-137 binds with low affinity at D2-like (D2, D3 and D4) dopamine and muscarinic receptors. LS-1-137 was found to partially reverse the learning deficits associated with scopolamine administration using a water maze test and an active avoidance task. LS-1-137 treatment was also found to trigger the release of brain-derived neurotrophic factor from rat astrocytes. CONCLUSIONS AND IMPLICATIONS: The σ1 receptor-selective compound LS-1-137 may represent a novel candidate cognitive enhancer for the treatment of muscarinic receptor-dependent cognitive deficits.


Subject(s)
Acetanilides/pharmacology , Acetanilides/therapeutic use , Cognition Disorders/chemically induced , Cognition Disorders/drug therapy , Muscarinic Antagonists/pharmacology , Piperidines/pharmacology , Piperidines/therapeutic use , Receptors, sigma/agonists , Receptors, sigma/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Avoidance Learning/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Ligands , Male , Maze Learning/drug effects , Mice , Rats , Scopolamine/pharmacology , Sigma-1 Receptor
12.
Pharmacology ; 92(1-2): 84-9, 2013.
Article in English | MEDLINE | ID: mdl-23942137

ABSTRACT

SV 293 [1-([5-methoxy-1H-indol-3-yl]methyl)-4-(4-[methylthio]​phenyl)piperidin-4-ol] binds with 100-fold higher affinity to human D2 receptors compared to the human D3 and D4 dopamine receptor subtypes. We investigated the intrinsic efficacy of this compound at the D2 dopamine receptor subtype using both: (1) a forskolin-dependent adenylyl cyclase inhibition assay and (2) an electrophysiological assay for evaluating coupling to G-protein-coupled inwardly rectifying potassium channels. In both assays SV 293 was found to be a neutral antagonist capable of blocking the effects of the full D2-like receptor agonist quinpirole. Based upon these results we propose that SV 293 is a useful pharmacological tool that can be used for both in vitro and in vivo studies to investigate the role of D2-like dopamine receptor subtypes in neurological, neuropsychiatric and movement disorders where dopaminergic pathways have been implicated.


Subject(s)
Dopamine Antagonists/pharmacology , Dopamine D2 Receptor Antagonists , Indoles/pharmacology , Piperidines/pharmacology , Adenylyl Cyclase Inhibitors , Animals , Cell Line, Tumor , Colforsin/pharmacology , Dopamine Agonists/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Humans , Mice , Quinpirole/pharmacology , Receptors, Dopamine D2/physiology
13.
Neurobiol Dis ; 59: 18-25, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23845275

ABSTRACT

Vascular dementia ranks as the second leading cause of dementia in the United States. However, its underlying pathophysiological mechanism is not fully understood and no effective treatment is available. The purpose of the current study was to evaluate long-term cognitive deficits induced by transient middle cerebral artery occlusion (tMCAO) in rats and to investigate the underlying mechanism. Sprague-Dawley rats were subjected to tMCAO or sham surgery. Behavior tests for locomotor activity and cognitive function were conducted at 7 or 30days after stroke. Hippocampal long term potentiation (LTP) and involvement of GABAergic neurotransmission were evaluated at 30days after sham surgery or stroke. Immunohistochemistry and Western blot analyses were conducted to determine the effect of tMCAO on cell signaling in the hippocampus. Transient MCAO induced a progressive deficiency in spatial performance. At 30days after stroke, no neuron loss or synaptic marker change in the hippocampus were observed. LTP in both hippocampi was reduced at 30days after stroke. This LTP impairment was prevented by blocking GABAA receptors. In addition, ERK activity was significantly reduced in both hippocampi. In summary, we identified a progressive decline in spatial learning and memory after ischemic stroke that correlates with suppression of hippocampal LTP, elevation of GABAergic neurotransmission, and inhibition of ERK activation. Our results indicate that the attenuation of GABAergic activity or enhancement of ERK/MAPK activation in the hippocampus might be potential therapeutic approaches to prevent or attenuate cognitive impairment after ischemic stroke.


Subject(s)
Cognition Disorders/etiology , Gene Expression Regulation/physiology , Infarction, Middle Cerebral Artery/complications , Signal Transduction/physiology , Animals , Central Nervous System Stimulants/pharmacology , Cognition Disorders/pathology , Disease Models, Animal , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Functional Laterality , Hippocampus/physiopathology , In Vitro Techniques , Male , Maze Learning/physiology , Membrane Proteins/metabolism , Motor Activity/physiology , Nerve Tissue Proteins/metabolism , Picrotoxin/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors , Visual Perception/physiology
14.
Neuropsychopharmacology ; 38(12): 2373-84, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23702798

ABSTRACT

Anecdotal reports have surfaced concerning misuse of the HIV antiretroviral medication efavirenz ((4S)-6-chloro-4-(2-cyclopropylethynyl)-4-(trifluoromethyl)-2,4-dihydro-1H-3,1-benzoxazin-2-one) by HIV patients and non-infected teens who crush the pills and smoke the powder for its psychoactive effects. Molecular profiling of the receptor pharmacology of efavirenz pinpointed interactions with multiple established sites of action for other known drugs of abuse including catecholamine and indolamine transporters, and GABAA and 5-HT(2A) receptors. In rodents, interaction with the 5-HT(2A) receptor, a primary site of action of lysergic acid diethylamine (LSD), appears to dominate efavirenz's behavioral profile. Both LSD and efavirenz reduce ambulation in a novel open-field environment. Efavirenz occasions drug-lever responding in rats discriminating LSD from saline, and this effect is abolished by selective blockade of the 5-HT(2A) receptor. Similar to LSD, efavirenz induces head-twitch responses in wild-type, but not in 5-HT(2A)-knockout, mice. Despite having GABAA-potentiating effects (like benzodiazepines and barbiturates), and interactions with dopamine transporter, serotonin transporter, and vesicular monoamine transporter 2 (like cocaine and methamphetamine), efavirenz fails to maintain responding in rats that self-administer cocaine, and it fails to produce a conditioned place preference. Although its molecular pharmacology is multifarious, efavirenz's prevailing behavioral effect in rodents is consistent with LSD-like activity mediated via the 5-HT(2A) receptor. This finding correlates, in part, with the subjective experiences in humans who abuse efavirenz and with specific dose-dependent adverse neuropsychiatric events, such as hallucinations and night terrors, reported by HIV patients taking it as a medication.


Subject(s)
Anti-HIV Agents/toxicity , Benzoxazines/toxicity , Hallucinogens/toxicity , Lysergic Acid Diethylamide/toxicity , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin 5-HT2 Receptor Agonists/metabolism , Alkynes , Animals , Behavior, Animal/drug effects , Conditioning, Psychological/drug effects , Cyclopropanes , Discrimination, Psychological , Male , Mice , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/metabolism
15.
ACS Chem Neurosci ; 3(12): 1050-62, 2012 Dec 19.
Article in English | MEDLINE | ID: mdl-23259040

ABSTRACT

We previously reported on the synthesis of substituted phenyl-4-hydroxy-1-piperidyl indole analogues with nanomolar affinity at D2 dopamine receptors, ranging from 10- to 100-fold selective for D2 compared to the D3 dopamine receptor subtype. More recently, we evaluated a panel of aripiprazole analogues, identifying several analogues that also exhibit D2 vs D3 dopamine receptor binding selectivity. These studies further characterize the intrinsic efficacy of the compound with the greatest binding selectivity from each chemical class, 1-((5-methoxy-1H-indol-3-yl)methyl)-4-(4-(methylthio)phenyl)piperidin-4-ol (SV 293) and 7-(4-(4-(2-methoxyphenyl)piperazin-1-yl)butoxy)-3,4-dihydroquinolin-2(1H)-one (SV-III-130s), using an adenylyl cyclase inhibition assay, a G-protein-coupled inward-rectifying potassium (GIRK) channel activation assay, and a cell based phospho-MAPK (pERK1/2) assay. SV 293 was found to be a neutral antagonist at D2 dopamine receptors using all three assays. SV-III-130s is a partial agonist using an adenylyl cyclase inhibition assay but an antagonist in the GIRK and phospho ERK1/2 assays. To define the molecular basis for the binding selectivity, the affinity of these two compounds was evaluated using (a) wild type human D2 and D3 receptors and (b) a panel of chimeric D2/D3 dopamine receptors. Computer-assisted modeling techniques were used to dock these compounds to the human D2 and D3 dopamine receptor subtypes. It is hoped that these studies on D2 receptor selective ligands will be useful in the future design of (a) receptor selective ligands used to define the function of D2-like receptor subtypes, (b) novel pharmacotherapeutic agents, and/or (c) in vitro and in vivo imaging agents.


Subject(s)
Dopamine Antagonists/chemical synthesis , Dopamine D2 Receptor Antagonists , Receptors, Dopamine D3/antagonists & inhibitors , Dopamine Antagonists/pharmacology , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Transfection
16.
Neuropharmacology ; 58(8): 1246-51, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20303997

ABSTRACT

The protein tyrosine kinase (PTK) inhibitor genistein has been widely used to examine potential effects of tyrosine phosphorylation on neurotransmitter function. We report here that genistein inhibits N-methyl-d-aspartate (NMDA) receptors through a direct effect. Whole-cell NMDA-activated current was recorded in native receptors from mouse hippocampal slice culture and rat recombinant NR1aNR2A and NR1aNR2B receptors transiently expressed in HEK293 cells. Extracellular application of genistein and NMDA reversibly inhibited NMDA-activated current. The inhibition of NMDA-activated current by genistein applied externally was not affected when genistein was also pre-equilibrated in the intracellular solution. Daidzein, an analog of genistein that does not block PTK, also inhibited NMDA-activated current. Coapplication of lavendustin A, a specific inhibitor of PTK, had no effect on the NMDA response. Moreover, genistein-induced inhibition of NMDA-activated current displayed concentration- and voltage-dependence. Our results demonstrate that genistein has a direct inhibitory effect on NMDA receptors that is not mediated via inhibition of tyrosine kinase. Thus, other PTK inhibitors may be more suitable for studying involvement of PTKs in NMDA receptor-mediated events.


Subject(s)
Genistein/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Animals, Newborn , Hippocampus/drug effects , Hippocampus/physiology , In Vitro Techniques , Ion Channel Gating , Isoflavones/pharmacology , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Phenols/pharmacology , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics
17.
Proc Natl Acad Sci U S A ; 105(39): 15148-53, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18815371

ABSTRACT

L-type voltage-gated Ca(2+)channels (VGCC) play an important role in dendritic development, neuronal survival, and synaptic plasticity. Recent studies have demonstrated that the gonadal steroid estrogen rapidly induces Ca(2+) influx in hippocampal neurons, which is required for neuroprotection and potentiation of LTP. The mechanism by which estrogen rapidly induces this Ca(2+) influx is not clearly understood. We show by electrophysiological studies that extremely low concentrations of estrogens acutely potentiate VGCC in hippocampal neurons, hippocampal slices, and HEK-293 cells transfected with neuronal L-type VGCC, in a manner that was estrogen receptor (ER)-independent. Equilibrium, competitive, and whole-cell binding assays indicate that estrogen directly interacts with the VGCC. Furthermore, a L-type VGCC antagonist to the dihydropyridine site displaced estrogen binding to neuronal membranes, and the effects of estrogen were markedly attenuated in a mutant, dihydropyridine-insensitive L-type VGCC, demonstrating a direct interaction of estrogens with L-type VGCC. Thus, estrogen-induced potentiation of calcium influx via L-type VGCC may link electrical events with rapid intracellular signaling seen with estrogen exposure leading to modulation of synaptic plasticity, neuroprotection, and memory formation.


Subject(s)
Calcium Channels, L-Type/metabolism , Estrogens/metabolism , Neurons/metabolism , Animals , Calcium Channels, L-Type/genetics , Cell Line , Estradiol/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Humans , Mutation , Neurons/drug effects , Rats , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
18.
Invest Ophthalmol Vis Sci ; 49(11): 4993-5002, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18641291

ABSTRACT

PURPOSE: The sigma-1 receptor belongs to a recently discovered family of transmembrane proteins expressed in the central nervous system, including the eye, and mediates the regulation of ion channels. The exact function of sigma receptors remains to be elucidated. The purpose of this study was to investigate the effect of sigma-1 receptor ligands on calcium homeostasis in a retinal ganglion cell line (RGC)-5 and rat primary RGCs. METHODS: Calcium imaging was used to assess the effect of sigma-1 receptor agonist (+)-N-allylnormetazocine ((+)-SKF10047) on potassium chloride (KCl)-induced calcium influx in RGC-5. The whole-cell patch clamp technique was used to analyze the effect of (+)-SKF10047 on calcium currents in primary RGCs. Coimmunoprecipitation assessed the interaction between the sigma-1 receptor and the L-type voltage-gated calcium channel. RESULTS: The sigma-1 receptor agonist (+)-SKF10047 inhibited potassium chloride (KCl)-induced calcium influx. The sigma-1 receptor antagonist, BD1047, reversed the inhibitory effect of (+)-SKF10047. Whole-cell patch clamp recordings of rat cultured primary RGCs demonstrated that (+)-SKF10047 inhibited calcium currents. Coimmunoprecipitation studies demonstrated an association between L-type calcium channels and the sigma-1 receptors. CONCLUSIONS: These results suggest that sigma-1 receptor activation can regulate calcium homeostasis and signaling in RGCs, likely by directly influencing the activity of L-type voltage-gated calcium channels. Regulation of calcium influx in RGCs by sigma-1 receptor ligands may represent in part the neuroprotective effect of sigma-1 receptors.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Receptors, sigma/biosynthesis , Retinal Ganglion Cells/metabolism , Animals , Blotting, Western , Calcium Channels, L-Type/drug effects , Cells, Cultured , DNA/genetics , Ethylenediamines/pharmacology , Gene Expression , Intracellular Fluid/metabolism , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Microscopy, Fluorescence , Molecular Sequence Data , Patch-Clamp Techniques , Phenazocine/analogs & derivatives , Phenazocine/pharmacology , Potassium Chloride/pharmacology , Rats , Receptors, sigma/antagonists & inhibitors , Receptors, sigma/drug effects , Receptors, sigma/genetics , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Signal Transduction/drug effects , Sigma-1 Receptor
19.
Neurosci Lett ; 431(2): 184-9, 2008 Jan 31.
Article in English | MEDLINE | ID: mdl-18162311

ABSTRACT

The presence of phenylalanine (F) at the 6' position of transmembrane domain 2 (TM2) in the alpha4 subunit of alpha4beta2 nicotinic receptors enhances desensitization. As the GABA A receptor affords the ability to study the influence of as few as one and as many as five Fs at this position, we have used it to investigate potential subunit- and stoichiometry-dependent effects of the TM2 6'F mutation on desensitization. Whereas the presence of one F at this position decreased extent of desensitization, desensitization was increased in all configurations that included two or more Fs at the TM2 6' position; desensitization was particularly rapid with 3 or 4 F residues present. Our results demonstrate the ability of F residues at the TM2 6' position to modulate desensitization is likely conserved in the cys-loop family of ligand-gated ion channels. Moreover, our findings demonstrate both stoichiometric- and subunit-dependent effects of the ability of this mutation to regulate desensitization in GABA A receptors.


Subject(s)
Mutation/physiology , Phenylalanine/genetics , Receptors, GABA-A/physiology , Stochastic Processes , Amino Acid Sequence , Animals , Cell Line, Transformed , Electric Stimulation/methods , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Patch-Clamp Techniques/methods , Protein Structure, Tertiary/physiology , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Receptors, GABA-A/genetics , Transfection/methods , gamma-Aminobutyric Acid/pharmacology
20.
Biochemistry ; 46(41): 11484-93, 2007 Oct 16.
Article in English | MEDLINE | ID: mdl-17887775

ABSTRACT

Little is known regarding the mechanism(s) by which glycine receptors are endocytosed. Here we examined the endocytosis of homomeric alpha1 glycine receptors expressed in HEK 293 cells using immunofluorescence/confocal microscopy and whole-cell patch-clamp recordings. Our studies demonstrate that constitutive endocytosis of glycine receptors is blocked by the dominant negative dynamin construct K44A and that intracellular dialysis with peptide P4, a dynamin/amphiphysin-disrupting peptide, increased whole-cell glycine-gated chloride currents. To examine whether receptor endocytosis could be regulated by PKC, experiments with the PKC activator PMA (phorbol 12-myristate 13-acetate) were performed. PMA, but not its inactive analogue PMM (phorbol 12-monomyristate), stimulated receptor endocytosis and inhibited glycine-gated chloride currents. Similar to constitutive endocytosis, PKC-stimulated endocytosis was blocked by dynamin K44A. Mutation of a putative AP2 adaptin dileucine motif (L314A, L315A) present in the receptor cytoplasmic loop blocked PMA-stimulated receptor endocytosis and also prevented PMA inhibition of glycine receptor currents. In patch-clamp experiments, intracellular dialysis of a 12-amino acid peptide corresponding to the region of the receptor containing the dileucine motif prevented PKC modulation of wild-type glycine receptors. Unlike PKC modulation of the receptor, constitutive endocytosis was not affected by mutation of this dileucine motif. These results demonstrate that PKC activation stimulates glycine receptor endocytosis, that both constitutive endocytosis and PKC-stimulated endocytosis are dynamin-dependent, and that PKC-stimulated endocytosis, but not constitutive endocytosis, occurs via the dileucine motif (L314A, L315A) within the cytoplasmic loop of the receptor.


Subject(s)
Endocytosis/physiology , Receptors, Glycine/physiology , Cell Line , Dynamins/physiology , Humans , Kinetics , Microscopy, Confocal , Patch-Clamp Techniques , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...