Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
Neurobiol Dis ; 158: 105473, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34371144

RESUMO

CalDAG-GEFI (CDGI) is a protein highly enriched in the striatum, particularly in the principal spiny projection neurons (SPNs). CDGI is strongly down-regulated in two hyperkinetic conditions related to striatal dysfunction: Huntington's disease and levodopa-induced dyskinesia in Parkinson's disease. We demonstrate that genetic deletion of CDGI in mice disrupts dendritic, but not somatic, M1 muscarinic receptors (M1Rs) signaling in indirect pathway SPNs. Loss of CDGI reduced temporal integration of excitatory postsynaptic potentials at dendritic glutamatergic synapses and impaired the induction of activity-dependent long-term potentiation. CDGI deletion selectively increased psychostimulant-induced repetitive behaviors, disrupted sequence learning, and eliminated M1R blockade of cocaine self-administration. These findings place CDGI as a major, but previously unrecognized, mediator of cholinergic signaling in the striatum. The effects of CDGI deletion on the self-administration of drugs of abuse and its marked alterations in hyperkinetic extrapyramidal disorders highlight CDGI's therapeutic potential.


Assuntos
Dendritos , Fatores de Troca do Nucleotídeo Guanina/genética , Neostriado/fisiopatologia , Plasticidade Neuronal , Sistema Nervoso Parassimpático/fisiopatologia , Sinapses , Animais , Doenças dos Gânglios da Base/genética , Doenças dos Gânglios da Base/fisiopatologia , Doenças dos Gânglios da Base/psicologia , Estimulantes do Sistema Nervoso Central/farmacologia , Potenciais Pós-Sinápticos Excitadores/genética , Hipercinese/genética , Hipercinese/psicologia , Potenciação de Longa Duração , Masculino , Camundongos , Camundongos Knockout , Atividade Motora , Polimorfismo de Nucleotídeo Único , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/fisiologia , Transtornos Relacionados ao Uso de Substâncias/genética , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/psicologia
2.
Neural Plast ; 2021: 8813734, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33510779

RESUMO

The cholinergic system plays a fundamental role in learning and memory. Pharmacological activation of the muscarinic receptor M1R potentiates NMDA receptor activity and induces short-term potentiation at the synapses called muscarinic LTP, mLTP. Dysfunction of cholinergic transmission has been detected in the settings of cognitive impairment and dementia. Systemic inflammation as well as neuroinflammation has been shown to profoundly alter synaptic transmission and LTP. Indeed, intervention which is aimed at reducing neuroinflammatory changes in the brain has been associated with an improvement in cognitive functions. While cognitive impairment caused either by cholinergic dysfunction and/or by systemic inflammation suggests a possible connection between the two, so far whether systemic inflammation affects mLTP has not been extensively studied. In the present work, we explored whether an acute versus persistent systemic inflammation induced by LPS injections would differently affect the ability of hippocampal synapses to undergo mLTP. Interestingly, while a short exposure to LPS resulted in a transient deficit in mLTP expression, a longer exposure persistently impaired mLTP. We believe that these findings may be involved in cognitive dysfunctions following sepsis and possibly neuroinflammatory processes.


Assuntos
Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Plasticidade Neuronal/fisiologia , Receptor Muscarínico M1/fisiologia , Animais , Agonistas Colinérgicos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/fisiopatologia , Lipopolissacarídeos/toxicidade , Potenciação de Longa Duração/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Plasticidade Neuronal/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Receptor Muscarínico M1/agonistas
3.
Pharmacol Rep ; 71(6): 1108-1114, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31634798

RESUMO

BACKGROUND: To compare pharmacologic effects of pirenzepine and AF-DX116, a selective competitive antagonist for M1 and M2 subtype muscarinic cholinergic receptors (mAChRs), respectively, with atropine, a non-selective competitive antagonist for mAChRs, on Lipopolysaccharide (LPS). METHODS: Male C57BL/6 mice were used to establish models of LPS-induced experimental endotoxemia. Mice were intraperitoneally injected 10 min prior to LPS injection with control (saline), atropine, pirenzepine and AF-DX116, respectively. Overall survival time was estimated using Kaplan-Meier plots. Inflammatory cytokine tumor necrosis factor-α (TNF-α) was monitored at various intervals after LPS injection and individual reagent administration. Pathological alternations in lungs and liver were analyzed. RESULTS: Pirenzepine and atropine pretreatment improved survival rate of LPS-induced septic shock; in contrast, AF-DX116 accelerated death from sepsis. Moreover, TNF-α plasma level was decreased in response to pirenzepine or atropine, whereas increased in response to AF-DX116. Pirenzepine and atropine relieved whereas AF-DX116 accelerated LPS-induced pulmonary and hepatic injury. Pirenzepine reduced proportion of M1 subtype of macrophages, while AF-DX116 promoted polarization of macrophages to M1 subtype. Pirenzepine pretreatment reduced while AF-DX116 enhanced expression of SOCS3 at mRNA level. CONCLUSIONS: The administration of pirenzepine and atropine may have beneficial effects on septic shock.


Assuntos
Atropina/farmacologia , Pirenzepina/análogos & derivados , Pirenzepina/farmacologia , Receptor Muscarínico M1/fisiologia , Receptor Muscarínico M2/fisiologia , Choque Séptico/tratamento farmacológico , Choque Séptico/fisiopatologia , Animais , Citocinas/metabolismo , Lipopolissacarídeos , Fígado/patologia , Pulmão/patologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/efeitos dos fármacos , Receptor Muscarínico M1/antagonistas & inibidores , Receptor Muscarínico M2/antagonistas & inibidores , Choque Séptico/induzido quimicamente , Choque Séptico/mortalidade , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/metabolismo
4.
J Neurosci ; 39(40): 7840-7852, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31451581

RESUMO

Transient receptor potential melastatin 3 (TRPM3) is a nonselective cation channel that is inhibited by Gßγ subunits liberated following activation of Gαi/o protein-coupled receptors. Here, we demonstrate that TRPM3 channels are also inhibited by Gßγ released from Gαs and Gαq Activation of the Gs-coupled adenosine 2B receptor and the Gq-coupled muscarinic acetylcholine M1 receptor inhibited the activity of TRPM3 heterologously expressed in HEK293 cells. This inhibition was prevented when the Gßγ sink ßARK1-ct (C terminus of ß-adrenergic receptor kinase-1) was coexpressed with TRPM3. In neurons isolated from mouse dorsal root ganglion (DRG), native TRPM3 channels were inhibited by activating Gs-coupled prostaglandin-EP2 and Gq-coupled bradykinin B2 (BK2) receptors. The Gi/o inhibitor pertussis toxin and inhibitors of PKA and PKC had no effect on EP2- and BK2-mediated inhibition of TRPM3, demonstrating that the receptors did not act through Gαi/o or through the major protein kinases activated downstream of G-protein-coupled receptor (GPCR) activation. When DRG neurons were dialyzed with GRK2i, which sequesters free Gßγ protein, TRPM3 inhibition by EP2 and BK2 was significantly reduced. Intraplantar injections of EP2 or BK2 agonists inhibited both the nocifensive response evoked by TRPM3 agonists, and the heat hypersensitivity produced by Freund's Complete Adjuvant (FCA). Furthermore, FCA-induced heat hypersensitivity was completely reversed by the selective TRPM3 antagonist ononetin in WT mice and did not develop in Trpm3-/- mice. Our results demonstrate that TRPM3 is subject to promiscuous inhibition by Gßγ protein in heterologous expression systems, primary neurons and in vivo, and suggest a critical role for this ion channel in inflammatory heat hypersensitivity.SIGNIFICANCE STATEMENT The ion channel TRPM3 is widely expressed in the nervous system. Recent studies showed that Gαi/o-coupled GPCRs inhibit TRPM3 through a direct interaction between Gßγ subunits and TRPM3. Since Gßγ proteins can be liberated from other Gα subunits than Gαi/o, we examined whether activation of Gs- and Gq-coupled receptors also influence TRPM3 via Gßγ. Our results demonstrate that activation of Gs- and Gq-coupled GPCRs in recombinant cells and sensory neurons inhibits TRPM3 via Gßγ liberation. We also demonstrated that Gs- and Gq-coupled receptors inhibit TRPM3 in vivo, thereby reducing pain produced by activation of TRPM3, and inflammatory heat hypersensitivity. Our results identify Gßγ inhibition of TRPM3 as an effector mechanism shared by the major Gα subunits.


Assuntos
Subunidades beta da Proteína de Ligação ao GTP/fisiologia , Subunidades gama da Proteína de Ligação ao GTP/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Canais de Cátion TRPM/fisiologia , Animais , Comportamento Animal , Feminino , Subunidades beta da Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidades gama da Proteína de Ligação ao GTP/antagonistas & inibidores , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Células HEK293 , Humanos , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Hiperalgesia/psicologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/fisiologia , Nociceptores/efeitos dos fármacos , Toxina Pertussis/farmacologia , Receptor A2B de Adenosina/fisiologia , Receptor Muscarínico M1/fisiologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Transdução de Sinais/fisiologia , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética
5.
Biochem Pharmacol ; 169: 113613, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31445019

RESUMO

Previously, we reported that hepatic muscarinic receptors modulate both acute and chronic liver injury, however, the role of muscarinic receptors in fatty liver disease is unclear. We observed in patients who underwent weight loss surgery, a decrease in hepatic expression of M3 muscarinic receptors (M3R). We also observed that fat loading of hepatocytes, increased M3R expression. Based on these observations, we tested the hypothesis that M3R regulate hepatocyte lipid accumulation. Incubation of AML12 hepatocytes with 1 mM oleic acid resulted in lipid accumulation that was significantly reduced by co-treatment with a muscarinic agonist (pilocarpine or carbachol), an effect blocked by atropine (a muscarinic antagonist). Similar treatment of Hepa 1-6 cells, a mouse hepatoblastoma cell line, showed comparable results. In both, control and fat-loaded AML12 cells, pilocarpine induced time-dependent AMPKα phosphorylation and significantly up-regulated lipolytic genes (ACOX1, CPT1, and PPARα). Compound C, a selective and reversible AMPK inhibitor, significantly blunted pilocarpine-mediated reduction of lipid accumulation and pilocarpine-mediated up-regulation of lipolytic genes. BAPTA-AM, a calcium chelator, and STO-609, a calcium/calmodulin-dependent protein kinase kinase inhibitor, attenuated agonist-induced AMPKα phosphorylation. Finally, M3R siRNA attenuated agonist-induced AMPKα phosphorylation as well as agonist-mediated reduction of hepatocyte steatosis. In conclusion, this proof-of-concept study demonstrates that M3R has protective effects against hepatocyte lipid accumulation by activating AMPK pathway and is a potential therapeutic target for non-alcoholic fatty liver disease.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/fisiologia , Hepatócitos/metabolismo , Metabolismo dos Lipídeos , Receptor Muscarínico M3/fisiologia , Animais , Células Cultivadas , Humanos , Camundongos , PPAR alfa/fisiologia , Fosforilação , Receptor Muscarínico M1/fisiologia , Transdução de Sinais/fisiologia
6.
Neuron ; 103(3): 432-444.e3, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31221559

RESUMO

Subtypes of nucleus accumbens medium spiny neurons (MSNs) promote dichotomous outcomes in motivated behaviors. However, recent reports indicate enhancing activity of either nucleus accumbens (NAc) core MSN subtype augments reward, suggesting coincident MSN activity may underlie this outcome. Here, we report a collateral excitation mechanism in which high-frequency, NAc core dopamine 1 (D1)-MSN activation causes long-lasting potentiation of excitatory transmission (LLP) on dopamine receptor 2 (D2)-MSNs. Our mechanistic investigation demonstrates that this form of plasticity requires release of the excitatory peptide substance P from D1-MSNs and robust cholinergic interneuron activation through neurokinin receptor stimulation. We also reveal that D2-MSN LLP requires muscarinic 1 receptor activation, intracellular calcium signaling, and GluR2-lacking AMPAR insertion. This study uncovers a mechanism for shaping NAc core activity through the transfer of excitatory information from D1-MSNs to D2-MSNs and may provide a means for altering goal-directed behavior through coordinated MSN activity.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Potenciação de Longa Duração/fisiologia , Núcleo Accumbens/fisiologia , Substância P/metabolismo , Potenciais de Ação/fisiologia , Animais , Aprepitanto/farmacologia , Sinalização do Cálcio/fisiologia , Neurônios Colinérgicos/fisiologia , Neurônios Dopaminérgicos/efeitos da radiação , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Interneurônios/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Motivação , Antagonistas dos Receptores de Neurocinina-1/farmacologia , Núcleo Accumbens/citologia , Estimulação Luminosa , Piperidinas/farmacologia , Receptor Muscarínico M1/fisiologia , Receptores de AMPA/fisiologia , Receptores de Dopamina D1/análise , Receptores de Dopamina D2/análise , Receptores da Neurocinina-1/fisiologia
7.
Naunyn Schmiedebergs Arch Pharmacol ; 392(9): 1071-1083, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31049606

RESUMO

The search for new drugs remains an important focus for the safe and effective treatment of cardiovascular diseases. Previous evidence has shown that choline analogs can offer therapeutic benefit for cardiovascular complications. The current study investigates the effects of 2-(4-((1-phenyl-1H-pyrazol-4-yl)methyl)piperazin-1-yl)ethan-1-ol (LQFM032) on cardiovascular function and cholinergic-nitric oxide signaling. Synthesized LQFM032 (0.3, 0.6, or 1.2 mg/kg) was administered by intravenous and intracerebroventricular routes to evaluate the potential alteration of mean arterial pressure, heart rate, and renal sympathetic nerve activity of normotensive and hypertensive rats. Vascular function was further evaluated in isolated vessels, while pharmacological antagonists and computational studies of nitric oxide synthase and muscarinic receptors were performed to assess possible mechanisms of LQFM032 activity. The intravenous and intracerebroventricular administration of LQFM032 elicited a temporal reduction in mean arterial pressure, heart rate, and renal sympathetic nerve activity of rats. The cumulative addition of LQFM032 to isolated endothelium-intact aortic rings reduced vascular tension and elicited a concentration-dependent relaxation. Intravenous pretreatment with L-NAME (nitric oxide synthase inhibitor), atropine (nonselective muscarinic receptor antagonist), pirenzepine, and 4-DAMP (muscarinic M1 and M3 subtype receptor antagonist, respectively) attenuated the cardiovascular effects of LQFM032. These changes may be due to a direct regulation of muscarinic signaling as docking data shows an interaction of choline analog with M1 and M3 but not nitric oxide synthase. Together, these findings demonstrate sympathoinhibitory, hypotensive, and antihypertensive effects of LQFM032 and suggest the involvement of muscarinic receptors.


Assuntos
Anti-Hipertensivos/farmacologia , Hipotensão/fisiopatologia , Piperazinas/farmacologia , Pirazóis/farmacologia , Receptor Muscarínico M1/fisiologia , Receptor Muscarínico M3/fisiologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiologia , Atropina/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Hipertensão/induzido quimicamente , Masculino , Antagonistas Muscarínicos/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Piperidinas/farmacologia , Pirenzepina/farmacologia , Ratos Endogâmicos SHR , Ratos Wistar
8.
J Nippon Med Sch ; 86(2): 117-121, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31130562

RESUMO

Sialorrhea is often treated with anticholinergic agents, but they can have undesirable side effects such as drowsiness, sedation, and constipation. Effective medication that acts selectively on the salivary glands is needed. We report the case of a patient with sialorrhea who was successfully treated by the combined use of pirenzepine and solifenacin (M1 and M3 muscarinic receptor antagonists, respectively). The patient was a 51-year-old man with mean unstimulated and stimulated salivary flow rates per 10 min of 6.1 mL and 41.7 mL, respectively (both were measured three times). 99mTcO4- salivary gland scintigraphy revealed characteristic spontaneous saliva secretion without stimulation. He was treated with Scopolia extract, escitalopram, solifenacin succinate, and the combined administration of solifenacin succinate and pirenzepine. A statistically significant decrease was observed from the pre-medication unstimulated and stimulated salivary flow rates only following the combined administration of solifenacin and pirenzepine. The major muscarinic receptor subtype expressed in the salivary glands is M3; however, M1 is also present. A study using knockout mice demonstrated that the presence of either M1 or M3 receptors was sufficient for salivation. Thus, the combined use of selective M1 and M3 antagonists could provide a good treatment option for sialorrhea.


Assuntos
Antagonistas Muscarínicos/administração & dosagem , Pirenzepina/administração & dosagem , Receptor Muscarínico M1/antagonistas & inibidores , Receptor Muscarínico M3/antagonistas & inibidores , Sialorreia/tratamento farmacológico , Succinato de Solifenacina/administração & dosagem , Animais , Quimioterapia Combinada , Humanos , Masculino , Camundongos Knockout , Pessoa de Meia-Idade , Cintilografia , Receptor Muscarínico M1/fisiologia , Receptor Muscarínico M3/fisiologia , Glândulas Salivares/diagnóstico por imagem , Glândulas Salivares/fisiopatologia , Salivação , Sialorreia/diagnóstico por imagem , Sialorreia/fisiopatologia , Resultado do Tratamento
9.
Biol Psychiatry ; 85(12): 989-1000, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31003787

RESUMO

BACKGROUND: The prefrontal cortex (PFC) integrates information from multiple inputs to exert top-down control allowing for appropriate responses in a given context. In psychiatric disorders such as posttraumatic stress disorder, PFC hyperactivity is associated with inappropriate fear in safe situations. We previously reported a form of muscarinic acetylcholine receptor (mAChR)-dependent long-term depression in the PFC that we hypothesize is involved in appropriate fear responding and could serve to reduce cortical hyperactivity following stress. However, it is unknown whether this long-term depression occurs at fear-related inputs. METHODS: Using optogenetics with extracellular and whole-cell electrophysiology, we assessed the effect of mAChR activation on the synaptic strength of specific PFC inputs. We used selective pharmacological tools to assess the involvement of M1 mAChRs in conditioned fear extinction in control mice and in the stress-enhanced fear-learning model. RESULTS: M1 mAChR activation induced long-term depression at inputs from the ventral hippocampus and basolateral amygdala but not from the mediodorsal nucleus of the thalamus. We found that systemic M1 mAChR antagonism impaired contextual fear extinction. Treatment with an M1 positive allosteric modulator enhanced contextual fear extinction consolidation in stress-enhanced fear learning-conditioned mice. CONCLUSIONS: M1 mAChRs dynamically modulate synaptic transmission at two PFC inputs whose activity is necessary for fear extinction, and M1 mAChR function is required for proper contextual fear extinction. Furthermore, an M1 positive allosteric modulator enhanced the consolidation of fear extinction in the stress-enhanced fear-learning model, suggesting that M1 positive allosteric modulators may provide a novel treatment strategy to facilitate exposure therapy in the clinic for the treatment of posttraumatic stress disorder.


Assuntos
Medo/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Receptor Muscarínico M1/fisiologia , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Animais , Complexo Nuclear Basolateral da Amígdala/fisiologia , Condicionamento Clássico , Extinção Psicológica/fisiologia , Hipocampo/fisiologia , Depressão Sináptica de Longo Prazo , Núcleo Mediodorsal do Tálamo/fisiologia , Camundongos Endogâmicos C57BL , Vias Neurais/fisiologia , Transmissão Sináptica/fisiologia
10.
Pharmacol Biochem Behav ; 176: 43-52, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30444985

RESUMO

The non-selective muscarinic receptor agonist oxotremorine-M has been found to decrease impulsive choice in high-impulsive (HI) rats and increase impulsive choice in low-impulsive (LI) rats, but little is known about the muscarinic M1 receptor agonist N-desmethylclozapine (NDMC). This study investigated effects of NDMC on impulsive choice, and the effect of co-administration of NDMC with the dopamine D1-like receptor antagonist SCH 23390 or D2-like receptor antagonist raclopride on impulsive choice in HI and LI rats, characterized by basal levels of impulsive choice in a delay-discounting task. The results revealed that NDMC (1 and 2 mg/kg) significantly increased impulsive choice in HI, but not LI rats. SCH 23390 significantly promoted impulsive choice in HI rats at 0.01 mg/kg, and in LI rats at 0.0075 and 0.01 mg/kg. Moreover, SCH 23390 (0.005 and 0.0075 mg/kg) significantly inhibited the increase in impulsive choice induced by NDMC (1 mg/kg) in HI rats, whereas the increase in impulsive choice produced by SCH 23390 (0.0075 mg/kg) was significantly reversed by NDMC (1 mg/kg) in LI rats. Raclopride (0.04, 0.08, and 0.12 mg/kg) did not affect choice in both HI and LI rats, but significantly antagonized the increase in impulsive choice induced by NDMC (1 mg/kg) in HI rats. These findings suggest that D1- and D2-like receptors might be involved in different effects of the M1 receptor agonist on impulsive choice between HI and LI rats.


Assuntos
Comportamento Impulsivo/fisiologia , Receptor Muscarínico M1/fisiologia , Receptores de Dopamina D1/fisiologia , Receptores de Dopamina D2/fisiologia , Análise de Variância , Animais , Área Sob a Curva , Comportamento Animal/efeitos dos fármacos , Benzazepinas/administração & dosagem , Benzazepinas/farmacologia , Clozapina/administração & dosagem , Clozapina/análogos & derivados , Clozapina/farmacologia , Desvalorização pelo Atraso/efeitos dos fármacos , Desvalorização pelo Atraso/fisiologia , Antagonistas dos Receptores de Dopamina D2/administração & dosagem , Antagonistas dos Receptores de Dopamina D2/farmacologia , Comportamento Impulsivo/efeitos dos fármacos , Masculino , Racloprida/administração & dosagem , Racloprida/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor Muscarínico M1/agonistas , Receptores de Dopamina D1/antagonistas & inibidores
11.
ACS Chem Neurosci ; 10(3): 1035-1042, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30086237

RESUMO

This Letter describes the chemical optimization of a new series of muscarinic acetylcholine receptor subtype 1 (M1) positive allosteric modulators (PAMs) based on novel tricyclic triazolo- and imidazopyridine lactam cores, devoid of M1 agonism, e.g., no M1 ago-PAM activity, in high expressing recombinant cell lines. While all the new tricyclic congeners afforded excellent rat pharmacokinetic (PK) properties (CLp < 8 mL/min/kg and t1/2 > 5 h), regioisomeric triazolopyridine analogues were uniformly not CNS penetrant ( Kp < 0.05), despite a lack of hydrogen bond donors. However, removal of a single nitrogen atom to afford imidazopyridine derivatives proved to retain the excellent rat PK and provide high CNS penetration ( Kp > 2), despite inclusion of a basic nitrogen. Moreover, 24c was devoid of M1 agonism in high expressing recombinant cell lines and did not induce cholinergic seizures in vivo in mice. Interestingly, all of the new M1 PAMs across the diverse tricyclic heterocyclic cores possessed equivalent CNS MPO scores (>4.5), highlighting the value of both "medicinal chemist's eye" and experimental data, e.g., not sole reliance (or decision bias) on in silico calculated properties, for parameters as complex as CNS penetration.


Assuntos
Descoberta de Drogas/métodos , Imidazóis/farmacologia , Lactamas/farmacologia , Agonistas Muscarínicos/farmacologia , Piridinas/farmacologia , Receptor Muscarínico M1/agonistas , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Animais , Descoberta de Drogas/tendências , Humanos , Imidazóis/química , Lactamas/química , Camundongos , Agonistas Muscarínicos/química , Piridinas/química , Ratos , Receptor Muscarínico M1/fisiologia
12.
Pharmacology ; 103(1-2): 38-49, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30380560

RESUMO

BACKGROUND/AIMS: In this study, we evaluated the functional impact of facilitatory presynaptic adenosine A2A and muscarinic M1 receptors in the recovery of neuromuscular tetanic depression caused by the blockage of high-affinity choline transporter (HChT) by hemicholinium-3 (HC-3), a condition that mimics a myasthenia-like condition. METHODS: Rat diaphragm preparations were indirectly stimulated via the phrenic nerve trunk with 50-Hz frequency trains, each consisting of 500-750 supramaximal intensity pulses. The tension at the beginning (A) and at the end (B) of the tetanus was recorded and the ratio (R) B/A calculated. RESULTS: Activation of A2A and M1 receptors with CGS21680 (CGS; 2 nmol/L) and McN-A-343c (McN; 3 µmol/L) increased R values. Similar facilitatory effects were obtained with forskolin (FSK; 3 µmol/L) and phorbol 12-myristate 13-acetate (PMA; 10 µmol/L), which activate adenylate cyclase and protein kinase C respectively. HC-3 (4 µmol/L) decreased transmitter exocytosis measured by real-time videomicroscopy with the FM4-64 fluorescent dye and prevented the facilitation of neuromuscular transmission caused by CGS, McN, and FSK, with a minor effect on PMA. The acetylcholinesterase inhibitor, neostigmine (NEO; 0.5 µmol/L), also decreased transmitter exocytosis. The paradoxical neuromuscular tetanic fade caused by NEO (0.5 µmol/L) was also prevented by HC-3 (4 µmol/L) and might result from the rundown of the positive feedback mechanism operated by neuronal nicotinic receptors (blocked by hexamethonium, 120 µmol/L). CONCLUSION: Data suggest that the recovery of tetanic neuromuscular facilitation by adenosine A2A and M1 receptors is highly dependent on HChT activity and may be weakened in myasthenic patients when HChT is inoperative.


Assuntos
Proteínas de Membrana Transportadoras/fisiologia , Receptor A2A de Adenosina/fisiologia , Receptor Muscarínico M1/fisiologia , Período Refratário Eletrofisiológico/efeitos dos fármacos , Cloreto de (4-(m-Clorofenilcarbamoiloxi)-2-butinil)trimetilamônio/farmacologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Colforsina/farmacologia , Diafragma/efeitos dos fármacos , Diafragma/fisiologia , Hemicolínio 3/farmacologia , Neostigmina/farmacologia , Fenetilaminas/farmacologia , Nervo Frênico/efeitos dos fármacos , Nervo Frênico/fisiologia , Ratos , Ratos Wistar , Transmissão Sináptica , Tétano/tratamento farmacológico , Tétano/fisiopatologia , Acetato de Tetradecanoilforbol/farmacologia
13.
Eur J Pharmacol ; 843: 104-112, 2019 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-30452911

RESUMO

Muscarinic receptor stimulation induces depolarizing inward currents and catecholamine secretion in adrenal medullary (AM) cells from various mammals. In guinea-pig AM cells muscarine and oxotremorine at concentrations ≤ 1 µM produce activation of nonselective cation channels with a similar potency and efficacy, whereas muscarine at higher concentrations produces not only nonselective cation channel activation, but also TASK1 channel inhibition. In rat AM cells, the muscarinic M1 receptor is involved in TASK1 channel inhibition in response to muscarinic agonists, and the efficacy of oxotremorine is half that of muscarine. These pharmacological findings might indicate that different muscarinic receptor subtypes are responsible for the regulation of nonselective cation and TASK1 channel activities. The present study aimed to determine the muscarinic receptor subtypes involved in nonselective cation channel activation in guinea-pig and mouse AM cells. The inward current evoked by 1 µM muscarine was completely suppressed by 100 µM quinine, whereas 30 µM muscarine-induced inward currents were comprised of quinine-sensitive and -insensitive components. The electrophysiological and pharmacological properties of the muscarine-induced currents indicated that the quinine-sensitive and insensitive components are due to nonselective cation channel activation and TASK1 channel inhibition, respectively. Muscarine at 30 µM failed to induce any current in AM cells treated with muscarinic toxin 7 or genetically deleted of the M1 receptor. The KD value of VU0255035 against the muscarinic receptor mediating nonselective cation channel activation was 17.5 nM. These results indicate that the M1 receptor mediates nonselective cation channel activation as well as TASK1 channel inhibition.


Assuntos
Medula Suprarrenal/citologia , Canais Iônicos/fisiologia , Agonistas Muscarínicos/farmacologia , Receptor Muscarínico M1/fisiologia , Animais , Cobaias , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muscarina/farmacologia , Oxotremorina/farmacologia , Quinina/farmacologia
14.
ACS Chem Neurosci ; 10(3): 1099-1114, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30547573

RESUMO

Targeting allosteric sites of the M1 muscarinic acetylcholine receptor (mAChR) is an enticing approach to overcome the lack of receptor subtype selectivity observed with orthosteric ligands. This is a promising strategy for obtaining novel therapeutics to treat cognitive deficits observed in Alzheimer's disease and schizophrenia, while reducing the peripheral side effects such as seen in the current treatment regimes, which are non-subtype selective. We previously described compound 2, the first positive allosteric modulator (PAM) of the M1 mAChR based on a 6-phenylpyrimidin-4-one scaffold, which has been further developed in this study. Herein, we present the synthesis, characterization, and pharmacological evaluation of a series of 6-phenylpyrimidin-4-ones with modifications to the 4-(1-methylpyrazol-4-yl)benzyl pendant. Selected compounds, BQCA, 1, 2, 9i, 13, 14b, 15c, and 15d, were further profiled in terms of their allosteric affinity, cooperativity with acetylcholine (ACh), and intrinsic efficacy. Additionally, 2 and 9i were tested in mouse primary cortical neurons, displaying various degrees of intrinsic agonism and potentiation of the acetylcholine response. Overall, the results suggest that the pendant moiety is important for allosteric binding affinity and the direct agonistic efficacy of the 6-phenylpyrimidin-4-one based M1 mAChR PAMs.


Assuntos
Pirimidinas/síntese química , Pirimidinas/farmacologia , Quinolinas/síntese química , Quinolinas/farmacologia , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/fisiologia , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Cristalografia por Raios X/métodos , Camundongos
15.
PLoS One ; 13(11): e0206532, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30395575

RESUMO

BACKGROUND: Electrical vagal nerve stimulation (VNS) has been used for years to treat patients with drug-resistant epilepsy. This technique also remains under investigation as a specific treatment of patients with Alzheimer's disease. Recently we discovered that VNS induced hippocampal formation (HPC) type II theta rhythm, which is involved in memory consolidation. In the present study, we have extended our previous observation and addressed the neuronal substrate and pharmacological profile of HPC type II theta rhythm induced by VNS in anesthetized rats. METHODS: Male Wistar rats were implanted with a VNS cuff electrode around the left vagus nerve, a tungsten microelectrode for recording the HPC field activity, and a medial septal (MS) cannula for the injection of a local anesthetic, procaine, and muscarinic agents. A direct, brief effect of VNS on the HPC field potential was evaluated before and after medial-septal drug injection. RESULTS: Medial septal injection of local anesthetic, procaine, reversibly abolished VNS-induced HPC theta rhythm. With the use of cholinergic muscarinic agonist and antagonists, we demonstrated that medial septal M1 receptors are involved in the mediation of the VNS effect on HPC theta field potential. CONCLUSION: The MS cholinergic M1 receptor mechanism integrates not only central inputs from the brainstem synchronizing pathway, which underlies the production of HPC type II theta rhythm, but also the input from the vagal afferents in the brain stem.


Assuntos
Hipocampo/fisiologia , Ritmo Teta/fisiologia , Estimulação do Nervo Vago , Cloreto de (4-(m-Clorofenilcarbamoiloxi)-2-butinil)trimetilamônio/administração & dosagem , Anestésicos Locais/administração & dosagem , Animais , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/fisiologia , Colinérgicos/administração & dosagem , Fibras Colinérgicas/efeitos dos fármacos , Fibras Colinérgicas/fisiologia , Hipocampo/efeitos dos fármacos , Masculino , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Procaína/administração & dosagem , Ratos , Ratos Wistar , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/fisiologia , Núcleos Septais/efeitos dos fármacos , Núcleos Septais/fisiologia , Ritmo Teta/efeitos dos fármacos
16.
J Neurosci ; 38(43): 9252-9262, 2018 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-30242046

RESUMO

Hebbian synaptic plasticity at hippocampal Schaffer collateral synapses is tightly regulated by postsynaptic small conductance (SK) channels that restrict NMDA receptor activity. SK channels are themselves modulated by G-protein-coupled signaling pathways, but it is not clear under what conditions these are activated to enable synaptic plasticity. Here, we show that muscarinic M1 receptor (M1R) and type 1 metabotropic glutamate receptor (mGluR1) signaling pathways, which are known to inhibit SK channels and thereby disinhibit NMDA receptors, converge to facilitate spine calcium transients during the induction of long-term potentiation (LTP) at hippocampal Schaffer collateral synapses onto CA1 pyramidal neurons of male rats. Furthermore, mGluR1 activation is required for LTP induced by reactivated place-cell firing patterns that occur in sharp-wave ripple events during rest or sleep. In contrast, M1R activation is required for LTP induced by place-cell firing patterns during exploration. Thus, we describe a common mechanism that enables synaptic plasticity during both encoding and consolidation of memories within hippocampal circuits.SIGNIFICANCE STATEMENT Memory ensembles in the hippocampus are formed during active exploration and consolidated during rest or sleep. These two distinct phases each require strengthening of synaptic connections by long-term potentiation (LTP). The neuronal activity patterns in each phase are very different, which makes it hard to map generalized rules for LTP induction onto both formation and consolidation phases. In this study, we show that inhibition of postsynaptic SK channels is a common necessary feature of LTP induction and that SK channel inhibition is achieved by separate but convergent metabotropic signaling pathways. Thus, we reveal a common mechanism for enabling LTP under distinct behavioral conditions.


Assuntos
Hipocampo/fisiologia , Plasticidade Neuronal/fisiologia , Receptor Muscarínico M1/fisiologia , Transdução de Sinais/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia , Animais , Apamina/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/química , Hipocampo/efeitos dos fármacos , Masculino , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Plasticidade Neuronal/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Receptor Muscarínico M1/agonistas , Transdução de Sinais/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Baixa/química
17.
BMC Res Notes ; 11(1): 681, 2018 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-30261922

RESUMO

OBJECTIVES: We examined whether two G protein-coupled receptors (GPCRs), muscarinic M1 receptors (M1Rs) and dopaminergic D2 receptors (D2Rs), utilize endogenously released fatty acid to inhibit L-type Ca2+ channels, CaV1.3. HEK-293 cells, stably transfected with M1Rs, were used to transiently transfect D2Rs and CaV1.3b with different CaVß-subunits, allowing for whole-cell current measurement from a pure channel population. RESULTS: M1R activation with Oxotremorine-M inhibited currents from CaV1.3b coexpressed with α2δ-1 and a ß1b, ß2a, ß3, or ß4-subunit. Surprisingly, the magnitude of inhibition was less with ß2a than with other CaVß-subunits. Normalizing currents revealed kinetic changes after modulation with ß1b, ß3, or ß4, but not ß2a-containing channels. We then examined if D2Rs modulate CaV1.3b when expressed with different CaVß-subunits. Stimulation with quinpirole produced little inhibition or kinetic changes for CaV1.3b coexpressed with ß2a or ß3. However, quinpirole inhibited N-type Ca2+ currents in a concentration-dependent manner, indicating functional expression of D2Rs. N-current inhibition by quinpirole was voltage-dependent and independent of phospholipase A2 (PLA2), whereas a PLA2 antagonist abolished M1R-mediated N-current inhibition. These findings highlight the specific regulation of Ca2+ channels by different GPCRs. Moreover, tissue-specific and/or cellular localization of CaV1.3b with different CaVß-subunits could fine tune the response of Ca2+ influx following GPCR activation.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Receptor Muscarínico M1/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Cálcio , Células HEK293 , Humanos , Receptores de Dopamina D2
18.
Cancer Discov ; 8(11): 1458-1473, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30185628

RESUMO

In many solid tumors, parasympathetic input is provided by the vagus nerve, which has been shown to modulate tumor growth. However, whether cholinergic signaling directly regulates progression of pancreatic ductal adenocarcinoma (PDAC) has not been defined. Here, we found that subdiaphragmatic vagotomy in LSL-Kras +/G12D;Pdx1-Cre (KC) mice accelerated PDAC development, whereas treatment with the systemic muscarinic agonist bethanechol restored the normal KC phenotype, thereby suppressing the accelerated tumorigenesis caused by vagotomy. In LSL-Kras +/G12D;LSL-Trp53 +/R172H;Pdx1-Cre mice with established PDAC, bethanechol significantly extended survival. These effects were mediated in part through CHRM1, which inhibited downstream MAPK/EGFR and PI3K/AKT pathways in PDAC cells. Enhanced cholinergic signaling led to a suppression of the cancer stem cell (CSC) compartment, CD11b+ myeloid cells, TNFα levels, and metastatic growth in the liver. Therefore, these data suggest that cholinergic signaling directly and indirectly suppresses growth of PDAC cells, and therapies that stimulate muscarinic receptors may be useful in the treatment of PDAC.Significance: Subdiaphragmatic vagotomy or Chrm1 knockout accelerates pancreatic tumorigenesis, in part via expansion of the CSC compartment. Systemic administration of a muscarinic agonist suppresses tumorigenesis through MAPK and PI3K/AKT signaling, in early stages of tumor growth and in more advanced, metastatic disease. Therefore, CHRM1 may represent a potentially attractive therapeutic target. Cancer Discov; 8(11); 1458-73. ©2018 AACR. This article is highlighted in the In This Issue feature, p. 1333.


Assuntos
Carcinoma Ductal Pancreático/prevenção & controle , Transformação Celular Neoplásica/efeitos dos fármacos , Colinérgicos/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias Pancreáticas/prevenção & controle , Receptor Muscarínico M1/fisiologia , Animais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Genes ras , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Transdução de Sinais , Neoplasias Pancreáticas
19.
J Pharmacol Sci ; 137(2): 233-236, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30078433

RESUMO

The role of the GABAB receptor in the anterior cingulate cortex (ACC) of neuropathic pain is unclear. Injection of a GABAB receptor antagonist CGP35348 into the ACC induced mechanical hypersensitivity in normal rats. Activation of the GABAB receptor injected by a GABAB receptor agonist baclofen into the ACC attenuated mechanical hypersensitivity in partial sciatic nerve ligation (PSNL) rats. Co-microinjection of CGP35348 with a muscarinic M1 receptor agonist McN-A-343 into the ACC significantly inhibited McN-A-343-induced antihypersensitivity in PSNL rats. These results suggest that the GABAB receptor in the ACC contributes to mechanical hypersensitivity and is involved in muscarinic M1 receptor-mediated antihypersensitivity.


Assuntos
Giro do Cíngulo , Hiperalgesia/genética , Neuralgia/genética , Receptores de GABA-B/fisiologia , Nervo Isquiático , Animais , Baclofeno/uso terapêutico , Modelos Animais de Doenças , Agonistas dos Receptores de GABA-B/uso terapêutico , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Ligadura , Masculino , Neuralgia/tratamento farmacológico , Ratos Wistar , Receptor Muscarínico M1/fisiologia
20.
J Pharmacol Exp Ther ; 364(1): 28-37, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29025977

RESUMO

Activation of muscarinic M1 receptor (M1R) is a promising approach for improving cognitive impairment in Alzheimer's disease. However, an M1R-selective positive allosteric modulator (PAM), benzyl quinolone carboxylic acid (BQCA), at 30 mg/kg, induced diarrhea in wild-type mice, but not in M1R knockout mice. Moreover, BQCA (0.1-1000 nM) augmented electric field stimulation (EFS)-induced ileum contraction in an in vitro Magnus assay. Thus, we decided to establish a drug-screening strategy to discover novel M1 PAMs producing potent cognitive improvement with minimized gastrointestinal (GI) dysfunction. We assessed PAM parameters of various M1 PAMs with ≥100-fold selectivity over other muscarinic receptor subtypes by using in vitro binding and functional analysis. Evaluation of these M1 PAMs in the Magnus assay revealed a significant correlation between percentage of ileum contractions at 1 µM and their α-value, a PAM parameter associated with the binding cooperativity between acetylcholine and M1 PAM. M1 PAMs with lower α-value showed lower impact on EFS-induced ileum contraction. Next, we characterized in vivo profiles of two M1 PAMs: compound A (log α = 1.18) and compound B (log α = 3.30). Compound A, at 30 mg/kg, significantly improved scopolamine-induced cognitive deficits without prominent signs of diarrhea at up to 1000 mg/kg in mice. In contrast, compound B, at 10 mg/kg, showed both significant improvement of scopolamine-induced cognitive deficits and severe diarrhea. Thus, fine adjustment of the α-values could be a key to discovering M1 PAMs yielding potent cognitive improvement with a lower risk of GI effects.


Assuntos
Transtornos Cognitivos/tratamento farmacológico , Diarreia/prevenção & controle , Descoberta de Drogas/métodos , Íleo/efeitos dos fármacos , Nootrópicos/uso terapêutico , Receptor Muscarínico M1/agonistas , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Animais , Células CHO , Transtornos Cognitivos/fisiopatologia , Cricetinae , Cricetulus , Diarreia/induzido quimicamente , Diarreia/fisiopatologia , Relação Dose-Resposta a Droga , Humanos , Íleo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Nootrópicos/efeitos adversos , Nootrópicos/farmacologia , Técnicas de Cultura de Órgãos , Quinolinas/efeitos adversos , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Distribuição Aleatória , Receptor Muscarínico M1/fisiologia , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA