Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Psychiatry Res ; 341: 116156, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39236366

RESUMO

We are studying the molecular pathology of a sub-group within schizophrenia (∼ 25 %: termed Muscarinic Receptor Deficit subgroup of Schizophrenia (MRDS)) who can be separated because they have very low levels of cortical muscarinic M1 receptors (CHRM1). Based on our transcriptomic data from Brodmann's area ((BA) 9, 10 and 33 (controls, schizophrenia and mood disorders) and the cortex of the CHRM1-/- mouse (a molecular model of aberrant CHRM1 signaling), we predicted levels of AKT interacting protein (AKTIP), but not tubulin alpha 1b (TUBA1B) or AKT serine/threonine kinase 1 (AKT1) and pyruvate dehydrogenase kinase 1 (PDK1) (two AKTIP-functionally associated proteins), would be changed in MRDS. Hence, we used Western blotting to measure AKTIP (BA 10: controls, schizophrenia and mood disorders; BA 9: controls and schizophrenia) plus TUBA1B, AKT1 and PDK1 (BA 10: controls and schizophrenia) proteins. The only significant change with diagnosis was higher levels of AKTIP protein in BA 10 (Cohen's d = 0.73; p = 0.02) in schizophrenia compared to controls due to higher levels of AKTIP only in people with MRDS (Cohen's d = 0.80; p = 0.03). As AKTIP is involved in AKT1 signaling, our data suggests that signaling pathway is particularly disturbed in BA 10 in MRDS.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Receptor Muscarínico M1 , Esquizofrenia , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Lobo Frontal/metabolismo , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil/metabolismo , Receptor Muscarínico M1/metabolismo , Esquizofrenia/metabolismo , Tubulina (Proteína)/metabolismo
2.
Br J Pharmacol ; 181(17): 3064-3081, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38689378

RESUMO

BACKGROUND AND PURPOSE: Current pharmacotherapies for Tourette syndrome (TS) are often unsatisfactory and poorly tolerated, underscoring the need for novel treatments. Insufficient striatal acetylcholine has been suggested to contribute to tic ontogeny. Thus, we tested whether activating M1 and/or M4 receptors-the two most abundant muscarinic receptors in the striatum-reduced tic-related behaviours in mouse models of TS. EXPERIMENTAL APPROACH: Studies were conducted using CIN-d and D1CT-7 mice, two TS models characterized by early-life depletion of striatal cholinergic interneurons and cortical neuropotentiation, respectively. First, we tested the effects of systemic and intrastriatal xanomeline, a selective M1/M4 receptor agonist, on tic-like and other TS-related responses. Then, we examined whether xanomeline effects were reduced by either M1 or M4 antagonists or mimicked by the M1/M3 agonist cevimeline or the M4 positive allosteric modulator (PAM) VU0467154. Finally, we measured striatal levels of M1 and M4 receptors and assessed the impact of VU0461754 on the striatal expression of the neural marker activity c-Fos. KEY RESULTS: Systemic and intrastriatal xanomeline reduced TS-related behaviours in CIN-d and D1CT-7 mice. Most effects were blocked by M4, but not M1, receptor antagonists. VU0467154, but not cevimeline, elicited xanomeline-like ameliorative effects in both models. M4, but not M1, receptors were down-regulated in the striatum of CIN-d mice. Additionally, VU0467154 reduced striatal c-Fos levels in these animals. CONCLUSION AND IMPLICATIONS: Activation of striatal M4, but not M1, receptors reduced tic-like manifestations in mouse models, pointing to xanomeline and M4 PAMs as novel putative therapeutic strategies for TS.


Assuntos
Corpo Estriado , Modelos Animais de Doenças , Agonistas Muscarínicos , Receptor Muscarínico M4 , Síndrome de Tourette , Animais , Síndrome de Tourette/metabolismo , Síndrome de Tourette/tratamento farmacológico , Receptor Muscarínico M4/metabolismo , Receptor Muscarínico M4/agonistas , Receptor Muscarínico M4/antagonistas & inibidores , Camundongos , Corpo Estriado/metabolismo , Corpo Estriado/efeitos dos fármacos , Masculino , Agonistas Muscarínicos/farmacologia , Comportamento Animal/efeitos dos fármacos , Piridinas/farmacologia , Tiques/tratamento farmacológico , Tiques/metabolismo , Tiofenos/farmacologia , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M1/agonistas , Dioxóis/farmacologia , Camundongos Endogâmicos C57BL , Tiadiazóis
3.
Brain Behav ; 14(5): e3507, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38688895

RESUMO

INTRODUCTION: Alzheimer's disease (AD) is a neurodegenerative condition characterized by gradual loss of cognitive abilities (dementia) and is a major public health problem. Here, we aimed at investigating the effects of Rosa damascena essential oil (RDEO) on learning and memory functions in a rat model of amnesia induced by scopolamine, as well as on changes in acetylcholinesterase (AChE) activity, M1 muscarinic acetylcholine receptor (mAChR) expression, and brain-derived neurotrophic factor (BDNF) levels in the extracted brain tissues. METHODS: The control, amnesia (scopolamine, 1 mg/kg/i.p.) and treatment (RDEO, 100 µL/kg/p.o. or galantamine, 1.5 mg/kg/i.p.) groups were subjected to Morris water maze and new object recognition tests. AChE activity was assayed by ELISA, and M1 mAChR and BDNF concentration changes were determined by western blotting. Also, using computational tools, human M1 mAChR was modeled in an active conformation, and the major components of RDEO were docked onto this receptor. RESULTS: According to our behavioral tests, RDEO was able to mitigate the learning and memory impairments caused by scopolamine in vivo. Our in vitro assays showed that the observed positive effects correlated well with a decrease in AChE activity and an increase in M1 mAChR and BDNF levels in amnestic rat brains. We also demonstrated in an in silico setting that the major components of RDEO, specifically -citronellol, geraniol, and nerol, could be accommodated favorably within the allosteric binding pocket of active-state human M1 mAChR and anchored here chiefly by hydrogen-bonding and alkyl-π interactions. CONCLUSION: Our findings offer a solid experimental foundation for future RDEO-based medicinal product development for patients suffering from AD.


Assuntos
Acetilcolinesterase , Amnésia , Fator Neurotrófico Derivado do Encéfalo , Óleos Voláteis , Rosa , Escopolamina , Animais , Ratos , Amnésia/induzido quimicamente , Amnésia/tratamento farmacológico , Amnésia/metabolismo , Óleos Voláteis/farmacologia , Óleos Voláteis/administração & dosagem , Masculino , Rosa/química , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Acetilcolinesterase/metabolismo , Receptor Muscarínico M1/metabolismo , Ratos Wistar , Nootrópicos/farmacologia , Modelos Animais de Doenças , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Cognição/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos
4.
Proc Natl Acad Sci U S A ; 119(24): e2201103119, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35671422

RESUMO

The quaternary organization of rhodopsin-like G protein-coupled receptors in native tissues is unknown. To address this we generated mice in which the M1 muscarinic acetylcholine receptor was replaced with a C-terminally monomeric enhanced green fluorescent protein (mEGFP)-linked variant. Fluorescence imaging of brain slices demonstrated appropriate regional distribution, and using both anti-M1 and anti-green fluorescent protein antisera the expressed transgene was detected in both cortex and hippocampus only as the full-length polypeptide. M1-mEGFP was expressed at levels equal to the M1 receptor in wild-type mice and was expressed throughout cell bodies and projections in cultured neurons from these animals. Signaling and behavioral studies demonstrated M1-mEGFP was fully active. Application of fluorescence intensity fluctuation spectrometry to regions of interest within M1-mEGFP-expressing neurons quantified local levels of expression and showed the receptor was present as a mixture of monomers, dimers, and higher-order oligomeric complexes. Treatment with both an agonist and an antagonist ligand promoted monomerization of the M1-mEGFP receptor. The quaternary organization of a class A G protein-coupled receptor in situ was directly quantified in neurons in this study, which answers the much-debated question of the extent and potential ligand-induced regulation of basal quaternary organization of such a receptor in native tissue when present at endogenous expression levels.


Assuntos
Córtex Cerebral , Hipocampo , Receptor Muscarínico M1 , Animais , Córtex Cerebral/metabolismo , Proteínas de Fluorescência Verde , Hipocampo/metabolismo , Ligantes , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Imagem Óptica , Receptor Muscarínico M1/química , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/metabolismo
5.
Bioengineered ; 13(3): 4898-4910, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35156515

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder with prominent individual morbidity and mortality among elderly people. Germacrone (Germ) has been reported to exert dominant protective roles in multiple human diseases, and neurological diseases are also included. The intention of this paper is to determine the impacts of Germ on okadaic acid (OA)-treated PC12 cells and confirm the hidden regulatory mechanism. First, PC12 cells were induced by OA in the absence or presence of Germ. Cell counting kit-8 assay was to monitor cell proliferation. Western blot was to test the protein levels of cholinergic muscarinic M1 receptor (CHRM1), Galphaq (Gq), phospholipase C beta (PLCß) and protein kinase C (PKC). The levels of reactive oxygen species (ROS) and other oxidative stress markers were evaluated using corresponding kits. ELISA was used to estimate the levels of AD markers. RT-qPCR was used to examine the mRNA levels of beta-site amyloid-precursor-protein-cleaving enzyme 1 (BACE-1) and apolipoprotein E (APOE). The results uncovered that Germ enhanced the proliferation of OA-insulted PC12 cells, elevated the protein level of CHRM1 and activated the Gq/PLCß/PKC signaling. Moreover, after OA-induced PC12 cells were administered with Germ, insufficiency of CHRM1 impeded cell proliferation, enhanced oxidative stress and neuron injury and inactivated the Gq/PLCß/PKC signaling. Furthermore, the addition of Gq inhibitor UBO-QIC, PLCß inhibitor U73122 or PKC inhibitor Go6983 reversed the enhanced proliferation, the reduced oxidative stress and neuron injury in OA-treated PC12 cells caused by Germ. Collectively, Germ modulated M1 muscarinic receptor-mediated Gq/PLCß/PKC signaling, thereby alleviating OA-induced PC12 cell injury.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Fosfolipase C beta , Proteína Quinase C , Sesquiterpenos de Germacrano , Animais , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Ácido Okadáico , Células PC12 , Fosfolipase C beta/metabolismo , Proteína Quinase C/metabolismo , Ratos , Receptor Muscarínico M1/metabolismo , Sesquiterpenos de Germacrano/farmacologia
6.
Epigenetics ; 17(2): 133-146, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33491544

RESUMO

Neural tube defects (NTDs) are a group of common and severe congenital malformations. The PI3K-AKT signalling pathway plays a crucial role in the neural tube development. There is limited evidence concerning any possible association between aberrant methylation in PI3K-AKT signalling pathway genes and NTDs. Therefore, we aimed to investigate potential associations between aberrant methylation of PI3K-AKT pathway genes and NTDs. Methylation studies of PI3K-AKT pathway genes utilizing microarray genome-methylation data derived from neural tissues of ten NTD cases and eight non-malformed controls were performed. Targeted DNA methylation analysis was subsequently performed in an independent cohort of 73 NTD cases and 32 controls to validate the methylation levels of identified genes. siRNAs were used to pull-down the target genes in human embryonic stem cells (hESCs) to examine the effects of the aberrant expression of target genes on neural cells. As a result, 321 differentially hypermethylated CpG sites in the promoter regions of 30 PI3K-AKT pathway genes were identified in the microarray data. In target methylation analysis, CHRM1, FGF19, and ITGA7 were confirmed to be significantly hypermethylated in NTD cases and were associated with increased risk for NTDs. The down-regulation of FGF19, CHRM1, and ITGA7 impaired the formation of rosette-like cell aggregates. The down-regulation of those three genes affected the expression of PAX6, SOX2 and MAP2, implying their influence on the differentiation of neural cells. This study for the first time reported that hypermethylation of PI3K-AKT pathway genes such as CHRM1, FGF19, and ITGA7 is associated with human NTDs.


Assuntos
Defeitos do Tubo Neural , Proteínas Proto-Oncogênicas c-akt , Antígenos CD/genética , Antígenos CD/metabolismo , Metilação de DNA , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Cadeias alfa de Integrinas/genética , Cadeias alfa de Integrinas/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/metabolismo , Transdução de Sinais
7.
Neurobiol Learn Mem ; 181: 107445, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33895349

RESUMO

In rodent models of smoking during pregnancy, early postnatal nicotine exposure results in impaired hippocampus-dependent memory, but the underlying mechanism remains elusive. Given that hippocampal cholinergic systems modulate memory and rapid development of hippocampal cholinergic systems occurs during nicotine exposure, here we investigated its impacts on cholinergic function. Both nicotinic and muscarinic activation produce transient or long-lasting depression of excitatory synaptic transmission in the hippocampal CA1 region. We found that postnatal nicotine exposure impairs both the induction and nicotinic modulation of NMDAR-dependent long-term depression (LTD). Activation of muscarinic receptors decreases excitatory synaptic transmission and CA1 network activity in both wild-type and α2 knockout mice. These muscarinic effects are still observed in nicotine-exposed mice. M1 muscarinic receptor activity is required for mGluR-dependent LTD. Early postnatal nicotine exposure has no effect on mGluR-dependent LTD induction, suggesting that it has no effect on the function of m1 muscarinic receptors involved in this form of LTD. Our results demonstrate that early postnatal nicotine exposure has more pronounced effects on nicotinic function than muscarinic function in the hippocampal CA1 region. Thus, impaired hippocampus-dependent memory may arise from the developmental disruption of nicotinic cholinergic systems in the hippocampal CA1 region.


Assuntos
Região CA1 Hipocampal/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Receptor Muscarínico M1/efeitos dos fármacos , Receptores Nicotínicos/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Região CA1 Hipocampal/crescimento & desenvolvimento , Região CA1 Hipocampal/metabolismo , Fumar Cigarros , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Lactação , Depressão Sináptica de Longo Prazo/fisiologia , Masculino , Exposição Materna , Memória/efeitos dos fármacos , Memória/fisiologia , Camundongos , Camundongos Knockout , Receptor Muscarínico M1/metabolismo , Receptores de Glutamato Metabotrópico/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/metabolismo , Receptores Muscarínicos/efeitos dos fármacos , Receptores Muscarínicos/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Nicotínicos/metabolismo
8.
Psychopharmacology (Berl) ; 238(7): 1953-1964, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33735392

RESUMO

RATIONALE: In addition to the disease-defining motor symptoms, patients with Parkinson's disease (PD) exhibit gait dysfunction, postural instability, and a propensity for falls. These dopamine (DA) replacement-resistant symptoms in part have been attributed to loss of basal forebrain (BF) cholinergic neurons and, in interaction with striatal dopamine (DA) loss, to the resulting disruption of the attentional control of balance and complex movements. Rats with dual cholinergic-DA losses ("DL rats") were previously demonstrated to model PD falls and associated impairments of gait and balance. OBJECTIVES: We previously found that the muscarinic M1-positive allosteric modulator (PAM) TAK-071 improved the attentional performance of rats with BF cholinergic losses. Here, we tested the hypotheses that TAK-071 reduces fall rates in DL rats. RESULTS: Prior to DL surgery, female rats were trained to traverse a rotating straight rod as well as a rod with two zigzag segments. DL rats were refamiliarized with such traversals post-surgery and tested over 7 days on increasingly demanding testing conditions. TAK-071 (0.1, 0.3 mg/kg, p.o.) was administered prior to daily test sessions over this 7-day period. As before, DL rats fell more frequently than sham-operated control rats. Treatment of DL rats with TAK-071 reduced falls from the rotating rod and the rotating zigzag rod, specifically when the angled part of the zigzag segment, upon entering, was at a steep, near vertical angle. CONCLUSIONS: TAK-071 may benefit complex movement control, specifically in situations which disrupt the patterning of forward movement and require the interplay between cognitive and motor functions to modify movement based on information about the state of dynamic surfaces, balance, and gait.


Assuntos
Acidentes por Quedas/prevenção & controle , Agonistas Muscarínicos/uso terapêutico , Oxidopamina/toxicidade , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/tratamento farmacológico , Receptor Muscarínico M1/agonistas , Administração Oral , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Animais , Prosencéfalo Basal/efeitos dos fármacos , Prosencéfalo Basal/metabolismo , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios Colinérgicos/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Dopamina/metabolismo , Feminino , Agonistas Muscarínicos/farmacologia , Transtornos Parkinsonianos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Muscarínico M1/metabolismo
9.
Free Radic Res ; 55(1): 11-25, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33222572

RESUMO

In the present study, we examined whether glutathione peroxidase-1 (GPx-1), a major H2O2 scavenger in the brain, affects memory deficits induced by Aß (1-42) in mice. Treatment with 400 pmol/5 µl Aß (1-42) (i.c.v.) resulted in a reduction of GPx-1 expression in wild-type (WT) mice. An Aß (1-42)-induced reduction in acetylcholine (ACh) level was observed in the hippocampus. Treatment with Aß (1-42) consistently resulted in reduced expression and activity of choline acetyltransferase (ChAT) and in an increase in expression and activity of acetylcholinesterase (AChE). Upon examining each of the muscarinic acetylcholine receptors (mAChRs) and nicotinic AChRs, we noted that Aß (1-42) treatment selectively reduced the levels of M1 mAChR. In addition, Aß (1-42) induced a significant reduction in phospho-cAMP response element-binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) expression. The cholinergic impairments induced by Aß (1-42) were more pronounced in GPx-1 knockout mice than in WT mice. Importantly, an adenoviral vector encoded with the GPx-1 gene (Ad-GPx-1) significantly rescued Aß (1-42)-induced cholinergic impairments in GPx-1 knockout mice. In addition, M1 mAChR antagonist dicyclomine significantly counteracted Ad-GPx-1-mediated increases in p-CREB and BDNF expression, as well as memory-enhancing effects in GPx-1 knockout mice, thus indicating that M1 mAChR might be a critical mediator for the rescue effects of Ad-GPx-1. Combined, our results suggest that GPx-1 gene protected against Aß (1-42)-induced memory impairments via activation of M1 mAChR-dependent CREB/BDNF signalling.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Glutationa Peroxidase/genética , Transtornos da Memória/induzido quimicamente , Fragmentos de Peptídeos/farmacologia , Receptor Muscarínico M1/metabolismo , Acetilcolina/metabolismo , Adenoviridae/genética , Animais , Modelos Animais de Doenças , Vetores Genéticos/genética , Glutationa Peroxidase/administração & dosagem , Glutationa Peroxidase/biossíntese , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Transtornos da Memória/genética , Camundongos , Camundongos Knockout , Transdução de Sinais/efeitos dos fármacos , Glutationa Peroxidase GPX1
10.
Anticancer Drugs ; 31(9): 908-917, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32097138

RESUMO

Hepatocellular carcinoma is the second leading cause of cancer-related death worldwide. Neural regulation plays an important role in the development of hepatocellular carcinoma, and activation of sympathetic nervous system can promote the migration and invasion of cancer cells. However, little research has been conducted on how the vagus nerve influences hepatocellular carcinoma. In this study, we found that the expression of vesicular acetylcholine transporter, a biomarker of vagus nerve, was associated with hepatocellular carcinoma patients' clinicopathological characteristics by immunohistochemistry. Further, activation of muscarinic acetylcholine receptor 1 (M1R) promoted HepG2 and SMMC-7721 cells migration and invasion and epithelial-mesenchymal transition via PI3K/Akt pathway. Moreover, inhibition of M1R by antagonist or shRNA suppressed hepatocellular carcinoma cells migration and invasion in vitro and in vivo, inhibited epithelial-mesenchymal transition and PI3K/Akt pathway. Therefore, these results indicate that activation of M1R promotes invasion of hepatocellular carcinoma through epithelial-mesenchymal transition and PI3K/Akt pathway.


Assuntos
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Receptor Muscarínico M1/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Transição Epitelial-Mesenquimal , Feminino , Células Hep G2 , Xenoenxertos , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Nervo Vago/patologia
11.
J Cell Physiol ; 235(10): 7056-7066, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31994734

RESUMO

TRPC5 channels are nonselective cation channels activated by G-protein-coupled receptors. It was previously found that recombinant TRPC5 currents are inhibited by intracellular ATP, when studied by whole-cell patch-clamp recording. In the present study, we investigated the mechanism of ATP inhibition at the single-channel level using patches from HEK-293 cells transiently transfected with TRPC5 and the M1 muscarinic receptor. In inside-out patches, application of ATP to the intracellular face of the membrane reduced TRPC5 channel activity at both positive and negative potentials without affecting the unitary current amplitude or open dwell time of the channel. The effect of ATP was rapidly reversible. These results suggest that ATP may bind to the channel protein and affect the ability of the channel to open or to remain in an open, nondesensitized state. The activity of TRPC5 channels may be influenced by cellular metabolism via changes in ATP levels.


Assuntos
Trifosfato de Adenosina/metabolismo , Potenciais da Membrana/fisiologia , Canais de Cátion TRPC/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Células HEK293 , Humanos , Técnicas de Patch-Clamp/métodos , Receptor Muscarínico M1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
12.
Cell Mol Neurobiol ; 40(4): 547-554, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31721013

RESUMO

M1 muscarinic acetylcholine receptors (M1 mAChRs) have long been an attractive target for the treatment of Alzheimer's disease (AD), the most common cause of dementia in the elderly. M1 mAChR agonists show desirably preclinical activities; however, most have not gone further into late clinical trials due to ineffectiveness or side effects. Thus, to understand the signaling pathways involved in M1 mAChR-mediated memory improvement may be important for design of biased agonists with on-target therapeutic effects. M1 mAChRs are classically coupled to Gαq or ectopically to Gαs to activate multiple kinases such as protein kinase C (PKC), Ras and protein kinase A (PKA). Our previous studies have found that M1 mAChRs could improve learning and memory through modulating AMPA receptor GluA1 subunit via PKA-PI3K-Akt signaling. Here, we further investigated whether PKC and Ras were involved in M1 mAChR-mediated modulation of GluA1. We demonstrated the role of PKC and Ras in the signaling pathway, as both PKC inhibitors Ro-31-8425 or Gö6983 and Ras inhibitor salirasib abolished the membrane insertion of GluA1 and enhancement of its phosphorylation at Ser845 induced by M1 mAChRs in the primary cultured neurons and hippocampus in vivo. We further showed that PKC and Ras modulated PKA-PI3K-Akt signaling since the increases of PKA, Akt and mTOR activities by M1 mAChR activation were blocked by PKC and Ras inhibitors. These data demonstrated the detailed mechanism underlying M1 mAChR-mediated modulation of GluA1 through Gαq/11 coupling, broadening the knowledge of the downstream signaling after M1 mAChR-Gαq/11 coupling.


Assuntos
Proteína Quinase C/metabolismo , Receptor Muscarínico M1/metabolismo , Receptores de AMPA/metabolismo , Proteínas ras/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Fosfosserina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais
13.
Curr Neuropharmacol ; 17(10): 990-1002, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30819085

RESUMO

Far-infrared ray (FIR) is an electromagnetic wave that produces various health benefits against pathophysiological conditions, such as diabetes mellitus, renocardiovascular disorders, stress, and depression etc. However, the therapeutic application on the FIR-mediated protective potentials remains to be further extended. To achieve better understanding on FIR-mediated therapeutic potentials, we summarized additional findings in the present study that exposure to FIR ameliorates stressful condition, memory impairments, drug dependence, and mitochondrial dysfunction in the central nervous system. In this review, we underlined that FIR requires modulations of janus kinase 2 / signal transducer and activator of transcription 3 (JAK2/STAT3), nuclear factor E2- related factor 2 (Nrf-2), muscarinic M1 acetylcholine receptor (M1 mAChR), dopamine D1 receptor, protein kinase C δ gene, and glutathione peroxidase-1 gene for exerting the protective potentials in response to neuropsychotoxic conditions.


Assuntos
Antioxidantes/uso terapêutico , Raios Infravermelhos/uso terapêutico , Doenças do Sistema Nervoso/radioterapia , Animais , Humanos , Janus Quinase 2/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Receptor Muscarínico M1/metabolismo , Receptores de Dopamina D1/metabolismo , Transdução de Sinais/fisiologia
14.
Clin Otolaryngol ; 44(3): 356-365, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30762948

RESUMO

OBJECTIVE: We investigated the difference in ciliary beat responsiveness to acetylcholine in ex vivo and the difference in the expressions of associated molecules (M1/M3 muscarinic receptors, pannexin-1 and P2X7 purinergic receptor) between the nasal polyp and turbinate mucosa. STUDY DESIGN: Laboratorial study. PARTICIPANTS: Nasal polyp and inferior turbinate were collected from patients with hypertrophic rhinitis and/or nasal polyp during endoscopic sinonasal surgery. MAIN OUTCOME MEASURES: The mucosa was cut into thin strips, and ciliary movement was observed under a phase-contrast light microscope equipped with a high-speed digital video camera. The samples were also examined by scanning electron microscopy, fluorescence immunohistochemistry, and quantitative reverse transcription-polymerase chain reaction. RESULTS: Cilia were well preserved in both tissues at the ultrastructural level. The baseline ciliary beat frequency (CBF) was not different between the two tissues. The CBF of the turbinate was significantly increased by stimulation with acetylcholine (P < 0.001), but that of the polyp was not. The ratio of the acetylcholine-stimulated CBF to the baseline CBF was significantly lower in the polyp than in the turbinate (P < 0.001). Immunohistochemical study revealed that immunoreactivities for M3, pannexin-1 and P2X7 were weaker in the polyp than in the turbinate. The mRNA expressions of M1, M3 and P2X7 were significantly lower and that of pannexin-1 tended to be lower in the polyp than in the turbinate. CONCLUSIONS: These results indicate that ciliary beat responsiveness to acetylcholine is decreased in the nasal polyp. This may be explained by the decreased expressions of M3, P2X7 and probably pannexin-1 in this tissue.


Assuntos
Acetilcolina/farmacologia , Cílios/efeitos dos fármacos , Mucosa Nasal/efeitos dos fármacos , Mucosa Nasal/metabolismo , Pólipos Nasais/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Cílios/ultraestrutura , Conexinas/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Depuração Mucociliar/efeitos dos fármacos , Pólipos Nasais/cirurgia , Proteínas do Tecido Nervoso/metabolismo , RNA Mensageiro/metabolismo , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M2/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Rinite/cirurgia , Conchas Nasais/efeitos dos fármacos , Conchas Nasais/metabolismo , Conchas Nasais/ultraestrutura
15.
FASEB J ; 33(5): 6622-6631, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30794430

RESUMO

M1 muscarinic acetylcholine receptors are highly expressed in key areas that control cognition, such as the cortex and hippocampus, representing one potential therapeutic target for cognitive dysfunctions of Alzheimer's disease and schizophrenia. We have reported that M1 receptors facilitate cognition by promoting membrane insertion of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor AMPA receptor subunit 1 (GluA1) through phosphorylation at Ser845. However, the signaling pathway is still unclear. Here we showed that adenylyl cyclase inhibitor 2',5'-dideoxyadenosine and PKA inhibitor KT5720 inhibited enhancement of phosphorylation of Ser845 and membrane insertion of GluA1 induced by M1 receptor activation. Furthermore, PI3K inhibitor LY294002 and protein kinase B (Akt) inhibitor IV blocked the effects of M1 receptors as well. Remarkably, the increase of the activity of PI3K-Akt signaling induced by M1 receptor activation could be abolished by cAMP-PKA inhibitors. Moreover, inhibiting the mammalian target of rapamycin (mTOR) complex 1, an important downstream effector of PI3K-Akt, by short-term application of rapamycin attenuated the effects of M1 receptors on GluA1. Furthermore, such effect was unrelated to possible protein synthesis promoted by mTOR. Taken together, these data demonstrate that M1 receptor activation induces membrane insertion of GluA1 via a signaling linking cAMP-PKA and PI3K-Akt-mTOR pathways but is irrelevant to protein synthesis.-Zhao, L.-X., Ge, Y.-H., Li, J.-B., Xiong, C.-H., Law, P.-Y., Xu, J.-R., Qiu, Y., Chen, H.-Z. M1 muscarinic receptors regulate the phosphorylation of AMPA receptor subunit GluA1 via a signaling pathway linking cAMP-PKA and PI3K-Akt.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Muscarínico M1/metabolismo , Receptores de AMPA/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , Animais , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Ratos , Ratos Sprague-Dawley
16.
ACS Chem Neurosci ; 10(3): 1753-1764, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30480428

RESUMO

Preclinical and clinical data suggest that muscarinic acetylcholine receptor activation may be therapeutically beneficial for the treatment of schizophrenia and Alzheimer's diseases. This is best exemplified by clinical observations with xanomeline, the efficacy of which is thought to be mediated through co-activation of the M1 and M4 muscarinic acetylcholine receptors (mAChRs). Here we examined the impact of treatment with xanomeline and compared it to the actions of selective M1 and M4 mAChR activators on in vivo intracellular signaling cascades in mice, including 3'-5'-cyclic adenosine monophosphate response element binding protein (CREB) phosphorylation and inositol phosphate-1 (IP1) accumulation in the striatum, hippocampus, and prefrontal cortex. We additionally assessed the effects of xanomeline on hippocampal electrophysiological signatures in rats using ex vivo recordings from CA1 (Cornu Ammonis 1) as well as in vivo hippocampal theta. As expected, xanomeline's effects across these readouts were consistent with activation of both M1 and M4 mAChRs; however, differences were observed across different brain regions, suggesting non-uniform activation of these receptor subtypes in the central nervous system. Interestingly, despite having nearly equal in vitro potency at the M1 and the M4 mAChRs, during in vivo assays xanomeline produced M4-like effects at significantly lower brain exposures than those at which M1-like effects were observed. Our results raise the possibility that clinical efficacy observed with xanomeline was driven, in part, through its non-uniform activation of mAChR subtypes in the central nervous system and, at lower doses, through preferential agonism of the M4 mAChR.


Assuntos
Hipocampo/efeitos dos fármacos , Agonistas Muscarínicos/farmacologia , Piridinas/farmacologia , Receptor Muscarínico M1/metabolismo , Tiadiazóis/farmacologia , Acetilcolina/metabolismo , Acetilcolina/farmacologia , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Hipocampo/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo
17.
Toxicology ; 394: 54-62, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29253600

RESUMO

Cadmium, an environmental neurotoxic compound, produces cognitive disorders, although the mechanism remains unknown. Previously, we described that cadmium induces a more pronounced cell death on cholinergic neurons from basal forebrain (BF). This effect, partially mediated by M1 receptor blockade, triggering it through AChE splices variants alteration, may explain cadmium effects on learning and memory processes. Cadmium has been also reported to induce oxidative stress generation leading to M2 and M4 muscarinic receptors alteration, in hippocampus and frontal cortex, which are necessary to maintain cell viability and cognitive regulation, so their alteration in BF could also mediate this effect. Moreover, it has been reported that antioxidant treatment could reverse cognitive disorders, muscarinic receptor and AChE variants alterations induced by cadmium. Thus, we hypothesized that cadmium induced cell death of BF cholinergic neurons is mediated by oxidative stress generation and this mechanism could produce this effect, in part, through AChE variants altered by muscarinic receptors disruption. To prove this, we evaluated in BF SN56 cholinergic neurons, whether cadmium induces oxidative stress and alters muscarinic receptors, and their involvement in the induction of cell death through alteration of AChE variants. Our results show that cadmium induces oxidative stress, which mediates partially the alteration of AChE variants and M2 to M4 muscarinic receptors expression and blockage of M1 receptor. In addition, cadmium induced oxidative stress generation by M1 and M3 receptors alteration through AChE variants disruption, leading to cell death. These results provide new understanding of the mechanisms contributing to cadmium harmful effects on cholinergic neurons.


Assuntos
Acetilcolinesterase/metabolismo , Cloreto de Cádmio/toxicidade , Neurônios Colinérgicos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M2/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Neurônios Colinérgicos/metabolismo , Neurônios Colinérgicos/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos , Doenças Neurodegenerativas/induzido quimicamente , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Estresse Oxidativo/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo , Prosencéfalo/patologia
18.
Low Urin Tract Symptoms ; 10(3): 315-319, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28681521

RESUMO

OBJECTIVES: To investigate the effect of high-fat diet (HFD) on bladder M1,3 muscarinic receptor expression and contractile function in the rat. METHODS: Eight-week-old male rats were divided into two groups including one with HFD for 8 weeks (short-term) and the other for 24 weeks (long-term). Each group was compared to age-matched rats fed with normal chow as controls. The body weight, food intake amount and blood biochemistry were monitored. Bladder muscle contractile responses to acetylcholine (0.1-10 µM), bethanechol (10 µM) and KCl (50 mM) were studied in an organ bath set-up. Bladder M1 and M3 muscarinic receptor protein expressions were measured by Western blotting analysis. RESULTS: Increase in body weight as well as blood triglyceride, cholesterol and sugar levels compared to controls were noted in both 8- and 24-week HFD rats. Eating appetite change with increased food and water intakes was noted in the HFD rats. Significantly decreased bladder contractile responses to acetylcholine and bethanechol were shown in both HFD groups. On the other hand, decreased bladder contractile response to KCl was demonstrated in the 24-week group but not the 8-week group. The expressions of bladder M1 and M3 muscarinic receptor proteins were significantly and progressively decreased by HFD feeding from 8 to 24 weeks. CONCLUSIONS: High-fat diet induces obesity and polyphagia in rats. Short-term and long-term HFD feeding decrease rat bladder M1 and M3 receptor expressions as well as contractile responses to the agonistic stimulation. In addition, bladder muscle dysfunction develops after long-term HFD feeding.


Assuntos
Gorduras na Dieta/administração & dosagem , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M3/metabolismo , Bexiga Urinária/metabolismo , Acetilcolina/farmacologia , Animais , Betanecol/farmacologia , Agonistas Colinérgicos/farmacologia , Comportamento Alimentar , Masculino , Agonistas Muscarínicos/farmacologia , Contração Muscular/efeitos dos fármacos , Músculo Liso/fisiopatologia , Ratos , Fatores de Tempo , Bexiga Urinária/fisiopatologia
19.
Bull Exp Biol Med ; 162(6): 750-753, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28429224

RESUMO

Experiments on random-bred albino mice showed that M1 muscarinic acetylcholine receptor agonist (TBPB) and α7n-acetylcholine receptor agonist (GTS-21) significantly reduced mortality of mice with experimental sepsis (intraperitoneally administration of E. coli) in 4 and 24 h after modeling by reducing blood concentration of proinflammatory cytokines TNF-α, IL-1ß, and IL-6. Combined treatment with TBPB and GTS-21 determined their additive effect.


Assuntos
Benzimidazóis/farmacologia , Compostos de Benzilideno/farmacologia , Infecções por Escherichia coli/tratamento farmacológico , Agonistas Muscarínicos/farmacologia , Agonistas Nicotínicos/farmacologia , Piperidinas/farmacologia , Piridinas/farmacologia , Sepse/tratamento farmacológico , Animais , Quimioterapia Combinada , Escherichia coli/patogenicidade , Escherichia coli/fisiologia , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/mortalidade , Infecções por Escherichia coli/patologia , Feminino , Regulação da Expressão Gênica , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/sangue , Interleucina-6/antagonistas & inibidores , Interleucina-6/sangue , Masculino , Camundongos , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/metabolismo , Sepse/microbiologia , Sepse/mortalidade , Sepse/patologia , Análise de Sobrevida , Resultado do Tratamento , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/sangue , Receptor Nicotínico de Acetilcolina alfa7/agonistas , Receptor Nicotínico de Acetilcolina alfa7/genética , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
20.
Neuropharmacology ; 117: 376-386, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28257887

RESUMO

Dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32) play essential roles in dopamine (DA) transmission in the striatum. It is suggested that a link exists between muscarinic acetylcholine receptors (mAChRs) and DA/DARPP-32 signaling, but the molecular mechanisms mediating this relationship have not been elucidated. The predominant mAChRs subtypes in the striatum are M1 and M4. In this study, we investigated the functions of these two receptors, particularly M4, in regulating cAMP production and DARPP-32 phosphorylation in rat striatal medium spiny neurons (MSNs). We used time-resolved fluorescence resonance energy transfer, immunofluorescence confocal microscopy, and western blot assays. In cultured intact MSNs, we confirmed that muscarinic M1 and M4 receptors were highly expressed. Notably, M4 receptors were co-expressed with D1 receptors in only a portion of the cultured MSNs. The nonselective muscarinic agonist oxotremorine M (OX) slightly enhanced cAMP production, but this effect was independent of M1 or M4 receptors. However, OX directly participated in DARPP-32 phosphorylation, phosphorylating DARPP-32 at Thr75 (the CDK5 site) and concomitantly de-phosphorylating DARPP-32 at Thr34 (the PKA site) in virtually cultured MSNs, whereas APO phosphorylated DARPP-32 at both Thr34 and Thr75. The OX-induced time-dependent increase in DARPP-32 phosphorylation at Thr75 was accompanied by increased p35 and CDK5 activity. Specifically, elevated immunoreactivity for phospho-DARPP-32-Thr75 and p35 was detected in M4 receptor-expressing MSNs. Both genetic knockdown and pharmacologic inhibition of M4 receptors with MT3, an M4 receptor-selective antagonist, decreased the OX-induced DARPP-32-Thr75 phosphorylation in MSNs. These results indicate that the M4 muscarinic receptor plays a critical role in modulating phosphorylation of DARPP-32-Thr75 in MSNs. The results suggest that M4 receptor activation acts antagonistically with dopamine D1-like receptors within the striatum, and indicate that M4 receptors may be a potential target for the treatment of Parkinson's disease and other relevant central nervous system disorders.


Assuntos
Corpo Estriado/efeitos dos fármacos , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Agonistas Muscarínicos/farmacologia , Neurônios/efeitos dos fármacos , Oxotremorina/farmacologia , Receptor Muscarínico M4/metabolismo , Animais , Células Cultivadas , Corpo Estriado/citologia , Corpo Estriado/metabolismo , AMP Cíclico/metabolismo , Técnicas de Silenciamento de Genes , Antagonistas Muscarínicos/farmacologia , Neurônios/citologia , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos , Ratos Sprague-Dawley , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M4/agonistas , Receptor Muscarínico M4/antagonistas & inibidores , Receptor Muscarínico M4/genética , Receptores de Dopamina D1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA