Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 328
Filtrar
1.
J Clin Hypertens (Greenwich) ; 26(1): 36-46, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38010846

RESUMEN

Recent studies have reported the role of the M3 muscarinic acetylcholine receptor (M3R), a member of the G-protein coupled receptor superfamily, encoded by the CHRM3 gene, in cardiac function and the regulation of blood pressure (BP). The aim of this study was to investigate the associations of CHRM3 genetic variants with salt sensitivity, longitudinal BP changes, and the development of hypertension in a Chinese population. We conducted a chronic dietary salt intervention experiment in a previously established Chinese cohort to analyze salt sensitivity of BP. Additionally, a 14-year follow-up was conducted on all participants in the cohort to evaluate the associations of CHRM3 polymorphisms with longitudinal BP changes, as well as the incidence of hypertension. The single nucleotide polymorphism (SNP) rs10802811 within the CHRM3 gene displayed significant associations with low salt-induced changes in systolic blood pressure (SBP), diastolic blood pressure (DBP), and mean arterial pressure (MAP), while rs373288072, rs114677844, and rs663148 exhibited significant associations with SBP and MAP responses to a high-salt diet. Furthermore, the SNP rs58359377 was associated with changes in SBP and pulse pressure (PP) over the course of 14 years. Additionally, the 14-year follow-up revealed a significant association between the rs619288 polymorphism and an increased risk of hypertension (OR = 1.74, 95% CI: 1.06-2.87, p = .029). This study provides evidence that CHRM3 may have a role in salt sensitivity, BP progression, and the development of hypertension.


Asunto(s)
Hipertensión , Adulto , Humanos , Presión Sanguínea/genética , Hipertensión/epidemiología , Hipertensión/genética , Cloruro de Sodio Dietético/efectos adversos , Incidencia , Polimorfismo de Nucleótido Simple , China/epidemiología , Receptor Muscarínico M3/genética
2.
FEBS Lett ; 597(8): 1164-1175, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36754848

RESUMEN

Cholinergic receptor muscarinic 3 (CHRM3)-mediated focal adhesion kinase/YES-associated protein (YAP) signalling is essential for the growth of castration-resistant prostate cancer (CRPC) cells. Here, we evaluated the molecular mechanisms through which CHRM3 overexpression facilitates castration-resistant growth. Small RNA sequencing combined with in silico analyses revealed that CHRM3 was a putative target of miR-15b-5p. Notably, androgen deprivation suppressed miR-15b-5p expression and increased CHRM3 expression. Moreover, miR-15b-5p bound directly to CHRM3 and inhibited YAP activation induced by CHRM3 stimulation. Furthermore, miR-15b-5p abolished the growth of CRPC cells induced by CHRM3 stimulation. We conclude that the miR-15b-5p/CHRM3/YAP signalling axis promotes the castration-resistant growth of prostate cancer.


Asunto(s)
MicroARNs , Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , MicroARNs/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Antagonistas de Andrógenos , Proliferación Celular/fisiología , Castración , Línea Celular Tumoral , Receptores Colinérgicos/metabolismo , Colinérgicos , Regulación Neoplásica de la Expresión Génica , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo
3.
Mol Neurobiol ; 60(1): 317-328, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36264433

RESUMEN

Emerging evidence suggested that long non-coding RNAs (lncRNAs) were involved in Parkinson's disease (PD) pathogenesis. Herein, we used gene expression profiles from GEO database to construct a PD-specific ceRNA network. Functional enrichment analysis suggested that ceRNA network might participate in the development of PD. PPI networks were constructed, and the ceRNA subnetwork based on five hub genes was set up. In a cohort of 32 PD patients and 31 healthy controls, the expression of 10 DElncRNAs (TTC3-AS1, LINC01259, ZMYND10-AS1, CHRM3-AS1, MYO16-AS1, AGBL5-IT1, HOTAIRM1, RABGAP1L-IT1, HLCS-IT1, and LINC00393) were further verified. Consistent with the microarray data, LINC01259 expression was significantly lower in PD patients compared with controls (P = 0.008). Intriguingly, such a difference was only observed among male patients and male controls when dividing study participants based on their gender (P = 0.016). However, the expression of other lncRNAs did not differ significantly between the two groups. Receiver operating characteristic (ROC) curve analysis revealed that the diagnostic power of LINC01259 was 0.694 for PD and 0.677 for early-stage PD. GSEA enrichment analysis revealed that LINC01259 was mainly enriched in biological processes associated with immune function and inflammatory response. Moreover, LINC01259 expression was not correlated with age of patients, disease duration, disease stage, MDS-UPDRS score, MDS-UPDRS III score, MMSE score, and MOCA score. The current study provides further evidence for the dysregulation of lncRNAs in circulating leukocytes of PD patients, revealing that LINC01259 has clinical potential as a novel immune and inflammatory biomarker for PD and early-stage PD diagnosis.


Asunto(s)
Enfermedad de Parkinson , ARN Largo no Codificante , Humanos , Masculino , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Enfermedad de Parkinson/genética , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Femenino
4.
Dig Dis Sci ; 68(2): 439-450, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35947306

RESUMEN

BACKGROUND: The specific role of the M3 muscarinic acetylcholine receptor in gastrointestinal motility under physiological conditions is unclear, due to a lack of subtype-selective compounds. AIMS: The objective of this study was to determine the region-specific role of the M3 receptor in gastrointestinal motility. METHODS: We developed a novel positive allosteric modulator (PAM) for the M3 receptor, PAM-369. The effects of PAM-369 on the carbachol-induced contractile response of porcine esophageal smooth muscle and mouse colonic smooth muscle (ex vivo) and on the transit in mouse small intestine and rat colon (in vivo) were examined. RESULTS: PAM-369 selectively potentiated the M3 receptor under the stimulation of its orthosteric ligands without agonistic or antagonistic activity. Half-maximal effective concentrations of PAM activity for human, mouse, and rat M3 receptors were 0.253, 0.345, and 0.127 µM, respectively. PAM-369 enhanced carbachol-induced contraction in porcine esophageal smooth muscle and mouse colonic smooth muscle without causing any contractile responses by itself. The oral administration of 30 mg/kg PAM-369 increased the small intestinal transit in both normal motility and loperamide-induced intestinal dysmotility mice but had no effects on the colonic transit, although the M3 receptor mRNA expression is higher in the colon than in the small intestine. CONCLUSIONS: This study provided the first direct evidence that the M3 receptor has different region-specific roles in the motility function between the small intestine and colon in physiological and pathophysiological contexts. Selective PAMs designed for targeted subtypes of muscarinic receptors are useful for elucidating the subtype-specific function.


Asunto(s)
Motilidad Gastrointestinal , Receptor Muscarínico M3 , Animales , Humanos , Ratones , Ratas , Carbacol/farmacología , Motilidad Gastrointestinal/genética , Motilidad Gastrointestinal/fisiología , Contracción Muscular , Receptor Muscarínico M2/genética , Receptor Muscarínico M2/metabolismo , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Receptores Muscarínicos/fisiología , Porcinos
5.
Eur J Gastroenterol Hepatol ; 34(10): 1031-1041, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36052691

RESUMEN

OBJECTIVE: The aim of this study was to find differentially expressed long noncoding RNAs (lncRNAs), microRNAs (miRNAs) and mRNAs and related signaling pathways, contributing to understanding the molecular mechanism of acute recurrent pancreatitis (ARP). METHODS: First, peripheral whole blood samples from five acute pancreatitis (AP) patients, five ARP patients and five healthy individuals ( N ) were collected for RNA sequencing. Second, differentially/specifically expressed lncRNAs, miRNAs and mRNAs were identified in AP vs. N , ARP vs. N and ARP. Third, the ceRNA (lncRNA-miRNA-mRNA) networks of common/specifical lncRNAs, miRNAs and mRNAs were constructed in AP vs. N , ARP vs. N and ARP. Finally, functional analysis of common mRNAs in AP vs. N and ARP vs. N was performed. RESULTS: A total of 315 common lncRNAs, 12 common miRNAs and 909 common mRNAs were identified between AP and ARP. Ninety-four specifically expressed lncRNAs, one specifically expressed miRNAs and 286 specifically expressed mRNAs were found in ARP. Some interaction pairs were identified in AP and ARP, such as LUCAT1/NEAT1-hsa-miR-16-2-3p-HK2, CHRM3-AS2-hsa-miR-122-5p/hsa-miR-145-3p-DBH/CACNA1C, CHRM3-AS2-hsa-miR-200a-3p-PDGFD, RBM26-AS1-hsa-miR-200b-3p-FHIT and LINC00891/KTN1-AS1-hsa-miR-143-3p-tyrosine kinase (TXK). ASAP1-IT2/DGCR9-hsa-miR-342-5p-ABCC5/MAP2K6 was the only one specific interaction pair identified in ARP. Four significantly enriched signaling pathways were identified in AP vs. N and ARP vs. N , including amino sugar and nucleotide sugar metabolism (involved NPL and HK2), MAPK signaling pathway (involved CACNA1C and PDGFD), metabolic pathways (involved DBH and FHIT) and leukocyte transendothelial migration (involved TXK). CONCLUSION: The identified altered lncRNAs, miRNAs, mRNAs and related signaling pathways may be involved in the AP development and recurrence.


Asunto(s)
MicroARNs , Pancreatitis , ARN Largo no Codificante , Enfermedad Aguda , Redes Reguladoras de Genes , Humanos , Proteínas de la Membrana/genética , MicroARNs/genética , MicroARNs/metabolismo , Pancreatitis/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , Receptor Muscarínico M3/genética
6.
PLoS One ; 17(6): e0269618, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35657974

RESUMEN

Overexpression of M3 subtype muscarinic receptors (M3R) hastens colon cancer progression. As microRNA (miRNA) expression is commonly dysregulated in cancer, we used microarrays to examine miRNA profiles in muscarinic receptor agonist-treated human colon cancer cells. We used quantitative RT-PCR (qPCR) to validate microarray results and examine miRNA expression in colon cancers and adjacent normal colon. These assays revealed that acetylcholine (ACh) treatment robustly induced miR-222 expression; miR-222 levels were three-fold higher in cancer compared to normal colon. In kinetic studies, ACh induced a 4.6-fold increase in pri-miR-222 levels within 1 h, while mature miR-222 increased gradually to 1.8-fold within 4 h. To identify post-M3R signaling mediating these actions, we used chemical inhibitors and agonists. ACh-induced increases in pri-miR-222 were attenuated by pre-incubating cells with atropine and inhibitors of protein kinase C (PKC) and p38 MAPK. Treatment with a PKC agonist, phorbol 12-myristate 13-acetate, increased pri-miR-222 levels, an effect blocked by PKC and p38 MAPK inhibitors, but not by atropine. Notably, treatment with ACh or transfection with miR-222 mimics increased cell proliferation; atropine blocked the effects of ACh but not miR-222. These findings identify a novel mechanism whereby post-M3R PKC/p38 MAPK signaling stimulates miR-222 expression and colon cancer cell proliferation.


Asunto(s)
Neoplasias del Colon , MicroARNs , Acetilcolina/metabolismo , Acetilcolina/farmacología , Derivados de Atropina , Colinérgicos , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Humanos , Cinética , MicroARNs/genética , Antagonistas Muscarínicos/farmacología , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Nat Commun ; 12(1): 6112, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34671051

RESUMEN

Stroke profoundly disrupts cortical excitability which impedes recovery, but how it affects the function of specific inhibitory interneurons, or subpopulations therein, is poorly understood. Interneurons expressing vasoactive intestinal peptide (VIP) represent an intriguing stroke target because they can regulate cortical excitability through disinhibition. Here we chemogenetically augmented VIP interneuron excitability in a murine model of photothrombotic stroke and show that it enhances somatosensory responses and improves recovery of paw function. Using longitudinal calcium imaging, we discovered that stroke primarily disrupts the fidelity (fraction of responsive trials) and predictability of sensory responses within a subset of highly active VIP neurons. Partial recovery of responses occurred largely within these active neurons and was not accompanied by the recruitment of minimally active neurons. Importantly, chemogenetic stimulation preserved sensory response fidelity and predictability in highly active neurons. These findings provide a new depth of understanding into how stroke and prospective therapies (chemogenetics), can influence subpopulations of inhibitory interneurons.


Asunto(s)
Interneuronas/fisiología , Accidente Cerebrovascular/terapia , Péptido Intestinal Vasoactivo/metabolismo , Animales , Clozapina/análogos & derivados , Clozapina/uso terapéutico , Humanos , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Ratones , Inhibición Neural/efectos de los fármacos , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Recuperación de la Función , Corteza Somatosensorial/citología , Corteza Somatosensorial/efectos de los fármacos , Corteza Somatosensorial/fisiología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología
8.
Nat Commun ; 12(1): 5475, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34531380

RESUMEN

Acetylcholine release in the hippocampus plays a central role in the formation of new memory representations. An influential but largely untested theory proposes that memory formation requires acetylcholine to enhance responses in CA1 to new sensory information from entorhinal cortex whilst depressing inputs from previously encoded representations in CA3. Here, we show that excitatory inputs from entorhinal cortex and CA3 are depressed equally by synaptic release of acetylcholine in CA1. However, feedforward inhibition from entorhinal cortex exhibits greater depression than CA3 resulting in a selective enhancement of excitatory-inhibitory balance and CA1 activation by entorhinal inputs. Entorhinal and CA3 pathways engage different feedforward interneuron subpopulations and cholinergic modulation of presynaptic function is mediated differentially by muscarinic M3 and M4 receptors, respectively. Thus, our data support a role and mechanisms for acetylcholine to prioritise novel information inputs to CA1 during memory formation.


Asunto(s)
Acetilcolina/metabolismo , Región CA1 Hipocampal/fisiología , Corteza Entorrinal/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Retroalimentación Fisiológica/fisiología , Transmisión Sináptica/fisiología , Animales , Región CA1 Hipocampal/citología , Carbacol/farmacología , Agonistas Colinérgicos/farmacología , Corteza Entorrinal/citología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Retroalimentación Fisiológica/efectos de los fármacos , Interneuronas/metabolismo , Interneuronas/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Células Piramidales/metabolismo , Células Piramidales/fisiología , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Transmisión Sináptica/efectos de los fármacos
9.
Oxid Med Cell Longev ; 2021: 3260789, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34367461

RESUMEN

The consumption of hypercaloric diets is related to the development of obesity, favoring the etiology of gastrointestinal disorders. In this context, Spirulina platensis (SP), some blue-green algae with antioxidant action, appears as a potential therapeutic alternative to prevent obesity and associated intestinal disorders. Thus, the present study is aimed at evaluating the deleterious effects of the hypercaloric diet on the contractile and relaxing reactivity of the ileum of rats, as well as the possible preventive mechanisms of dietary supplementation with SP. Wistar rats were divided into three groups: fed a standard diet (SD), a hypercaloric diet (HCD), and/or supplemented with 25 mg/kg SP (HCD + SP25) for 8 weeks. The hypercaloric diet was effective in promoting obesity in rats, as well as decreasing potency and ileal relaxing and contractile efficacy. In contrast, dietary supplementation with SP was able to prevent some of the parameters of experimental obesity. In addition, SP prevented the reduction of intestinal reactivity, possibly due to a positive modulation of voltage-gated calcium channels (CaV) and negative regulation of muscarinic receptors (M3). Thus, food supplementation with Spirulina platensis becomes a promising alternative in the prevention of gastrointestinal diseases induced and/or aggravated by obesity.


Asunto(s)
Antioxidantes/farmacología , Canales de Calcio/metabolismo , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Obesidad/prevención & control , Receptor Muscarínico M3/metabolismo , Spirulina/fisiología , Animales , Canales de Calcio/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Ratas , Ratas Wistar , Receptor Muscarínico M3/genética , Spirulina/química
10.
Mol Neurobiol ; 58(11): 5635-5648, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34382160

RESUMEN

Numerous human clinical studies have suggested that decreased locomotor activity is a common symptom of major depressive disorder (MDD), as well as other psychiatric diseases. In MDD, the midbrain ventral tegmental area (VTA) dopamine (DA) neurons are closely related to regulate the information processing of reward, motivation, cognition, and aversion. However, the neural circuit mechanism that underlie the relationship between VTA-DA neurons and MDD-related motor impairments, especially hypolocomotion, is still largely unknown. Herein, we investigate how the VTA-DA neurons contribute to the hypolocomotion performance in chronic social defeat stress (CSDS), a mouse model of depression-relevant neurobehavioral states. The results show that CSDS could affect the spontaneous locomotor activity of mice, but not the grip strength and forced locomotor ability. Chemogenetic activation of VTA-DA neurons alleviated CSDS-induced hypolocomotion. Subsequently, quantitative whole-brain mapping revealed decreased projections from VTA-DA neurons to substantia nigra pars reticulata (SNr) after CSDS treatment. Optogenetic activation of dopaminergic projection from VTA to SNr with the stimulation of phasic firing, but not tonic firing, could significantly increase the locomotor activity of mice. Moreover, chemogenetic activation of VTA-SNr dopaminergic circuit in CSDS mice could also rescued the decline of locomotor activity. Taken together, our data suggest that the VTA-SNr dopaminergic projection mediates CSDS-induced hypolocomotion, which provides a theoretical basis and potential therapeutic target for MDD.


Asunto(s)
Dopamina/fisiología , Neuronas Dopaminérgicas/fisiología , Locomoción , Vías Nerviosas/fisiopatología , Porción Reticular de la Sustancia Negra/fisiopatología , Derrota Social , Estrés Psicológico/fisiopatología , Área Tegmental Ventral/fisiopatología , Animales , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Enfermedad Crónica , Clozapina/análogos & derivados , Clozapina/farmacología , Trastorno Depresivo Mayor/fisiopatología , Modelos Animales de Enfermedad , Genes Reporteros , Vectores Genéticos/administración & dosificación , Fuerza de la Mano , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/efectos de los fármacos , Optogenética , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Proteínas Recombinantes/metabolismo , Prueba de Desempeño de Rotación con Aceleración Constante , Estrés Psicológico/etiología , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
11.
J Pharmacol Exp Ther ; 379(1): 64-73, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34244231

RESUMEN

Muscarinic M3 (M3) receptors mediate a wide range of acetylcholine (ACh)-induced functions, including visceral smooth-muscle contraction and glandular secretion. Positive allosteric modulators (PAMs) can avoid various side effects of muscarinic agonists with their spatiotemporal receptor activation control and potentially better subtype selectivity. However, the mechanism of allosteric modulation of M3 receptors is not fully understood, presumably because of the lack of a potent and selective PAM. In this study, we investigated the pharmacological profile of ASP8302, a novel PAM of M3 receptors, and explored the principal site of amino-acid sequences in the human M3 receptor required for the potentiation of receptor activation. In cells expressing human M3 and M5 receptors, ASP8302 shifted the concentration-response curve (CRC) for carbachol to the lower concentrations with no significant effects on other subtypes. In a binding study with M3 receptor-expressing membrane, ASP8302 also shifted the CRC for ACh without affecting the binding of orthosteric agonists. Similar shifts in the CRC of contractions by multiple stimulants were also confirmed in isolated human bladder strips. Mutagenesis analysis indicated no interaction between ASP8302 and previously reported allosteric sites; however, it identified threonine 230 as the amino acid essential for the PAM effect of ASP8302. These results demonstrate that ASP8302 enhances the activation of human M3 receptors by interacting with a single amino acid distinct from the reported allosteric sites. Our findings suggest not only a novel allosteric site of M3 receptors but also the potential application of ASP8302 to diseases caused by insufficient M3 receptor activation. SIGNIFICANCE STATEMENT: The significance of this study is that the novel M3 receptor positive allosteric modulator ASP8302 enhances the activation of human M3 receptor by interacting with a residue distinct from the reported allosteric sites. The finding of Thr230 as a novel amino acid involved in the allosteric modulation of M3 receptors provides significant insight into further research of the mechanism of allosteric modulation of M3 and other muscarinic receptors.


Asunto(s)
Sitio Alostérico/efectos de los fármacos , Agonistas Muscarínicos/química , Agonistas Muscarínicos/metabolismo , Receptor Muscarínico M3/agonistas , Receptor Muscarínico M3/metabolismo , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Sitio Alostérico/fisiología , Secuencia de Aminoácidos , Animales , Células CHO , Cricetulus , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Agonistas Muscarínicos/farmacología , Técnicas de Cultivo de Órganos , Receptor Muscarínico M3/genética , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/metabolismo
12.
Life Sci Alliance ; 4(9)2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34244422

RESUMEN

Acetylcholine (ACh) signaling through activation of nicotinic and muscarinic ACh receptors regulates expression of specific genes that mediate and sustain proliferation, differentiation, and homeostasis in the intestinal crypts. This signaling plays a pivotal role in the regulation of intestinal stem cell function, but the details have not been clarified. Here, we performed experiments using type 3 muscarinic acetylcholine receptor (M3) knockout mice and their intestinal organoids and report that endogenous ACh affects the size of the intestinal stem niche via M3 signaling. RNA sequencing of crypts identified up-regulation of the EphB/ephrin-B signaling pathway. Furthermore, using an MEK inhibitor (U0126), we found that mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling, which is downstream of EphB/ephrin-B signaling, is activated in M3-deficient crypts. Collectively, M3, EphB/ephrin-B, and the MAPK/ERK signaling cascade work together to maintain the homeostasis of intestinal epithelial cell growth and differentiation following modifications of the cholinergic intestinal niche.


Asunto(s)
Autorrenovación de las Células/genética , Intestinos/citología , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Receptores de la Familia Eph/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Animales , Biomarcadores , Diferenciación Celular/genética , Proliferación Celular , Femenino , Técnica del Anticuerpo Fluorescente , Expresión Génica , Inmunohistoquímica , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Organoides
13.
JCI Insight ; 6(17)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34314386

RESUMEN

Hyperstimulation of the cholecystokinin 1 receptor (CCK1R), a G protein-coupled receptor (GPCR), in pancreatic acinar cells is commonly used to induce pancreatitis in rodents. Human pancreatic acinar cells lack CCK1R but express cholinergic receptor muscarinic 3 (M3R), another GPCR. To test whether M3R activation is involved in pancreatitis, a mutant M3R was conditionally expressed in pancreatic acinar cells in mice. This mutant receptor loses responsiveness to its native ligand, acetylcholine, but can be activated by an inert small molecule, clozapine-N-oxide (CNO). Intracellular calcium and amylase were elicited by CNO in pancreatic acinar cells isolated from mutant M3R mice but not WT mice. Similarly, acute pancreatitis (AP) could be induced by a single injection of CNO in the transgenic mice but not WT mice. Compared with the cerulein-induced AP, CNO caused more widespread acinar cell death and inflammation. Furthermore, chronic pancreatitis developed at 4 weeks after 3 episodes of CNO-induced AP. In contrast, in mice with 3 recurrent episodes of cerulein-included AP, pancreas histology was restored in 4 weeks. Furthermore, the M3R antagonist ameliorated the severity of cerulein-induced AP in WT mice. We conclude that M3R activation can cause the pathogenesis of pancreatitis. This model may provide an alternative approach for pancreatitis research.


Asunto(s)
Células Acinares/metabolismo , Regulación de la Expresión Génica , Pancreatitis/genética , ARN/genética , Receptor Muscarínico M3/genética , Células Acinares/patología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Ratones , Ratones Mutantes , Ratones Transgénicos , Pancreatitis/metabolismo , Pancreatitis/patología , Receptor Muscarínico M3/biosíntesis , Transducción de Señal
14.
Int J Mol Sci ; 22(2)2021 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-33477687

RESUMEN

Parasympathetic signalling via muscarinic acetylcholine receptors (mAChRs) regulates gastrointestinal smooth muscle function. In most instances, the mAChR population in smooth muscle consists mainly of M2 and M3 subtypes in a roughly 80% to 20% mixture. Stimulation of these mAChRs triggers a complex array of biochemical and electrical events in the cell via associated G proteins, leading to smooth muscle contraction and facilitating gastrointestinal motility. Major signalling events induced by mAChRs include adenylyl cyclase inhibition, phosphoinositide hydrolysis, intracellular Ca2+ mobilisation, myofilament Ca2+ sensitisation, generation of non-selective cationic and chloride currents, K+ current modulation, inhibition or potentiation of voltage-dependent Ca2+ currents and membrane depolarisation. A lack of ligands with a high degree of receptor subtype selectivity and the frequent contribution of multiple receptor subtypes to responses in the same cell type have hampered studies on the signal transduction mechanisms and functions of individual mAChR subtypes. Therefore, novel strategies such as genetic manipulation are required to elucidate both the contributions of specific AChR subtypes to smooth muscle function and the underlying molecular mechanisms. In this article, we review recent studies on muscarinic function in gastrointestinal smooth muscle using mAChR subtype-knockout mice.


Asunto(s)
Tracto Gastrointestinal/metabolismo , Músculo Liso/metabolismo , Receptor Muscarínico M2/genética , Receptor Muscarínico M3/genética , Animales , Proteínas de Unión al GTP/genética , Tracto Gastrointestinal/crecimiento & desarrollo , Tracto Gastrointestinal/patología , Ratones Noqueados/genética , Contracción Muscular/genética , Músculo Liso/crecimiento & desarrollo , Transducción de Señal/genética
15.
Int J Mol Sci ; 22(2)2021 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-33450835

RESUMEN

Despite great advances in our understanding of the pathobiology of colorectal cancer and the genetic and environmental factors that mitigate its onset and progression, a paucity of effective treatments persists. The five-year survival for advanced, stage IV disease remains substantially less than 20%. This review examines a relatively untapped reservoir of potential therapies to target muscarinic receptor expression, activation, and signaling in colorectal cancer. Most colorectal cancers overexpress M3 muscarinic receptors (M3R), and both in vitro and in vivo studies have shown that activating these receptors stimulates cellular programs that result in colon cancer growth, survival, and spread. In vivo studies using mouse models of intestinal neoplasia have shown that using either genetic or pharmacological approaches to block M3R expression and activation, respectively, attenuates the development and progression of colon cancer. Moreover, both in vitro and in vivo studies have shown that blocking the activity of matrix metalloproteinases (MMPs) that are induced selectively by M3R activation, i.e., MMP1 and MMP7, also impedes colon cancer growth and progression. Nonetheless, the widespread expression of muscarinic receptors and MMPs and their importance for many cellular functions raises important concerns about off-target effects and the safety of employing similar strategies in humans. As we highlight in this review, highly selective approaches can overcome these obstacles and permit clinicians to exploit the reliance of colon cancer cells on muscarinic receptors and their downstream signal transduction pathways for therapeutic purposes.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/metabolismo , Receptores Muscarínicos/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Biomarcadores , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/patología , Manejo de la Enfermedad , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Metaloproteinasas de la Matriz/metabolismo , Terapia Molecular Dirigida , Agonistas Muscarínicos/farmacología , Agonistas Muscarínicos/uso terapéutico , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Receptores Muscarínicos/clasificación , Receptores Muscarínicos/genética
16.
Brain Behav Immun ; 91: 89-104, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32927021

RESUMEN

Microglia are the immune cells of the brain and become activated during any type of brain injury. In the middle cerebral artery occlusion (MCAo) model, a mouse model for ischemic stroke, we have previously shown that microglia and invaded monocytes upregulate the expression of the muscarinic acetylcholine receptor 3 (M3R) in the ischemic lesion. Here we tested whether this upregulation has an impact on the pathogenesis of MCAo. We depleted the m3R receptor in microglia, but not in circulating monocytes by giving tamoxifen to CX3CR1-CreERT+/+M3Rflox/flox (M3RKOmi) animals 3 weeks prior to MCAo. We found that M3RKOmi male mice had bigger lesions, more pronounced motor deficits after one week and cognitive deficits after about one month compared to control males. The density of Iba1+ cells was lower in the lesions of M3RKO male mice in the early, but not in the late disease phase. In females, these differences were not significant. By giving tamoxifen 1 week prior to MCAo, we depleted m3R in microglia and in circulating monocytes (M3RKOmi/mo). Male M3RKOmi/mo did not differ in lesion size, but had a lower survival rate, showed motor deficits and a reduced accumulation of Iba1+ positive cells into the lesion site. In conclusion, our data suggest that the upregulation of m3R in microglia and monocytes in stroke has a beneficial effect on the clinical outcome in male mice.


Asunto(s)
Isquemia Encefálica , Microglía , Receptor Muscarínico M3/genética , Accidente Cerebrovascular , Animales , Encéfalo , Modelos Animales de Enfermedad , Femenino , Infarto de la Arteria Cerebral Media , Masculino , Ratones , Ratones Endogámicos C57BL
17.
Nature ; 586(7829): 417-423, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32999463

RESUMEN

Microglia, the brain's resident macrophages, help to regulate brain function by removing dying neurons, pruning non-functional synapses, and producing ligands that support neuronal survival1. Here we show that microglia are also critical modulators of neuronal activity and associated behavioural responses in mice. Microglia respond to neuronal activation by suppressing neuronal activity, and ablation of microglia amplifies and synchronizes the activity of neurons, leading to seizures. Suppression of neuronal activation by microglia occurs in a highly region-specific fashion and depends on the ability of microglia to sense and catabolize extracellular ATP, which is released upon neuronal activation by neurons and astrocytes. ATP triggers the recruitment of microglial protrusions and is converted by the microglial ATP/ADP hydrolysing ectoenzyme CD39 into AMP; AMP is then converted into adenosine by CD73, which is expressed on microglia as well as other brain cells. Microglial sensing of ATP, the ensuing microglia-dependent production of adenosine, and the adenosine-mediated suppression of neuronal responses via the adenosine receptor A1R are essential for the regulation of neuronal activity and animal behaviour. Our findings suggest that this microglia-driven negative feedback mechanism operates similarly to inhibitory neurons and is essential for protecting the brain from excessive activation in health and disease.


Asunto(s)
Retroalimentación Fisiológica , Microglía/fisiología , Inhibición Neural , Neuronas/fisiología , 5'-Nucleotidasa/metabolismo , Potenciales de Acción , Adenosina/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antígenos CD/metabolismo , Apirasa/metabolismo , Calcio/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Inhibición Neural/genética , Receptor de Adenosina A1/metabolismo , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Factores de Tiempo
18.
Pancreas ; 49(10): 1335-1341, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33122522

RESUMEN

OBJECTIVES: Pancreatic acinar necrosis is a typical feature in the early phase of severe acute pancreatitis (SAP). Muscarinic acetylcholine receptor M3 (CHRM3) has been reported to play important roles in promoting insulin secretion and tumor cell proliferation, but its effect on necrosis remains unknown. This study revealed the important role of CHRM3 in regulating L-arginine-induced SAP and the molecular mechanisms. METHODS: To verify the function of CHRM3, pancreatic tissues and primary acinar cells of CRISPR/Cas9-mediated Chrm3 knockout mice were used in CHRM3 knockdown experiments, and to ascertain the CHRM3 overexpression, PLV-EGFP-Chrm3 plasmids were transfected in acinar cells in vitro. RESULTS: In L-arginine-induced SAP, CHRM3 is activated and regulates SAP through the mitogen-activated protein kinase/p38 pathway. Moreover, the expression of miR-31-5p decreased in the SAP model both in vitro and in vivo. Mir-31-5p effects the necrosis of acinar cells in SAP by upregulating the target gene RIP3, and miR-31-5p is a downstream miRNA of CHRM3. CONCLUSIONS: Necrosis in L-arginine-induced SAP is promoted by CHRM3 through the mitogen-activated protein kinase-p38/miR-31-5p/RIP3 axis.


Asunto(s)
Células Acinares/enzimología , MicroARNs/metabolismo , Páncreas/enzimología , Pancreatitis/prevención & control , Receptor Muscarínico M3/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células Acinares/patología , Animales , Arginina , Células Cultivadas , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Necrosis , Páncreas/patología , Pancreatitis/inducido químicamente , Pancreatitis/enzimología , Pancreatitis/patología , Fosforilación , Receptor Muscarínico M3/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal
19.
Nat Methods ; 17(11): 1139-1146, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32989318

RESUMEN

The ability to directly measure acetylcholine (ACh) release is an essential step toward understanding its physiological function. Here we optimized the GRABACh (GPCR-activation-based ACh) sensor to achieve substantially improved sensitivity in ACh detection, as well as reduced downstream coupling to intracellular pathways. The improved version of the ACh sensor retains the subsecond response kinetics, physiologically relevant affinity and precise molecular specificity for ACh of its predecessor. Using this sensor, we revealed compartmental ACh signals in the olfactory center of transgenic flies in response to external stimuli including odor and body shock. Using fiber photometry recording and two-photon imaging, our ACh sensor also enabled sensitive detection of single-trial ACh dynamics in multiple brain regions in mice performing a variety of behaviors.


Asunto(s)
Acetilcolina/metabolismo , Técnicas Biosensibles/métodos , Encéfalo/metabolismo , Animales , Animales Modificados Genéticamente , Conducta Animal/fisiología , Colinérgicos/farmacología , Drosophila/genética , Drosophila/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Ratones , Cuerpos Pedunculados/metabolismo , Neuronas/metabolismo , Corteza Olfatoria/metabolismo , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Corteza Somatosensorial/metabolismo
20.
Arterioscler Thromb Vasc Biol ; 40(11): 2649-2664, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32938216

RESUMEN

OBJECTIVE: Phenotypic switching of vascular smooth muscle cells (VSMCs) plays a critical role in atherosclerosis, vascular restenosis, and hypertension. Choline exerts cardioprotective effects; however, little is known about its effects on VSMC phenotypic switching and vascular remodeling. Here, we investigated whether choline modulates VSMC phenotypic changes and explored the underlying mechanisms. Approach and Results: In cultured VSMCs, choline promoted Nrf2 (nuclear factor erythroid 2-related factor 2) nuclear translocation, inducing the expression of HO-1 (heme oxygenase-1) and NQO-1 (NAD[P]H quinone oxidoreductase-1). Consequently, choline ameliorated Ang II (angiotensin II)-induced increases in NOX (NAD[P]H oxidase) expression and the mitochondrial reactive oxygen species level, thereby attenuating Ang II-induced VSMC phenotypic switching, proliferation, and migration, presumably via M3AChRs (type 3 muscarinic acetylcholine receptors). Downregulation of M3AChR or Nrf2 diminished choline-mediated upregulation of Nrf2, HO-1, and NQO-1 expression, as well as inhibition of VSMC phenotypic transformation, suggesting that M3AChR and Nrf2 activation are responsible for the protective effects of choline. Moreover, activation of the Nrf2 pathway by sulforaphane suppressed Ang II-induced VSMC phenotypic switching and proliferation, indicating that Nrf2 is a key regulator of VSMC phenotypic switching and vascular homeostasis. In a rat model of abdominal aortic constriction in vivo, choline attenuated VSMC phenotypic transformation and vascular remodeling in a manner related to activation of the Nrf2 pathway. CONCLUSIONS: These results reveal that choline impedes VSMC phenotypic switching, proliferation, migration, and vascular remodeling by activating M3AChR and Nrf2-antioxidant signaling and suggest a novel role for Nrf2 in VSMC phenotypic modulation.


Asunto(s)
Plasticidad de la Célula/efectos de los fármacos , Colina/farmacología , Agonistas Muscarínicos/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Receptor Muscarínico M3/agonistas , Remodelación Vascular/efectos de los fármacos , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Factor 2 Relacionado con NF-E2/genética , Fenotipo , Ratas Sprague-Dawley , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA