Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Pharmacol Rep ; 71(6): 1108-1114, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31634798

RESUMEN

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.


Asunto(s)
Atropina/farmacología , Pirenzepina/análogos & derivados , Pirenzepina/farmacología , Receptor Muscarínico M1/fisiología , Receptor Muscarínico M2/fisiología , Choque Séptico/tratamiento farmacológico , Choque Séptico/fisiopatología , Animales , Citocinas/metabolismo , Lipopolisacáridos , Hígado/patología , Pulmón/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/efectos de los fármacos , Receptor Muscarínico M1/antagonistas & inhibidores , Receptor Muscarínico M2/antagonistas & inhibidores , Choque Séptico/inducido químicamente , Choque Séptico/mortalidad , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/metabolismo
2.
J Neurosci ; 39(40): 7840-7852, 2019 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-31451581

RESUMEN

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.


Asunto(s)
Subunidades beta de la Proteína de Unión al GTP/fisiología , Subunidades gamma de la Proteína de Unión al GTP/fisiología , Receptores Acoplados a Proteínas G/fisiología , Canales Catiónicos TRPM/fisiología , Animales , Conducta Animal , Femenino , Subunidades beta de la Proteína de Unión al GTP/antagonistas & inhibidores , Subunidades gamma de la Proteína de Unión al GTP/antagonistas & inhibidores , Ganglios Espinales/citología , Ganglios Espinales/fisiología , Células HEK293 , Humanos , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Hiperalgesia/psicología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Nociceptores/efectos de los fármacos , Toxina del Pertussis/farmacología , Receptor de Adenosina A2B/fisiología , Receptor Muscarínico M1/fisiología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal/fisiología , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/genética
3.
Biochem Pharmacol ; 169: 113613, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31445019

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/fisiología , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Receptor Muscarínico M3/fisiología , Animales , Células Cultivadas , Humanos , Ratones , PPAR alfa/fisiología , Fosforilación , Receptor Muscarínico M1/fisiología , Transducción de Señal/fisiología
4.
Pharmacology ; 103(1-2): 38-49, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30380560

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Membrana/fisiología , Receptor de Adenosina A2A/fisiología , Receptor Muscarínico M1/fisiología , Periodo Refractario Electrofisiológico/efectos de los fármacos , Cloruro de (4-(m-Clorofenilcarbamoiloxi)-2-butinil)trimetilamonio/farmacología , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Colforsina/farmacología , Diafragma/efectos de los fármacos , Diafragma/fisiología , Hemicolinio 3/farmacología , Neostigmina/farmacología , Fenetilaminas/farmacología , Nervio Frénico/efectos de los fármacos , Nervio Frénico/fisiología , Ratas , Ratas Wistar , Transmisión Sináptica , Tétanos/tratamiento farmacológico , Tétanos/fisiopatología , Acetato de Tetradecanoilforbol/farmacología
5.
Cancer Discov ; 8(11): 1458-1473, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30185628

RESUMEN

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.


Asunto(s)
Carcinoma Ductal Pancreático/prevención & control , Transformación Celular Neoplásica/efectos de los fármacos , Colinérgicos/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Pancreáticas/prevención & control , Receptor Muscarínico M1/fisiología , Animales , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Genes ras , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Transducción de Señal , Neoplasias Pancreáticas
6.
Free Radic Biol Med ; 78: 66-81, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25452146

RESUMEN

The role of muscarinic receptor subtypes in modulating acute liver injury is unknown. We detected M1 muscarinic receptor (M1R) expression in human and murine hepatocytes, and investigated the consequences of M1R deficiency on acute liver injury in vivo and inhibiting M1R activation on hepatocyte injury in vitro. Age-matched wild-type (WT) and M1R-deficient (Chrm1(-/-)) male mice were injected intraperitoneally with 200mg/kg acetaminophen (APAP) and euthanized 0, 2, 4, 16, 24, and 36h later. Biochemical and histological parameters indicated that liver injury peaked within 16h after APAP treatment and resolved by 24h. Compared to WT, M1R-deficient mice had reduced intrahepatic hemorrhage and hepatocyte necrosis, reflected by an attenuated rise in serum alanine aminotransferase levels. Livers of M1R-deficient mice showed reduced hepatocyte DNA fragmentation and attenuated expression of injury cytokines (Il-1α, Il-1ß, Il-6, and Fasl). In all mice hepatic glutathione levels decreased after APAP injection, but they recovered more quickly in M1R-deficient mice. During the course of APAP-induced liver injury in M1R-deficient compared to WT mice, hepatic Nrf-2, Gclc, and Nqo1 expressions increased and nitrotyrosine generation decreased. APAP metabolic pathways were not altered by M1R deficiency; expression of hepatic Cyp2e1, Cyp1a2, Cyp3a11, Cyp3a13, Car, and Pxr was similar in Chrm1(-/-) and WT mice. Finally, treatment of murine AML12 hepatocytes with a novel M1R antagonist, VU0255035, attenuated H2O2-induced oxidative stress, prevented GSH depletion, and enhanced viability. We conclude that M1R modify hepatocyte responses to oxidative stress and that targeting M1R has therapeutic potential for toxic liver injury.


Asunto(s)
Acetaminofén/toxicidad , Analgésicos no Narcóticos/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Estrés Oxidativo/efectos de los fármacos , Receptor Muscarínico M1/fisiología , Animales , Apoptosis , Western Blotting , Proliferación Celular , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Glutatión/metabolismo , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Noqueados , Oxidantes/farmacología , ARN Mensajero/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Neurosci ; 34(5): 1570-8, 2014 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-24478341

RESUMEN

Neuromodulators released during and after a fearful experience promote the consolidation of long-term memory for that experience. Because overconsolidation may contribute to the recurrent and intrusive memories of post-traumatic stress disorder, neuromodulatory receptors provide a potential pharmacological target for prevention. Stimulation of muscarinic receptors promotes memory consolidation in several conditioning paradigms, an effect primarily associated with the M1 receptor (M1R). However, neither inhibiting nor genetically disrupting M1R impairs the consolidation of cued fear memory. Using the M1R agonist cevimeline and antagonist telenzepine, as well as M1R knock-out mice, we show here that M1R, along with ß2-adrenergic (ß2AR) and D5-dopaminergic (D5R) receptors, regulates the consolidation of cued fear memory by redundantly activating phospholipase C (PLC) in the basolateral amygdala (BLA). We also demonstrate that fear memory consolidation in the BLA is mediated in part by neuromodulatory inhibition of the M-current, which is conducted by KCNQ channels and is known to be inhibited by muscarinic receptors. Manipulating the M-current by administering the KCNQ channel blocker XE991 or the KCNQ channel opener retigabine reverses the effects on consolidation caused by manipulating ß2AR, D5R, M1R, and PLC. Finally, we show that cAMP and protein kinase A (cAMP/PKA) signaling relevant to this stage of consolidation is upstream of these neuromodulators and PLC, suggesting an important presynaptic role for cAMP/PKA in consolidation. These results support the idea that neuromodulatory regulation of ion channel activity and neuronal excitability is a critical mechanism for promoting consolidation well after acquisition has occurred.


Asunto(s)
Miedo/fisiología , Canales de Potasio KCNQ/metabolismo , Memoria/fisiología , Receptor Muscarínico M1/fisiología , Fosfolipasas de Tipo C/metabolismo , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Clásico/fisiología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Antagonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica/efectos adversos , Inhibidores Enzimáticos/farmacología , Miedo/efectos de los fármacos , Femenino , Canales de Potasio KCNQ/efectos de los fármacos , Masculino , Moduladores del Transporte de Membrana/farmacología , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Agonistas Muscarínicos/farmacología , Procaterol/farmacología , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/antagonistas & inhibidores , Receptor Muscarínico M1/deficiencia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
8.
Biochim Biophys Acta ; 1842(4): 635-45, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24365239

RESUMEN

Impairment of epithelial barrier is observed in various intestinal disorders including inflammatory bowel diseases (IBD). Numerous factors may cause temporary damage of the intestinal epithelium. A complex network of highly divergent factors regulates healing of the epithelium to prevent inflammatory response. However, the exact repair mechanisms involved in maintaining homeostatic intestinal barrier integrity remain to be clarified. In this study, we demonstrate that activation of M1 muscarinic acetylcholine receptor (mAChR) augments the restitution of epithelial barrier function in T84 cell monolayers after ethanol-induced epithelial injury, via ERK-dependent phosphorylation of focal adhesion kinase (FAK). We have shown that ethanol injury decreased the transepithelial electrical resistance (TER) along with the reduction of ERK and FAK phosphorylation. Carbachol (CCh) increased ERK and FAK phosphorylation with enhanced TER recovery, which was completely blocked by either MT-7 (M1 antagonist) or atropine. The CCh-induced enhancement of TER recovery was also blocked by either U0126 (ERK pathway inhibitor) or PF-228 (FAK inhibitor). Treatment of T84 cell monolayers with interferon-γ (IFN-γ) impaired the barrier function with the reduction of FAK phosphorylation. The CCh-induced ERK and FAK phosphorylation were also attenuated by the IFN-γ treatment. Immunological and binding experiments exhibited a significant reduction of M1 mAChR after IFN-γ treatment. The reduction of M1 mAChR in inflammatory area was also observed in surgical specimens from IBD patients, using immunohistochemical analysis. These findings provide important clues regarding mechanisms by which M1 mAChR participates in the maintenance of intestinal barrier function under not only physiological but also pathological conditions.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/fisiología , Mucosa Intestinal/metabolismo , Receptor Muscarínico M1/fisiología , Línea Celular Tumoral , Impedancia Eléctrica , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/análisis , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Proteína-Tirosina Quinasas de Adhesión Focal/análisis , Humanos , Inmunohistoquímica , Interferón gamma/farmacología , Fosforilación , Receptor Muscarínico M1/análisis
9.
Int J Neuropsychopharmacol ; 16(4): 721-31, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22932339

RESUMEN

Episodic memory deficits are a core feature of neurodegenerative disorders. Muscarinic M(1) receptors play a critical role in modulating learning and memory and are highly expressed in the hippocampus. We examined the effect of GSK1034702, a potent M(1) receptor allosteric agonist, on cognitive function, and in particular episodic memory, in healthy smokers using the nicotine abstinence model of cognitive dysfunction. The study utilized a randomized, double-blind, placebo-controlled, cross-over design in which 20 male nicotine abstained smokers were tested following single doses of placebo, 4 and 8 mg GSK1034702. Compared to the baseline (nicotine on-state), nicotine abstinence showed statistical significance in reducing immediate (p=0.019) and delayed (p=0.02) recall. GSK1034702 (8 mg) significantly attenuated (i.e. improved) immediate recall (p=0.014) but not delayed recall. None of the other cognitive domains was modulated by either nicotine abstinence or GSK1034702. These findings suggest that stimulating M(1) receptor mediated neurotransmission in humans with GSK1034702 improves memory encoding potentially by modulating hippocampal function. Hence, selective M(1) receptor allosteric agonists may have therapeutic benefits in disorders of impaired learning including Alzheimer's disease.


Asunto(s)
Conducta Adictiva/psicología , Bencimidazoles/uso terapéutico , Trastornos del Conocimiento/psicología , Memoria Episódica , Receptor Muscarínico M1/agonistas , Cese del Hábito de Fumar/psicología , Adulto , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Conducta Adictiva/tratamiento farmacológico , Bencimidazoles/farmacología , Trastornos del Conocimiento/tratamiento farmacológico , Estudios Cruzados , Método Doble Ciego , Humanos , Masculino , Persona de Mediana Edad , Nicotina , Receptor Muscarínico M1/fisiología , Fumar/psicología , Adulto Joven
10.
J Neurochem ; 120(4): 631-40, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22146060

RESUMEN

Recent evidence indicates that supplementation with a specific combination of nutrients may affect cell membrane synthesis and composition. To investigate whether such nutrients may also modify the physical properties of membranes, and affect membrane-bound processes involved in signal transduction pathways, we studied the effects of nutrient supplementation on G protein-coupled receptor activation in vitro. In particular, we investigated muscarinic receptors, which are important for the progression of memory deterioration and pathology of Alzheimer's disease. Nerve growth factor differentiated pheochromocytoma cells that were supplemented with specific combinations of nutrients showed enhanced responses to muscarinic receptor agonists in a membrane potential assay. The largest effects were obtained with a combination of nutrients known as Fortasyn™ Connect, comprising docosahexaenoic acid, eicosapentaenoic acid, uridine monophosphate as a uridine source, choline, vitamin B6, vitamin B12, folic acid, phospholipids, vitamin C, vitamin E, and selenium. In subsequent experiments, it was shown that the effects of supplementation could not be attributed to single nutrients. In addition, it was shown that the agonist-induced response and the supplement-induced enhancement of the response were blocked with the muscarinic receptor antagonists atropine, telenzepine, and AF-DX 384. In order to determine whether the effects of Fortasyn™ Connect supplementation were receptor subtype specific, we investigated binding properties and activation of human muscarinic M1, M2 and M4 receptors in stably transfected Chinese hamster ovary cells after supplementation. Multi-nutrient supplementation did not change M1 receptor density in plasma membranes. However, M1 receptor-mediated G protein activation was significantly enhanced. In contrast, supplementation of M2- or M4-expressing cells did not affect receptor signaling. Taken together, these results indicate that a specific combination of nutrients acts synergistically in enhancing muscarinic M1 receptor responses, probably by facilitating receptor-mediated G protein activation.


Asunto(s)
Micronutrientes/farmacología , Receptor Muscarínico M1/fisiología , Regulación hacia Arriba/fisiología , Animales , Células CHO , Carbacol/farmacología , Cricetinae , Cricetulus , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/fisiología , Humanos , Potenciales de la Membrana/fisiología , Micronutrientes/química , Células PC12 , Unión Proteica , Ratas , Receptor Muscarínico M1/agonistas
11.
J Neurochem ; 110(3): 1107-19, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19558456

RESUMEN

In this study, we provide evidence that the muscarinic M1 receptor targets NF-E2-related factor-2 (Nrf2), a transcription factor that regulates the expression of genes containing antioxidant response elements (AREs) in their promoters and that collectively constitute the phase II antioxidant response. In hippocampal primary and cerebellar granule neuron cultures expressing endogenous M1 receptor, carbachol increased the levels of a prototypical phase II antioxidant enzyme, heme oxygenase-1. Moreover, in a heterologous system, based on lentiviral expression of M1 receptor in PC12 pheochromocytoma cells, we found that M1 increased total and nuclear Nrf2 protein levels and heme oxygenase-1 messenger RNA and protein levels. Luciferase reporter constructs for AREs and the use of two inhibitors of protein kinase C (PKC), chelerythrine and 2-aminoethyl diphenylborinate, or transfection with relevant expression vectors allowed us to identify Galphaq, phospholipase C-beta and the classical PKC-gamma isoenzyme, as responsible for the regulation of Nrf2. A PKC-insensitive Nrf2S40A single-point mutant partially channeled M1 signaling to AREs, therefore suggesting the participation of additional intermediates. Inhibition of glycogen synthase kinase-3beta (GSK-3beta) augmented M1-dependent activation of AREs while a PKC-insensitive mutant of GSK-3beta (GSK-3beta-Delta9) blocked this effect and prevented M1-induced accumulation of Nrf2 in the nucleus. Our results demonstrate a previously unidentified role of the Galphaq/phospholipase C-beta/PKC/GSK-3beta axis in regulation of Nrf2 by M1. Such role provides additional conceptual support for the use of cholinemimetics in the treatment of pathologies that, like Alzheimer's disease, require a reinforcement of the cell antioxidant capacity.


Asunto(s)
Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/metabolismo , Proteína Quinasa C/fisiología , Receptor Muscarínico M1/fisiología , Transducción de Señal/fisiología , Transmisión Sináptica/fisiología , Animales , Línea Celular , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Glucógeno Sintasa Quinasa 3/fisiología , Glucógeno Sintasa Quinasa 3 beta , Humanos , Factor 2 Relacionado con NF-E2/fisiología , Neuronas/enzimología , Neuronas/metabolismo , Fármacos Neuroprotectores/metabolismo , Células PC12 , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M1/biosíntesis , Receptor Muscarínico M1/genética
12.
J Physiol Sci ; 59(4): 291-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19343482

RESUMEN

This study was performed to determine whether spinal cholinergic systems mediate the relieving effects of electroacupuncture (EA) on cold and warm allodynia in a rat model of neuropathic pain. For neuropathic surgery, the right superior caudal trunk was resected at the level between the S1 and S2 spinal nerves innervating the tail. Two weeks after the injury, the intrathecal (i.t.) catheter was implanted. Five days after the catheterization, the rats were injected with atropine (non-selective muscarinic antagonist, 30 microg), mecamylamine (non-selective nicotinic antagonist, 50 microg), pirenzepine (M(1) muscarinic antagonist, 10 microg), methoctramine (M(2) antagonist, 10 microg) or 4-diphenylacetoxy-N-methylpiperidine methiodide (4-DAMP) (M(3) antagonist, 10 microg). Ten minutes after the injection, EA was applied to the ST36 acupoint for 30 min. The cold and warm allodynia were assessed by the tail immersion test [i.e., immersing the tail in cold (4 degrees C) or warm (40 degrees C) water and measuring the latency of an abrupt tail movement] before and after the treatments. The i.t. atropine, but not mecamylamine, blocked the relieving effects of EA on cold and warm allodynia. Furthermore, i.t. pirenzepine attenuated the antiallodynic effects of EA, whereas methoctramine and 4-DAMP did not. These results suggest that spinal muscarinic receptors, especially M(1) subtype, mediate the EA-induced antiallodynia in neuropathic rats.


Asunto(s)
Fibras Colinérgicas/fisiología , Electroacupuntura , Neuralgia/fisiopatología , Neuralgia/terapia , Nervios Espinales/fisiopatología , Animales , Atropina/farmacología , Colinérgicos/farmacología , Fibras Colinérgicas/efectos de los fármacos , Frío , Diaminas/farmacología , Modelos Animales de Enfermedad , Calor , Hiperestesia/fisiopatología , Hiperestesia/terapia , Masculino , Mecamilamina/farmacología , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Piperidinas/farmacología , Pirenzepina/farmacología , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M1/efectos de los fármacos , Receptor Muscarínico M1/fisiología , Nervios Espinales/efectos de los fármacos
13.
J Agric Food Chem ; 56(22): 10544-51, 2008 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-18954071

RESUMEN

Research suggests that polyphenolic compounds contained in fruits and vegetables that are rich in color may have potent antioxidant and anti-inflammatory activities. The present studies determined if stilbene (e.g., resveratrol) compounds would be efficacious in reversing the deleterious effects of aging in 19 month old Fischer 344 rats. Experiment I utilized resveratrol and six resveratrol analogues and examined their efficacies in preventing dopamine-induced decrements in calcium clearance following oxotremorine-induced depolarization in COS-7 cells transfected with M1 muscarinic receptors (MAChR) that we have shown previously to be sensitive to oxidative stressors. Experiment II utilized the most efficacious analogue (pterostilbene) from experiment I and fed aged rats a diet with a low (0.004%) or a high (0.016%) concentration of pterostilbene. Results indicated that pterostilbene was effective in reversing cognitive behavioral deficits, as well as dopamine release, and working memory was correlated with pterostilbene levels in the hippocampus.


Asunto(s)
Envejecimiento/efectos de los fármacos , Estilbenos/farmacología , Animales , Células COS , Calcio/metabolismo , Chlorocebus aethiops , Trastornos del Conocimiento/tratamiento farmacológico , Dieta , Dopamina/metabolismo , Dopamina/farmacología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/fisiología , Resveratrol , Estilbenos/administración & dosificación , Transfección
14.
Mol Cell Neurosci ; 37(4): 673-81, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18272392

RESUMEN

Extracellular tau promotes an increase in the level of intracellular calcium in cultured neuronal cells. We have found that such increase is impaired in the presence of antagonists of muscarinic receptors. In order to identify the nature of those receptors, we have tested the effect of different specific muscarinic receptor antagonists on tau promoted calcium increase. Our results indicate that the increase does not take place in the presence of antagonists of muscarinic (mainly M1 and M3) receptors. A similar increase in intracellular calcium was found in non-neuronal cells transfected with cDNA of M1 and M3 muscarinic receptors when tau was added. These results suggest that observed effect of tau protein on neuronal (neuroblastoma and primary cultures of hippocampal and cortical neurons) cells is through M1 and M3 muscarinic receptors. Therefore blocking M1 and for M3 receptors, by using specific receptor antagonists, can prevent that tau toxic effect that could take place in tauopathies.


Asunto(s)
Calcio/metabolismo , Líquido Extracelular/fisiología , Líquido Intracelular/metabolismo , Neuronas/metabolismo , Receptor Muscarínico M1/fisiología , Receptor Muscarínico M3/fisiología , Proteínas tau/fisiología , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular Tumoral , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Chlorocebus aethiops , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Datos de Secuencia Molecular , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neuronas/fisiología
15.
J Pharmacol Exp Ther ; 324(1): 196-205, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17540859

RESUMEN

We determined the functional role of a small domain in the third intracellular loop of the human muscarinic M(1) (hM(1)) receptor. Using site-directed mutagenesis, several mutant hM(1) receptors were made possessing either a deletion or point mutations within the third intracellular loop domain (252)PETPPGRCCRCC(263). Wild-type and mutant hM(1) receptors were transiently expressed in Chinese hamster ovary cells, and the effects of each mutation on radioligand binding, agonist-mediated phosphoinositide hydrolysis, and agonist-induced internalization were determined. The mutant receptors exhibited a modest reduction in affinity for [(3)H]N-methylscopolamine (pK(D) = approximately 9.0) and a moderately increased binding capacity relative to the wild-type receptor. This moderate increase in binding capacity was associated with small increases in the maximal response and potency of carbachol for eliciting phosphoinositide hydrolysis through the mutant receptors (pEC(50) = approximately 5.5) relative to wild-type (pEC(50) = 5.35 +/- 0.05). In contrast, carbachol-induced internalization of mutant hM(1) receptors possessing either C259A/C260A or C262A/C263A or both double point mutations was significantly reduced compared to the wild-type hM(1) receptor. Of the hM(1) receptor mutants tested, those possessing a C262D/C263D double point mutation had the least carbachol-induced internalization. The desensitization and down-regulation of receptors possessing either Cys/Ala or Cys/Asp double point mutations were similar to those observed for the wild-type hM(1) receptor. Collectively, these observations suggest that Cys pairs Cys259/Cys260 and Cys262/Cys263 play an important role in the agonist-induced internalization of hM(1) receptors.


Asunto(s)
Cisteína/fisiología , Receptor Muscarínico M1/química , Receptor Muscarínico M1/fisiología , Animales , Células CHO , Carbacol/farmacología , Cricetinae , Cricetulus , Cisteína/química , Humanos , Hidrólisis , Mutagénesis Sitio-Dirigida , N-Metilescopolamina/metabolismo , Fosfatidilinositoles/metabolismo , Estructura Terciaria de Proteína , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/antagonistas & inhibidores , Transfección
16.
J Neurosci ; 27(15): 4083-92, 2007 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-17428986

RESUMEN

The cellular prion protein (PrP(c)) undergoes a physiological processing yielding the N-terminal fragment referred to as N1, the production of which can be constitutive or protein kinase C regulated. We show that activation of endogenous muscarinic receptors by carbachol and by the M1-selective agonist AF267B increases N1 recovery in an atropine-sensitive manner, in mouse embryonic primary neurons. To identify the muscarinic receptor subtype involved, we used human embryonic kidney HEK293 (HEK) cells stably overexpressing M1, M2, M3, or M4 receptor subtype. Carbachol and the selective M1 agonist AF267B dose dependently increased N1 release by HEK-M3 and HEK-M1 cells, respectively, whereas carbachol did not modify N1 production by HEK-M2 or HEK-M4 cells. We demonstrate that the increase of N1 was not attributable to modified trafficking to the membrane of either PrP(c) or the disintegrin metalloproteases ADAM10 or ADAM17. Furthermore, we establish that carbachol affects the overall phosphorylation of ADAM17 on its threonine and tyrosine but not serine residues, whereas levels of phosphorylated ADAM9 were not affected. Interestingly, carbachol also increases the hydrolysis of the fluorimetric substrate JMV2770, which mimicked the sequence encompassing the N1 site cleavage and was shown previously to behave as an ADAM protease substrate. Mutations of threonine 735 but not of tyrosine 702 of the ADAM17 cytoplasmic tail abolishes the carbachol-induced increase of N1, ADAM17 phosphorylation, and JMV2770-hydrolyzing activity in M1- and M3-expressing HEK293 cells. Thus, our data provide strong evidence that muscarinic receptor activation increases the physiological processing of PrP(c) by upregulating the phosphorylation state and activity of ADAM17 protease.


Asunto(s)
Proteínas ADAM/metabolismo , Priones/fisiología , Receptor Muscarínico M1/fisiología , Receptor Muscarínico M3/fisiología , Proteínas ADAM/antagonistas & inhibidores , Proteína ADAM17 , Animales , Carbacol/farmacología , Línea Celular , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Fosforilación/efectos de los fármacos , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M3/agonistas
17.
Nitric Oxide ; 16(1): 110-7, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16843016

RESUMEN

We investigated whether vagal cardiac cholinergic facilitation by nitric oxide (NO) is mediated by cardiac muscarinic receptor subtypes in the vagally innervated rat right atrium in vitro. Experiments were carried out in the presence of atenolol (4 microM). The right vagus was stimulated at 4, 8, 16, 32 Hz; pulse duration 1 ms at 20 V for 20s; vagal postganglionic activation was achieved using nicotine (0.1, 0.3, 0.5, 1mM) and the effect on cardiac interval (ms) assessed. Pirenzepine (1 microM), a M1 antagonist, attenuated vagally induced increase in cardiac interval. L-Arginine (0.34 mM) superfused with pirenzepine failed to reverse this attenuation, however, L-arginine applied alone reversed the reduction vagal cardiac slowing. Similarly, sodium nitroprusside (10 microM) applied alone, and not together with pirenzepine, was able to reverse the attenuation of vagal effects caused by pirenzepine. Synthetic MT7 (1 nM) toxin, a selective M1 antagonist confirmed these results. M3 antagonism using para-fluorohexahydrosiladifenidol (p-F-HHSiD) (300 nM) and M4 antagonism with PD 102807 (200 nM) did not affect the vagally induced increase in cardiac interval. Nicotine induced increase in cardiac interval was not altered by pirenzepine. These results show that antagonism of M1 receptors on cardiac vagal preganglionic fibres reduces vagal efficacy which can be recovered by either a nitric oxide synthase substrate or a NO donor.


Asunto(s)
Atrios Cardíacos/inervación , Óxido Nítrico/fisiología , Receptor Muscarínico M1/fisiología , Análisis de Varianza , Animales , Atrios Cardíacos/efectos de los fármacos , Técnicas In Vitro , Antagonistas Muscarínicos/farmacología , Nicotina/farmacología , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología , Pirenzepina/farmacología , Ratas , Ratas Wistar
18.
Brain Res ; 1085(1): 102-10, 2006 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-16580648

RESUMEN

Certain organophosphate (OP) cholinesterase inhibitors (ChEIs) are also known to bind to the muscarinic acetylcholine receptor (mAChR). The functional consequences of such binding were investigated here using the following OP compounds: VX, echothiophate, sarin, and soman. VX (charged at physiological pH) and echothiophate (formally charged) inhibited a specific signal transduction pathway in CHO cells expressing either the M(1) or M(3) mAChR. Hence, they blocked carbamylcholine (CCh)-induced cyclic adenosine monophosphate (cAMP) synthesis (muM) and had almost no effect on CCh-induced phosphoinositide (PI) hydrolysis. These substances were inactive on forskolin-induced cAMP inhibition signaling in CHO cells expressing M(2) mAChR. In binding studies, using [(3)H]-N-methyl scopolamine ([(3)H]NMS) as the competitor ligand, the ChEIs, VX and echothiophate exhibited binding to rat cortical mAChR with K(i) values in the muM range. The non-charged compounds, sarin and soman, were inert in modulating both cAMP metabolism and PI hydrolysis in CHO cells expressing M(1), M(2), and M(3) mAChRs, and no binding was observed in presence of [(3)H]NMS. These data suggest that VX and echothiophate act as function-specific blockers via a non-classical path of antagonistic activity, implying the involvement of allosteric/ectopic-binding site in M(1) and M(3) mAChRs. The functionally selective antagonistic behavior of echothiophate and VX makes them potential tools for dissecting the interactions of the mAChR with different G proteins.


Asunto(s)
Inhibidores de la Colinesterasa/farmacología , Yoduro de Ecotiofato/farmacología , Compuestos Organotiofosforados/farmacología , Receptor Muscarínico M1/efectos de los fármacos , Receptor Muscarínico M3/efectos de los fármacos , Animales , Atropina/farmacología , Células CHO , Inhibidores de la Colinesterasa/química , Colforsina/farmacología , Cricetinae , AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Yoduro de Ecotiofato/química , Agonistas Muscarínicos/farmacocinética , Antagonistas Muscarínicos/farmacología , N-Metilescopolamina/farmacocinética , Compuestos Organotiofosforados/química , Oxotremorina/farmacocinética , Fosfatidilinositoles/metabolismo , Unión Proteica/efectos de los fármacos , Receptor Muscarínico M1/fisiología , Receptor Muscarínico M3/fisiología , Transfección/métodos , Tritio/farmacocinética
19.
Neurosci Lett ; 397(3): 214-8, 2006 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-16406345

RESUMEN

The release of soluble amyloid precursor protein alpha (sAPPalpha), produced during alpha-secretase processing by cleavage within the beta amyloid peptide domain of APP, is highly regulated by several external and internal signals. Because evidence suggests the involvement of inflammatory processes in the pathology of Alzheimer's disease and APP formation, we examined the involvement of the phospholipase A2 (PLA2) pathway and of its downstream cyclooxygenase (COX) and lipoxygenase (LOX) pathways in the regulation of sAPPalpha, release induced by muscarinic receptor activation in SH-SY5Y cells. The amount of sAPP released into the culture medium was analyzed using a monoclonal 6E10 antibody detecting sAPPalpha. Treatment with the PLA2 inhibitor, manoalide, blocked the release of oxoM (muscarinic receptor agonist)-stimulated sAPPalpha, and the muscarinic receptor-mediated sAPPalpha release was increased by the non-selective PLA2 activator mellitin. COX and LOX inhibitors inhibited exogenous AA-induced sAPPalpha release, but upregulated basal constitutive sAPPalpha release. However, treatment with COX or LOX inhibitors failed to significantly change oxoM-stimulated sAPPalpha release, and furthermore, muscarinic receptor activation inhibited AA-stimulated COX activity. Our results suggest that sAPPalpha release induced by muscarinic receptor activation is regulated by AA generation via PLA2 activation independently of COX and LOX activities, but that the COX and LOX pathways are possibly involved in the constitutive release of sAPPalpha.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Lipooxigenasa/metabolismo , Fosfolipasas A/fisiología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Receptor Muscarínico M1/fisiología , Receptor Muscarínico M3/fisiología , Línea Celular Tumoral , Inhibidores de la Ciclooxigenasa/farmacología , Humanos , Inhibidores de la Lipooxigenasa/farmacología , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A2 , Solubilidad
20.
J Biol Chem ; 280(36): 32035-47, 2005 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-15994335

RESUMEN

In this study we showed that stimulation of M1 muscarinic acetylcholine receptors (mAChRs) activates endogenous transient receptor potential-canonical, subtype 6 (TRPC6), channels in neuronal PC12D cells. Activation of TRPC6 channels is correlated with the formation of a multiprotein complex containing M1 mAChRs, TRPC6 channels, and protein kinase C (PKC). Formation of the M1 mAChR-TRPC6-PKC complex is transient, with highest levels reached approximately 2 min after stimulation of M1 mAChRs. PKC in the complex phosphorylates TRPC6 on a conserved serine residue in the carboxyl-terminal domain (Ser768 in the TRPC6A isoform and Ser714 in the TRPC6B isoform). The immunophilin FKBP12, the phosphatase calcineurin, and Ca2+-binding protein calmodulin are also recruited to the M1 mAChR-TRPC6-PKC complex following activation of M1 mAChRs and remain stably associated with the TRPC6 channels after M1 mAChRs and PKC have disassociated. Binding of FKBP12, calcineurin, and calmodulin to TRPC6 channels is blocked by the following: 1) inhibition of PKC; 2) mutation of the PKC phosphorylation site (Ser(7168/714)) in the channels; or 3) pretreatment with FK506 or rapamycin, immunosuppressants that directly bind FKBP12. Inhibition of FKBP12 binding blocks the dephosphorylation of TRPC6 channels and the disassociation of M1 mAChRs, without affecting disassociation of PKC. The calcineurin inhibitor cyclosporin A also blocks the dephosphorylation of TRPC6 and prevents the disassociation of M1 mAChRs. Together, these results show that activated TRPC6 channels form the center of a dynamic multiprotein complex that includes PKC and calcineurin, which respectively phosphorylate and dephosphorylate the channels. Phosphorylation of the TRPC6 channels by PKC is required for the binding of FKBP12, which in turn is required for the binding of calcineurin and calmodulin. Subsequent dephosphorylation of the channels by calcineurin is required for the disassociation of M1 mAChRs.


Asunto(s)
Canales de Calcio/metabolismo , Receptor Muscarínico M1/fisiología , Animales , Calcineurina/metabolismo , Calmodulina/metabolismo , Células PC12 , Fosforilación , Proteína Quinasa C/metabolismo , Estructura Secundaria de Proteína , Ratas , Serina/metabolismo , Canales Catiónicos TRPC , Proteína 1A de Unión a Tacrolimus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA