Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Eur J Pharm Sci ; 150: 105329, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32360768

ABSTRACT

Overactive bladder is a troublesome disease that affects 15% of the population in developed countries. Since pharmacotherapy of this condition is frequently associated with side effects, the better tolerated drugs are being searched for. The main objective of our study was to check whether activation of the atypical cannabinoid receptor GPR55 would normalize the changes in cystometric, cardiovascular and biochemical parameters in the hypertensive female Wistar-Kyoto rats presenting the symptoms of overactive bladder accompanied by inflammation and oxidative damage in the urinary tracts. A 14-day intra-arterial administration of O-1602 (0.25 mg/kg/day), a potent agonist of GRP55 receptors, was able to abolish the signs of detrusor overactivity, inflammation and oxidative damage in the urinary bladder of the spontaneously hypertensive animals. Moreover, it increased their heart rate, reduced the mean blood pressure, and normalized the levels of several proteins that play a significant role in the proper functioning of the urinary bladder (i.e., calcitonin gene related peptide, organic cation transporter 3, extracellular signal-regulated kinase 1/2, vesicular acetylcholine transporter, RhoA). Based on the outcomes of our experiments, the atypical cannabinoid receptor GPR55 has emerged as a potential drug target for the treatment of overactive bladder in female subjects. It could be particularly attractive in the cases in which this condition is accompanied with elevated blood pressure, though further studies on this subject are needed.


Subject(s)
Cannabidiol/analogs & derivatives , Hypertension/drug therapy , Receptors, G-Protein-Coupled/agonists , Urinary Bladder, Overactive/drug therapy , Urinary Bladder/drug effects , Animals , Aorta/drug effects , Aorta/physiology , Calcitonin Gene-Related Peptide/physiology , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , Extracellular Signal-Regulated MAP Kinases/physiology , Female , Hypertension/physiopathology , Octamer Transcription Factor-3/physiology , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Cannabinoid/physiology , Receptors, G-Protein-Coupled/physiology , Urinary Bladder/physiopathology , Urinary Bladder, Overactive/physiopathology , Vesicular Acetylcholine Transport Proteins/physiology
2.
Dev Biol ; 444 Suppl 1: S325-S336, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29792856

ABSTRACT

Although the basic schema of the body plan is similar among different species of amniotes (mammals, birds, and reptiles), the lung is an exception. Here, anatomy and physiology are considerably different, particularly between mammals and birds. In mammals, inhaled and exhaled airs mix in the airways, whereas in birds the inspired air flows unidirectionally without mixing with the expired air. This bird-specific respiration system is enabled by the complex tubular structures called parabronchi where gas exchange takes place, and also by the bellow-like air sacs appended to the main part of the lung. That the lung is predominantly governed by the parasympathetic nervous system has been shown mostly by physiological studies in mammals. However, how the parasympathetic nervous system in the lung is established during late development has largely been unexplored both in mammals and birds. In this study, by combining immunocytochemistry, the tissue-clearing CUBIC method, and ink-injection to airways, we have visualized the 3-D distribution patterns of parasympathetic nerves and ganglia in the lung at late developmental stages of mice and chickens. These patterns were further compared between these species, and three prominent similarities emerged: (1) parasympathetic postganglionic fibers and ganglia are widely distributed in the lung covering the proximal and distal portions, (2) the gas exchange units, alveoli in mice and parabronchi in chickens, are devoid of parasympathetic nerves, (3) parasympathetic nerves are in close association with smooth muscle cells, particularly at the base of the gas exchange units. These observations suggest that despite gross differences in anatomy, the basic mechanisms underlying parasympathetic control of smooth muscles and gas exchange might be conserved between mammals and birds.


Subject(s)
Lung/embryology , Lung/physiology , Parasympathetic Nervous System/physiology , Animals , Chick Embryo , Chickens , Ganglia/embryology , Mammals/physiology , Mice , Mice, Inbred ICR , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Parasympathetic Nervous System/embryology , Pulmonary Alveoli/embryology , Vesicular Acetylcholine Transport Proteins/metabolism , Vesicular Acetylcholine Transport Proteins/physiology
3.
Behav Brain Res ; 330: 127-132, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28461009

ABSTRACT

Depression is extremely harmful to modern society. Despite its complex spectrum of symptoms, previous studies have mostly focused on the monaminergic system in search of pharmacological targets. However, other neurotransmitter systems have also been linked to the pathophysiology of depression. In this study, we provide evidence for a role of the cholinergic system in depressive-like behavior of female mice. We evaluated mice knockdown for the vesicular acetylcholine transporter (VAChT KD mice), which have been previously shown to exhibit reduced cholinergic transmission. Animals were subjected to the tail suspension and marble burying tests, classical paradigms to assess depressive-like behaviors and to screen for novel antidepressant drugs. In addition, brain levels of serotonin and dopamine were measured by high performance liquid chromatography. We found that female homozygous VAChT KD mice spent less time immobile during tail suspension and buried less marbles, indicating a less depressive phenotype. These differences in behavior were reverted by central, but not peripheral, acetylcholinesterase inhibition. Moreover, female homozygous VAChT KD mice exhibited higher levels of dopamine and serotonin in the striatum, and increased dopamine in the hippocampus. Our study thus shows a connection between depressive-like behaviors and the cholinergic system, and that the latter interacts with the monoaminergic system.


Subject(s)
Depression/metabolism , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/metabolism , Acetylcholine/metabolism , Animals , Antidepressive Agents/pharmacology , Brain/metabolism , Corpus Striatum/metabolism , Depressive Disorder/physiopathology , Disease Models, Animal , Dopamine/metabolism , Dopamine/pharmacology , Female , Gene Knockdown Techniques , Hippocampus/metabolism , Mice , Motor Activity/drug effects , Neurotransmitter Agents/pharmacology , Serotonin/metabolism , Serotonin/pharmacology , Vesicular Acetylcholine Transport Proteins/physiology
4.
J Neurochem ; 140(5): 787-798, 2017 03.
Article in English | MEDLINE | ID: mdl-27889925

ABSTRACT

Postural instability and gait disturbances, common disabilities in the elderly and frequently present in Parkinson's disease (PD), have been suggested to be related to dysfunctional cholinergic signaling in the brainstem. We investigated how long-term loss of cholinergic signaling from mesopontine nuclei influence motor behaviors. We selectively eliminated the vesicular acetylcholine transporter (VAChT) in pedunculopontine and laterodorsal tegmental nuclei cholinergic neurons to generate mice with selective mesopontine cholinergic deficiency (VAChTEn1-Cre-flox/flox ). VAChTEn1-Cre-flox/flox mice did not show any gross health or neuromuscular abnormality on metabolic cages, wire-hang and grip-force tests. Young VAChTEn1-Cre-flox/flox mice (2-5 months-old) presented motor learning/coordination deficits on the rotarod; moved slower, and had smaller steps on the catwalk, but showed no difference in locomotor activity on the open field. Old VAChTEn1-Creflox/flox mice (13-16 months-old) showed more pronounced motor learning/balance deficits on the rotarod, and more pronounced balance deficits on the catwalk. Furthermore, old mutants moved faster than controls, but with similar step length. Additionally, old VAChT-deficient mice were hyperactive. These results suggest that dysfunction of cholinergic neurons from mesopontine nuclei, which is commonly seen in PD, has causal roles in motor functions. Prevention of mesopontine cholinergic failure may help to prevent/improve postural instability and falls in PD patients. Read the Editorial Highlight for this article on page 688.


Subject(s)
Gait Disorders, Neurologic/genetics , Neurons/physiology , Pedunculopontine Tegmental Nucleus/metabolism , Vesicular Acetylcholine Transport Proteins/genetics , Animals , Gait Disorders, Neurologic/psychology , Gene Deletion , Hand Strength , Learning Disabilities/genetics , Locomotion , Male , Mice , Motor Skills Disorders/genetics , Mutation/genetics , Parasympathetic Nervous System/cytology , Parasympathetic Nervous System/physiology , Pedunculopontine Tegmental Nucleus/cytology , Postural Balance , Psychomotor Performance , Tegmentum Mesencephali/cytology , Tegmentum Mesencephali/metabolism , Vesicular Acetylcholine Transport Proteins/physiology
5.
Skelet Muscle ; 6: 31, 2016.
Article in English | MEDLINE | ID: mdl-27713817

ABSTRACT

BACKGROUND: Cholinergic dysfunction occurs during aging and in a variety of diseases, including amyotrophic lateral sclerosis (ALS). However, it remains unknown whether changes in cholinergic transmission contributes to age- and disease-related degeneration of the motor system. Here we investigated the effect of moderately increasing levels of synaptic acetylcholine (ACh) on the neuromuscular junction (NMJ), muscle fibers, and motor neurons during development and aging and in a mouse model for amyotrophic lateral sclerosis (ALS). METHODS: Chat-ChR2-EYFP (VAChTHyp) mice containing multiple copies of the vesicular acetylcholine transporter (VAChT), mutant superoxide dismutase 1 (SOD1G93A), and Chat-IRES-Cre and tdTomato transgenic mice were used in this study. NMJs, muscle fibers, and α-motor neurons' somata and their axons were examined using a light microscope. Transcripts for select genes in muscles and spinal cords were assessed using real-time quantitative PCR. Motor function tests were carried out using an inverted wire mesh and a rotarod. Electrophysiological recordings were collected to examine miniature endplate potentials (MEPP) in muscles. RESULTS: We show that VAChT is elevated in the spinal cord and at NMJs of VAChTHyp mice. We also show that the amplitude of MEPPs is significantly higher in VAChTHyp muscles, indicating that more ACh is loaded into synaptic vesicles and released into the synaptic cleft at NMJs of VAChTHyp mice compared to control mice. While the development of NMJs was not affected in VAChTHyp mice, NMJs prematurely acquired age-related structural alterations in adult VAChTHyp mice. These structural changes at NMJs were accompanied by motor deficits in VAChTHyp mice. However, cellular features of muscle fibers and levels of molecules with critical functions at the NMJ and in muscle fibers were largely unchanged in VAChTHyp mice. In the SOD1G93A mouse model for ALS, increasing synaptic ACh accelerated degeneration of NMJs caused motor deficits and resulted in premature death specifically in male mice. CONCLUSIONS: The data presented in this manuscript demonstrate that increasing levels of ACh at the synaptic cleft promote degeneration of adult NMJs, contributing to age- and disease-related motor deficits. We thus propose that maintaining normal cholinergic signaling in muscles will slow degeneration of NMJs and attenuate loss of motor function caused by aging and neuromuscular diseases.


Subject(s)
Acetylcholine/metabolism , Aging , Amyotrophic Lateral Sclerosis/physiopathology , Neuromuscular Junction/physiology , Vesicular Acetylcholine Transport Proteins/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic , Miniature Postsynaptic Potentials , Motor Neurons/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Neuromuscular Junction/metabolism , RNA, Messenger/metabolism , Spinal Cord/metabolism , Survival Analysis , Vesicular Acetylcholine Transport Proteins/physiology
6.
PLoS One ; 10(7): e0133989, 2015.
Article in English | MEDLINE | ID: mdl-26226617

ABSTRACT

BACKGROUND: Despite rigorous characterization of the role of acetylcholine in retinal development, long-term effects of its absence as a neurotransmitter are unknown. One of the unanswered questions is how acetylcholine contributes to the functional capacity of mature retinal circuits. The current study investigates the effects of disrupting cholinergic signalling in mice, through deletion of vesicular acetylcholine transporter (VAChT) in the developing retina, pigmented epithelium, optic nerve and optic stalk, on electrophysiology and structure of the mature retina. METHODS & RESULTS: A combination of electroretinography, optical coherence tomography imaging and histological evaluation assessed retinal integrity in mice bearing retina- targeted (embryonic day 12.5) deletion of VAChT (VAChTSix3-Cre-flox/flox) and littermate controls at 5 and 12 months of age. VAChTSix3-Cre-flox/flox mice did not show any gross changes in nuclear layer cellularity or synaptic layer thickness. However, VAChTSix3-Cre-flox/flox mice showed reduced electrophysiological response of the retina to light stimulus under scotopic conditions at 5 and 12 months of age, including reduced a-wave, b-wave, and oscillatory potential (OP) amplitudes and decreased OP peak power and total energy. Reduced a-wave amplitude was proportional to the reduction in b-wave amplitude and not associated with altered a-wave 10%-90% rise time or inner and outer segment thicknesses. SIGNIFICANCE: This study used a novel genetic model in the first examination of function and structure of the mature mouse retina with disruption of cholinergic signalling. Reduced amplitude across the electroretinogram wave form does not suggest dysfunction in specific retinal cell types and could reflect underlying changes in the retinal and/or extraretinal microenvironment. Our findings suggest that release of acetylcholine by VAChT is essential for the normal electrophysiological response of the mature mouse retina.


Subject(s)
Acetylcholine/physiology , Neurotransmitter Agents/physiology , Retina/physiology , Vesicular Acetylcholine Transport Proteins/physiology , Animals , Blotting, Western , Electroretinography , Gene Deletion , Male , Mice , Mice, Knockout , Optic Nerve/physiology , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/physiology , Tomography, Optical Coherence , Vesicular Acetylcholine Transport Proteins/genetics
7.
Neurobiol Aging ; 36(5): 1881-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25813545

ABSTRACT

Aging is marked by progressive impairments in the process of adult neurogenesis and spatial memory performance. The underlying mechanisms for these impairments have not been fully established; however, they may coincide with decline of cholinergic signaling in the hippocampus. This study investigates whether augmenting cholinergic neurotransmission, by enhancing the expression of the vesicular acetylcholine transporter (VAChT), influences the age-related decline in the development of newborn hippocampal cells and spatial memory. We found that enhanced VAChT expression in the hippocampus of mice contributes to lifelong increases in the dendritic complexity of newborn neurons. Furthermore, enhanced VAChT expression improved memory acquisition through an increased use of spatially precise search strategies in the Morris water maze through the course of the aging process. These data suggest that VAChT overexpression contributes to increases in dendritic complexity and improved spatial memory during aging.


Subject(s)
Dendrites/physiology , Hippocampus/cytology , Spatial Memory , Vesicular Acetylcholine Transport Proteins/physiology , Aging , Animals , Cells, Cultured , Cholinergic Neurons/physiology , Gene Expression , Hippocampus/physiology , Hippocampus/physiopathology , Male , Mice, Inbred C57BL , Neurogenesis , Spatial Memory/physiology , Synaptic Transmission , Vesicular Acetylcholine Transport Proteins/genetics
8.
PLoS One ; 10(3): e0120441, 2015.
Article in English | MEDLINE | ID: mdl-25816137

ABSTRACT

Acetylcholine (ACh) plays a crucial role in physiological responses of both the central and the peripheral nervous system. Moreover, ACh was described as an anti-inflammatory mediator involved in the suppression of exacerbated innate response and cytokine release in various organs. However, the specific contributions of endogenous release ACh for inflammatory responses in the lung are not well understood. To address this question we have used mice with reduced levels of the vesicular acetylcholine transporter (VAChT), a protein required for ACh storage in secretory vesicles. VAChT deficiency induced airway inflammation with enhanced TNF-α and IL-4 content, but not IL-6, IL-13 and IL-10 quantified by ELISA. Mice with decreased levels of VAChT presented increased collagen and elastic fibers deposition in airway walls which was consistent with an increase in inflammatory cells positive to MMP-9 and TIMP-1 in the lung. In vivo lung function evaluation showed airway hyperresponsiveness to methacholine in mutant mice. The expression of nuclear factor-kappa B (p65-NF-kB) in lung of VAChT-deficient mice were higher than in wild-type mice, whereas a decreased expression of janus-kinase 2 (JAK2) was observed in the lung of mutant animals. Our findings show the first evidence that cholinergic deficiency impaired lung function and produce local inflammation. Our data supports the notion that cholinergic system modulates airway inflammation by modulation of JAK2 and NF-kB pathway. We proposed that intact cholinergic pathway is necessary to maintain the lung homeostasis.


Subject(s)
Edema/physiopathology , Lung/pathology , Pneumonia/physiopathology , Vesicular Acetylcholine Transport Proteins/physiology , Acetylcholine/genetics , Acetylcholine/metabolism , Animals , Blotting, Western , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Edema/etiology , Immunoenzyme Techniques , Lung/metabolism , Male , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Pneumonia/etiology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
J Neurosci ; 33(25): 10427-38, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23785154

ABSTRACT

Acetylcholine (ACh) is an important neuromodulator in the nervous system implicated in many forms of cognitive and motor processing. Recent studies have used bacterial artificial chromosome (BAC) transgenic mice expressing channelrhodopsin-2 (ChR2) protein under the control of the choline acetyltransferase (ChAT) promoter (ChAT-ChR2-EYFP) to dissect cholinergic circuit connectivity and function using optogenetic approaches. We report that a mouse line used for this purpose also carries several copies of the vesicular acetylcholine transporter gene (VAChT), which leads to overexpression of functional VAChT and consequently increased cholinergic tone. We demonstrate that these mice have marked improvement in motor endurance. However, they also present severe cognitive deficits, including attention deficits and dysfunction in working memory and spatial memory. These results suggest that increased VAChT expression may disrupt critical steps in information processing. Our studies demonstrate that ChAT-ChR2-EYFP mice show altered cholinergic tone that fundamentally differentiates them from wild-type mice.


Subject(s)
Attention/physiology , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Cognition/physiology , Physical Endurance/genetics , Physical Endurance/physiology , Animals , Anxiety/psychology , Blotting, Western , Channelrhodopsins , Fluorescent Antibody Technique , Glucose Tolerance Test , Hand Strength/physiology , Hindlimb Suspension , Maze Learning/physiology , Metabolism/genetics , Metabolism/physiology , Mice , Mice, Transgenic , Parasympathetic Nervous System/physiology , Polymerase Chain Reaction , Postural Balance/physiology , Psychomotor Performance/physiology , Reaction Time/physiology , Swimming/physiology , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/physiology
10.
J Neurosci ; 33(26): 10667-75, 2013 Jun 26.
Article in English | MEDLINE | ID: mdl-23804090

ABSTRACT

The embryonic sympathetic nervous system consists of predominantly noradrenergic neurons and a very small population of cholinergic neurons. Postnatal development further allows target-dependent switch of a subset of noradrenergic neurons into cholinergic phenotype. How embryonic cholinergic neurons are specified at the prenatal stages remains largely unknown. In this study, we found that the expression of transcription factor Tlx3 was progressively restricted to a small population of embryonic sympathetic neurons in mice. Immunostaining for vesicular acetylcholine transporter (VAChT) showed that Tlx3 was highly expressed in cholinergic neurons at the late embryonic stage E18.5. Deletion of Tlx3 resulted in the loss of Vacht expression at E18.5 but not E12.5. By contrast, Tlx3 was required for expression of the cholinergic peptide vasoactive intestinal polypeptide (VIP), and somatostatin (SOM) at both E12.5 and E18.5. Furthermore, we found that, at E18.5 these putative cholinergic neurons expressed glial cell line-derived neurotrophic factor family coreceptor Ret but not tyrosine hydroxylase (Ret(+)/TH(-)). Deletion of Tlx3 also resulted in disappearance of high-level Ret expression. Last, unlike Tlx3, Ret was required for the expression of VIP and SOM at E18.5 but not E12.5. Together, these results indicate that transcription factor Tlx3 is required for the acquisition of cholinergic phenotype at the late embryonic stage as well as the expression and maintenance of cholinergic peptides VIP and SOM throughout prenatal development of mouse sympathetic neurons.


Subject(s)
Homeodomain Proteins/physiology , Neurons/physiology , Neuropeptides/physiology , Neurotransmitter Agents/physiology , Parasympathetic Nervous System/physiology , Sympathetic Nervous System/physiology , Animals , Cell Count , Female , Fetus , Gene Deletion , Immunohistochemistry , Mice , Mice, Knockout , Mutation/physiology , Pregnancy , Proto-Oncogene Proteins c-ret/biosynthesis , Proto-Oncogene Proteins c-ret/genetics , Somatostatin/genetics , Somatostatin/physiology , Stellate Ganglion/cytology , Stellate Ganglion/growth & development , Sympathetic Nervous System/cytology , Sympathetic Nervous System/embryology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/physiology , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/physiology , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/physiology
11.
Mol Aspects Med ; 34(2-3): 360-72, 2013.
Article in English | MEDLINE | ID: mdl-23506877

ABSTRACT

The exocytotic release of neurotransmitters requires active transport into synaptic vesicles and other types of secretory vesicles. Members of the SLC18 family perform this function for acetylcholine (SLC18A3, the vesicular acetylcholine transporter or VAChT) and monoamines such as dopamine and serotonin (SLC18A1 and 2, the vesicular monoamine transporters VMAT1 and 2, respectively). To date, no specific diseases have been attributed to a mutation in an SLC18 family member; however, polymorphisms in SLC18A1 and SLC18A2 may confer risk for some neuropsychiatric disorders. Additional members of this family include SLC18A4, expressed in insects, and SLC18B1, the function of which is not known. SLC18 is part of the Drug:H(+) Antiporter-1 Family (DHA1, TCID 2.A.1.2) within the Major Facilitator Superfamily (MFS, TCID 2.A.1).


Subject(s)
Gene Expression Regulation/physiology , Models, Molecular , Multigene Family/genetics , Nervous System Diseases/metabolism , Protein Conformation , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/physiology , Animals , Biogenic Monoamines/metabolism , Biological Transport/physiology , Caenorhabditis elegans , Drosophila , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Mice , Models, Biological , Mutation/genetics , Phosphorylation , Phylogeny , Vesicular Acetylcholine Transport Proteins/biosynthesis , Vesicular Acetylcholine Transport Proteins/metabolism , Vesicular Monoamine Transport Proteins/metabolism
12.
Nat Commun ; 3: 1154, 2012.
Article in English | MEDLINE | ID: mdl-23093191

ABSTRACT

The molecular mechanism responsible for capturing, sorting and retrieving vesicle membrane proteins following triggered exocytosis is not understood. Here we image the post-fusion release and then capture of a vesicle membrane protein, the vesicular acetylcholine transporter, from single vesicles in living neuroendocrine cells. We combine these measurements with super-resolution interferometric photo-activation localization microscopy and electron microscopy, and modelling to map the nanometer-scale topography and architecture of the structures responsible for the transporter's capture following exocytosis. We show that after exocytosis, the transporter rapidly diffuses into the plasma membrane, but most travels only a short distance before it is locally captured over a dense network of membrane-resident clathrin-coated structures. We propose that the extreme density of these structures acts as a short-range diffusion trap. They quickly sequester diffusing vesicle material and limit its spread across the membrane. This system could provide a means for clathrin-mediated endocytosis to quickly recycle vesicle proteins in highly excitable cells.


Subject(s)
Membrane Fusion/physiology , Membrane Proteins/physiology , Vesicular Acetylcholine Transport Proteins/physiology , Animals , Cell Membrane/physiology , Cell Membrane/ultrastructure , Clathrin/physiology , Clathrin/ultrastructure , Endocytosis/physiology , Exocytosis/physiology , Membrane Proteins/ultrastructure , Microscopy, Electron , Microscopy, Interference/methods , PC12 Cells/physiology , Rats , Synaptic Vesicles/physiology , Synaptic Vesicles/ultrastructure , Vesicular Acetylcholine Transport Proteins/ultrastructure
13.
Eur J Neurosci ; 36(5): 2679-88, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22708650

ABSTRACT

Cholinergic input modulates excitability of motoneurons and plays an important role in the control of locomotion in both intact and spinalized animals. However, spinal cord transection in adult rats affects cholinergic innervation of only some hindlimb motoneurons, suggesting that specificity of this response is related to functional differences between motoneurons. Our aim was therefore to compare cholinergic input to motoneurons innervating the soleus (Sol) and tibialis anterior (TA) motoneurons following spinal cord transection at a low-thoracic level. The second aim was to investigate whether deficits in cholinergic input to these motoneurons could be modified by locomotor training. The Sol and TA motoneurons were identified by retrograde labelling with fluorescent dyes injected intramuscularly. Cholinergic terminals were detected using anti-vesicular acetylcholine transporter (VAChT) antibody. Overall innervation of motoneurons was evaluated with anti-synaptophysin antibody. After spinalization we found a decrease in the number of VAChT-positive boutons apposing perikarya of the Sol (to 49%) but not TA motoneurons. Locomotor training, resulting in moderate functional improvement, partly reduced the deficit in cholinergic innervation of Sol motoneurons by increasing the number of VAChT-positive boutons. However, the optical density of VAChT-positive boutons terminating on various motoneurons, which decreased after spinalization, continued to decrease despite the training, suggesting an impairment of acetylcholine availability in the terminals. Different effects of spinal cord transection on cholinergic innervation of motoneurons controlling the ankle extensor and flexor muscles point to different functional states of these muscles in paraplegia as a possible source of activity-dependent signaling regulating cholinergic input to the motoneurons.


Subject(s)
Cholinergic Neurons/physiology , Locomotion/physiology , Motor Neurons/physiology , Spinal Cord/physiology , Acetylcholine/physiology , Animals , Male , Muscle, Skeletal/innervation , Presynaptic Terminals/physiology , Rats , Rats, Wistar , Spinal Cord/surgery , Tarsus, Animal/innervation , Vesicular Acetylcholine Transport Proteins/physiology
14.
Nat Med ; 17(7): 888-92, 2011 Jun 19.
Article in English | MEDLINE | ID: mdl-21685896

ABSTRACT

Acetylcholine is a neurotransmitter that has a major role in the function of the insulin-secreting pancreatic beta cell. Parasympathetic innervation of the endocrine pancreas, the islets of Langerhans, has been shown to provide cholinergic input to the beta cell in several species, but the role of autonomic innervation in human beta cell function is at present unclear. Here we show that, in contrast to the case in mouse islets, cholinergic innervation of human islets is sparse. Instead, we find that the alpha cells of human islets provide paracrine cholinergic input to surrounding endocrine cells. Human alpha cells express the vesicular acetylcholine transporter and release acetylcholine when stimulated with kainate or a lowering in glucose concentration. Acetylcholine secretion by alpha cells in turn sensitizes the beta cell response to increases in glucose concentration. Our results demonstrate that in human islets acetylcholine is a paracrine signal that primes the beta cell to respond optimally to subsequent increases in glucose concentration. Cholinergic signaling within islets represents a potential therapeutic target in diabetes, highlighting the relevance of this advance to future drug development.


Subject(s)
Acetylcholine/metabolism , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Acetylcholine/physiology , Alkenes/pharmacology , Animals , Dose-Response Relationship, Drug , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/physiology , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mice , Physostigmine/pharmacology , Piperidines/pharmacology , Receptors, Cholinergic/physiology , Secretory Vesicles/physiology , Signal Transduction/physiology , Vesicular Acetylcholine Transport Proteins/physiology
15.
Neuroscience ; 186: 1-12, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21557989

ABSTRACT

We investigated the role of the vesicular acetylcholine transporter in the mechanism of non-quantal (non-vesicular) secretion of neurotransmitter in the neuromuscular synapse of the rat diaphragm muscle. Non-quantal secretion was estimated electrophysiologically by the amplitude of end-plate hyperpolarization after inhibition of cholinesterase and nicotinic receptors (H-effect) or measured by the optical detection of acetylcholine in the bathing solution. It was shown that 1 mM methyl-ß-cyclodextrin (MCD) reduced both endocytosis and, to much lesser extent, exocytosis of synaptic vesicles (SV) thereby increasing non-quantal secretion of acetylcholine with a concurrent decrease in axoplasm pH. During high-frequency stimulation of the motor nerve, that substantially increases vesicles exocytosis, the non-quantal secretion was further enhanced if the endocytosis of SV was blocked by MCD. In contrast, non-quantal secretion of acetylcholine did not increase when the MCD-treated neuromuscular preparations were superfused with either vesamicol, an inhibitor of vesicular transporter of acetylcholine, or sodium propionate, which decreases intracellular pH. These results suggest that the proton-dependent, vesamicol-sensitive vesicular transporters of acetylcholine, which become inserted into the presynaptic membrane during SV exocytosis and removed during endocytotic recycling of SV, play the major role in the process of non-quantal secretion of neurotransmitter.


Subject(s)
Acetylcholine/metabolism , Endocytosis/physiology , Neuromuscular Junction/metabolism , Presynaptic Terminals/metabolism , Vesicular Acetylcholine Transport Proteins/physiology , beta-Cyclodextrins/pharmacology , Animals , Endocytosis/drug effects , Male , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neuromuscular Junction/drug effects , Presynaptic Terminals/drug effects , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Up-Regulation/drug effects , Up-Regulation/physiology , Vesicular Acetylcholine Transport Proteins/antagonists & inhibitors
16.
Arch Oral Biol ; 54(10): 909-16, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19647228

ABSTRACT

The parasympathetic vasodilatory fibres are known to innervate vessels in a rat masseter muscle via both cholinergic and non-cholinergic mechanisms. However, the non-cholinergic mechanisms are still unclear. Recently, vasoactive intestinal polypeptide (VIP) was convincingly shown to be involved in the parasympathetic vasodilatation in orofacial areas, such as submandibular glands and lower lip. However, very little is known about the rat masseter muscle. The present study was designed in the rat masseter muscle to assess (1) whether the parasympathetic nerve innervating vessels have VIP immunoreactivities, (2) whether intravenous administration of VIP induces the vasodilatation, and (3) effects of selective VIP receptor antagonist ([4Cl-d-Phe(6), Leu(17)] VIP) in the presence or absence of atropine on the parasympathetic vasodilatation. The VIP immunoreactivities were found at two sites of the parasympathetic otic ganglion and nerve fibres located around vessels. The intravenous administration of VIP induced the vasodilatation, and [4Cl-d-Phe(6), Leu(17)] VIP markedly decreased the vasodilatation evoked by VIP administration. The parasympathetic vasodilatation was not inhibited by [4Cl-d-Phe(6), Leu(17)] VIP. However, treatment with [4Cl-d-Phe(6), Leu(17)] VIP markedly decreased the parasympathetic vasodilatation when [4Cl-d-Phe(6), Leu(17)] VIP was administered together with atropine. These results suggest that (1) VIP exists in the postganglionic parasympathetic nerve innervating the vessels in the masseter muscle, (2) the intravenous administration of VIP induces the vasodilatation in the masseter muscle, and (3) VIP may be involved in the parasympathetic vasodilatation in the masseter muscle when muscarinic cholinergic receptors are deactivated by either atropine or the suppression of the ACh release.


Subject(s)
Masseter Muscle/blood supply , Masseter Muscle/innervation , Parasympathetic Fibers, Postganglionic/physiology , Vasoactive Intestinal Peptide/physiology , Vasodilation/physiology , Animals , Atropine/pharmacology , Fluorescent Antibody Technique , Ganglia, Parasympathetic/physiology , Laser-Doppler Flowmetry , Male , Parasympathetic Fibers, Postganglionic/drug effects , Parasympatholytics/pharmacology , Rats , Rats, Wistar , Receptors, Vasoactive Intestinal Peptide/antagonists & inhibitors , Vasoactive Intestinal Peptide/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Vesicular Acetylcholine Transport Proteins/physiology
17.
Brain Res ; 1246: 167-77, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-18848922

ABSTRACT

Traumatic brain injury (TBI) is one of the leading causes of death and disability. Cognitive deficits are believed to be connected with impairments of the cholinergic system. The present study was conducted to evaluate the cholinergic system in a model of focal brain injury with special attention to the time course of posttraumatic events in critical brain regions. Three groups of male Sprague-Dawley rats (post-TBI survival time: 2 h, 24 h and 72 h) were subjected to sham-operation (control) or controlled cortical impact injury. Receptor densities were determined on frozen ipsilateral sagittal brain sections with [(3)H]epibatidine (nicotinic acetylcholine receptors) and [(3)H]QNB (muscarinic acetylcholine receptors). The density of the vesicular acetylcholine transporter (vAChT) was evaluated with (-)[(3)H]vesamicol. Compared to control, vAChT was lowered (up to 50%) at each time point after trauma, with reductions in olfactory tubercle, basal forebrain, motor cortex, putamen, thalamic and hypothalamic areas and the gigantocellular reticular nucleus. Time-dependent reductions of about 20% of nAChR-density in the thalamus, hypothalamus, olfactory tubercle, gigantocellular reticular nucleus and motor cortex were observed post-TBI at 24 and 72 h. The same brain regions showed reductions of mAChR at 24 and 72 h after trauma with additional decreases in the corpus callosum, basal forebrain and anterior olfactory nucleus. In conclusion, cholinergic markers showed significant time-dependent impairments after TBI. Considering the role of the cholinergic system for cognitive processes in the brain, it seems likely that these impairments contribute to clinically relevant cognitive deficits.


Subject(s)
Brain Injuries/physiopathology , Receptors, Cholinergic/physiology , Animals , Autoradiography , Brain/pathology , Brain Injuries/pathology , Choline/metabolism , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Receptors, Muscarinic/physiology , Receptors, Nicotinic/physiology , Time Factors , Vesicular Acetylcholine Transport Proteins/physiology
20.
J Neurosci ; 27(6): 1411-21, 2007 Feb 07.
Article in English | MEDLINE | ID: mdl-17287516

ABSTRACT

In this study, we address why metabotropic and ionotropic cholinergic signaling pathways are used to facilitate motor behaviors. We demonstrate that a G alpha(q)-coupled muscarinic acetylcholine receptor (mAChR) signaling pathway enhances nicotinic acetylcholine receptor (nAChR) signaling to facilitate the insertion of the Caenorhabditis elegans male copulatory spicules into the hermaphrodite during mating. Previous studies showed that ACh (acetylcholine) activates nAChRs on the spicule protractor muscles to induce the attached spicules to extend from the tail. Using the mAChR agonist Oxo M (oxotremorine M), we identified a GAR-3(mAChR)-G alpha(q) pathway that promotes protractor muscle contraction by upregulating nAChR signaling before mating. GAR-3(mAChR) is expressed in the protractor muscles and in the spicule-associated SPC and PCB cholinergic neurons. However, ablation of these neurons or impairing cholinergic transmission reduces drug-induced spicule protraction, suggesting that drug-stimulated neurons directly activate muscle contraction. Behavioral analysis of gar-3 mutants indicates that, in wild-type males, GAR-3(mAChR) expression in the SPC and PCB neurons is required for the male to sustain rhythmic spicule muscle contractions during attempts to breach the vulva. We propose that the GAR-3(mAChR)/G alpha(q) pathway sensitizes the spicule neurons and muscles before and during mating so that the male can respond to hermaphrodite vulva efficiently.


Subject(s)
Acetylcholine/physiology , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Receptors, Muscarinic/physiology , Receptors, Nicotinic/physiology , Sexual Behavior, Animal/physiology , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/drug effects , Caenorhabditis elegans Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/physiology , Disorders of Sex Development , GTP-Binding Protein alpha Subunits, Gq-G11/deficiency , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Genitalia/innervation , Genitalia/physiology , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/physiology , Levamisole/pharmacology , Muscarinic Agonists/pharmacology , Muscle Contraction/physiology , Mutation, Missense , Neurons/physiology , Oxotremorine/pharmacology , Periodicity , Phospholipase C beta , Potassium Channels/deficiency , Potassium Channels/genetics , Potassium Channels/physiology , Receptors, Muscarinic/deficiency , Receptors, Muscarinic/genetics , Recombinant Fusion Proteins/physiology , Ryanodine Receptor Calcium Release Channel/drug effects , Signal Transduction/physiology , Syntaxin 1/deficiency , Syntaxin 1/genetics , Syntaxin 1/physiology , Type C Phospholipases/deficiency , Type C Phospholipases/genetics , Type C Phospholipases/physiology , Vesicular Acetylcholine Transport Proteins/deficiency , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...