Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 190
Filter
1.
J Neurosci ; 44(17)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38438258

ABSTRACT

Acetylcholine (ACh) is released from basal forebrain cholinergic neurons in response to salient stimuli and engages brain states supporting attention and memory. These high ACh states are associated with theta oscillations, which synchronize neuronal ensembles. Theta oscillations in the basolateral amygdala (BLA) in both humans and rodents have been shown to underlie emotional memory, yet their mechanism remains unclear. Here, using brain slice electrophysiology in male and female mice, we show large ACh stimuli evoke prolonged theta oscillations in BLA local field potentials that depend upon M3 muscarinic receptor activation of cholecystokinin (CCK) interneurons (INs) without the need for external glutamate signaling. Somatostatin (SOM) INs inhibit CCK INs and are themselves inhibited by ACh, providing a functional SOM→CCK IN circuit connection gating BLA theta. Parvalbumin (PV) INs, which can drive BLA oscillations in baseline states, are not involved in the generation of ACh-induced theta, highlighting that ACh induces a cellular switch in the control of BLA oscillatory activity and establishes an internally BLA-driven theta oscillation through CCK INs. Theta activity is more readily evoked in BLA over the cortex or hippocampus, suggesting preferential activation of the BLA during high ACh states. These data reveal a SOM→CCK IN circuit in the BLA that gates internal theta oscillations and suggest a mechanism by which salient stimuli acting through ACh switch the BLA into a network state enabling emotional memory.


Subject(s)
Acetylcholine , Cholecystokinin , Mice, Inbred C57BL , Theta Rhythm , Theta Rhythm/drug effects , Theta Rhythm/physiology , Animals , Male , Mice , Female , Acetylcholine/pharmacology , Acetylcholine/metabolism , Cholecystokinin/pharmacology , Cholecystokinin/metabolism , Interneurons/physiology , Interneurons/drug effects , Somatostatin/metabolism , Somatostatin/pharmacology , Amygdala/physiology , Amygdala/drug effects , Basolateral Nuclear Complex/physiology , Basolateral Nuclear Complex/drug effects , Nerve Net/physiology , Nerve Net/drug effects , Receptor, Muscarinic M3/physiology , Receptor, Muscarinic M3/metabolism , Parvalbumins/metabolism
2.
Low Urin Tract Symptoms ; 14(4): 289-300, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35150075

ABSTRACT

OBJECTIVES: Muscarinic M3 (M3 ) receptors mediate cholinergic smooth muscle contraction of the bladder. Current drugs targeting bladder M3 receptors for micturition disorders have a risk of cholinergic side effects due to excessive receptor activation and insufficient selectivity. We investigated the effect of ASP8302, a novel positive allosteric modulator (PAM) of M3 receptors, on bladder function in rats. METHODS: Modulation of carbachol-induced increases in intracellular Ca2+ was assessed in cells expressing rat muscarinic receptors. Potentiation of bladder contractions was evaluated using isolated rat bladder strips and by measuring intravesical pressure in anesthetized rats. Conscious cystometry was performed to investigate the effects on residual urine volume and voiding efficiency in rat voiding dysfunction models induced by the α1 -adrenoceptor agonist midodrine and muscarinic receptor antagonist atropine, and bladder outlet obstruction. To assess potential side effects, the number of stools and tracheal insufflation pressure were measured in conscious and anesthetized rats, respectively. RESULTS: ASP8302 demonstrated PAM effects on the rat M3 receptor in cell assays, and augmented cholinergic bladder contractions both in vivo and in vitro. ASP8302 improved voiding efficiency and reduced residual urine volume in two voiding dysfunction models as effectively as distigmine bromide, but unlike distigmine bromide did not affect the number of stools or tracheal insufflation pressure. CONCLUSIONS: Our results in rats indicate that ASP8302 improves voiding dysfunction by potentiating bladder contraction with fewer effects on cholinergic responses in other organs, and suggest a potential advantage over current cholinomimetic drugs for treating micturition disorders caused by insufficient bladder contraction.


Subject(s)
Muscarinic Agonists , Muscarinic Antagonists , Receptor, Muscarinic M3 , Urinary Bladder , Animals , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Muscle Contraction , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M3/physiology
3.
Int J Mol Sci ; 22(11)2021 Jun 07.
Article in English | MEDLINE | ID: mdl-34200187

ABSTRACT

The parasympathetic nervous system is critically involved in the regulation of tear secretion by activating muscarinic acetylcholine receptors. Hence, various animal models targeting parasympathetic signaling have been developed to induce dry eye disease (DED). However, the muscarinic receptor subtype (M1-M5) mediating tear secretion remains to be determined. This study was conducted to test the hypothesis that the M3 receptor subtype regulates tear secretion and to evaluate the ocular surface phenotype of mice with targeted disruption of the M3 receptor (M3R-/-). The experimental techniques included quantification of tear production, fluorescein staining of the ocular surface, environmental scanning electron microscopy, assessment of proliferating cells in the corneal epithelium and of goblet cells in the conjunctiva, quantification of mRNA for inflammatory cytokines and prooxidant redox enzymes and quantification of reactive oxygen species. Tear volume was reduced in M3R-/- mice compared to age-matched controls at the age of 3 months and 15 months, respectively. This was associated with mild corneal epitheliopathy in the 15-month-old but not in the 3-month-old M3R-/- mice. M3R-/- mice at the age of 15 months also displayed changes in corneal epithelial cell texture, reduced conjunctival goblet cell density, oxidative stress and elevated mRNA expression levels for inflammatory cytokines and prooxidant redox enzymes. The findings suggest that the M3 receptor plays a pivotal role in tear production and its absence leads to ocular surface changes typical for DED at advanced age.


Subject(s)
Conjunctiva/pathology , Dry Eye Syndromes/pathology , Epithelium, Corneal/pathology , Goblet Cells/pathology , Receptor, Muscarinic M3/physiology , Animals , Conjunctiva/metabolism , Disease Models, Animal , Dry Eye Syndromes/etiology , Dry Eye Syndromes/metabolism , Epithelium, Corneal/metabolism , Goblet Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reactive Oxygen Species/metabolism , Tears/metabolism
4.
J Ethnopharmacol ; 274: 114048, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-33781875

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Plectranthus vettiveroides (Jacob) N.P. Singh & B.D. Sharma is a traditional medicinal plant used in Siddha System of Medicine and its aromatic root is used to reduce the elevated blood pressure. AIM: The aim of the present study was to study vasorelaxant property of the root essential oil nanoemulsion (EON) of P. vettiveroides. METHODS: The EON was formulated to enhance the solubility and bioavailability and characterized. The preliminary screening was performed by treating the EON with aortic rings pre-contracted with phenylephrine (1 µM) and potassium chloride (80 mM). The role of K⁺ channels in EON induced vasorelaxation was investigated by pre-incubating the aortic rings with different K⁺ channel inhibitors namely, glibenclamide (a non-specific ATP sensitive K⁺ channel blocker, 10 µM), TEA (a Ca2⁺ activated non-selective K⁺ channel blocker, 10-2 M), 4-AP (a voltage-activated K⁺ channel blocker, 10-3 M) and barium chloride (inward rectifier K⁺ channel blocker, 1 mM). The involvement of extracellular Ca2+ was performed by adding cumulative dose of extracellular calcium in the presence and absence of EON and the concentration-response curve (CRC) obtained is compared. Similarly, the role of nitric oxide synthase, muscarinic and prostacyclin receptors on EON induced vasorelaxation were evaluated by pre-incubating the aortic rings with their inhibitors and the CRC obtained in the presence and absence of inhibitor were compared. RESULTS: The GC-MS and GC-FID analyses of the root essential oil revealed the presence of 62 volatile compounds. The EON exhibited significant vasorelaxant effect through nitric oxide-mediated pathway, G-protein coupled muscarinic (M3) receptor pathway, involvement of K+ channels (KATP, KIR, KCa), and blocking of the calcium influx by receptor-operated calcium channel. CONCLUSION: It is concluded that the root essential oil of P. vettiveroides is possessing marked vasorelaxant property. The multiple mechanisms of action of the essential oil of P. vettiveroides make it a potential source of antihypertensive drug.


Subject(s)
Antihypertensive Agents/pharmacology , Aorta, Thoracic/drug effects , Oils, Volatile/pharmacology , Plectranthus , Vasodilator Agents/pharmacology , Animals , Antihypertensive Agents/chemistry , Aorta, Thoracic/physiology , Calcium/physiology , Calcium Channels/physiology , Emulsions , Inositol 1,4,5-Trisphosphate Receptors/physiology , KATP Channels/physiology , Male , Nitric Oxide/physiology , Oils, Volatile/chemistry , Phytochemicals/analysis , Phytochemicals/pharmacology , Plant Roots , Potassium Channels, Inwardly Rectifying/physiology , Rats, Wistar , Receptor, Muscarinic M3/physiology , Vasodilation/drug effects , Vasodilator Agents/chemistry
5.
Sci Rep ; 10(1): 14181, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32843670

ABSTRACT

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.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Hyperalgesia/prevention & control , Inflammation/physiopathology , Neuroglia/enzymology , Nociception/physiology , Receptor, Adenosine A1/physiology , Receptor, Muscarinic M3/physiology , Animals , Benzilates/pharmacology , Clozapine/analogs & derivatives , Clozapine/pharmacology , Freund's Adjuvant/toxicity , Genes, Synthetic , Hot Temperature , Hyperalgesia/physiopathology , Inflammation/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscarinic Agonists/pharmacology , Neuroglia/physiology , Nortropanes/pharmacology , Promoter Regions, Genetic , Purinergic P1 Receptor Agonists/pharmacology , Purinergic P1 Receptor Antagonists/pharmacology , Receptor, Adenosine A1/drug effects , Receptor, Muscarinic M3/drug effects , Receptor, Muscarinic M3/genetics , Receptors, G-Protein-Coupled , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Theophylline/analogs & derivatives , Theophylline/pharmacology , Touch , Xanthines/pharmacology
6.
J Clin Invest ; 130(1): 83-93, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31738186

ABSTRACT

The mineralocorticoid aldosterone is produced in the adrenal zona glomerulosa (ZG) under the control of the renin-angiotensin II (AngII) system. Primary aldosteronism (PA) results from renin-independent production of aldosterone and is a common cause of hypertension. PA is caused by dysregulated localization of the enzyme aldosterone synthase (Cyp11b2), which is normally restricted to the ZG. Cyp11b2 transcription and aldosterone production are predominantly regulated by AngII activation of the Gq signaling pathway. Here, we report the generation of transgenic mice with Gq-coupled designer receptors exclusively activated by designer drugs (DREADDs) specifically in the adrenal cortex. We show that adrenal-wide ligand activation of Gq DREADD receptors triggered disorganization of adrenal functional zonation, with induction of Cyp11b2 in glucocorticoid-producing zona fasciculata cells. This result was consistent with increased renin-independent aldosterone production and hypertension. All parameters were reversible following termination of DREADD-mediated Gq signaling. These findings demonstrate that Gq signaling is sufficient for adrenocortical aldosterone production and implicate this pathway in the determination of zone-specific steroid production within the adrenal cortex. This transgenic mouse also provides an inducible and reversible model of hyperaldosteronism to investigate PA therapeutics and the mechanisms leading to the damaging effects of aldosterone on the cardiovascular system.


Subject(s)
Adrenal Cortex/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Hyperaldosteronism/etiology , Zona Glomerulosa/physiology , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Cytochrome P-450 CYP11B2/physiology , Designer Drugs/pharmacology , Female , Hyperaldosteronism/drug therapy , Hypertension/etiology , Mice , Mice, Transgenic , Receptor, Muscarinic M3/physiology , Signal Transduction/physiology
7.
J Cell Biochem ; 121(3): 2618-2631, 2020 03.
Article in English | MEDLINE | ID: mdl-31692054

ABSTRACT

Acinar cells in acute pancreatitis (AP) die through apoptosis and necrosis, the impacts of which are quite different. Early clinical interference strategies on preventing the progress of AP to severe acute pancreatitis (SAP) are the elimination of inflammation response and inhibition of necrosis. Muscarinic acetylcholine receptor M3 was encoded by Chrm3 gene. It is one of the best-characterized receptors of pancreatic ß cells and regulates insulin secretion, but its function in AP remains unclear. In this study, we explored the function of Chrm3 gene in the regulation of cell death in l-arginine-induced SAP animal models. We found that Chrm3 was upregulated in pancreatitis, and we further confirmed the localization of Chrm3 resided in both pancreatic islets and acinar cell membranes. The reduction of Chrm3 decreased the pathological lesion of SAP and reduced amylase activities in serum. Consistently, Chrm3 can suppress acinar cells necrosis markedly, but has no effect on regulating apoptosis after l-arginine treatment. It was shown that Chrm3 attenuated acinar cells necrosis at least in part by stabilizing caspase-8. Thus, this study indicates that Chrm3 is critical participants in SAP, and regulation of Chrm3 expression might be a useful therapeutic strategy for preventing pathologic necrosis.


Subject(s)
Acinar Cells/pathology , Caspase 8/metabolism , Necrosis , Pancreatitis/prevention & control , Protective Agents/pharmacology , Receptor, Muscarinic M3/physiology , Transcriptome , Acinar Cells/metabolism , Animals , Arginine/toxicity , Caspase 8/chemistry , Caspase 8/genetics , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis/chemically induced , Pancreatitis/pathology
8.
Sci Rep ; 9(1): 14051, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31575899

ABSTRACT

Neuromodulation ensures that neural circuits produce output that is flexible whilst remaining within an optimal operational range. The neuromodulator acetylcholine is released during locomotion to regulate spinal motor circuits. However, the range of receptors and downstream mechanisms by which acetylcholine acts have yet to be fully elucidated. We therefore investigated metabotropic acetylcholine receptor-mediated modulation by using isolated spinal cord preparations from neonatal mice in which locomotor-related output can be induced pharmacologically. We report that M2 receptor blockade decreases the frequency and amplitude of locomotor-related activity, whilst reducing its variability. In contrast, M3 receptor blockade destabilizes locomotor-related bursting. Motoneuron recordings from spinal cord slices revealed that activation of M2 receptors induces an outward current, decreases rheobase, reduces the medium afterhyperpolarization, shortens spike duration and decreases synaptic inputs. In contrast, M3 receptor activation elicits an inward current, increases rheobase, extends action potential duration and increases synaptic inputs. Analysis of miniature postsynaptic currents support that M2 and M3 receptors modulate synaptic transmission via different mechanisms. In summary, we demonstrate that M2 and M3 receptors have opposing modulatory actions on locomotor circuit output, likely reflecting contrasting cellular mechanisms of action. Thus, intraspinal cholinergic systems mediate balanced, multimodal control of spinal motor output.


Subject(s)
Acetylcholine/metabolism , Locomotion/physiology , Motor Neurons/metabolism , Receptor, Muscarinic M2/metabolism , Receptor, Muscarinic M3/metabolism , Spinal Cord/metabolism , Acetylcholine/physiology , Action Potentials/drug effects , Animals , Animals, Newborn , Diamines/pharmacology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Motor Neurons/physiology , Muscarine/pharmacology , Piperidines/pharmacology , Receptor, Muscarinic M2/antagonists & inhibitors , Receptor, Muscarinic M2/physiology , Receptor, Muscarinic M3/antagonists & inhibitors , Receptor, Muscarinic M3/physiology , Spinal Cord/physiology
9.
Biochem Pharmacol ; 169: 113613, 2019 11.
Article in English | MEDLINE | ID: mdl-31445019

ABSTRACT

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


Subject(s)
AMP-Activated Protein Kinases/physiology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/physiology , Hepatocytes/metabolism , Lipid Metabolism , Receptor, Muscarinic M3/physiology , Animals , Cells, Cultured , Humans , Mice , PPAR alpha/physiology , Phosphorylation , Receptor, Muscarinic M1/physiology , Signal Transduction/physiology
10.
Cell Signal ; 63: 109358, 2019 11.
Article in English | MEDLINE | ID: mdl-31295519

ABSTRACT

Calcium influx via store-operated calcium entry (SOCE) has an important role for regulation of vast majority of cellular physiological events. MAPK signalling is also another pivotal modulator of many cellular functions. However, the relationship between SOCE and MAPK is not well understood. In this study, we elucidated the involvement of SOCE in Gαq/11 protein-mediated activation of p38 MAPK in an intestinal epithelial cell line HT-29/B6. In this cell line, we previously showed that the stimulation of M3 muscarinic acetylcholine receptor (M3-mAChR) but not histamine H1 receptor (H1R) led to phosphorylation of p38 MAPK which suppressed tumor necrosis factor-α (TNF-α)-induced NF-κB signalling through ADAM17 protease-mediated shedding of TNF receptor-1 (TNFR1). First, we found that stimulation of M3-mAChR and protease-activated receptor-2 (PAR-2) but not H1R induced persistent upregulation of cytosolic Ca2+ concentration through SOCE. Activation of M3-mAChR or PAR-2 also suppressed TNF-α-induced NF-κB phosphorylation, which was dependent on the p38 MAPK activity. Time course experiments revealed that M3-mAChR stimulation evoked intracellular Ca2+-dependent early phase p38 MAPK phosphorylation and extracellular Ca2+-dependent later phase p38 MAPK phosphorylation. This later phase p38 MAPK phosphorylation, evoked by M3-mAChRs or PAR-2, was abolished by inhibition of SOCE. Thapsigargin or ionomycin also phosphorylate p38 MAPK by Ca2+ influx through SOCE, leading to suppression of TNF-α-induced NF-κB phosphorylation. Finally, we showed that p38 MAPK was essential for thapsigargin-induced cleavage of TNFR1 and suppression of TNF-α-induced NF-κB phosphorylation. In conclusion, SOCE is important for p38 MAPK phosphorylation and is involved in TNF-α signalling suppression.


Subject(s)
Calcium/physiology , Receptor, Muscarinic M3/physiology , Receptor, PAR-2/physiology , Receptors, Histamine H1/physiology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , HT29 Cells , Humans , NF-kappa B/metabolism
11.
J Nippon Med Sch ; 86(2): 117-121, 2019.
Article in English | MEDLINE | ID: mdl-31130562

ABSTRACT

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


Subject(s)
Muscarinic Antagonists/administration & dosage , Pirenzepine/administration & dosage , Receptor, Muscarinic M1/antagonists & inhibitors , Receptor, Muscarinic M3/antagonists & inhibitors , Sialorrhea/drug therapy , Solifenacin Succinate/administration & dosage , Animals , Drug Therapy, Combination , Humans , Male , Mice, Knockout , Middle Aged , Radionuclide Imaging , Receptor, Muscarinic M1/physiology , Receptor, Muscarinic M3/physiology , Salivary Glands/diagnostic imaging , Salivary Glands/physiopathology , Salivation , Sialorrhea/diagnostic imaging , Sialorrhea/physiopathology , Treatment Outcome
12.
Naunyn Schmiedebergs Arch Pharmacol ; 392(9): 1071-1083, 2019 09.
Article in English | MEDLINE | ID: mdl-31049606

ABSTRACT

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


Subject(s)
Antihypertensive Agents/pharmacology , Hypotension/physiopathology , Piperazines/pharmacology , Pyrazoles/pharmacology , Receptor, Muscarinic M1/physiology , Receptor, Muscarinic M3/physiology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Atropine/pharmacology , Blood Pressure/drug effects , Heart Rate/drug effects , Hypertension/chemically induced , Male , Muscarinic Antagonists/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Piperidines/pharmacology , Pirenzepine/pharmacology , Rats, Inbred SHR , Rats, Wistar
13.
FASEB J ; 33(6): 7252-7260, 2019 06.
Article in English | MEDLINE | ID: mdl-30860868

ABSTRACT

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.


Subject(s)
Adrenergic Neurons/physiology , Anesthesia, General , Arousal/physiology , Delayed Emergence from Anesthesia/physiopathology , Intralaminar Thalamic Nuclei/physiopathology , Nerve Endings/physiology , Receptors, Adrenergic, alpha-1/physiology , Adrenergic Neurons/drug effects , Anesthetics, Intravenous , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Designer Drugs/pharmacology , Electroencephalography , Genetic Vectors/administration & dosage , Intralaminar Thalamic Nuclei/drug effects , Mice , Mice, Inbred C57BL , Nerve Endings/drug effects , Norepinephrine/pharmacology , Patch-Clamp Techniques , Propofol , Random Allocation , Receptor, Muscarinic M3/drug effects , Receptor, Muscarinic M3/physiology , Receptors, Adrenergic, alpha-1/drug effects , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Reflex, Righting/drug effects , Single-Blind Method , Specific Pathogen-Free Organisms , Synaptic Potentials/drug effects , Synaptic Potentials/physiology
14.
Eur J Pharmacol ; 843: 240-250, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30502343

ABSTRACT

Several antimuscarinics have been commonly used for overactive bladder patients, but dry mouth as a major anticholinergic side effect remains a shortcoming to limit long-term use. The aim of this study was to elucidate the pharmacological properties of DA-8010, a novel muscarinic receptor antagonist selective for urinary bladder over salivary gland. DA-8010 exhibited a high binding affinity for human muscarinic M3 receptor with pKi of 8.81 ±â€¯0.05 and great potencies for human M3 receptor and rat bladder preparation. The potency of DA-8010 for bladder smooth muscle cells was 3.6-fold higher than that for salivary gland cells isolated from mice. Intravenous administration of DA-8010 dose-dependently inhibited rhythmic urinary bladder contractions induced by distension in rats, indicating the most potent activity (ID30 = 0.08 mg/kg) among the antimuscarinics tested. Taken together with the inhibitory effects of DA-8010 and other antimuscarinics on carbachol-induced salivary secretion in rats, the in vivo functional selectivity of DA-8010 for urinary bladder over salivary gland was 3.1-fold, 3.2-fold and 5.2-fold greater than those observed for solifenacin, oxybutynin and darifenacin, respectively. Furthermore, oral administration of DA-8010 in mice resulted in more selective and persistent binding for muscarinic receptors in the bladder rather than in the submaxillary gland, in comparison with other antimuscarinics. These findings suggest that DA-8010 is a potent muscarinic M3 receptor antagonist to be highly selective for bladder over salivary gland, which might be a promising agent with greater efficacy and less dry mouth in the treatment of overactive bladder.


Subject(s)
Muscarinic Antagonists/pharmacology , Receptor, Muscarinic M3/antagonists & inhibitors , Salivary Glands/drug effects , Urinary Bladder/drug effects , Animals , Female , Male , Mice, Inbred ICR , Rats, Sprague-Dawley , Receptor, Muscarinic M3/physiology , Salivary Glands/physiology , Urinary Bladder/physiology
15.
Auton Neurosci ; 216: 33-38, 2019 01.
Article in English | MEDLINE | ID: mdl-30196037

ABSTRACT

The mechanisms underlying bowel dysfunction after high-level spinal cord injury (SCI) are poorly understood. However, impaired supraspinal sympathetic and parasympathetic control is likely a major contributing factor. Disruption of the descending autonomic pathways traversing the spinal cord was achieved by a T3 complete spinal cord transection, and colonic function was examined in vivo and ex vivo four weeks post-injury. Total gastrointestinal transit time (TGTT) was reduced and contractility of the proximal and distal colon was impaired due to reduced M3 receptor sensitivity. These data describe a clinically relevant model of bowel dysfunction after SCI.


Subject(s)
Colon/physiopathology , Gastrointestinal Motility/physiology , Muscle, Smooth/physiopathology , Spinal Cord Injuries/physiopathology , Animals , Gastrointestinal Transit/physiology , Male , Rats , Receptor, Muscarinic M3/physiology , Time Factors
16.
J Neurosci ; 38(31): 6921-6932, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29959237

ABSTRACT

Muscarinic receptor antagonists act as potent inducers of oligodendrocyte differentiation and accelerate remyelination. However, the use of muscarinic antagonists in the clinic is limited by poor understanding of the operant receptor subtype, and questions regarding possible species differences between rodents and humans. Based on high selective expression in human oligodendrocyte progenitor cells (OPCs), we hypothesized that M3R is the functionally relevant receptor. Lentiviral M3R knockdown in human primary CD140a/PDGFαR+ OPCs resulted in enhanced differentiation in vitro and substantially reduced the calcium response following muscarinic agonist treatment. Importantly, following transplantation in hypomyelinating shiverer/rag2 mice, M3R knockdown improved remyelination by human OPCs. Furthermore, conditional M3R ablation in adult NG2-expressing OPCs increased oligodendrocyte differentiation and led to improved spontaneous remyelination in mice. Together, we demonstrate that M3R receptor mediates muscarinic signaling in human OPCs that act to delay differentiation and remyelination, suggesting that M3 receptors are viable targets for human demyelinating disease.SIGNIFICANCE STATEMENT The identification of drug targets aimed at improving remyelination in patients with demyelination disease is a key step in development of effective regenerative therapies to treat diseases, such as multiple sclerosis. Muscarinic receptor antagonists have been identified as effective potentiators of remyelination, but the receptor subtypes that mediate these receptors are unclear. In this study, we show that genetic M3R ablation in both mouse and human cells results in improved remyelination and is mediated by acceleration of oligodendrocyte commitment from oligodendrocyte progenitor cells. Therefore, M3R represents an attractive target for induced remyelination in human disease.


Subject(s)
Myelin Sheath/physiology , Neurogenesis/physiology , Oligodendrocyte Precursor Cells/physiology , Receptor, Muscarinic M3/physiology , Remyelination/physiology , Animals , Brain Tissue Transplantation , Calcium Signaling , Cells, Cultured , Fetal Tissue Transplantation , Gene Knockdown Techniques , Humans , Mice , Mice, Neurologic Mutants , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Prosencephalon/embryology , Prosencephalon/transplantation , RNA Interference , Receptor, Muscarinic M3/agonists , Receptor, Muscarinic M3/antagonists & inhibitors , Spinal Cord/chemistry , Spinal Cord/ultrastructure
17.
Pharmacol Res ; 133: 152-159, 2018 07.
Article in English | MEDLINE | ID: mdl-29787869

ABSTRACT

The aim of this study was to better understand the role of TRPV4 in the regulation of blood vessel dilatation by blood flow and activation of GPCRs. Using pressure myography, the dilator responses to the TRPV4 agonist GSK1016790A and to acetylcholine, were examined in rat cremaster arterioles exposed to either no shear stress or to 200 µl/min flow for 6 min. In control vessels GSK1016709A caused vasodilatation (pEC50 7.73 ±â€¯0.12 M, ΔDmax 97 ±â€¯3%) which was significantly attenuated by the TRPV4 antagonists GSK2193874 (100 nM) (pEC50 6.19 ±â€¯0.11 M, p < 0.05) and HC067047 (300 nM) (pEC50 6.44 ±â€¯0.12 M) and abolished by removal of the endothelium. Shear conditioned arterioles were significantly more sensitive to GSK1016790A (pEC50 8.34 ±â€¯0.11, p < 0.05). Acetylcholine-induced vasodilatation (pEC50 7.02 ±â€¯0.07 M, ΔDmax 93 ±â€¯2%) was not affected by shear forces (pEC50 7.08 ±â€¯0.07 M, ΔDmax 95 ±â€¯1%). The dilator response to acetylcholine was unaffected by the TRPV4 antagonist GSK2193874 in control arterioles (pEC50 7.24 ±â€¯0.07 M, ΔDmax 97 ±â€¯2%). However, in shear treated arterioles, the acetylcholine-response was significantly attenuated by GSK2193874 (pEC50 6.25 ±â€¯0.12 M, p < 0.05) indicating an induced interaction between TRPV4 and muscarinic receptors. TRPV4 antibodies localized TRPV4 to the endothelium and shear stress had no effect on its localisation. Finally, agonist activation of the M3 muscarinic receptor opened TRPV4 in HEK293 cells. We concluded that shear stress increases endothelial TRPV4 agonist sensitivity and links TRPV4 activation to muscarinic receptor mediated endothelium-dependent vasodilatation, providing strong evidence that blood flow modulates downstream signalling from at least one but not all GPCRs expressed in the endothelium.


Subject(s)
Abdominal Muscles/blood supply , Arterioles/physiology , TRPV Cation Channels/physiology , Vasodilation/physiology , Animals , Endothelium, Vascular/physiology , HEK293 Cells , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Male , Rats, Wistar , Receptor, Muscarinic M3/physiology , Stress, Mechanical , Sulfonamides/pharmacology , TRPV Cation Channels/agonists
18.
Neurourol Urodyn ; 37(7): 2128-2134, 2018 09.
Article in English | MEDLINE | ID: mdl-29675856

ABSTRACT

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.


Subject(s)
Receptor, Muscarinic M3/physiology , Receptors, Adrenergic, alpha-1/physiology , Urinary Tract/innervation , Adrenergic alpha-Agonists/pharmacology , Aged , Aged, 80 and over , Carbachol/pharmacology , Female , Humans , Male , Middle Aged , Muscarinic Agonists/pharmacology , Muscle Contraction/drug effects , Phenylephrine/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Receptor, Muscarinic M3/drug effects , Receptors, Adrenergic, alpha-1/drug effects , Sex Characteristics , Urethra/drug effects , Urethra/physiology , rho-Associated Kinases/antagonists & inhibitors
19.
Neurosci Lett ; 649: 62-69, 2017 05 10.
Article in English | MEDLINE | ID: mdl-28408330

ABSTRACT

Muscarinic cholinoreceptors regulate the neurosecretion process in vertebrate neuromuscular junctions. The diversity of muscarinic effects on acetylcholine (ACh) secretion may be attributed to the different muscarinic subtypes involved in this process. In the present study, the location of five muscarinic receptor subtypes (M1, M2, M3, M4 and M5) on the motor nerve terminals of frog cutaneous pectoris muscle was shown using specific polyclonal antibodies. The modulatory roles of these receptors were investigated via assessment of the effects of muscarine and specific muscarinic antagonists on the quantal content of endplate currents (EPCs) and the time course of secretion, which was estimated from the distribution of "real" synaptic delays of EPCs recorded in a low Ca2+/high Mg2+ solution. The agonist muscarine decreased the EPC quantal content and synchronized the release process. The depressing action of muscarine on the EPC quantal content was abolished only by pretreatment of the preparation with the M3 blockers 4-DAMP (1,1-Dimethyl-4-diphenylacetoxypiperidinium iodide) and J 104129 fumarate ((αR)-α-Cyclopentyl-α-hydroxy-N-[1-(4-methyl-3-pentenyl)-4-piperidinyl]benzeneacetamide fumarate). Moreover, antagonists of the M1, M2, M3 and M4 receptors per se diminished the intensity of secretion, which suggests a putative up-regulation of the release by endogenous ACh.


Subject(s)
Acetylcholine/metabolism , Motor Endplate/metabolism , Receptors, Muscarinic/physiology , Animals , Female , Male , Motor Endplate/physiology , Rana ridibunda , Receptor, Muscarinic M1/physiology , Receptor, Muscarinic M2/physiology , Receptor, Muscarinic M3/physiology , Receptor, Muscarinic M4/physiology
20.
Behav Brain Res ; 323: 111-116, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28143769

ABSTRACT

The ventral tegmental area (VTA) and the rostromedial tegmental nucleus (RMTg) each contribute to opiate reward and each receive inputs from the laterodorsal tegmental and pedunculopontine tegmental nuclei, the two principle brainstem cholinergic cell groups. We compared the contributions of VTA or RMTg muscarinic cholinergic receptors to locomotion induced by morphine infusions into the same sites. VTA co-infusion of atropine completely blocked VTA morphine-induced locomotion providing additional support for the important role of VTA muscarinic cholinergic receptors in the stimulant effects of opiates. By contrast, RMTg co-infusion of atropine increased RMTg morphine-induced locomotion. Furthermore, RMTg co-infusion of the M3-selective antagonist 4-DAMP, but not the M4-selective antagonist Tropicamide, strongly increased RMTg morphine-induced locomotion. RMTg infusions of 4-DAMP, but not of Tropicamide, by themselves strongly increased drug-free locomotion. Muscarinic cholinergic receptors in the RMTg thus also contribute to the stimulant effects of morphine, but in a way opposite to those in VTA. We suggest that the net effect of endogenous cholinergic input to the RMTg on drug-free and on RMTg morphine-induced locomotion is inhibitory.


Subject(s)
Locomotion/drug effects , Morphine/administration & dosage , Muscarinic Antagonists/administration & dosage , Receptor, Muscarinic M3/physiology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology , Acetylcholine/physiology , Animals , Atropine/administration & dosage , Mice , Mice, Inbred C57BL , Piperidines/administration & dosage , Receptor, Muscarinic M3/antagonists & inhibitors , Receptor, Muscarinic M4/antagonists & inhibitors , Receptor, Muscarinic M4/physiology , Tropicamide/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...