Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Sci Rep ; 10(1): 17581, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-33067534

RESUMEN

SPARC-deficient mice have been shown to exhibit impaired glucose tolerance and insulin secretion, but the underlying mechanism remains unknown. Here, we showed that SPARC enhanced the promoting effect of Muscarinic receptor agonist oxotremorine-M on insulin secretion in cultured mouse islets. Overexpression of SPARC down-regulated RGS4, a negative regulator of ß-cell M3 muscarinic receptors. Conversely, knockdown of SPARC up-regulated RGS4 in Min6 cells. RGS4 was up-regulated in islets from sparc -/- mice, which correlated with decreased glucose-stimulated insulin secretion (GSIS). Furthermore, inhibition of RGS4 restored GSIS in the islets from sparc -/- mice, and knockdown of RGS4 partially decreased the promoting effect of SPARC on oxotremorine-M-stimulated insulin secretion. Phosphoinositide 3-kinase (PI3K) inhibitor LY-294002 abolished SPARC-induced down-regulation of RGS4. Taken together, our data revealed that SPARC promoted GSIS by inhibiting RGS4 in pancreatic ß cells.


Asunto(s)
Secreción de Insulina/efectos de los fármacos , Osteonectina/metabolismo , Proteínas RGS/metabolismo , Animales , Diabetes Mellitus Experimental/metabolismo , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteonectina/genética , Oxotremorina/análogos & derivados , Oxotremorina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas RGS/fisiología , Receptor Muscarínico M3/efectos de los fármacos , Receptor Muscarínico M3/metabolismo
2.
Sci Rep ; 10(1): 14181, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32843670

RESUMEN

Glial fibrillary acidic protein expressing (GFAP+) glia modulate nociceptive neuronal activity in both the peripheral nervous system (PNS) and the central nervous system (CNS). Resident GFAP+ glia in dorsal root ganglia (DRG) known as satellite glial cells (SGCs) potentiate neuronal activity by releasing pro-inflammatory cytokines and neuroactive compounds. In this study, we tested the hypothesis that SGC Gq-coupled receptor (Gq-GPCR) signaling modulates pain sensitivity in vivo using Gfap-hM3Dq mice. Complete Freund's adjuvant (CFA) was used to induce inflammatory pain, and mechanical sensitivity and thermal sensitivity were used to assess the neuromodulatory effect of glial Gq-GPCR activation in awake mice. Pharmacogenetic activation of Gq-GPCR signaling in sensory SGCs decreased heat-induced nociceptive responses and reversed inflammation-induced mechanical allodynia via peripheral adenosine A1 receptor activation. These data reveal a previously unexplored role of sensory SGCs in decreasing afferent excitability. The identified molecular mechanism underlying the analgesic role of SGCs offers new approaches for reversing peripheral nociceptive sensitization.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Hiperalgesia/prevención & control , Inflamación/fisiopatología , Neuroglía/enzimología , Nocicepción/fisiología , Receptor de Adenosina A1/fisiología , Receptor Muscarínico M3/fisiología , Animales , Bencilatos/farmacología , Clozapina/análogos & derivados , Clozapina/farmacología , Adyuvante de Freund/toxicidad , Genes Sintéticos , Calor , Hiperalgesia/fisiopatología , Inflamación/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Agonistas Muscarínicos/farmacología , Neuroglía/fisiología , Nortropanos/farmacología , Regiones Promotoras Genéticas , Agonistas del Receptor Purinérgico P1/farmacología , Antagonistas de Receptores Purinérgicos P1/farmacología , Receptor de Adenosina A1/efectos de los fármacos , Receptor Muscarínico M3/efectos de los fármacos , Receptor Muscarínico M3/genética , Receptores Acoplados a Proteínas G , Proteínas Recombinantes de Fusión/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Teofilina/análogos & derivados , Teofilina/farmacología , Tacto , Xantinas/farmacología
3.
Sleep Breath ; 24(1): 329-337, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31898190

RESUMEN

OBJECTIVE: The objective of this study was to explore the effect of Alpiniae oxyphyllae Fructus (AOF) on a rat model of chronic intermittent hypoxia (CIH)-induced enuresis. Findings of this study may help identify therapeutic targets in children with nocturnal enuresis (NE). METHODS: Female rats were randomly divided into a control group (saline gavage, 4 weeks of normal air), CIH group (saline gavage, 4 weeks of CIH), and AOF group (AOF gavage, 4 weeks of CIH). The variables measured in this study included water intake, urine output, bladder leak point pressure (BLPP), malondialdehyde (MDA) levels, and superoxide dismutase (SOD) activity. The expression levels of the purinergic P2X3 receptor, muscarinic M3 receptor, and ß3-adrenergic receptor (ß3-AR) in the bladder were also measured. The bladder was subjected to haematoxylin and eosin (HE) and Weigert staining, and histological changes were observed under a light microscope to evaluate the morphological changes in the bladder in each group. RESULTS: Compared with the control group, urine output was increased, and the BLPP was decreased in the CIH group, but AOF administration decreased urine output and increased BLPP. In addition, the serum MDA level increased and the SOD activity decreased in the CIH group compared with the control group. Administration of AOF decreased the MDA level and increased the SOD activity. Additionally, compared with the control group, HE and Weigert staining in the CIH group showed that the bladder detrusor muscle bundles were disordered and loose, some muscle bundles were broken, the content of collagen fibres in the gap was reduced, and the gap was significantly widened. However, following the administration of AOF, the bladder detrusor muscle bundles were neatly arranged, and the content of collagen fibres in the gap was increased. Furthermore, compared with the control group, the purinergic P2X3 receptor and muscarinic M3 receptor were expressed at higher levels, and ß3-AR was expressed at lower levels in the CIH group, but AOF administration decreased the expression of the purinergic P2X3 receptor and muscarinic M3 receptor and increased the expression of the ß3-AR. CONCLUSIONS: AOF improves enuresis by inhibiting oxidative stress and regulating the expression of the purinergic P2X3 receptor, muscarinic M3 receptor, and ß3 adrenergic receptor.


Asunto(s)
Modelos Animales de Enfermedad , Enuresis/prevención & control , Hipoxia/complicaciones , Extractos Vegetales/farmacología , Alpinia , Animales , Enuresis/sangre , Femenino , Hipoxia/sangre , Malondialdehído/sangre , Estrés Oxidativo/efectos de los fármacos , Ratas , Receptor Muscarínico M3/efectos de los fármacos , Receptores Adrenérgicos beta 3/efectos de los fármacos , Receptores Purinérgicos P2X3/efectos de los fármacos , Superóxido Dismutasa/sangre , Vejiga Urinaria/efectos de los fármacos , Micción/efectos de los fármacos
4.
Int J Neurosci ; 130(2): 204-211, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31517560

RESUMEN

Aim: Food intake regulated by a complex of physiologic mechanisms in the nervous system. Muscarinergic system has an important role in the central regulation of appetite in mammals, but there is no information for Muscarinic receptors in avian. The purpose of this study was to examine the effects of intracerebroventricular injection of carbachol (cholinergic agonist), Telenzepine (M1 receptor antagonist), AF-DX116 (M2 receptor antagonist), 4-DAMP (M3 receptor antagonist), and PD102807 (M4 receptor antagonist) on feeding behavior in 3-h food-deprived (FD3) neonatal broiler chicken.Materials and Methods: In experiment 1, chicken intracerebroventricular injected with carbachol (125, 250, and 500 nmol). In experiment 2, birds intracerebroventricular injected with telenzepine (125, 250, and 500 nmol). In experiments 3-5, birds intracerebroventricular injected with AF-DX 116 (125, 250, and 500 nmol), 4-DAMP (125, 250, and 500 nmol), and PD102807 (125, 250, and 500 nmol), respectively. In experiment 6, broilers intracerebroventricular injected with carbacol (500 nmol), co-injection of telenzepine (125 nmol)+carbacol (500 nmol), and 4-DAMP (125 nmol)+carbacol (500 nmol). In experiment 7, injection procedure was carbacol (500 nmol), co-injection of AF-DX116 (125 nmol)+carbacol (500 nmol), and PD102807 (125 nmol)+carbacol (500 nmol). Then, food intake measured until 120 min after injection.Results: According to the data, carbachol (250 and 500 nmol) significantly decreased food intake in comparison with control group (P < 0.05). Intracerebroventricular injection of telenzepine (250 and 500 nmol) and 4-DAMP (250 and 500 nmol) significantly increased food intake (P < 0.05). In addition, carbacol-induced hypophagia was significantly attenuated by co-injection of telenzepine + carbacol (P < 0.05). Also, co-injection of 4-DAMP + carbacol decreased the effect of carbacol on food intake (P < 0.05). However, AF-DX116 and PD102807 had no effect on hypophagia induced by carbacol (P > 0.05).Conclusion: These results suggest, hypophagic effect of muscarinergic system is mediated via M1 and M3 receptors in neonatal chicken.


Asunto(s)
Conducta Animal/efectos de los fármacos , Carbacol/farmacología , Ingestión de Alimentos/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Receptor Muscarínico M1/efectos de los fármacos , Receptor Muscarínico M3/efectos de los fármacos , Animales , Animales Recién Nacidos , Carbacol/administración & dosificación , Pollos , Modelos Animales de Enfermedad , Inyecciones Intraventriculares , Agonistas Muscarínicos/administración & dosificación , Antagonistas Muscarínicos/administración & dosificación , Piperidinas/farmacología , Pirenzepina/análogos & derivados , Pirenzepina/farmacología , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/antagonistas & inhibidores , Receptor Muscarínico M3/agonistas , Receptor Muscarínico M3/antagonistas & inhibidores
5.
Proc Natl Acad Sci U S A ; 116(37): 18684-18690, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31451647

RESUMEN

Given the global epidemic in type 2 diabetes, novel antidiabetic drugs with increased efficacy and reduced side effects are urgently needed. Previous work has shown that M3 muscarinic acetylcholine (ACh) receptors (M3Rs) expressed by pancreatic ß cells play key roles in stimulating insulin secretion and maintaining physiological blood glucose levels. In the present study, we tested the hypothesis that a positive allosteric modulator (PAM) of M3R function can improve glucose homeostasis in mice by promoting insulin release. One major advantage of this approach is that allosteric agents respect the ACh-dependent spatiotemporal control of M3R activity. In this study, we first demonstrated that VU0119498, a drug known to act as a PAM at M3Rs, significantly augmented ACh-induced insulin release from cultured ß cells and mouse and human pancreatic islets. This stimulatory effect was absent in islets prepared from mice lacking M3Rs, indicative of the involvement of M3Rs. VU0119498 treatment of wild-type mice caused a significant increase in plasma insulin levels, accompanied by a striking improvement in glucose tolerance. These effects were mediated by ß-cell M3Rs, since they were absent in mutant mice selectively lacking M3Rs in ß cells. Moreover, acute VU0119498 treatment of obese, glucose-intolerant mice triggered enhanced insulin release and restored normal glucose tolerance. Interestingly, doses of VU0119498 that led to pronounced improvements in glucose homeostasis did not cause any significant side effects due to activation of M3Rs expressed by other peripheral cell types. Taken together, the data from this proof-of-concept study strongly suggest that M3R PAMs may become clinically useful as novel antidiabetic agents.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Islotes Pancreáticos/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Receptor Muscarínico M3/efectos de los fármacos , Acetilcolina/metabolismo , Adulto , Regulación Alostérica/efectos de los fármacos , Animales , Glucemia/análisis , Glucemia/metabolismo , Línea Celular Tumoral , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Femenino , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/tratamiento farmacológico , Intolerancia a la Glucosa/metabolismo , Humanos , Hipoglucemiantes/uso terapéutico , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Obesos , Ratones Transgénicos , Persona de Mediana Edad , Agonistas Muscarínicos/uso terapéutico , Obesidad/sangre , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Cultivo Primario de Células , Prueba de Estudio Conceptual , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Adulto Joven
6.
FASEB J ; 33(6): 7252-7260, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30860868

RESUMEN

Electroencephalogram monitoring during propofol (PRO) anesthesia typically features low-frequency oscillations, which may be involved with thalamic reticular nucleus (TRN) modulation. TRN receives noradrenergic inputs from the locus coeruleus (LC). We hypothesized that specific noradrenergic connections in the TRN may contribute to the emergence from PRO anesthesia. Intranuclei norepinephrine (NE) injections (n = 10) and designer receptors exclusively activated by designer drugs (DREADDs) (n = 10) were used to investigate the role of noradrenergic inputs from the LC to the TRN during PRO anesthesia. Whole-cell recording in acute brain slice preparations was used to identify the type of adrenoceptor that regulates noradrenergic innervation in the TRN. An intracerebral injection of NE into the TRN delays arousal in mice recovering from PRO anesthesia (means ± sd; 486.6 ± 57.32 s for the NE injection group vs. 422.4 ± 48.19 s for the control group; P = 0.0143) and increases the cortical-δ (0.1-4 Hz, 25.4 ± 2.9 for the NE injection group vs. 21.0 ± 1.7 for the control group; P = 0.0094) oscillation. An intra-TRN injection of NE also decreased the EC50 of PRO-induced unconsciousness (57.05 ± 1.78 mg/kg for the NE injection group vs. 72.44 ± 3.23 mg/kg for the control group; P = 0.0096). Moreover, the activation of LC-noradrenergic nerve terminals in the TRN using DREADDs increased the recovery time [466.1 ± 44.57 s for the clozapine N-oxide (CNO) injection group vs. 426.1 ± 38.75 s for the control group; P = 0.0033], decreased the EC50 of PRO-induced unconsciousness (64.77 ± 3.40 mg/kg for the CNO injection group vs. 74.00 ± 2.08 mg/kg for the control group; P = 0.0081), and increased the cortical-δ oscillation during PRO anesthesia (23.29 ± 2.58 for the CNO injection group vs. 19.56 ± 1.9 for the control group; P = 0.0213). In addition, whole-cell recording revealed that NE augmented the inhibitory postsynaptic currents in the TRN neurons via the α1-adrenoceptor. Our data indicated that enhanced NE signaling at the noradrenergic terminals of the LC-TRN projection delays arousal from general anesthesia, which is likely mediated by the α1-adrenoceptor activation. Our findings open a door for further understanding of the functions of various LC targets in both anesthesia and arousal.-Zhang, Y., Fu, B., Liu, C., Yu, S., Luo, T., Zhang, L., Zhou, W., Yu, T. Activation of noradrenergic terminals in the reticular thalamus delays arousal from propofol anesthesia in mice.


Asunto(s)
Neuronas Adrenérgicas/fisiología , Anestesia General , Nivel de Alerta/fisiología , Retraso en el Despertar Posanestésico/fisiopatología , Núcleos Talámicos Intralaminares/fisiopatología , Terminaciones Nerviosas/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Neuronas Adrenérgicas/efectos de los fármacos , Anestésicos Intravenosos , Animales , Clozapina/análogos & derivados , Clozapina/farmacología , Drogas de Diseño/farmacología , Electroencefalografía , Vectores Genéticos/administración & dosificación , Núcleos Talámicos Intralaminares/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Terminaciones Nerviosas/efectos de los fármacos , Norepinefrina/farmacología , Técnicas de Placa-Clamp , Propofol , Distribución Aleatoria , Receptor Muscarínico M3/efectos de los fármacos , Receptor Muscarínico M3/fisiología , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Reflejo de Enderezamiento/efectos de los fármacos , Método Simple Ciego , Organismos Libres de Patógenos Específicos , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/fisiología
7.
Neurourol Urodyn ; 37(8): 2560-2570, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30252154

RESUMEN

AIMS: We explored the therapeutic potential of intragastric administration traditional Chinese medicine Glycine tomentella Hayata (I-Tiao-Gung, ITG) extract and its active component Daidzin on cyclophosphamide (CYP)-induced cystitis and bladder hyperactivity in rats. METHODS: Female Wistar rats were divided into control, CYP (200 mg/kg), CYP + ITG (1.17 g/kg/day), CYP + Daidzin (12.5 mg/kg/day), and 1 week of ITG preconditioning with CYP (ITG + CYP) groups. We determined the trans cystometrogram associated with external urethral sphincter electromyogram, and the expression of M2 and M3 muscarinic and P2 × 2 and P2 × 3 purinergic receptors by Western blot in these animals. RESULTS: ITG extract contains 1.07% of Daidzin and 0.77% of Daidzein by high-performance liquid chromatography. Daidzin was more efficient than Daidzein in scavenging H2 O2 activity by a chemiluminescence analyzer. CYP induced higher frequency, shorter intercontraction interval, lower maximal voiding pressure, lower threshold pressure, and Phase-2 emptying contraction with a depressed external urethral sphincter electromyogram activity, and hemorrhagic cystitis in the bladders. The altered parameters by CYP were significantly improved in CYP + ITG, CYP + Daidzin, and ITG + CYP groups. The P2 × 2 and P2 × 3 expressions were significantly upregulated in CYP group, but were depressed in CYP + ITG, CYP + Daidzin, and ITG + CYP groups. The M2 expression was not significantly different among these five groups. The M3 expression was significantly upregulated in CYP group, but was significantly depressed in CYP + ITG, CYP + Daidzin, and ITG + CYP groups. CONCLUSIONS: These data suggest that ITG extract through its active component Daidzin effectively improved CYP-induced cystitis by the action of restoring Phase 2 activity and inhibiting the expressions of P2 × 2, P2 × 3, and M3 receptors.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Isoflavonas/farmacología , Vejiga Urinaria/efectos de los fármacos , Animales , Ciclofosfamida/toxicidad , Cistitis/inducido químicamente , Cistitis/fisiopatología , Electromiografía , Femenino , Ratas , Ratas Wistar , Receptor Muscarínico M2/efectos de los fármacos , Receptor Muscarínico M2/metabolismo , Receptor Muscarínico M3/efectos de los fármacos , Receptor Muscarínico M3/metabolismo , Receptores Purinérgicos P2/efectos de los fármacos , Receptores Purinérgicos P2/metabolismo , Uretra/efectos de los fármacos , Uretra/fisiopatología , Vejiga Urinaria/metabolismo , Vejiga Urinaria/fisiopatología , Vejiga Urinaria Hiperactiva/inducido químicamente , Vejiga Urinaria Hiperactiva/fisiopatología , Micción/efectos de los fármacos
8.
Neurourol Urodyn ; 37(7): 2128-2134, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29675856

RESUMEN

AIM: To examine putative interaction between adrenergic and muscarinic contractile activation in the human urinary outflow tract. METHODS: Tissue from the trigone and prostatic urethra was obtained from 12 cystectomy and 16 prostatectomy specimen. Contractions were elicited by exposure to exogenous agonists before and after inhibition of Rho kinase and protein kinase c (PKC). Immunofluorescence and Western-blot studies were performed using antibodies to muscarinic M3-receptors (M3-R) and alpha1A-adrenoreceptors (alpha1A-AR). The study is registered with ClinicalTrials.gov, number NCT01227447. RESULTS: There was strong co-localization of M3-R and alpha1A-AR on trigonal and urethral myocytes. Western blot analysis revealed a significantly higher expression of alpha1A-AR in the superficial than in the deep trigone. Phenylephrine (PE, 1 µm) augmented contractions induced by carbachol (CA, 3 µm) to more than threefold control in the male superficial trigone, and to about sevenfold control in the proximal urethra. No such potentiation could be detected in female bladder outlet. Both PKC inhibitor GF 109203X and Rho kinase inhibitor Y-27632 reduced responses to 1 µM PE as well as to 3 µM CA significantly. However, the synergistic effect of the combined intervention remained proportionally unaffected. CONCLUSIONS: Muscarinic and adrenergic receptor activation exerts a strong synergistic effect in the male human bladder trigone and proximal urethra.


Asunto(s)
Receptor Muscarínico M3/fisiología , Receptores Adrenérgicos alfa 1/fisiología , Sistema Urinario/inervación , Agonistas alfa-Adrenérgicos/farmacología , Anciano , Anciano de 80 o más Años , Carbacol/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Agonistas Muscarínicos/farmacología , Contracción Muscular/efectos de los fármacos , Fenilefrina/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Receptor Muscarínico M3/efectos de los fármacos , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Caracteres Sexuales , Uretra/efectos de los fármacos , Uretra/fisiología , Quinasas Asociadas a rho/antagonistas & inhibidores
9.
BMC Gastroenterol ; 17(1): 21, 2017 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-28122499

RESUMEN

BACKGROUND: Uridine (Urd), which has been reported as a major component of RNA, plays an important role in various biological process including neuroprotection, biochemical modulation and glycolysis, although its role in constipation has yet to be established. Therefore, in this study, we investigated the laxative effects of Urd on chronic constipation. METHODS: The constipation phenotypes and their related mechanisms were investigated in the transverse colons of SD rats with loperamide (Lop)-induced constipation after treatment with 100 mg/kg of Urd. RESULTS: The number, weight and water contents of stools were significantly higher in the Lop + Urd treated group than the Lop + Vehicle treated group, while food intake and water consumption of the same group were maintained at a constant level. The thickness of the mucosa layer, muscle and flat luminal surface, as well as the number of goblet cells, paneth cells and lipid droplets were enhanced in the Lop + Urd treated group. Furthermore, the expression of the muscarinic acetylcholine receptors M2 and M3 (mAChR M2 and M3) at the transcriptional and translational level was recovered in the Lop + Urd treated group, while some markers such as Gα and inositol triphosphate (IP3) in their downstream signaling pathway were completely recovered by Urd treatment. Moreover, the ability for mucin secretion and the expression of membrane water channel (aquaporine 8, AQP8) were increased significantly in the Lop + Urd treated group compared with Lop + Vehicle treated group. Finally, the activity of Urd was confirmed in primary smooth muscle of rat intestine cells (pRISMC) based on Gα expression and IP3 concentration. CONCLUSIONS: The results of the present study provide the first strong evidence that Urd can be considered an important candidate for improving chronic constipation induced by Lop treatment in animal models.


Asunto(s)
Estreñimiento/tratamiento farmacológico , Estreñimiento/metabolismo , Laxativos/uso terapéutico , Mucinas/metabolismo , Receptor Muscarínico M2/metabolismo , Receptor Muscarínico M3/metabolismo , Uridina/uso terapéutico , Animales , Colon Transverso/efectos de los fármacos , Colon Transverso/patología , Colon Transverso/ultraestructura , Modelos Animales de Enfermedad , Conducta Alimentaria/efectos de los fármacos , Fosfatos de Inositol/metabolismo , Mucosa Intestinal/metabolismo , Mucinas/efectos de los fármacos , Músculo Liso/metabolismo , Ratas Sprague-Dawley , Receptor Muscarínico M2/efectos de los fármacos , Receptor Muscarínico M3/efectos de los fármacos , Transducción de Señal , Uridina/metabolismo , Uridina/farmacología
10.
Environ Toxicol ; 32(2): 371-381, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26916448

RESUMEN

Regarding the human health effects, airborne fine particulate matter 2.5 (PM2.5 ) is an important environmental risk factor. However, the underlying molecular mechanisms are largely unknown. The present study examined the hypothesis that PM2.5 causes bronchial hyperreactivity by upregulated muscarinic receptors via the mitogen-activated protein kinase (MAPK) pathway. The isolated rat bronchi segments were cultured with different concentration of PM2.5 for different time. The contractile response of the bronchi segments were recorded by a sensitive myograph. The mRNA and protein expression levels of M3 muscarinic receptors were studied by quantitative real-time PCR and immunohistochemistry, respectively. The muscarinic receptors agonist, carbachol induced a remarkable contractile response on fresh and DMSO cultured bronchial segments. Compared with the fresh or DMSO culture groups, 1.0 µg/mL of PM2.5 cultured for 24 h significantly enhanced muscarinic receptor-mediated contractile responses in bronchi with a markedly increased maximal contraction. In addition, the expression levels of mRNA and protein for M3 muscarinic receptors in bronchi of PM2.5 group were higher than that of fresh or DMSO culture groups. SB203580 (p38 inhibitor) and U0126 (MEK1/2 inhibitor) significantly inhibited the PM2.5 -induced enhanced contraction and increased mRNA and protein expression of muscarinic receptors. However, JNK inhibitor SP600125 had no effect on PM2.5 -induced muscarinic receptor upregulation and bronchial hyperreactivity. In conclusion, airborne PM2.5 upregulates muscarinic receptors, which causes subsequently bronchial hyperreactivity shown as enhanced contractility in bronchi. This process may be mediated by p38 and MEK1/2 MAPK pathways. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 371-381, 2017.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Hiperreactividad Bronquial/inducido químicamente , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Material Particulado/toxicidad , Receptor Muscarínico M3/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Bronquios/efectos de los fármacos , Hiperreactividad Bronquial/fisiopatología , Inmunohistoquímica , Masculino , Contracción Muscular/efectos de los fármacos , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos
11.
Cardiovasc Res ; 112(3): 669-676, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27702763

RESUMEN

AIMS: Although the nature of the humoral factor which mediates cardioprotection established by remote ischaemic conditioning (RIc) remains unknown, parasympathetic (vagal) mechanisms appear to play a critical role. As the production and release of many gut hormones is modulated by the vagus nerve, here we tested the hypothesis that RIc cardioprotection is mediated by the actions of glucagon-like peptide-1 (GLP-1). METHODS AND RESULTS: A rat model of myocardial infarction (coronary artery occlusion followed by reperfusion) was used. Remote ischaemic pre- (RIPre) or perconditioning (RIPer) was induced by 15 min occlusion of femoral arteries applied prior to or during the myocardial ischaemia. The degree of RIPre and RIPer cardioprotection was determined in conditions of cervical or subdiaphragmatic vagotomy, or following blockade of GLP-1 receptors (GLP-1R) using specific antagonist Exendin(9-39). Phosphorylation of PI3K/AKT and STAT3 was assessed. RIPre and RIPer reduced infarct size by ∼50%. In conditions of bilateral cervical or subdiaphragmatic vagotomy RIPer failed to establish cardioprotection. GLP-1R blockade abolished cardioprotection induced by either RIPre or RIPer. Exendin(9-39) also prevented RIPre-induced AKT phosphorylation. Cardioprotection induced by GLP-1R agonist Exendin-4 was preserved following cervical vagotomy, but was abolished in conditions of M3 muscarinic receptor blockade. CONCLUSIONS: These data strongly suggest that GLP-1 functions as a humoral factor of remote ischaemic conditioning cardioprotection. This phenomenon requires intact vagal innervation of the visceral organs and recruitment of GLP-1R-mediated signalling. Cardioprotection induced by GLP-1R activation is mediated by a mechanism involving M3 muscarinic receptors.


Asunto(s)
Arteria Femoral/cirugía , Péptido 1 Similar al Glucagón/metabolismo , Precondicionamiento Isquémico/métodos , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Nervio Vago/fisiopatología , Animales , Modelos Animales de Enfermedad , Exenatida , Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Receptor del Péptido 1 Similar al Glucagón/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Antagonistas de Hormonas/farmacología , Ligadura , Masculino , Antagonistas Muscarínicos/farmacología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Receptor Muscarínico M3/efectos de los fármacos , Receptor Muscarínico M3/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Vagotomía , Nervio Vago/efectos de los fármacos , Nervio Vago/cirugía , Ponzoñas/farmacología
12.
Psychopharmacology (Berl) ; 233(2): 267-80, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26518024

RESUMEN

RATIONALE: Basal forebrain cholinergic neurons modulate the activation of cortical neurons by several stimuli such as fear and anxiety. However, the role of the muscarinic receptor in the medial prefrontal cortex (MPFC) in the modulation of the conditioned emotional response (CER) evoked in the model contextual conditioned fear remains unclear. OBJECTIVES: The objective of this study is to test the hypothesis that inhibition of the muscarinic receptor in ventral MPFC modulates CER observed during animal's re-exposure to the aversive context. METHODS: Rats implanted with cannulae aimed at the prelimbic (PL) or the infralimbic (IL) were submitted to a high-intensity contextual fear conditioning protocol. Before the test session, they received microinjections of the hemicholinium (choline reuptake blocker), atropine (muscarinic antagonist), J104129 fumarate (M1-M3 muscarinic antagonists), pirenzepine (M1 muscarinic antagonist), neostigmine (inhibitor acetylcholinesterase enzyme), or the systemic administration of the FG7142 (inverse benzodiazepine agonist). Additional independent groups received the neostigmine or FG7142 before the ineffective doses of J104129 fumarate in the low-intensity protocol of contextual fear conditioning. RESULTS: In the high-intensity protocol, the administration of hemicholinium (1 nmol), atropine (0.06-6 nmol), J104129 fumarate (6 nmol), or pirenzepine (6 nmol) attenuated the expression of CER in rats. However, in the low-intensity protocol, only J10129 fumarate (0.06 nmol) reduced the expression of the CER. Finally, neostigmine (0.1-1 nmol) or FG7142 (8 mg/Kg) increased CER expression, an effect inhibited by the low dose of the J10129 fumarate. CONCLUSIONS: These results indicated that the blockade of M3 muscarinic receptor in the vMPFC attenuates the CER expression.


Asunto(s)
Condicionamiento Psicológico , Miedo/psicología , Corteza Prefrontal/metabolismo , Receptor Muscarínico M3/metabolismo , Animales , Benzodiazepinas/agonistas , Inhibidores de la Colinesterasa/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Emociones , Masculino , Antagonistas Muscarínicos/farmacología , Sistema Nervioso Parasimpático/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Wistar , Receptor Muscarínico M3/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
13.
J Dent Res ; 94(5): 722-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25784251

RESUMEN

The presence of functional autoantibodies against the muscarinic type 3 receptor (M3R) has been reported in primary Sjögren's syndrome (pSS). However, the pathogenic role of these autoantibodies in pSS development remains to be elucidated. In this experiment, we investigated a pathologic role of pSS autoantibodies (pSS IgG) associated with downregulation of the major histocompatibility complex I (MHC I) molecule with M3R through internalization. Anti-M3R autoantibodies in purified control and pSS IgG were detected using 4 synthesized cyclic M3R peptides by enzyme-linked immunosorbent assay. The binding reactivity of pSS IgG to M3R in situ was analyzed by a dual immunostaining method. Surface expression, interaction, and internalization of M3R with MHC I were analyzed by immunofluorescence confocal microscopy and biochemical assays. Synthetic cyclic peptides M3RP(205-221) and M3RP(520-527) showed significantly high reactivity with pSS IgG compared to the control IgG or the other 3 peptides (P < 0.05). Significantly high reactivity of pSS IgG to M3R in situ was observed. PSS IgG increased the interaction of membrane M3R with MHC I and induced their internalization in primary human submandibular gland cells. The pSS IgG-induced internalization of M3R with MHC I was significantly inhibited by the cholesterol-sequestering drug filipin. Our novel finding-namely, strong downregulation of the membrane MHC I with M3R through internalization of the cholesterol-rich microdomain associating with anti-M3R autoantibodies-could be an important mechanism contributing to the impaired salivation seen in pSS and linking secretory hypofunction to autoimmune pathogenesis.


Asunto(s)
Autoanticuerpos/inmunología , Receptor Muscarínico M3/inmunología , Síndrome de Sjögren/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Técnicas de Cultivo de Célula , Membrana Celular/inmunología , Células Cultivadas , Citosol/inmunología , Regulación hacia Abajo , Endocitosis/efectos de los fármacos , Endocitosis/inmunología , Femenino , Filipina/farmacología , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunoglobulina G/efectos de los fármacos , Inmunoglobulina G/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Masculino , Microscopía Confocal , Microscopía Fluorescente , Persona de Mediana Edad , Receptor Muscarínico M3/efectos de los fármacos , Glándula Submandibular/citología , Glándula Submandibular/inmunología , Adulto Joven
14.
ACS Chem Neurosci ; 6(3): 476-84, 2015 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-25587888

RESUMEN

Over the past decade, two independent technologies have emerged and been widely adopted by the neuroscience community for remotely controlling neuronal activity: optogenetics which utilize engineered channelrhodopsin and other opsins, and chemogenetics which utilize engineered G protein-coupled receptors (Designer Receptors Exclusively Activated by Designer Drugs (DREADDs)) and other orthologous ligand-receptor pairs. Using directed molecular evolution, two types of DREADDs derived from human muscarinic acetylcholine receptors have been developed: hM3Dq which activates neuronal firing, and hM4Di which inhibits neuronal firing. Importantly, these DREADDs were not activated by the native ligand acetylcholine (ACh), but selectively activated by clozapine N-oxide (CNO), a pharmacologically inert ligand. CNO has been used extensively in rodent models to activate DREADDs, and although CNO is not subject to significant metabolic transformation in mice, a small fraction of CNO is apparently metabolized to clozapine in humans and guinea pigs, lessening the translational potential of DREADDs. To effectively translate the DREADD technology, the next generation of DREADD agonists are needed and a thorough understanding of structure-activity relationships (SARs) of DREADDs is required for developing such ligands. We therefore conducted the first SAR studies of hM3Dq. We explored multiple regions of the scaffold represented by CNO, identified interesting SAR trends, and discovered several compounds that are very potent hM3Dq agonists but do not activate the native human M3 receptor (hM3). We also discovered that the approved drug perlapine is a novel hM3Dq agonist with >10 000-fold selectivity for hM3Dq over hM3.


Asunto(s)
Drogas de Diseño , Receptor Muscarínico M3/química , Receptor Muscarínico M3/efectos de los fármacos , Acetilcolina/farmacología , Animales , Agonistas Colinérgicos/farmacología , Clozapina/análogos & derivados , Clozapina/farmacología , Dibenzazepinas/farmacología , Humanos , Ligandos , Antagonistas de la Serotonina/farmacología , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
15.
Planta Med ; 81(2): 130-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25590366

RESUMEN

Dendrobium officinale is an herbal medicine that has been clinically used to promote body fluid production. Previous works demonstrated that D. officinale polysaccharides could ameliorate symptoms of salivary secretion of patients with Sjögren's syndrome and in a respective mice model. In the present study, we investigated the underlying mechanism by which D. officinale polysaccharides activate M3 muscarinic receptors and induce extracellular calcium influx, leading to the translocation of aquaporin 5, a water channel protein, to the apical membrane of human submandibular gland epithelial cells. Enzymatic treatment of D. officinale polysaccharides suggested that they are hydrolyzed but do not permeate cell membranes. This finding supports the pharmacological activity of D. officinale polysaccharides to promote salivary secretion.


Asunto(s)
Acuaporina 5/metabolismo , Dendrobium/química , Extractos Vegetales/farmacología , Polisacáridos/farmacología , Receptor Muscarínico M3/metabolismo , Síndrome de Sjögren/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Transporte de Proteínas/efectos de los fármacos , Receptor Muscarínico M3/efectos de los fármacos
16.
Int J Clin Pract ; 69(2): 242-50, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25363415

RESUMEN

OBJECTIVES: To evaluate the dose-response relationship of tarafenacin, an antimuscarinic agent in development phase, for efficacy and safety, at daily doses of 0.2 and 0.4 mg for the treatment of overactive bladder (OAB) PATIENTS AND METHODS: This multicentre, placebo-controlled, randomised, double-blind, phase 2b study was conducted. Patients were randomised to tarafenacin 0.2 mg, tarafenacin 0.4 mg or placebo daily for 12 weeks. Adult patients with OAB for at least 6 months, with an average of ≥ 8 micturitions per day and ≥ 3 incontinence episodes or a total of ≥ 6 urgency episodes per 3 days were enrolled. The primary objective was to compare the mean changes in the number of micturitions per 24 h of the two doses of tarafenacin compared with placebo from baseline to 12 weeks after treatment. RESULTS: A total of 334 patients were screened, of whom 235 patients were randomised. The mean decrease in the number of micturitions per 24 h from baseline to 12 weeks was statistically higher in the tarafenacin 0.4 mg group (-2.43 ± 2.21 times per day, p = 0.033) and non-statistically significant in the tarafenacin 0.2 mg group (-1.92 ± 2.45 times per day, p = 0.393) when compared with the placebo group (-1.77 ± 2.95 times per day). There were no statistically significant differences in the mean change of urgency episodes per 24 h among three groups. The most common adverse event was dry mouth. There were no significant differences in blurred vision and constipation compared with placebo. CONCLUSIONS: Tarafenacin 0.4 mg decreased the number of micturitions in patients with OAB after 12 weeks compared with placebo, and the dose-response relationship of tarafenacin 0.2 and 0.4 mg was confirmed. Both dose levels of tarafenacin were well tolerated.


Asunto(s)
Antagonistas Muscarínicos/uso terapéutico , Receptor Muscarínico M3/efectos de los fármacos , Vejiga Urinaria Hiperactiva/tratamiento farmacológico , Incontinencia Urinaria/tratamiento farmacológico , Adulto , Anciano , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Antagonistas Muscarínicos/efectos adversos , Placebos
17.
Neurochem Res ; 40(10): 2068-77, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24821386

RESUMEN

We have found earlier that changes in membrane cholesterol content have distinct impact on signaling via the M1, M2, or M3 receptors expressed in CHO cells (CHO-M1 through CHO-M3). Now we investigated whether gradual changes in membrane cholesterol exerts differential effects on coupling of the M1 and M3 muscarinic receptors to preferential signaling pathways through Gq/11 and non-preferential Gs G-proteins signaling. Changes in membrane cholesterol resulted in only marginal alterations of antagonist and agonist affinity of the M1 and M3 receptors, and did not influence precoupling of either subtype. Changes in membrane cholesterol did not influence parameters of carbachol-stimulated GTP-γ(35)S binding in CHO-M1 membranes while reduction as well as augmentation of membrane cholesterol lowered the efficacy but increased the potency of carbachol in CHO-M3 membranes. Gradual increase or decrease in membrane cholesterol concentration dependently attenuated agonist-induced inositolphosphates release while only cholesterol depletion increased basal values in both cell lines. Similarly, membrane cholesterol manipulation modified basal and agonist-stimulated cAMP synthesis via Gs in the same way in both cell lines. These results demonstrate that changes in membrane cholesterol concentration differentially impact preferential and non-preferential M1 and M3 receptor signaling. They point to the activated G-protein/effector protein interaction as the main site of action in alterations of M1 receptor-mediated stimulation of second messenger pathways. On the other hand, modifications in agonist-stimulated GTP-γ(35)S binding in CHO-M3 membranes indicate that in this case changes in ligand-activated receptor/G-protein interaction may also play a role.


Asunto(s)
Colesterol/metabolismo , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M3/metabolismo , Animales , Células CHO , Carbacol/farmacología , Cricetulus , Proteínas de Unión al GTP/metabolismo , Humanos , Receptor Muscarínico M1/efectos de los fármacos , Receptor Muscarínico M3/efectos de los fármacos , Sistemas de Mensajero Secundario/fisiología , Transducción de Señal
18.
Urol J ; 11(3): 1642-7, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-25015611

RESUMEN

PURPOSE: To locate the muscarinic (M) M2 and M3 receptors in bladder interstitial cells of Cajal (ICCs) and to determine the effects of M2 and M3 agonists on bladder ICCs. MATERIALS AND METHODS: A total of 30 adult male Sprague-Dawley rats weighing 225-250 g were used in this study. Double-labeled fluorescence of muscarinic receptors and c-kit was performed for co-localization. To evaluate the effect of muscarinic agents on the excitation of bladder ICCs, we analyzed the inward current of bladder ICCs using the whole-cell patch clamp. The effect of muscarinic agents on the carbachol-induced inward currents was evaluated with the whole-cell patch clamp. RESULTS: M2 and M3 receptors were confirmed in the stroma ICCs in rats' bladders with double-labeled immunofluorescence. Spontaneous action potential was observed in freshly isolated bladder ICCs. The carbachol-induced inward Ca2+ current in ICCs can be blocked by atropine. The M2 receptor antagonist methoctramine (1 µM) showed a weak inhibitory capability on the inward Ca2+ current [from 74.8 ± 9.6 to 63.3 ± 13.8 Pascal (pA), n = 12, P = .03]. While the M3 receptor antagonist 4-diphenyl-acetoxy-N-methyl-piperidine methiodide (4-DAMP) (1 µM) significantly inhibited the inward Ca2+ current (from 78.4 ± 11.2 to 17.3 ± 7.9 pA, n = 12, P < .001). CONCLUSION: Bladder ICCs express M2 and M3 cholinergic receptors. Most muscarinic cholinergic receptor antagonists, especially the M3 antagonists, can effectively inhibit the carbamylcholine- induced inward current of bladder ICCs.


Asunto(s)
Canales de Calcio/fisiología , Células Intersticiales de Cajal/fisiología , Receptor Muscarínico M2/fisiología , Receptor Muscarínico M3/fisiología , Potenciales de Acción , Animales , Atropina/farmacología , Canales de Calcio/efectos de los fármacos , Carbacol/farmacología , Agonistas Colinérgicos/farmacología , Diaminas/farmacología , Células Intersticiales de Cajal/química , Masculino , Antagonistas Muscarínicos/farmacología , Técnicas de Placa-Clamp , Piperidinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M2/análisis , Receptor Muscarínico M2/efectos de los fármacos , Receptor Muscarínico M3/análisis , Receptor Muscarínico M3/efectos de los fármacos , Vejiga Urinaria/química , Vejiga Urinaria/fisiología
19.
Br J Anaesth ; 112(2): 370-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24065730

RESUMEN

BACKGROUND: Signalling of several G-protein-coupled receptors of the Gq/11 family is time-dependently inhibited by local anaesthetics (LAs). Since LA-induced modulation of muscarinic m1 and m3 receptor function may explain their beneficial effects in clinical practice, such as decreased postoperative cognitive dysfunction or less bronchoconstriction, we studied how prolonged exposure affects muscarinic signalling (Wang D, Wu X, Li J, Xiao F, Liu X, Meng M. The effect of lidocaine on early postoperative cognitive dysfunction after coronary artery bypass surgery. Anesth Analg 2002; 95: 1134-41; Groeben H, Silvanus MT, Beste M, Peters J. Combined lidocaine and salbutamol inhalation for airway anesthesia markedly protects against reflex bronchoconstriction. Chest 2000; 118: 509-15). METHODS: A two-electrode voltage clamp was used to assess the effects of lidocaine or its permanently charged analogue QX314 on recombinantly expressed m1 and m3 receptors in Xenopus oocytes. Antisense knock-down of functional Gαq-protein and inhibition of protein kinase C (PKC) served to define mechanisms and sites of action. RESULTS: Lidocaine affected muscarinic signalling in a biphasic way: an initial decrease in methylcholine bromide-elicited m1 and m3 responses after 30 min, followed by a significant increase in muscarinic responses after 8 h. Intracellularly injected QX314 time-dependently inhibited muscarinic signalling, but had no effect in Gαq-depleted oocytes. PKC-antagonism enhanced m1 and m3 signalling, but completely abolished the LA-induced increase in muscarinic responses, unmasking an underlying time-dependent inhibition of m1 and m3 responses after 8 h. CONCLUSIONS: Lidocaine modulates muscarinic m1 and m3 receptors in a time- and Gαq-dependent manner, but this is masked by enhanced PKC activity. The biphasic time course may be due to interactions of LAs with an extracellular receptor domain, modulated by PKC activity. Prolonged exposure to LAs may not benefit pulmonary function, but may positively affect postoperative cognitive function.


Asunto(s)
Anestésicos Locales/farmacología , Lidocaína/farmacología , Receptor Muscarínico M1/efectos de los fármacos , Receptor Muscarínico M3/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Ratas , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M3/metabolismo , Factores de Tiempo , Xenopus laevis
20.
Mol Endocrinol ; 27(8): 1208-16, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23820900

RESUMEN

The release of insulin from pancreatic ß-cells is regulated by a considerable number of G protein-coupled receptors. During the past several years, we have focused on the physiological importance of ß-cell M3 muscarinic acetylcholine receptors (M3Rs). At the molecular level, the M3R selectively activates G proteins of the G(q) family. Phenotypic analysis of several M3R mutant mouse models, including a mouse strain that lacks M3Rs only in pancreatic ß-cells, indicated that ß-cell M3Rs play a key role in maintaining blood glucose levels within a normal range. Additional studies with transgenic M3R mouse models strongly suggest that strategies aimed to enhance signaling through ß-cell M3Rs may prove useful in the treatment of type 2 diabetes. More recently, we analyzed transgenic mice that expressed an M3R-based designer receptor in a ß-cell-specific fashion, which enabled us to chronically activate a ß-cell G(q)-coupled receptor by a drug that is otherwise pharmacologically inert. Drug-dependent activation of this designer receptor stimulated the sequential activation of G(q), phospholipase C, ERK1/2, and insulin receptor substrate 2 signaling, thus triggering a series of events that greatly improved ß-cell function. Most importantly, chronic stimulation of this pathway protected mice against experimentally induced diabetes and glucose intolerance, induced either by streptozotocin or by the consumption of an energy-rich, high-fat diet. Because ß-cells are endowed with numerous receptors that mediate their cellular effects via activation of G(q)-type G proteins, these findings provide a rational basis for the development of novel antidiabetic drugs targeting this class of receptors.


Asunto(s)
Diabetes Mellitus Tipo 2/prevención & control , Intolerancia a la Glucosa/prevención & control , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/metabolismo , Receptor Muscarínico M3/metabolismo , Animales , Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Unión al GTP/metabolismo , Intolerancia a la Glucosa/tratamiento farmacológico , Proteínas Sustrato del Receptor de Insulina/efectos de los fármacos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Ratones Noqueados , Receptor Muscarínico M3/efectos de los fármacos , Receptor Muscarínico M3/genética , Transducción de Señal , Fosfolipasas de Tipo C/efectos de los fármacos , Fosfolipasas de Tipo C/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...