Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 200
Filter
1.
Mar Drugs ; 20(2)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35200675

ABSTRACT

α6ß4 nAChR is expressed in the peripheral and central nervous systems and is associated with pain, addiction, and movement disorders. Natural α-conotoxins (α-CTxs) can effectively block different nAChR subtypes with higher efficacy and selectivity. However, the research on α6ß4 nAChR is relatively poor, partly because of the lack of available target-specific α-CTxs. In this study, we synthesized a novel α-4/7 conotoxin QuIA that was found from Conus quercinus. We investigated the efficacy of this peptide to different nAChR subtypes using a two-electrode voltage-clamp technique. Remarkably, we found α-QuIA inhibited the neuronal α3ß2 and α6/α3ß4 nAChR subtypes with significantly high affinity (IC50 was 55.7 nM and 90.68 nM, respectively), and did not block other nAChR subtypes even at a high concentration of 10 µM. In contrast, most α-CTxs have been determined so far to effectively block the α6/α3ß4 nAChR subtype while also maintaining a similar higher efficacy against the closely related α6ß2ß3 and/or α3ß4 subtypes, which are different from QuIA. In conclusion, α-QuIA is a novel α4/7-CTx, which has the potential to develop as an effective neuropharmacology tool to detect the function of α6ß4 nAChR.


Subject(s)
Conotoxins/pharmacology , Conus Snail/metabolism , Nicotinic Antagonists/pharmacology , Animals , Conotoxins/administration & dosage , Conotoxins/isolation & purification , Humans , Inhibitory Concentration 50 , Mice , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/isolation & purification , Patch-Clamp Techniques , Rats , Receptors, Nicotinic/drug effects , Xenopus laevis
2.
J Neuroinflammation ; 18(1): 291, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34920740

ABSTRACT

BACKGROUND: Stress-induced neuroinflammation was considered to play a critical role in the pathogenesis of depression. Transcutaneous auricular vagus nerve stimulation (taVNS) is a relatively non-invasive alternative treatment for patients suffering from major depressive disorder. The anti-inflammatory signal of vagus nerve is mediated by α7 nicotinic acetylcholine receptor (α7nAchR), and the hippocampus, the region with the most distribution of α7nAchR, regulates emotions. Here, we investigated the role of α7nAchR mediating hippocampal neuroinflammation in taVNS antidepressant effect though homozygous α7nAChR (-/-) gene knockout and α7nAchR antagonist (methyllycaconitine, MLA). METHODS: There were control, model, taVNS, α7nAChR(-/-) + taVNS, hippocampus (Hi) MLA + taVNS and Hi saline + taVNS groups. We used the chronic unpredicted mild stress (CUMS) method to establish depressive model rats for 42 days, excepting control group. After the successful modeling, except the control and model, the rats in the other groups were given taVNS, which was applied through an electroacupuncture apparatus at the auricular concha (2/15 Hz, 2 mA, 30 min/days) for 21 days. Behavioral tests were conducted at baseline, after modeling and after taVNS intervention, including sucrose preference test (SPT), open field test (OFT) and forced swimming test (FST). These tests are widely used to evaluate depression-like behavior in rats. The samples were taken after experiment, the expressions of α7nAchR, NF-κB p65, IL-1ß and the morphology of microglia were detected. RESULTS: Depression-like behavior and hippocampal neuroinflammation in CUMS model rats were manifested by down-regulated expression of α7nAchR, up-regulated expression of NF-κB p65 and IL-1ß, and the morphology of microglia was in amoebic-like activated state. TaVNS could significantly reverse the above-mentioned phenomena, but had rare improvement effect for α7nAChR(-/-) rats and Hi MLA rats. CONCLUSION: The antidepressant effect of taVNS is related to hippocampal α7nAchR/NF-κB signal pathway.


Subject(s)
Depressive Disorder, Major/metabolism , Hippocampus/metabolism , Stress, Psychological/metabolism , Transcription Factor RelA/metabolism , Vagus Nerve Stimulation/methods , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Chronic Disease , Depressive Disorder, Major/genetics , Depressive Disorder, Major/therapy , Gene Knockout Techniques/methods , Hippocampus/drug effects , Male , Nicotinic Antagonists/administration & dosage , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Stress, Psychological/genetics , Stress, Psychological/therapy , Transcription Factor RelA/genetics , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , alpha7 Nicotinic Acetylcholine Receptor/genetics
3.
Int Heart J ; 62(3): 607-615, 2021.
Article in English | MEDLINE | ID: mdl-34054001

ABSTRACT

The aim was to investigate the role of the α7nAChR-mediated cholinergic anti-inflammatory pathway in vagal nerve regulated atrial fibrillation (AF).18 beagles (standard dogs for testing) were used in this study, and the effective refractory period (ERP) of atrium and pulmonary veins and AF inducibility were measured hourly during rapid atrial pacing at 800 beats/minute for 6 hours in all beagles. After cessation of 3 hours of RAP, the low-level vagal nerve stimulation (LL-VNS) group (n = 6) was given LL-VNS and injection of salinne (0.5 mL/GP) into four GPs, the methyllycaconitine (MLA, the antagonist of α7nAChR) group (n = 6) was given LL-VNS and injection of MLA into four GPs, and the Control group (n = 6) was given saline into four GPs and the right cervical vagal nerve was exposed without stimulation. Then, the levels of the tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), acetylcholine (ACh), STAT3, and NF-κB proteins were measured. During the first 3 hours of RAP, the ERPs gradually decreased while the dispersion of ERPs (dERPs) and AF inducibility gradually increased in all three groups. During the last 3 hours of 6 hours' RAP in this study, the ERPs in the LL-VNS group were higher, while the dERPs and AF inducibility were significantly lower when compared with the Control and MLA groups at the same time points. The levels of ACh in the serum and atrium in the LL-VNS and MLA groups were higher than in the Control group, and the levels of TNF-α and IL-6 were higher in the Control and MLA groups than in the LL-VNS group. The concentrations of STAT3 in RA and LA tissues were higher in the LL-VNS group while those of NF-κB were lower.In conclusion, the cholinergic anti-inflammatory pathway mediated by α7nACh plays an important role in low-level vagal nerve-regulated AF.


Subject(s)
Aconitine/analogs & derivatives , Atrial Fibrillation/physiopathology , Neuroimmunomodulation/drug effects , Vagus Nerve/drug effects , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , Acetylcholine/blood , Aconitine/administration & dosage , Aconitine/pharmacology , Animals , Cardiac Pacing, Artificial/adverse effects , Cardiac Pacing, Artificial/methods , Case-Control Studies , Disease Models, Animal , Dogs , Heart Atria/innervation , Heart Atria/physiopathology , Interleukin-6/blood , NF-kappa B/blood , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/pharmacology , Pulmonary Veins/innervation , Pulmonary Veins/physiopathology , Refractory Period, Electrophysiological/drug effects , STAT3 Transcription Factor/blood , Tumor Necrosis Factor-alpha/blood , Vagus Nerve Stimulation/adverse effects , Vagus Nerve Stimulation/methods
4.
Nicotine Tob Res ; 22(2): 213-223, 2020 02 06.
Article in English | MEDLINE | ID: mdl-30958557

ABSTRACT

INTRODUCTION: Tobacco use improves mood states and smoking cessation leads to anhedonia, which contributes to relapse. Animal studies have shown that noncontingent nicotine administration enhances brain reward function and leads to dependence. However, little is known about the effects of nicotine self-administration on the state of the reward system. METHODS: To investigate the relationship between nicotine self-administration and reward function, rats were prepared with intracranial self-stimulation electrodes and intravenous catheters. The rats were trained on the intracranial self-stimulation procedure and allowed to self-administer 0.03 mg/kg/infusion of nicotine. All rats self-administered nicotine daily for 10 days (1 hour/day) and were then switched to an intermittent short access (ShA, 1 hour/day) or long access (LgA, 23 hour/day) schedule (2 days/week, 5 weeks). RESULTS: During the first 10 daily, 1-hour sessions, nicotine self-administration decreased the reward thresholds, which indicates that nicotine potentiates reward function. After switching to the intermittent LgA or ShA schedule, nicotine intake was lower in the ShA rats than the LgA rats. The LgA rats increased their nicotine intake over time and they gradually consumed a higher percentage of their nicotine during the light phase. The nicotinic acetylcholine receptor (nAChR) antagonist mecamylamine induced a larger increase in reward thresholds (ie, anhedonia) in the LgA rats than the ShA rats. In the LgA rats, nAChR blockade with mecamylamine decreased nicotine intake for 2 hours and this was followed by a rebound increase in nicotine intake. CONCLUSIONS: A brief period of nicotine self-administration enhances reward function and a high level of nicotine intake leads to dependence. IMPLICATIONS: These animal studies indicate that there is a strong relationship between the level of nicotine intake and brain reward function. A high level of nicotine intake was more rewarding than a low level of nicotine intake and nicotine dependence was observed after long, but not short, access to nicotine. This powerful combination of nicotine reward and withdrawal makes it difficult to quit smoking. Blockade of nAChRs temporarily decreased nicotine intake, but this was followed by a large rebound increase in nicotine intake. Therefore, nAChR blockade might not decrease the use of combustible cigarettes or electronic cigarettes.


Subject(s)
Anhedonia/drug effects , Nicotine/administration & dosage , Reward , Self Stimulation/drug effects , Anhedonia/physiology , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Electrodes, Implanted , Male , Mecamylamine/administration & dosage , Nicotinic Agonists/administration & dosage , Nicotinic Antagonists/administration & dosage , Rats , Rats, Wistar , Receptors, Nicotinic/physiology , Self Administration/methods , Self Stimulation/physiology , Time Factors , Tobacco Use Disorder/psychology
5.
Proc Natl Acad Sci U S A ; 114(10): E1825-E1832, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28223528

ABSTRACT

Opioids are first-line drugs for moderate to severe acute pain and cancer pain. However, these medications are associated with severe side effects, and whether they are efficacious in treatment of chronic nonmalignant pain remains controversial. Medications that act through alternative molecular mechanisms are critically needed. Antagonists of α9α10 nicotinic acetylcholine receptors (nAChRs) have been proposed as an important nonopioid mechanism based on studies demonstrating prevention of neuropathology after trauma-induced nerve injury. However, the key α9α10 ligands characterized to date are at least two orders of magnitude less potent on human vs. rodent nAChRs, limiting their translational application. Furthermore, an alternative proposal that these ligands achieve their beneficial effects by acting as agonists of GABAB receptors has caused confusion over whether blockade of α9α10 nAChRs is the fundamental underlying mechanism. To address these issues definitively, we developed RgIA4, a peptide that exhibits high potency for both human and rodent α9α10 nAChRs, and was at least 1,000-fold more selective for α9α10 nAChRs vs. all other molecular targets tested, including opioid and GABAB receptors. A daily s.c. dose of RgIA4 prevented chemotherapy-induced neuropathic pain in rats. In wild-type mice, oxaliplatin treatment produced cold allodynia that could be prevented by RgIA4. Additionally, in α9 KO mice, chemotherapy-induced development of cold allodynia was attenuated and the milder, temporary cold allodynia was not relieved by RgIA4. These findings establish blockade of α9-containing nAChRs as the basis for the efficacy of RgIA4, and that α9-containing nAChRs are a critical target for prevention of chronic cancer chemotherapy-induced neuropathic pain.


Subject(s)
Cancer Pain/drug therapy , Hyperalgesia/drug therapy , Peptides/administration & dosage , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Analgesics, Opioid/adverse effects , Animals , Cancer Pain/chemically induced , Cancer Pain/genetics , Cancer Pain/pathology , Humans , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Hyperalgesia/pathology , Ligands , Mice , Mice, Knockout , Neuralgia/chemically induced , Neuralgia/drug therapy , Neuralgia/genetics , Neuralgia/pathology , Nicotinic Antagonists/administration & dosage , Organoplatinum Compounds/adverse effects , Oxaliplatin , Receptors, GABA-B/genetics
6.
FASEB J ; 31(1): 192-202, 2017 01.
Article in English | MEDLINE | ID: mdl-27682206

ABSTRACT

Although α6-contaning (α6*) nicotinic acetylcholine receptors (nAChRs) are densely expressed in the visual system, their role is not well known. We have characterized a family of toxins that are antagonists for α6ß2* receptors and used one of these [RDP-MII(E11R)] to localize α6* nAChRs and investigate their impact on retinal function in adult Long-Evans rats. The α6*nAChRs in retinal tissue were localized using either a fluorescently tagged [RDP-MII(E11R)] or anti-α6-specific antibodies and found to be predominantly at the level of the ganglion cell layer. After intraocular injection of RDP-MII(E11R) in one eye and vehicle or inactive MII in contralateral eyes as controls, we recorded flash electroretinograms (F-ERGs), pattern ERGs (P-ERGs), and cortical visual-evoked potential (VEPs). There was no significant difference in F-ERG between the RDP-MII(E11R)-treated and control eyes. In contrast, P-ERG response amplitude was significantly reduced in the RDP-MII(E11R)-injected eye. Blocking α6* nAChRs at retinal level also decreased the VEP amplitude recorded in the visual cortex contralateral to the injected eye. Because both the cortical and inner retina output were affected by RDP-MII(E11R), whereas photoreceptor output was preserved, we conclude that the reduced visual response was due to an alteration in the function of α6* nAChRs present in the ganglion cell layer.-Barloscio, D., Cerri, E., Domenici, L., Longhi, R., Dallanoce, C., Moretti, M., Vilella, A., Zoli, M., Gotti, C., and Origlia, N. In vivo study of the role of α6-containing nicotinic acetylcholine receptor in retinal function using subtype-specific RDP-MII(E11R) toxin.


Subject(s)
Conotoxins/toxicity , Nicotinic Antagonists/toxicity , Receptors, Nicotinic/metabolism , Retina/physiology , Animals , Cerebral Cortex/physiology , Conotoxins/administration & dosage , Evoked Potentials, Visual/drug effects , Evoked Potentials, Visual/physiology , Male , Nicotinic Antagonists/administration & dosage , Rats , Rats, Long-Evans
7.
Addict Biol ; 23(5): 1046-1054, 2018 09.
Article in English | MEDLINE | ID: mdl-28940989

ABSTRACT

The aversive effect of nicotine withdrawal is greater in female versus male rats, and we postulate that this sex difference is mediated in the nucleus accumbens (NAc). Nicotine withdrawal induces decreases in NAc dopamine and increases in acetylcholine (ACh) levels in male rats. To our knowledge, these neurochemical markers of nicotine withdrawal have not been compared in female versus male rats. Given the role of amino acids in modulating NAc dopaminergic and cholinergic transmission, concomitant measures of gamma-aminobutyric acid (GABA) and glutamate levels were also compared across sex. Rats received continuous nicotine exposure for 14 days, and then NAc dialysate was collected during baseline and following administration of the nicotinic receptor antagonist mecamylamine to precipitate withdrawal. Chronic nicotine exposure was associated with larger increases in baseline dopamine, GABA and glutamate levels in the NAc of female versus male rats, whereas baseline ACh was only increased in male rats. During withdrawal, both sexes displayed equivalent increases in NAc ACh levels. As expected, male rats displayed decreases in dopamine, coupled with increases in GABA and decreases in glutamate levels, suggesting the possibility of increased inhibitory tone in the NAc during withdrawal. Relative to males, female rats displayed larger decreases in NAc dopamine and related increases in GABAergic transmission. As female rats also showed elevated glutamate levels that persist during withdrawal, it is suggested that sex differences may arise from increased glutamatergic drive of inhibitory tone in the NAc. The findings provide a potential mechanism whereby the aversive effects of nicotine withdrawal are enhanced in female rats.


Subject(s)
Dopamine/metabolism , Glutamic Acid/metabolism , Nucleus Accumbens/metabolism , Substance Withdrawal Syndrome/metabolism , Tobacco Use Disorder/metabolism , Amino Acids/metabolism , Animals , Disease Models, Animal , Female , Male , Mecamylamine/administration & dosage , Nicotine/metabolism , Nicotinic Antagonists/administration & dosage , Rats , Rats, Wistar , Sex Factors
8.
J Biol Chem ; 290(2): 1039-48, 2015 Jan 09.
Article in English | MEDLINE | ID: mdl-25411242

ABSTRACT

Activation of the α3ß4 nicotinic acetylcholine receptor (nAChR) subtype has recently been implicated in the pathophysiology of various conditions, including development and progression of lung cancer and in nicotine addiction. As selective α3ß4 nAChR antagonists, α-conotoxins are valuable tools to evaluate the functional roles of this receptor subtype. We previously reported the discovery of a new α4/7-conotoxin, RegIIA. RegIIA was isolated from Conus regius and inhibits acetylcholine (ACh)-evoked currents mediated by α3ß4, α3ß2, and α7 nAChR subtypes. The current study used alanine scanning mutagenesis to understand the selectivity profile of RegIIA at the α3ß4 nAChR subtype. [N11A] and [N12A] RegIIA analogs exhibited 3-fold more selectivity for the α3ß4 than the α3ß2 nAChR subtype. We also report synthesis of [N11A,N12A]RegIIA, a selective α3ß4 nAChR antagonist (IC50 of 370 nM) that could potentially be used in the treatment of lung cancer and nicotine addiction. Molecular dynamics simulations of RegIIA and [N11A,N12A]RegIIA bound to α3ß4 and α3ß2 suggest that destabilization of toxin contacts with residues at the principal and complementary faces of α3ß2 (α3-Tyr(92), Ser(149), Tyr(189), Cys(192), and Tyr(196); ß2-Trp(57), Arg(81), and Phe(119)) may form the molecular basis for the selectivity shift.


Subject(s)
Conotoxins/administration & dosage , Lung Neoplasms/drug therapy , Nicotinic Antagonists/administration & dosage , Receptors, Nicotinic/metabolism , Acetylcholine/metabolism , Alanine/chemistry , Alanine/isolation & purification , Animals , Conotoxins/chemical synthesis , Conotoxins/chemistry , Conus Snail/chemistry , Humans , Lung Neoplasms/metabolism , Molecular Dynamics Simulation , Mutagenesis , Nicotine/adverse effects , Nicotine/chemistry , Nicotine/metabolism , Nicotinic Antagonists/chemistry , Oocytes/drug effects , Oocytes/metabolism , Receptors, Nicotinic/drug effects , Xenopus , alpha7 Nicotinic Acetylcholine Receptor
9.
Neurobiol Learn Mem ; 128: 110-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26775017

ABSTRACT

Nicotine is a major psychoactive and addictive component of tobacco. Although cessation of tobacco use produces various somatic and affective symptoms, withdrawal-related cognitive deficits are considered to be a critical symptom that predict relapse. Therefore, delineating the cognitive mechanisms of nicotine withdrawal may likely provide gainful insights into the neurobiology of nicotine addiction. The present study was designed to examine the effects of nicotine withdrawal induced by mecamylamine, a non-specific nicotinic receptor (nAChR) antagonist, on cognitive control processes in mice using an operant strategy switching task. Brain-derived neurotrophic factor (BDNF) modulates synaptic transmission in frontostriatal circuits, and these circuits are critical for executive functions. Thus, we examined the effects of mecamylamine-precipitated nicotine withdrawal on prefrontal and striatal BDNF protein expression. Mice undergoing precipitated nicotine withdrawal required more trials to attain strategy switching criterion as compared to the controls. Error analysis indicated that impaired performance in these animals was mostly related to their inability to execute the new strategy. The striatal/prefrontal BDNF ratios robustly increased following precipitated nicotine withdrawal. Moreover, higher BDNF ratios were associated with longer task acquisition. Collectively, our findings illustrate that mecamylamine-induced nicotine withdrawal disrupts cognitive control processes and that these changes are possibly linked to perturbations in frontostriatal BDNF signaling.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Executive Function/physiology , Mecamylamine/administration & dosage , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Nicotinic Antagonists/administration & dosage , Substance Withdrawal Syndrome/metabolism , Animals , Cognition/drug effects , Cognition/physiology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Executive Function/drug effects , Male , Mice , Mice, Inbred C57BL , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism
10.
Dokl Biochem Biophys ; 468(1): 173-5, 2016 May.
Article in English | MEDLINE | ID: mdl-27417712

ABSTRACT

The present report describes development of hexamethonium complexes based on fullerene C60. Hexamethonium has a limited penetration into CNS and therefore can antagonize central effects of nicotine only when given at high doses. In the present studies conducted in laboratory rodents, intraperitoneal administration of hexamethonium-fullerene complexes blocked effects of nicotine (convulsions and locomotor stimulation). When compared to equimolar doses of hexamethonium, complexes of hexamethonium with derivatives of fullerene C60 were 40 times more potent indicating an enhanced ability to interact with central nicotine receptors. Thus, fullerene C60 derivatives should be explored further as potential carrier systems for polar drug delivery into CNS.


Subject(s)
Brain/drug effects , Fullerenes/pharmacokinetics , Hexamethonium Compounds/pharmacokinetics , Nicotinic Antagonists/pharmacokinetics , Aminocaproates/chemistry , Animals , Anticonvulsants/chemistry , Anticonvulsants/pharmacokinetics , Brain/metabolism , Dose-Response Relationship, Drug , Fullerenes/administration & dosage , Fullerenes/chemistry , Hexamethonium Compounds/administration & dosage , Hexamethonium Compounds/chemistry , Locomotion/drug effects , Male , Mice , Nicotine , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/chemistry , Rats, Wistar , Seizures/drug therapy
11.
Neurobiol Learn Mem ; 123: 239-49, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26188277

ABSTRACT

Rats display both conditioned flavor preference (CFP) for fructose, and conditioned flavor avoidance (CFA) following sweet adulteration with quinine. Previous pharmacological analyses revealed that fructose-CFP expression was significantly reduced by dopamine (DA) D1 or D2 antagonists, but not NMDA or opioid antagonists. Fructose-CFP acquisition was significantly reduced by DA D1, DA D2 or NMDA antagonists, but not opioid antagonists. Quinine-CFA acquisition was significantly enhanced and prolonged by DA D1, NMDA or opioid, but not DA D2 antagonists. Cholinergic interneurons and projections interact with DA systems in the nucleus accumbens and ventral tegmental area. Further, both muscarinic and nicotinic cholinergic receptor signaling have been implicated in sweet intake and development of food-related preferences. Therefore, the present study examined whether systemic administration of muscarinic (scopolamine: SCOP) or nicotinic (mecamylamine: MEC) cholinergic receptor antagonists mediated fructose-CFP expression, fructose-CFP acquisition and quinine-CFA acquisition. For fructose-CFP expression, rats were trained over 10 sessions with a CS+ flavor in 8% fructose and 0.2% saccharin and a CS- flavor in 0.2% saccharin. Two-bottle choice tests with CS+ and CS- flavors mixed in 0.2% saccharin occurred following vehicle, SCOP (0.1-10mg/kg) and MEC (1-8mg/kg). For fructose-CFP acquisition, six groups of rats received vehicle, SCOP (1 or 2.5mg/kg), MEC (4 or 6mg/kg) or a limited intake vehicle control 0.5h prior to 10 CS+ and CS- training sessions followed by six 2-bottle CS+ and CS- choice tests in 0.2% saccharin. For quinine-CFA acquisition, five groups of rats received vehicle, SCOP (1 or 2.5mg/kg) or MEC (4 or 6mg/kg) 0.5h prior to 8 one-bottle CS- (8% fructose+0.2% saccharin: FS) and CS+ (fructose+saccharin+quinine (0.030%: FSQ) training sessions followed by six 2-bottle CS- and CS+ choice tests in fructose-saccharin solutions. Fructose-CFP expression was significantly reduced by SCOP (2.5-10mg/kg: 65-68%) and MEC (4-8mg/kg: 67-73%) relative to vehicle (89-90%), that occurred only when antagonist doses reduced total saccharin intake but in which CS+ intake was still significantly higher than CS- intake. Fructose-CFP acquisition was eliminated by SCOP at doses of 1 (40-54%) and 2.5 (45-58%)mg/kg, and was accompanied by a failure to observe CS+ and CS- intake differences during testing relative to vehicle (85-92%) and limited control (74-88%) conditions. In contrast, MEC failed to alter fructose-CFP acquisition. Quinine-CFA acquisition was significantly enhanced and prolonged by MEC at 4 (18-24%) and 6 (11-13%) mg/kg relative to vehicle (34-48%). In contrast, SCOP failed to alter quinine-CFA acquisition. These data implicate the cholinergic receptor system in mediating acquisition (learning) of sugar-induced preferences and quinine-induced aversions with muscarinic receptor signaling controlling the former and nicotinic receptor signaling controlling the latter.


Subject(s)
Avoidance Learning/drug effects , Conditioning, Psychological/drug effects , Muscarinic Antagonists/pharmacology , Nicotinic Antagonists/pharmacology , Taste Perception/drug effects , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/pharmacology , Animals , Fructose/administration & dosage , Fructose/pharmacology , Male , Mecamylamine/administration & dosage , Mecamylamine/pharmacology , Muscarinic Antagonists/administration & dosage , Nicotinic Antagonists/administration & dosage , Quinine/administration & dosage , Quinine/pharmacology , Rats , Rats, Sprague-Dawley , Scopolamine/administration & dosage , Scopolamine/pharmacology , Sweetening Agents/administration & dosage , Sweetening Agents/pharmacology
12.
Mar Drugs ; 13(6): 3259-75, 2015 May 27.
Article in English | MEDLINE | ID: mdl-26023835

ABSTRACT

In the present study, we synthesized and, structurally and functionally characterized a novel α4/7-conotoxin Mr1.7 (PECCTHPACHVSHPELC-NH2), which was previously identified by cDNA libraries from Conus marmoreus in our lab. The NMR solution structure showed that Mr1.7 contained a 310-helix from residues Pro7 to His10 and a type I ß-turn from residues Pro14 to Cys17. Electrophysiological results showed that Mr1.7 selectively inhibited the α3ß2, α9α10 and α6/α3ß2ß3 neuronal nicotinic acetylcholine receptors (nAChRs) with an IC50 of 53.1 nM, 185.7 nM and 284.2 nM, respectively, but showed no inhibitory activity on other nAChR subtypes. Further structure-activity studies of Mr1.7 demonstrated that the PE residues at the N-terminal sequence of Mr1.7 were important for modulating its selectivity, and the replacement of Glu2 by Ala resulted in a significant increase in potency and selectivity to the α3ß2 nAChR. Furthermore, the substitution of Ser12 with Asn in the loop2 significantly increased the binding of Mr1.7 to α3ß2, α3ß4, α2ß4 and α7 nAChR subtypes. Taken together, this work expanded our knowledge of selectivity and provided a new way to improve the potency and selectivity of inhibitors for nAChR subtypes.


Subject(s)
Conotoxins/pharmacology , Nicotinic Antagonists/pharmacology , Receptors, Nicotinic/drug effects , Animals , Conotoxins/administration & dosage , Conotoxins/chemistry , Conus Snail/chemistry , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/chemistry , Receptors, Nicotinic/metabolism , Structure-Activity Relationship
13.
Gen Physiol Biophys ; 34(2): 189-200, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25504063

ABSTRACT

Organophosphorus nerve agents inhibit acetylcholinesterase (AChE) which causes the breakdown of the transmitter acetylcholine (ACh) in the synaptic cleft. Overstimulation of cholinergic receptors (muscarinic and nicotinic) by excessive amounts of ACh causes several health problems and may even cause death. Reversible AChE inhibitors play an important role in prophylaxis against nerve agents. The presented study investigated whether 7-methoxytacrine (7-MEOTA) and 7-MEOTA-donepezil derivatives can act as central and peripheral reversible AChE inhibitors and simultaneously antagonize muscarinic and nicotinic receptors. The possible mechanism of action was studied on cell cultures (patch clamp technique, calcium mobilization assay) and on isolated smooth muscle tissue (contraction study). Furthermore, the kinetics of the compounds were also examined. CNS availability was predicted by determining the passive blood-brain barrier penetration estimated via a modified PAMPA assay. In conclusion, this study provides promising evidence that the new synthesized 7-MEOTA-donepezil derivatives have the desired anticholinergic effect; they can inhibit AChE, and nicotinic and muscarinic receptors in the micromolar range. Furthermore, they seem to penetrate readily into the CNS. However, their real potency and benefit must be verified by in vivo experiments.


Subject(s)
Cholinergic Antagonists/administration & dosage , Indans/administration & dosage , Muscarinic Antagonists/administration & dosage , Muscle Fibers, Skeletal/metabolism , Nicotinic Antagonists/administration & dosage , Piperidines/administration & dosage , Tacrine/analogs & derivatives , Animals , Cells, Cultured , Donepezil , Dose-Response Relationship, Drug , Male , Muscle Fibers, Skeletal/drug effects , Rats , Rats, Wistar , Tacrine/administration & dosage
14.
Int J Neuropsychopharmacol ; 17(8): 1321-6, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24852262

ABSTRACT

Several lines of evidence indicate that ketamine has a rapid antidepressant-like effect in rodents and humans, but underlying mechanisms are unclear. In the present study, we investigated the effect of ketamine on serotonin (5-HT) release in the rat prefrontal cortex by in vivo microdialysis. A subcutaneous administration of ketamine (5 and 25 mg/kg) significantly increased the prefrontal 5-HT level in a dose-dependent manner, which was attenuated by local injection of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) antagonists into the dorsal raphe nucleus (DRN). Direct stimulation of AMPARs in the DRN significantly increased prefrontal 5-HT level, while intra-DRN injection of ketamine (36.5 nmol) had no effect. Furthermore, intra-DRN injection of an α 4 ß 2-nicotinic acetylcholine receptor (nAChR) antagonist, dihydro-ß-erythroidine (10 nmol), significantly attenuated the subcutaneous ketamine-induced increase in prefrontal 5-HT levels. These results suggest that AMPARs and α 4 ß 2-nAChRs in the DRN play a key role in the ketamine-induced 5-HT release in the prefrontal cortex.


Subject(s)
Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/metabolism , Ketamine/pharmacology , Prefrontal Cortex/drug effects , Receptors, AMPA/metabolism , Receptors, Nicotinic/metabolism , Serotonin/metabolism , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/pharmacology , Benzothiadiazines/administration & dosage , Benzothiadiazines/pharmacology , Dihydro-beta-Erythroidine/administration & dosage , Dihydro-beta-Erythroidine/pharmacology , Dose-Response Relationship, Drug , Injections, Subcutaneous , Ketamine/administration & dosage , Ketamine/antagonists & inhibitors , Male , Microdialysis , Microinjections , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/pharmacology , Prefrontal Cortex/metabolism , Quinoxalines/administration & dosage , Quinoxalines/pharmacology , Rats , Receptors, AMPA/agonists , Receptors, AMPA/antagonists & inhibitors , Spermine/administration & dosage , Spermine/analogs & derivatives , Spermine/pharmacology
15.
Behav Pharmacol ; 24(3): 229-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23625379

ABSTRACT

Although a body of evidence shows the crucial role of hippocampal nitrergic and cholinergic systems in the modulation of anxiety, little is known about their functional relationship with regard to anxiety. The present study investigated the relationship between intra-CA1 administration of a nicotinic acetylcholine receptor antagonist (mecamylamine) and a nitric oxide synthase inhibitor [Nω-nitro-L-arginine methyl ester (L-NAME)] or its precursor (L-arginine) in anxiety-related behaviors. Mice received bilateral intra-CA1 injections of either L-NAME or L-arginine in the presence of mecamylamine and were subsequently tested in the elevated plus maze. A dose of 0.5 µg/0.5 µl mecamylamine bilaterally administered into CA1 did not change the percentage of open arm time (%OAT) or the percentage of open arm entries (%OAE) in the elevated plus maze task and thus was considered as a subeffective dose. Intra-CA1 administration of either L-arginine (1 and 1.5 µg/0.5 µl, bilaterally) or L-NAME (at 60 ng/0.5 µl, bilaterally) decreased %OAT, which represents an anxiogenic-like effect. Coadministration of the subeffective dose of mecamylamine together with the lower doses of L-NAME (10 and 30 ng/0.5 µl, bilaterally) or L-arginine (0.5 µg/0.5 µl, bilaterally) led to a decrease in %OAT and %OAE. Thus, both L-NAME and L-arginine showed anxiogenic-like effects, but the effects of mecamylamine were too small to support a functional relationship between the hippocampal cholinergic and nitrergic systems.


Subject(s)
Anxiety/drug therapy , Anxiety/pathology , Cholinergic Agents/metabolism , Hippocampus/metabolism , Nitric Oxide/metabolism , Analysis of Variance , Animals , Arginine/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Hippocampus/drug effects , Male , Maze Learning/drug effects , Mecamylamine/administration & dosage , Mice , Microinjections , NG-Nitroarginine Methyl Ester/administration & dosage , Nicotinic Antagonists/administration & dosage
16.
J Neurosci ; 31(40): 14367-77, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21976522

ABSTRACT

Adolescent smoking is associated with auditory-cognitive deficits and structural alterations to auditory thalamocortical systems, suggesting that higher auditory function is vulnerable to nicotine exposure during adolescence. Although nicotinic acetylcholine receptors (nAChRs) regulate thalamocortical processing in adults, it is not known whether they regulate processing at earlier ages since their expression pattern changes throughout postnatal development. Here we investigate nicotinic regulation of tone-evoked current source density (CSD) profiles in mouse primary auditory cortex from just after hearing onset until adulthood. At the youngest ages, systemic nicotine did not affect CSD profiles. However, beginning in early adolescence nicotine enhanced characteristic frequency (CF)-evoked responses in layers 2-4 by enhancing thalamocortical, early intracortical, and late intracortical response components. Nicotinic responsiveness developed rapidly and peaked over the course of adolescence, then declined thereafter. Generally, responsiveness in females developed more quickly, peaked earlier, and declined more abruptly and fully than in males. In contrast to the enhancement of CF-evoked responses, nicotine suppressed shorter-latency intracortical responses to spectrally distant (non-CF) stimuli while enhancing longer-latency responses. Intracortical infusion of nAChR antagonists showed that enhancement of CF-evoked intracortical processing involves α4ß2*, but not α7, nAChRs, whereas both receptor subtypes regulate non-CF-evoked late intracortical responses. Notably, antagonist effects in females implied regulation by endogenous acetylcholine. Thus, nicotinic regulation of cortical processing varies with age and sex, with peak effects during adolescence that may contribute to the vulnerability of adolescents to smoking.


Subject(s)
Acoustic Stimulation/methods , Auditory Cortex/physiology , Evoked Potentials, Auditory/physiology , Nicotine/administration & dosage , Age Factors , Animals , Auditory Cortex/drug effects , Evoked Potentials, Auditory/drug effects , Female , Injections, Intraventricular , Male , Mice , Nicotinic Antagonists/administration & dosage , Receptors, Nicotinic/physiology , alpha7 Nicotinic Acetylcholine Receptor
17.
J Neurosci ; 31(38): 13546-61, 2011 Sep 21.
Article in English | MEDLINE | ID: mdl-21940446

ABSTRACT

Release of conventional neurotransmitters is mainly controlled by calcium (Ca²âº) influx via high-voltage-activated (HVA), Ca(v)2, channels ("N-, P/Q-, or R-types") that are opened by action potentials. Regulation of transmission by subthreshold depolarizations does occur, but there is little evidence that low-voltage-activated, Ca(v)3 ("T-type"), channels take part. GABA release from cortical perisomatic-targeting interneurons affects numerous physiological processes, and yet its underlying control mechanisms are not fully understood. We investigated whether T-type Ca²âº channels are involved in regulating GABA transmission from these cells in rat hippocampal CA1 using a combination of whole-cell voltage-clamp, multiple-fluorescence confocal microscopy, dual-immunolabeling electron-microscopy, and optogenetic methods. We show that Ca(v)3.1, T-type Ca²âº channels can be activated by α3ß4 nicotinic acetylcholine receptors (nAChRs) that are located on the synaptic regions of the GABAergic perisomatic-targeting interneuronal axons, including the parvalbumin-expressing cells. Asynchronous, quantal GABA release can be triggered by Ca²âº influx through presynaptic T-type Ca²âº channels, augmented by Ca²âº from internal stores, following focal microiontophoretic activation of the α3ß4 nAChRs. The resulting GABA release can inhibit pyramidal cells. The T-type Ca²âº channel-dependent mechanism is not dependent on, or accompanied by, HVA channel Ca²âº influx, and is insensitive to agonists of cannabinoid, µ-opioid, or GABA(B) receptors. It may therefore operate in parallel with the normal HVA-dependent processes. The results reveal new aspects of the regulation of GABA transmission and contribute to a deeper understanding of ACh and nicotine actions in CNS.


Subject(s)
Calcium Channels, T-Type/physiology , Calcium/metabolism , Interneurons/metabolism , Nerve Endings/physiology , Receptors, Nicotinic/physiology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Action Potentials/physiology , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Choline O-Acetyltransferase/genetics , In Vitro Techniques , Interneurons/physiology , Interneurons/ultrastructure , Mice , Mice, Transgenic , Microinjections , Nerve Endings/ultrastructure , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/pharmacology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
18.
Hippocampus ; 22(8): 1681-90, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22271264

ABSTRACT

Nicotine administration alters various forms of hippocampus-dependent learning and memory. Increasing work has found that the dorsal and ventral hippocampus differentially contribute to multiple behaviors. Thus, the present study examined whether the effects of nicotine in the dorsal and ventral hippocampus have distinct influences on contextual fear learning in male C57BL/6J mice. Direct infusion of nicotine into the dorsal hippocampus resulted in an enhancement of contextual fear learning, whereas nicotine infused into the ventral hippocampus resulted in deficits. Nicotine infusions into the ventral hippocampus did not alter hippocampus-independent cued fear conditioning or time spent in the open arm of the elevated plus maze, a measure of anxiety, suggesting that the effects are due to alterations in contextual learning and not other general processes. Finally, results from using direct infusions of MLA, a low-affinity α7 nicotinic acetylcholine receptor (nAChR) antagonist, in conjunction with systemic nicotine, provide evidence that α7-nAChRs in the ventral hippocampus mediate the detrimental effect of ventral hippocampal nicotine on contextual fear learning. These results suggest that with systemic nicotine administration, competition exists between the dorsal and ventral hippocampus for behavioral control over contextual learning.


Subject(s)
Conditioning, Psychological/drug effects , Fear/drug effects , Hippocampus/drug effects , Nicotine/administration & dosage , Receptors, Nicotinic/metabolism , Acetylcholine/metabolism , Aconitine/administration & dosage , Aconitine/analogs & derivatives , Analysis of Variance , Animals , Cues , Hippocampus/metabolism , Injections, Intraventricular , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Nicotinic Antagonists/administration & dosage , alpha7 Nicotinic Acetylcholine Receptor
19.
Nicotine Tob Res ; 14(6): 711-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22241831

ABSTRACT

INTRODUCTION: The smoking cessation aid, varenicline, has higher affinity for the alpha4beta2-subtype of the nicotinic acetylcholine receptor (α4ß2*-nAChR) than for other subtypes of nAChRs by in vitro assays. The mechanism of action of acute varenicline was studied in vivo to determine (a) subtype activation associated with physiological effects and (b) dose relationship as an antagonist of nicotine. METHODS: Acute doses of saline, nicotine, and varenicline were given to mice, and locomotor depression and hypothermia were measured. Subunit null mutant mice as well as selective antagonists were used to study mode of action of varenicline as an agonist. Varenicline as an antagonist of nicotine was also investigated. RESULTS: Varenicline evokes locomotor depression and hypothermia at higher doses than necessary for nicotine. Null mutation of the α7- or ß2-nAChR subunit did not decrease the effectiveness of varenicline; however, null mutation of the ß4 subunit significantly decreased the magnitude of the varenicline effect. Effects of the highest dose studied were blocked by mecamylamine (general nAChR antagonist) and partially antagonized by hexamethonium (largely peripheral nAChR antagonist). No significant block was seen with ondansetron antagonist of 5-hydroxytryptamine 3 receptor. Using a dose of nicotine selective for ß2*-nAChR subtype effects with these tests, dose-dependent antagonism by varenicline was seen. Effective inhibitory doses were determined and appear to be in a range consistent with binding affinity or desensitization of ß2*-nAChRs. CONCLUSIONS: Varenicline acts as a functional antagonist of ß2*-nAChRs, blocking certain effects of nicotine. At higher doses, varenicline is an agonist of ß4*-nAChRs producing physiological changes in mice.


Subject(s)
Benzazepines/pharmacology , Nerve Tissue Proteins/drug effects , Nicotinic Agonists/pharmacology , Quinoxalines/pharmacology , Receptors, Nicotinic/drug effects , Animals , Benzazepines/administration & dosage , Dose-Response Relationship, Drug , Female , Genotype , Hexamethonium/pharmacology , Hypothermia/chemically induced , Inhibitory Concentration 50 , Male , Mecamylamine/pharmacology , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nicotine/administration & dosage , Nicotine/pharmacology , Nicotinic Agonists/administration & dosage , Nicotinic Antagonists/administration & dosage , Nicotinic Antagonists/pharmacology , Ondansetron/antagonists & inhibitors , Quinoxalines/administration & dosage , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Varenicline
20.
Anesth Analg ; 114(1): 102-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22003222

ABSTRACT

BACKGROUND: We tested the hypothesis that a single injection of botulinum toxin not only has local, but also distant effects on muscle function, biochemistry, and pharmacodynamics of atracurium. METHODS: Botulinum toxin (2.5 U) was injected into the tibialis muscle of anesthetized rats (n = 26). The contralateral side with no injection served to study distant effects. Control animals (n = 25) received a saline injection. Neuromuscular function, pharmacology, and expression of acetylcholine receptors (nAChRs) were evaluated in the tibialis at 0, 4, and 16 days after injection and in comparison with saline- injected controls. RESULTS: On day 4, botulinum toxin caused complete paralysis of the tibialis, while its contralateral side showed a decrease in absolute twitch tension (1.8 N [1.6; 1.9] vs 3.0 N [2.8; 3.1], Newton, P < 0.05). On day 16, muscle weakness was only present on the toxin-injected side where absolute twitch tension was decreased (0.6 N [0.6, 0.7] vs 3.4 N [3.1, 3.7], P < 0.05). Tibialis mass was decreased on the toxin-injected side at day 4 (1.46 mg/g [1.43, 1.48] vs 1.74 mg/g [1.72; 1.75], P < 0.05) and on day 16 (0.78 mg/g [0.76, 0.79] vs 1.73 mg/g [1.69; 1.77], P < 0.05). Effects distant from the site of injection were seen on day 16, when muscle atrophy was also present in the adjacent gastrocnemius and soleus muscles. Normalized to tibialis mass, specific twitch tension (tension/g muscle) was reduced on the contralateral side at day 4 and on the toxin-injected side at day 16 in relation to saline controls. At day 16, an increased sensitivity to atracurium was seen on the toxin-injected side, evidenced as a decreased ED(50) (0.23 mg/kg [0.13, 0.33] vs 0.72 mg/kg [0.63, 0.82], P < 0.05) and a lower infusion rate (38 µL/kg/min [32, 43] vs135 µL/kg/min [126, 144], P < 0.05), together with a reduced plasma concentration requirement of atracurium (0.5 µg/mL [0.4, 0.7] vs 4.5 µg/mL [3.8, 5.2], P < 0.05) to achieve a steady state 50% reduction in baseline (absolute) twitch tension. ED(50) of atracurium was also decreased on the contralateral side at day 16 in relation to saline controls. The nAChRs in the tibialis were increased on the toxin-injected side to 123 fmol/mg [115, 131] vs 28 fmol/mg [25, 29] (P < 0.05) in time-matched saline-injected controls at day 4 and to 378 [341, 413] vs 27 fmol/mg [25, 29] (P < 0.05) at day 16. CONCLUSIONS: Botulinum toxin has local and distant effects on muscle. The decrease in specific twitch tension indicates that the muscle atrophy alone cannot explain the functional changes; neuromuscular transmission is also impaired. An increased sensitivity to atracurium on the toxin-injected side, despite up-regulation of nAChRs, seems unique to botulinum toxin.


Subject(s)
Botulinum Toxins/administration & dosage , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/innervation , Neuromuscular Blocking Agents/administration & dosage , Synaptic Transmission/drug effects , Animals , Atracurium/administration & dosage , Botulinum Toxins/toxicity , Dose-Response Relationship, Drug , Injections, Intramuscular , Male , Muscle Fatigue/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/chemically induced , Muscular Atrophy/physiopathology , Neuromuscular Blocking Agents/toxicity , Neuromuscular Nondepolarizing Agents/administration & dosage , Nicotinic Antagonists/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Cholinergic/drug effects , Receptors, Cholinergic/metabolism , Time Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL