Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Nature ; 585(7826): 591-596, 2020 09.
Article in English | MEDLINE | ID: mdl-32526765

ABSTRACT

Recent clinical and experimental evidence has evoked the concept of the gut-brain axis to explain mutual interactions between the central nervous system and gut microbiota that are closely associated with the bidirectional effects of inflammatory bowel disease and central nervous system disorders1-4. Despite recent advances in our understanding of neuroimmune interactions, it remains unclear how the gut and brain communicate to maintain gut immune homeostasis, including in the induction and maintenance of peripheral regulatory T cells (pTreg cells), and what environmental cues prompt the host to protect itself from development of inflammatory bowel diseases. Here we report a liver-brain-gut neural arc that ensures the proper differentiation and maintenance of pTreg cells in the gut. The hepatic vagal sensory afferent nerves are responsible for indirectly sensing the gut microenvironment and relaying the sensory inputs to the nucleus tractus solitarius of the brainstem, and ultimately to the vagal parasympathetic nerves and enteric neurons. Surgical and chemical perturbation of the vagal sensory afferents at the hepatic afferent level reduced the abundance of colonic pTreg cells; this was attributed to decreased aldehyde dehydrogenase (ALDH) expression and retinoic acid synthesis by intestinal antigen-presenting cells. Activation of muscarinic acetylcholine receptors directly induced ALDH gene expression in both human and mouse colonic antigen-presenting cells, whereas genetic ablation of these receptors abolished the stimulation of antigen-presenting cells in vitro. Disruption of left vagal sensory afferents from the liver to the brainstem in mouse models of colitis reduced the colonic pTreg cell pool, resulting in increased susceptibility to colitis. These results demonstrate that the novel vago-vagal liver-brain-gut reflex arc controls the number of pTreg cells and maintains gut homeostasis. Intervention in this autonomic feedback feedforward system could help in the development of therapeutic strategies to treat or prevent immunological disorders of the gut.


Subject(s)
Brain/cytology , Intestines/cytology , Intestines/innervation , Liver/cytology , Liver/innervation , Neurons/physiology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Afferent Pathways , Animals , Antigen-Presenting Cells/immunology , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Homeostasis , Humans , Intestines/immunology , Male , Mice , Rats , Receptors, Muscarinic/metabolism , Spleen/cytology , Spleen/immunology , Vagus Nerve/physiology
2.
Mod Rheumatol ; 30(5): 884-893, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31490711

ABSTRACT

Objective: We previously reported that Rag1-/- mice inoculated with splenocytes from M3 muscarinic acetylcholine receptor (M3R) knockout mice immunized with an M3R peptide mixture developed sialadenitis-like Sjögren's syndrome (M3R-induced sialadenitis [MIS]). We also found that intravenous administration of altered peptide ligand (APL) of N-terminal 1 (N1), which is one of the T-cell epitopes of M3R, suppressed MIS. In this study, we aimed to evaluate the suppressive ability and its mechanisms of rice seeds expressing N1-APL7 against MIS.Methods: Rice seeds expressing N1 and N1-APL7 were orally administered to MIS mice for 2 weeks. The changes in saliva flow and sialadenitis (salivary gland inflammation) were analyzed. The M3R-specific T-cell response in the spleen and the expression of regulatory molecules in the cervical lymph nodes and mesenteric lymph nodes were also analyzed.Results: Oral administration of N1-APL7-expressing rice seeds significantly recovered reduction in saliva flow and suppressed sialadenitis when compared with treatment with nontransgenic rice seeds and N1 rice seeds. IFNγ production from M3R-reactive T cells tended to decline in the N1-APL7 rice-treated group as compared with those in the other groups. In the N1-APL7 rice-treated group, the mRNA expression levels of Foxp3 in the cervical-lymph-node CD4+ T cells were higher than those in the other groups.Conclusion: Oral administration of N1-APL7-expressing rice suppressed MIS via suppression of M3R-specific IFNγ and IL-17 production and via enhancement of regulatory molecule expression.Key messagesWe generated N1-peptide- or N1-APL7-expressing rice seeds. Oral administration of N1-APL7-expressing rice seeds significantly recovered the reduction of saliva flow and suppressed sialadenitis via the suppression of M3R specific IFNγ and IL-17 production and via enhancement of regulatory T (Treg) cells.


Subject(s)
Peptides/therapeutic use , Plant Proteins/chemistry , Receptor, Muscarinic M3/metabolism , Sialadenitis/drug therapy , Sjogren's Syndrome/drug therapy , Animals , Humans , Ligands , Mice , Oryza/chemistry , Oryza/genetics , Peptides/administration & dosage , Peptides/chemistry , Plants, Genetically Modified/chemistry , Plants, Genetically Modified/genetics , Protein Binding , Seeds/chemistry , Sialadenitis/immunology , Sjogren's Syndrome/immunology , Spleen/immunology , T-Lymphocytes, Regulatory/immunology
3.
Eur J Immunol ; 42(5): 1152-63, 2012 May.
Article in English | MEDLINE | ID: mdl-22328321

ABSTRACT

Patients with dilated cardiomyopathy (DCM) often have autoantibodies against cardiac antigens including the M(2) muscarinic acetylcholine receptor (M(2)R). To elucidate the role of autoimmunity against M(2)R in disease development, we induced an immune response against M(2)R by adoptive transfer into Rag2(-/-) mice of splenocytes from M(2)R(-/-) mice immunized with a recombinant M(2)R protein. T lymphocytes transiently infiltrated the heart in recipient mice followed by morphological changes in cardiomyocytes. These mice produced IgG antibodies against M(2)R, which bound to cardiomyocytes in vivo and decreased the amplitude of calcium signals in isolated rat cardiomyocytes in vitro. Recipient mice showed increased heart weights associated with increased intraventricular diameter, decreased systolic function, and increased action potential duration, which are characteristics of DCM. Our results suggest that myocarditis and DCM associated with the presence of anti-M(2)R antibodies are autoimmune diseases with a risk of progressing to the terminal stage. Our mouse model will be useful in the analysis of the molecular mechanisms of disease progression and the development of new therapies for DCM.


Subject(s)
Autoimmunity , Cardiomyopathy, Dilated/immunology , Disease Models, Animal , Myocarditis/immunology , Receptor, Muscarinic M2/immunology , Action Potentials/physiology , Animals , Autoantibodies/biosynthesis , Autoantibodies/immunology , Calcium Signaling/immunology , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Cells, Cultured , Female , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred C57BL , Myocarditis/pathology , Myocarditis/physiopathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Organ Size/immunology , Rats , Rats, Wistar , T-Lymphocytes/immunology
4.
J Neurosci ; 30(12): 4408-18, 2010 Mar 24.
Article in English | MEDLINE | ID: mdl-20335477

ABSTRACT

Acetylcholine (ACh) plays important roles for higher brain functions, including arousal, attention, and cognition. These effects are mediated largely by muscarinic acetylcholine receptors (mAChRs). However, it remains inconclusive whether the mode of ACh-mAChR signaling is synaptic, so-called "wired," transmission mediated by ACh released into the synaptic cleft, or nonsynaptic, so-called "volume," transmission by ambient ACh. To address this issue, we examined cellular and subcellular distribution of M(1), the most predominant mAChR subtype in the cerebral cortex and hippocampus, and pursued its anatomical relationship with cholinergic varicosities in these regions of adult mice. M(1) was highly expressed in glutamatergic pyramidal neurons, whereas it was low or undetectable in various GABAergic interneuron subtypes. M(1) was preferentially distributed on the extrasynaptic membrane of pyramidal cell dendrites and spines. Cholinergic varicosities often made direct contact to pyramidal cell dendrites and synapses. At such contact sites, however, synapse-like specialization was infrequent, and no particular accumulation was found at around contact sites for both M(1) and presynpatic active zone protein Bassoon. These features contrasted with those of the glutamatergic system, in which AMPA receptor GluA2 and metabotropic receptor mGluR5 were recruited to the synaptic or perisynaptic membrane, respectively, and Bassoon was highly accumulated in the presynaptic terminals. These results suggest that M(1) is so positioned to sense ambient ACh released from cholinergic varicosities at variable distances, and to enhance the synaptic efficacy and excitability of pyramidal cells. These molecular-anatomical arrangements will provide the evidence for volume transmission, at least in M(1)-mediated cortical cholinergic signaling.


Subject(s)
Cerebral Cortex/cytology , Dendrites/ultrastructure , Dendritic Spines/metabolism , Pyramidal Cells/ultrastructure , Receptor, Muscarinic M1/metabolism , Animals , Calbindin 2 , Dendrites/metabolism , Glutamate Decarboxylase/metabolism , Hippocampus/cytology , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission/methods , Microscopy, Immunoelectron/methods , Microtubule-Associated Proteins/metabolism , Neuropeptide Y/metabolism , Nitric Oxide Synthase Type I/metabolism , Pyramidal Cells/metabolism , RNA, Messenger/metabolism , Receptor, Muscarinic M1/deficiency , Receptor, Muscarinic M1/genetics , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , S100 Calcium Binding Protein G/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism , Vesicular Glutamate Transport Protein 1/metabolism
5.
J Pharmacol Exp Ther ; 339(3): 870-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21885618

ABSTRACT

Increased smooth muscle tone in the human prostate contributes to the symptoms associated with benign prostatic hyperplasia. In the mouse prostate gland, cholinergic innervation is responsible for a component of the nerve-mediated contractile response. This study investigates the muscarinic receptor subtype responsible for the cholinergic contractile response in the mouse prostate gland. To characterize the muscarinic receptor subtype, mouse prostates taken from wild-type or M(3) muscarinic receptor knockout mice were mounted in organ baths. The isometric force that tissues developed in response to electrical-field stimulation or exogenously applied cholinergic agonists in the presence or absence of a range of muscarinic receptor antagonists was evaluated. Carbachol elicited reproducible and concentration-dependent contractions of the isolated mouse prostate, which were antagonized by the presence of muscarinic receptor antagonists. Calculation of antagonist affinities (pA(2) values) indicated a rank order of antagonist potencies in the mouse prostate of: darifenacin (9.08) = atropine (9.07) = 1,1-dimethyl-4-diphenylacetoxypiperidinium iodide (9.02) > cyclohexyl-hydroxy-phenyl-(3-piperidin-1-ylpropyl)silane (7.85) > cyclohexyl-(4-fluorophenyl)-hydroxy-(3-piperidin-1-ylpropyl)silane (7.39) > himbacine (7.19) > pirenzipine (6.88) > methoctramine (6.20). Furthermore, genetic deletion of the M(3) muscarinic receptor inhibited prostatic contractions to electrical-field stimulation or exogenous administration of acetylcholine. In this study we identified that the cholinergic component of contraction in the mouse prostate is mediated by the M(3) muscarinic receptor subtype. Pharmacological antagonism of the M(3) muscarinic receptor may be a beneficial additional target for the treatment of benign prostatic hyperplasia in the human prostate gland.


Subject(s)
Acetylcholine/pharmacology , Cholinergic Agonists/pharmacology , Mecamylamine/pharmacology , Muscarinic Antagonists/pharmacology , Muscle Contraction/drug effects , Nicotinic Antagonists/pharmacology , Prostate/physiology , Receptor, Muscarinic M3/antagonists & inhibitors , Receptor, Muscarinic M3/metabolism , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Animals , Benzofurans/pharmacology , Body Weight/drug effects , Carbachol/pharmacology , Drug Interactions , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction/physiology , Prazosin/pharmacology , Prostate/drug effects , Pyrrolidines/pharmacology , Receptor, Muscarinic M3/genetics
6.
J Neurosci ; 29(31): 9888-902, 2009 Aug 05.
Article in English | MEDLINE | ID: mdl-19657040

ABSTRACT

ACh release into the rodent prefrontal cortex is predictive of successful performance of cue detection tasks, yet the cellular mechanisms underlying cholinergic modulation of cortical function are not fully understood. Prolonged ("tonic") muscarinic ACh receptor (mAChR) activation increases the excitability of cortical pyramidal neurons, whereas transient ("phasic") mAChR activation generates inhibitory and/or excitatory responses, depending on neuron subtype. These cholinergic effects result from activation of "M1-like" mAChRs (M1, M3, and M5 receptors), but the specific receptor subtypes involved are not known. We recorded from cortical pyramidal neurons from wild-type mice and mice lacking M1, M3, and/or M5 receptors to determine the relative contribution of M1-like mAChRs to cholinergic signaling in the mouse prefrontal cortex. Wild-type neurons in layer 5 were excited by tonic mAChR stimulation, and had biphasic inhibitory followed by excitatory, responses to phasic ACh application. Pyramidal neurons in layer 2/3 were substantially less responsive to tonic and phasic cholinergic input. Cholinergic effects were largely absent in neurons from mice lacking M1 receptors, but most were robust in neurons lacking M3, M5, or both M3 and M5 receptors. The exception was tonic cholinergic suppression of the afterhyperpolarization in layer 5 neurons, which was absent in cells lacking either M1 or M3 receptors. Finally, we confirm a role for M1 receptors in behavior by demonstrating cue detection deficits in M1-lacking mice. Together, our results demonstrate that M1 receptors facilitate cue detection behaviors and are both necessary and sufficient for most direct effects of ACh on pyramidal neuron excitability.


Subject(s)
Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Receptor, Muscarinic M1/metabolism , Action Potentials/drug effects , Analysis of Variance , Animals , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Conditioning, Classical/physiology , Cues , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Patch-Clamp Techniques , Prefrontal Cortex/drug effects , Pyramidal Cells/drug effects , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M3/agonists , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/metabolism , Receptor, Muscarinic M5/agonists , Receptor, Muscarinic M5/genetics , Receptor, Muscarinic M5/metabolism
7.
J Neurosci ; 29(41): 12896-908, 2009 Oct 14.
Article in English | MEDLINE | ID: mdl-19828804

ABSTRACT

To examine the intrasynaptic arrangement of postsynaptic receptors in relation to the functional role of the synapse, we quantitatively analyzed the two-dimensional distribution of AMPA and NMDA receptors (AMPARs and NMDARs, respectively) using SDS-digested freeze-fracture replica labeling (SDS-FRL) and assessed the implication of distribution differences on the postsynaptic responses by simulation. In the dorsal lateral geniculate nucleus, corticogeniculate (CG) synapses were twice as large as retinogeniculate (RG) synapses but expressed similar numbers of AMPARs. Two-dimensional views of replicas revealed that AMPARs form microclusters in both synapses to a similar extent, resulting in larger AMPAR-lacking areas in the CG synapses. Despite the broad difference in the AMPAR distribution within a synapse, our simulations based on the actual receptor distributions suggested that the AMPAR quantal response at individual RG synapses is only slightly larger in amplitude, less variable, and faster in kinetics than that at CG synapses having a similar number of the receptors. NMDARs at the CG synapses were expressed twice as many as those in the RG synapses. Electrophysiological recordings confirmed a larger contribution of NMDAR relative to AMPAR-mediated responses in CG synapses. We conclude that synapse size and the density and distribution of receptors have minor influences on quantal responses and that the number of receptors acts as a predominant postsynaptic determinant of the synaptic strength mediated by both the AMPARs and NMDARs.


Subject(s)
Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , Animals, Newborn , Biophysics , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Freeze Fracturing/methods , Geniculate Bodies/cytology , Glutamic Acid/pharmacology , In Vitro Techniques , Male , Mice , Mice, Knockout , Microscopy, Electron/methods , Neural Pathways/metabolism , Neural Pathways/ultrastructure , Rats , Rats, Long-Evans , Receptor, Muscarinic M2/deficiency , Receptors, AMPA/classification , Receptors, AMPA/ultrastructure , Receptors, N-Methyl-D-Aspartate/classification , Receptors, N-Methyl-D-Aspartate/ultrastructure , Retina/cytology , Retina/physiology , Statistics, Nonparametric , Synapses/classification , Synapses/drug effects , Synapses/ultrastructure , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/metabolism
8.
J Autoimmun ; 35(4): 383-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20864316

ABSTRACT

The aim of this study was to clarify the role of the immune response to muscarinic type 3 receptor (M3R) in the pathogenesis of Sjögren's syndrome (SS). M3R(-/-) mice were immunized with murine M3R peptides and their splenocytes were inoculated into Rag1(-/-) (M3R(-/-)→Rag1(-/-)) mice. M3R(-/-)→Rag1(-/-) mice had high serum levels of anti-M3R antibodies and low saliva volume. Histological examination showed marked infiltration of mononuclear cells in the salivary glands and immunohistochemistry demonstrated that the majority of these cells were CD4(+) T cells with a few B cells and several IFN-γ- and IL-17-producing cells. Apoptotic cells were present in the salivary glands of M3R(-/-)→Rag1(-/-) mice. Moreover, transfer of only CD3(+) T cells from M3R(-/-) immunized with M3R peptides into Rag1(-/-) mice resulted in cell infiltration and destruction of epithelial cells in the salivary glands, suggesting that M3R reactive CD3(+) T cells play a pathogenic role in the development of autoimmune sialoadenitis. Our findings support the notion that the immune response to M3R plays a crucial role in the pathogenesis of SS-like autoimmune sialoadenitis.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Peptide Fragments/metabolism , Receptor, Muscarinic M3/metabolism , Sialadenitis/immunology , Sjogren's Syndrome/immunology , Adoptive Transfer , Animals , Apoptosis , Autoantibodies/blood , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/pathology , Humans , Immunization , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/genetics , Peptide Fragments/immunology , Receptor, Muscarinic M3/genetics , Receptor, Muscarinic M3/immunology , Salivary Glands/pathology , Sialadenitis/blood
10.
Mol Pharmacol ; 75(2): 381-96, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18996972

ABSTRACT

In prior work, we have shown that it is possible to estimate the product of observed affinity and intrinsic efficacy of an agonist expressed relative to that of a standard agonist simply through the analysis of their respective concentration-response curves. In this report, we show analytically and through mathematical modeling that this product, termed intrinsic relative activity (RA(i)), is equivalent to the ratio of microscopic affinity constants of the agonists for the active state of the receptor. We also compared the RA(i) estimates of selected muscarinic agonists with a relative estimate of the product of observed affinity and intrinsic efficacy determined independently through the method of partial receptor inactivation. There was good agreement between these two estimates when agonist-mediated inhibition of forskolin-stimulated cAMP accumulation was measured in Chinese hamster ovary cells stably expressing the human M(2) muscarinic receptor. Likewise, there was good agreement between the two estimates when agonist activity was measured on the ileum from M(2) muscarinic receptor knockout mice, a convenient assay for M(3) receptor activity. The RA(i) estimates of agonists in the mouse ileum were similar to those estimated at the human M(3) receptor with the exception of 4-(m-chlorophenyl-carbamoyloxy)-2-butynyltrimethylammonium (McN-A-343), which is known to be an M(1)- and M(4)-selective muscarinic agonist. Additional experiments showed that the response to McN-A-343 in the mouse ileum included a non-M(3) muscarinic receptor component. Our results show that the RA(i) estimate is a useful receptor-dependent measure of agonist activity and ligand-directed signaling.


Subject(s)
Protein Interaction Domains and Motifs , Receptor, Muscarinic M2/chemistry , Receptor, Muscarinic M3/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Female , Humans , Male , Mice , Physical Phenomena
11.
Eur J Pharmacol ; 843: 104-112, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30452911

ABSTRACT

Muscarinic receptor stimulation induces depolarizing inward currents and catecholamine secretion in adrenal medullary (AM) cells from various mammals. In guinea-pig AM cells muscarine and oxotremorine at concentrations ≤ 1 µM produce activation of nonselective cation channels with a similar potency and efficacy, whereas muscarine at higher concentrations produces not only nonselective cation channel activation, but also TASK1 channel inhibition. In rat AM cells, the muscarinic M1 receptor is involved in TASK1 channel inhibition in response to muscarinic agonists, and the efficacy of oxotremorine is half that of muscarine. These pharmacological findings might indicate that different muscarinic receptor subtypes are responsible for the regulation of nonselective cation and TASK1 channel activities. The present study aimed to determine the muscarinic receptor subtypes involved in nonselective cation channel activation in guinea-pig and mouse AM cells. The inward current evoked by 1 µM muscarine was completely suppressed by 100 µM quinine, whereas 30 µM muscarine-induced inward currents were comprised of quinine-sensitive and -insensitive components. The electrophysiological and pharmacological properties of the muscarine-induced currents indicated that the quinine-sensitive and insensitive components are due to nonselective cation channel activation and TASK1 channel inhibition, respectively. Muscarine at 30 µM failed to induce any current in AM cells treated with muscarinic toxin 7 or genetically deleted of the M1 receptor. The KD value of VU0255035 against the muscarinic receptor mediating nonselective cation channel activation was 17.5 nM. These results indicate that the M1 receptor mediates nonselective cation channel activation as well as TASK1 channel inhibition.


Subject(s)
Adrenal Medulla/cytology , Ion Channels/physiology , Muscarinic Agonists/pharmacology , Receptor, Muscarinic M1/physiology , Animals , Guinea Pigs , Male , Mice, Inbred C57BL , Mice, Knockout , Muscarine/pharmacology , Oxotremorine/pharmacology , Quinine/pharmacology
12.
J Neurosci ; 27(3): 496-506, 2007 Jan 17.
Article in English | MEDLINE | ID: mdl-17234582

ABSTRACT

Tonically active cholinergic interneurons in the striatum modulate activities of striatal outputs from medium spiny (MS) neurons and significantly influence overall functions of the basal ganglia. Cellular mechanisms of this modulation are not fully understood. Here we show that ambient acetylcholine (ACh) derived from tonically active cholinergic interneurons constitutively upregulates depolarization-induced release of endocannabinoids from MS neurons. The released endocannabinoids cause transient suppression of inhibitory synaptic inputs to MS neurons through acting retrogradely onto presynaptic CB1 cannabinoid receptors. The effects were mediated by postsynaptic M(1) subtype of muscarinic ACh receptors, because the action of a muscarinic agonist to release endocannabinoids and the enhancement of depolarization-induced endocannabinoid release by ambient ACh were both deficient in M1 knock-out mice and were blocked by postsynaptic infusion of guanosine-5'-O-(2-thiodiphosphate). Suppression of spontaneous firings of cholinergic interneurons by inhibiting Ih current reduced the depolarization-induced release of endocannabinoids. Conversely, elevation of ambient ACh concentration by inhibiting choline esterase significantly enhanced the endocannabinoid release. Paired recording from a cholinergic interneuron and an MS neuron revealed that the activity of single cholinergic neuron could influence endocannabinoid-mediated signaling in neighboring MS neurons. These results clearly indicate that striatal endocannabinoid-mediated modulation is under the control of cholinergic interneuron activity. By immunofluorescent and immunoelectron microscopic examinations, we demonstrated that M1 receptor was densely distributed in perikarya and dendrites of dopamine D1 or D2 receptor-positive MS neurons. Thus, we have disclosed a novel mechanism by which the muscarinic system regulates striatal output and may contribute to motor control.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Cholinergic Fibers/physiology , Corpus Striatum/physiology , Endocannabinoids , Interneurons/physiology , Neural Inhibition/physiology , Synaptic Transmission/physiology , Amino Acid Sequence , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Receptor, Cannabinoid, CB1/physiology
14.
J Neuroimmunol ; 188(1-2): 80-5, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17586055

ABSTRACT

Immunological activation of T cells enhances synthesis of acetylcholine (ACh) and transcription of choline acetyltransferase (ChAT), M5 muscarinic ACh receptor (mAChR) and acetylcholinesterase (AChE). Stimulation of mAChRs on T and B cells causes oscillating Ca(2+)-signaling and up-regulation of c-fos expression; moreover, M1 mAChRs play a crucial role in the differentiation of CD8(+) T cells into cytolytic T lymphocytes. Collectively, these findings suggest that immune cell function is regulated by its own cholinergic system. Bearing that in mind, we tested whether immune function can be regulated via mAChR-mediated pathways by immunizing combined M1 and M5 mAChR knockout (M1/M5 KO) and wild-type (WT) C57BL/6JJcl mice with ovalbumin (OVA) and measuring serum IgG1 and IgM 1 wk later. We found that serum levels of total and anti-OVA-specific IgG1 were significantly lower in M1/M5 KO than WT mice, though there was no difference in serum levels of total and anti-OVA-specific IgM between the two genotypes. Secretion of interleukin (IL)-6 from activated spleen cells was significantly reduced in M1/M5 KO mice, whereas there was no significant change in gamma interferon secretion. Expression of AChE mRNA was significantly reduced in activated spleen cells from M1/M5 KO mice. These results suggest that M1 and/or M5 mAChRs are involved in regulating cytokine (e.g., IL-6) production, leading to modulation of antibody class switching from IgM to IgG1, but are not involved in the initial generation of the antibody response. They also support the notion that a non-neuronal cholinergic system is involved in regulating immune cell function.


Subject(s)
Acetylcholinesterase/metabolism , Gene Expression Regulation, Enzymologic/genetics , Immunoglobulin G/blood , Interleukin-6/metabolism , Receptor, Muscarinic M1/deficiency , Receptor, Muscarinic M5/deficiency , Acetylcholinesterase/genetics , Animals , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Female , Interferon-gamma/metabolism , Lymphocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
15.
Neurosci Res ; 57(3): 399-410, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17196693

ABSTRACT

The vesl-1/homer1 gene encodes a scaffold protein that interacts with several receptors to modulate synaptic functions. The gene also encodes two shorter forms that counteract the functions of the long form of Vesl. Expression of the shorter forms is driven by neural activities such as long-term potentiation. Here we analyzed the mechanism regulating vesl-1 alternative splicing. Each functional poly(A) site was in a different part of the 3'-terminal exon, with promoter-proximal and promoter-distal sites at the end of exons corresponding to the short and long form Vesl-1, respectively. 3'-End-processing at proximal poly(A) site, specifically at the vesl-1M poly(A) site, was enhanced by extracellular stimuli, thereby switching transcription termination from promoter-distal to -proximal poly(A) site. This switch was not specifically coupled to the vesl-1 promoter and was independent of de novo protein synthesis. Analysis of transcripts from mini-genes that mimic the structure of endogenous vesl-1 revealed that the vesl-1M poly(A) region plays a crucial role in switching to the alternative pre-mRNA splicing that is triggered by extracellular stimuli. Therefore, a 3'-end-processing event regulates the neural activity-dependent alternative splicing of vesl-1. This is the first report of a gene in which alternative poly(A) site-selection regulates alternative splicing in a protein synthesis-independent manner.


Subject(s)
Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Central Nervous System/metabolism , Neurons/metabolism , Polyadenylation/genetics , RNA Splice Sites/genetics , Synaptic Membranes/metabolism , Alternative Splicing/genetics , Animals , Gene Expression Regulation/genetics , Homer Scaffolding Proteins , Humans , Mice , PC12 Cells , Promoter Regions, Genetic/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Regulatory Elements, Transcriptional/genetics , Synaptic Membranes/genetics
16.
J Neurosci ; 25(48): 11194-200, 2005 Nov 30.
Article in English | MEDLINE | ID: mdl-16319319

ABSTRACT

The muscarinic acetylcholine receptor (mAChR) has been considered one of the neurotransmitter receptors regulating hippocampal synaptic plasticity, which likely plays a critical role in learning and memory. In previous studies, however, muscarinic agonists were used at relatively high concentrations, and the subtype selectivity of muscarinic antagonists was not satisfactory. Thus, it remains to be answered whether physiological levels of ACh are involved in the regulation of synaptic plasticity and which mAChR subtypes are responsible for such effects. We found in this study that a low concentration (50 nM) of carbachol enhanced long-term potentiation (LTP) of excitatory synaptic transmission in mouse hippocampal slices. Notably, this enhancing effect was abolished in M1 mAChR knock-out (KO) but not in M3 mAChR KO mice, although LTP itself was intact in both mutant mice. Furthermore, we found that repetitive stimulation in the stratum oriens, which presumably triggered the release of endogenous ACh from cholinergic terminals, could enhance LTP in wild-type mice but not in M1 mAChR KO mice. These results suggest that physiologically released ACh from cholinergic fibers modulates hippocampal synaptic plasticity through the postsynaptic M1 mAChR activation.


Subject(s)
Hippocampus/metabolism , Neuronal Plasticity/physiology , Receptor, Muscarinic M1/physiology , Synapses/physiology , Acetylcholine/physiology , Animals , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Electric Stimulation , Electrophysiology , In Vitro Techniques , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Muscarinic M1/deficiency , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
17.
J Neurosci ; 22(24): 10627-32, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12486155

ABSTRACT

Cholinergic agents elicit prominent smooth muscle contractions via stimulation of muscarinic receptors that comprise five distinct subtypes (M1-M5). Although such contractions are important for autonomic organs, the role of each subtype has not been characterized precisely because of the poor selectivity of the currently available muscarinic ligands. Here, we generated a mutant mouse line (M2-/-M3-/- mice) lacking M2 and M3 receptors that are implicated in such cholinergic contractions. The relative contributions of M2 and M3 receptors in vitro was approximately 5 and 95% for the detrusor muscle contraction and approximately 25 and 75% for the ileal longitudinal muscle contraction, respectively. Thus, M1, M4, or M5 receptors do not seem to play a role in such contractions. Despite the complete lack of cholinergic contractions in vitro, M2-/-M3-/- mice were viable, fertile, and free of apparent intestinal complications. The urinary bladder was distended only in males, which excludes a major contribution by cholinergic mechanisms to the urination in females. Thus, cholinergic mechanisms are dispensable in gastrointestinal motility and female urination. After 10 Hz electrical field stimulation, noncholinergic inputs were found to be increased in the ileum of M2-/-M3-/- females, which may account for the lack of apparent functional deficits. Interestingly, the M2-/-M3-/- mice had smaller ocular pupils than M3-deficient mice. The results suggest a novel role of M2 in the pupillary dilation, contrary to the well known cholinergic constriction. These results collectively suggest that an additional mechanism operates in the control of pupillary constriction-dilatation.


Subject(s)
Muscle Contraction , Muscle, Smooth/physiology , Receptors, Muscarinic/physiology , Animals , Carbachol/pharmacology , Digestive System/anatomy & histology , Electric Stimulation , Female , Gene Targeting , Male , Mice , Mice, Knockout , Miosis/pathology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Phenotype , Pupil/physiology , Receptor, Muscarinic M2 , Receptor, Muscarinic M3 , Receptors, Muscarinic/genetics , Sex Factors , Survival Analysis , Urinary Retention/diagnosis , Urinary Retention/etiology
18.
Brain Res Mol Brain Res ; 133(1): 6-11, 2005 Jan 05.
Article in English | MEDLINE | ID: mdl-15661360

ABSTRACT

We have studied binding parameters (Kd, Bmax) of [3H]N-methylscopolamine ([3H]NMS) in various brain regions and spinal cord of wild-type (WT) and muscarinic acetylcholine receptor (mAChR) subtype (M1-M5) knockout (KO) mice. In the M1-M4 KO mice, the number of [3H]NMS binding sites (Bmax) was decreased throughout the central nervous system (CNS) with significant regional differences. Our results collectively suggest that M1 receptor was present in a relatively high density in the cerebral cortex and hippocampus, and the densities of M1 and M4 subtypes were highest in the corpus striatum. M2 receptor appeared to be the major subtype in the thalamus, hypothalamus, midbrain, pons-medulla, cerebellum and spinal cord. These findings may contribute significantly not only to the further understanding of the physiological roles of mAChR subtypes in the central cholinergic functions, but also to the development of selective therapeutic agents targeting specific subtype.


Subject(s)
Central Nervous System/metabolism , N-Methylscopolamine/pharmacokinetics , Receptors, Muscarinic/metabolism , Animals , Dose-Response Relationship, Drug , Mice , Mice, Knockout/metabolism , Parasympatholytics/pharmacokinetics , Protein Binding , Radioligand Assay/methods , Receptors, Muscarinic/classification , Receptors, Muscarinic/genetics , Tissue Distribution , Tritium
19.
Br J Pharmacol ; 172(5): 1348-59, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25393049

ABSTRACT

BACKGROUND AND PURPOSE: Activation of muscarinic receptors results in catecholamine secretion in adrenal chromaffin cells in many mammals, and muscarinic receptors partly mediate synaptic transmission from the splanchnic nerve, at least in guinea pigs. To elucidate the physiological functions of muscarinic receptors in chromaffin cells, it is necessary to identify the muscarinic receptor subtypes involved in excitation. EXPERIMENTAL APPROACH: To identify muscarinic receptors, pharmacological tools and strains of mice where one or several muscarinic receptor subtypes were genetically deleted were used. Cellular responses to muscarinic stimulation in isolated chromaffin cells were studied with the patch clamp technique and amperometry. KEY RESULTS: Muscarinic M1, M4 and M5 receptors were immunologically detected in mouse chromaffin cells, and these receptors disappeared after the appropriate gene deletion. Mouse cells secreted catecholamines in response to muscarinic agonists, angiotensin II and a decrease in external pH. Genetic deletion of M1, but not M3, M4 or M5, receptors in mice abolished secretion in response to muscarine, but not to other stimuli. The muscarine-induced secretion was suppressed by MT7, a snake peptide toxin specific for M1 receptors. Similarly, muscarine failed to induce an inward current in the presence of MT7 in mouse and rat chromaffin cells. The binding affinity of VU0255035 for the inhibition of muscarine-induced currents agreed with that for the M1 receptor. CONCLUSIONS AND IMPLICATIONS: Based upon the effects of genetic deletion of muscarinic receptors and MT7, it is concluded that the M1 receptor alone is responsible for muscarine-induced catecholamine secretion.


Subject(s)
Adrenal Medulla/cytology , Catecholamines/metabolism , Chromaffin Cells/metabolism , Receptors, Muscarinic/metabolism , Adrenal Medulla/metabolism , Animals , Cells, Cultured , Chromaffin Cells/cytology , Dose-Response Relationship, Drug , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscarine/antagonists & inhibitors , Muscarine/pharmacology , PC12 Cells , Rats , Rats, Wistar , Receptors, Muscarinic/deficiency , Receptors, Muscarinic/genetics , Structure-Activity Relationship , Sulfonamides/pharmacology , Thiadiazoles/pharmacology
20.
Arthritis Rheumatol ; 67(8): 2213-25, 2015 May.
Article in English | MEDLINE | ID: mdl-25891013

ABSTRACT

OBJECTIVE: Autoreactive CD4+ T cells are involved in the pathogenesis of Sjögren's syndrome (SS). The aim of the present study was to clarify the dominant T cell epitopes of M3 muscarinic acetylcholine receptor (M3R) and to establish a new antigen-specific therapy for SS using an experimental mouse model. METHODS: Production of cytokines from M3R-reactive CD4+ T cells, after culture with various M3R peptides, was analyzed by enzyme-linked immunosorbent assay. Adoptive cell transfer was performed using splenocytes from M3R(-/-) mice that were immunized with M3R peptides or phosphate buffered saline plus H37Ra as a control. Rag1(-/-) mice were inoculated with the splenocytes and examined for the development of sialadenitis. Altered peptide ligands (APLs) of the T cell epitopes, with substitutions in amino acid residues at T cell receptor contact sites, were synthesized, and the ability of the APLs to suppress sialadenitis was evaluated. The mechanisms underlying such effects were assessed. RESULTS: CD4+ M3R-reactive T cells produced interleukin-17 (IL-17) and interferon-γ (IFNγ) in response to the N-terminal 1 (N1) and 1st extracellular loop peptides of M3R, and Rag1(-/-) mice that received N1- and/or 1st peptide-immunized splenocytes developed sialadenitis. Among the designed APLs, N1-APL7 (N→S at amino acid 15) significantly suppressed IFNγ production in vitro, and also suppressed sialadenitis in vivo. Levels of early growth response 2 in CD4+ T cells from the cervical lymph nodes of N1-APL7-treated mice were significantly higher than those of control mice, and cell proliferation was reversed by administration of exogenous IL-2. Levels of the anergy-related molecules itchy homolog E3 ubiquitin-protein ligase, Casitas B-lineage lymphoma b, gene related to anergy in lymphocytes, and Deltex-1 were significantly higher in CD4+ T cells cultured with N1-APL7. CONCLUSION: The major T cell epitopes were from the N1 and 1st peptide regions. Moreover, N1-APL7, selected as the antagonistic APL in vitro, also suppressed sialadenitis through the induction of anergy. This is a potentially useful strategy for regulating pathogenic T cell infiltration in SS.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Receptor, Muscarinic M3/immunology , Sialadenitis/immunology , Sjogren's Syndrome/immunology , Adaptor Proteins, Signal Transducing/metabolism , Adoptive Transfer , Animals , Clonal Anergy/immunology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Homeodomain Proteins/genetics , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-2/pharmacology , Ligands , Mice , Mice, Knockout , Proto-Oncogene Proteins c-cbl/metabolism , Receptor, Muscarinic M3/genetics , Spleen/cytology , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL