Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Histochem Cell Biol ; 148(2): 189-206, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28337539

ABSTRACT

Homer1, a scaffolding protein of the postsynaptic density (PSD), enriched at excitatory synapses is known to anchor and modulate group I metabotropic glutamate receptors (mGluRs) and different channel- and receptor-proteins. Homer proteins are expressed in neurons of different brain regions, but also in non-neuronal tissues like skeletal muscle. Occurrence and location of Homer1 and mGluR5 in myenteric plexus and neuromuscular junctions (NMJ) of rat esophagus have yet not been characterized. We located Homer1 and mGluR5 immunoreactivity (-iry) in rat esophagus and focused on myenteric neurons, intraganglionic laminar endings (IGLEs) and NMJs, using double- and triple-label immunohistochemistry and confocal laser scanning microscopy. Homer1-iry was found in a subpopulation of vesicular glutamate transporter 2 (VGLUT2) positive IGLEs and cholinergic varicosities within myenteric ganglia, but neither in nitrergic nor cholinergic myenteric neuronal cell bodies. Homer1-iry was detected in 63% of esophageal and, for comparison, in 35% of sternomastoid NMJs. Besides the location in the PSD, Homer1-iry colocalized with cholinergic markers, indicating a presynaptic location in coarse VAChT/CGRP/NF200- immunoreactive (-ir) terminals of nucleus ambiguus neurons supplying striated esophageal muscle. mGluR5-iry was found in subpopulations of myenteric neuronal cell bodies, VGLUT2-ir IGLEs and cholinergic varicosities within the myenteric neuropil and NMJs of esophagus and sternomastoid muscles. Thus, Homer1 may anchor mGluR5 at presynaptic sites of cholinergic boutons at esophageal motor endplates, in a small subpopulation of VGLUT2-ir IGLEs and cholinergic varicosities within myenteric ganglia possibly modulating Ca2+-currents and neurotransmitter release.


Subject(s)
Esophagus/chemistry , Homer Scaffolding Proteins/analysis , Myenteric Plexus/chemistry , Neuromuscular Junction/chemistry , Animals , Esophagus/cytology , Esophagus/metabolism , Guinea Pigs , Homer Scaffolding Proteins/metabolism , Immunohistochemistry , Microscopy, Confocal , Myenteric Plexus/cytology , Myenteric Plexus/metabolism , Neuromuscular Junction/cytology , Neuromuscular Junction/metabolism , Rabbits , Rats , Rats, Wistar
2.
Neurogastroenterol Motil ; 36(1): e14693, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37882149

ABSTRACT

BACKGROUND: Accurately reporting the identity and representation of enteric nervous system (ENS) neuronal subtypes along the length of the gastrointestinal (GI) tract is critical to advancing our understanding of ENS control of GI function. Reports of varying proportions of subtype marker expression have employed different dissection techniques to achieve wholemount muscularis preparations of myenteric plexus. In this study, we asked whether differences in GI dissection methods could introduce variability into the quantification of marker expression. METHODS: We compared three commonly used methods of ENS wholemount dissection: two flat-sheet preparations that differed in the order of microdissection and fixation and a third rod-mounted peeling technique. We also tested a reversed orientation variation of flat-sheet peeling, two step-by-step variations of the rod peeling technique, and whole-gut fixation as a tube. We assessed marker expression using immunohistochemistry, genetic reporter lines, confocal microscopy, and automated image analysis. KEY RESULTS AND CONCLUSIONS: We found no significant differences between the two flat-sheet preparation methods in the expression of calretinin or neuronal nitric oxide synthase (nNOS) as a proportion of total neurons in ileum myenteric plexus. However, the rod-mounted peeling method resulted in decreased proportion of neurons labeled for both calretinin and nNOS. This method also resulted in decreased transgenic reporter fluorescent protein (tdTomato) for substance P in distal colon and choline acetyltransferase (ChAT) in both ileum and distal colon. These results suggest that labeling among some markers, both native protein and transgenic fluorescent reporters, is decreased by the rod-mounted mechanical method of peeling. The step-by-step variations of this method point to mechanical manipulation of the tissue as the likely cause of decreased labeling. Our study thereby demonstrates a critical variability in wholemount muscularis dissection methods.


Subject(s)
Enteric Nervous System , Myenteric Plexus , Mice , Animals , Myenteric Plexus/chemistry , Calbindin 2/metabolism , Enteric Nervous System/metabolism , Neurons/metabolism , Colon
3.
Am J Surg Pathol ; 48(7): 803-812, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38726836

ABSTRACT

Isolated hypoganglionosis (IHG) is histologically characterized by small numbers of myenteric ganglion cells and small myenteric ganglia; however, no numerical diagnostic criteria for IHG have been established. Therefore, this study aimed to develop quantitative pathologic criteria for IHG. We evaluated 160 resected intestinal tissue specimens from 29 pediatric autopsies and 10 IHG cases. These specimens were obtained from the jejunum, ileum, ascending colon, transverse colon, and rectum. Morphologic features of the myenteric ganglion cells and myenteric ganglia were quantified and analyzed in digitized HuC/HuD-immunostained and CD56-immunostained sections, respectively. Quantitative criteria were developed with a scoring system that used parameters with the area under the receiver operating characteristic curve (AUC) values >0.7 and sensitivity and specificity exceeding 70%. The selected parameters were the number of myenteric ganglion cells per cm and the number of myenteric ganglia with an area >2500 µm 2 per cm. The score for each parameter ranged from -1 to 2, and the total score of the scoring system ranged from -2 to 4. With a cutoff value of ≥2 (AUC, 0.98; 95% CI: 0.96-1.00), the scoring system had a sensitivity of 96% (95% CI: 0.82-1.00) and a specificity of 99% (95% CI: 0.95-1.00). We devised a novel pathologic criterion based on the quantification of the number of myenteric ganglion cells and ganglia. Furthermore, this criterion showed high diagnostic accuracy and could lead to a definitive diagnosis of IHG in clinical practice.


Subject(s)
Myenteric Plexus , Humans , Male , Female , Myenteric Plexus/pathology , Myenteric Plexus/chemistry , Infant , Child, Preschool , Child , Predictive Value of Tests , Immunohistochemistry , Infant, Newborn , Autopsy , Biomarkers/analysis , ROC Curve , Adolescent , Cell Count , Area Under Curve
4.
Colorectal Dis ; 15(10): e592-8, 2013.
Article in English | MEDLINE | ID: mdl-23810202

ABSTRACT

AIM: Megacolon, chronic dilation of a colonic segment,is accompanied by extensive myenteric neuron loss. However, this fails to explain unequivocally the formation of megacolon. We aimed to study further enteric structures that are directly or indirectly involved in colonic motility. METHOD: From surgically removed megacolon segments of seven Chagasic patients, three sets of cryosections from oral, megacolonic and anal zones were immunohistochemically quadruple-stained for smooth-muscle actin (SMA), synaptophysin (SYN, for nerve fibres), S100 (glia) and c-Kit (interstitial cells of Cajal, ICCs). Values of area measurements were related to the appropriate muscle layer areas and these proportions were compared with those of seven non-Chagasic control patients. RESULTS: Whereas nerve and glia profile proportions did not mirror unequivocally the changes of Chagasic colon calibre (nondilation/dilation/nondilation), the proportions of SMA (i.e. muscle tissue density) and c-Kit (i.e. ICC density) did so: they decreased from the oral to the megacolonic segment but increased to the anal zones (muscle tissue density: control 68.3%, oral 54.3%, mega 42.1%, anal 47.6%; ICC-density: control 1.8%, oral 1.1%, mega 0.4, anal 0.8%). CONCLUSION: Of the parameters evaluated, muscle tissue and ICC densities may be involved in the formation of Chagasic megacolon, although the mechanism of destruction cannot be deduced.


Subject(s)
Chagas Disease/complications , Colon/chemistry , Interstitial Cells of Cajal/chemistry , Megacolon/pathology , Muscle, Smooth/chemistry , Actins/analysis , Aged , Case-Control Studies , Colon/innervation , Female , Humans , Interstitial Cells of Cajal/pathology , Male , Megacolon/parasitology , Middle Aged , Muscle, Smooth/pathology , Myenteric Plexus/chemistry , Neuroglia/chemistry , Proto-Oncogene Proteins c-kit/analysis , S100 Proteins/analysis , Synaptophysin/analysis
5.
J Anat ; 221(4): 303-10, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22803609

ABSTRACT

HCN2 channels are involved in the spontaneous rhythmic activities of some CNS neurons and act by generating I(f) current. The gastrointestinal (GI) tract is known to be capable of spontaneous rhythmic activity; however, the possible role of HCN2 channels in this organ has not yet been elucidated. This study investigated the distribution of HCN2-positive cells in the mouse GI tract using immunohistochemistry. To identify the nature of these HCN2 cells, anti-ChAT and anti-Kit antibodies were used to co-label neurons and the interstitial cells of Cajal (ICCs), respectively. Additionally, differences in the distribution of HCN2-positive cells within the GI tract were also analyzed. Our results showed that HCN2 channels were mainly located within the myenteric neurons of the enteric nervous system in the GI tract. Double-staining revealed that HCN2-positive neurons were labeled by ChAT, indicating that these HCN2-positive cells are also cholinergic neurons. Although the HCN2-positive cells were not stained by the anti-Kit antibody, their processes were in close proximity to ICCs around the myenteric plexus region. Moreover, several differences in the distribution of HCN2 in the stomach, small intestine and colon were partly consistent with the regional differences in the spontaneous rhythmic activities of these organs. Basing on the role HCN2, we suggested that HCN2 channels facilitate the release of Ach from cholinergic neurons to affect the GI peristalsis by acting on M receptors on the ICCs. However, the HCN2 channels are not directly involved in spontaneous slow-wave initiation by ICCs.


Subject(s)
Gastrointestinal Tract/cytology , Ion Channels/analysis , Myenteric Plexus/chemistry , Animals , Enteric Nervous System , Gastrointestinal Tract/anatomy & histology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Immunohistochemistry , Interstitial Cells of Cajal/chemistry , Interstitial Cells of Cajal/cytology , Ion Channels/physiology , Male , Mice/anatomy & histology , Mice, Inbred BALB C , Myenteric Plexus/cytology , Neurons/chemistry , Potassium Channels
6.
Nat Neurosci ; 24(1): 34-46, 2021 01.
Article in English | MEDLINE | ID: mdl-33288908

ABSTRACT

Autonomous regulation of the intestine requires the combined activity of functionally distinct neurons of the enteric nervous system (ENS). However, the variety of enteric neuron types and how they emerge during development remain largely unknown. Here, we define a molecular taxonomy of 12 enteric neuron classes within the myenteric plexus of the mouse small intestine using single-cell RNA sequencing. We present cell-cell communication features and histochemical markers for motor neurons, sensory neurons and interneurons, together with transgenic tools for class-specific targeting. Transcriptome analysis of the embryonic ENS uncovers a novel principle of neuronal diversification, where two neuron classes arise through a binary neurogenic branching and all other identities emerge through subsequent postmitotic differentiation. We identify generic and class-specific transcriptional regulators and functionally connect Pbx3 to a postmitotic fate transition. Our results offer a conceptual and molecular resource for dissecting ENS circuits and predicting key regulators for directed differentiation of distinct enteric neuron classes.


Subject(s)
Myenteric Plexus/chemistry , Neurons/chemistry , RNA/chemistry , RNA/genetics , Single-Cell Analysis , Animals , Cell Communication , Enteric Nervous System/physiology , Homeodomain Proteins/genetics , Interneurons/physiology , Mice , Mice, Inbred C57BL , Motor Neurons/physiology , Myenteric Plexus/cytology , Neurons/classification , Neurons/ultrastructure , Proto-Oncogene Proteins/genetics , Sensory Receptor Cells/physiology , Sequence Analysis, RNA , Transcriptome
7.
Methods Mol Biol ; 1952: 117-125, 2019.
Article in English | MEDLINE | ID: mdl-30825171

ABSTRACT

The cellular components of the enteric nervous system (ENS), namely enteric neurons and glia, display plasticity and respond to environmental cues deriving from growth factors, extracellular matrix (ECM) molecules, and cell-surface molecules, both in physiological and pathological conditions. ECM, in particular, provides an important framework for the enteric microenvironment and influences the homeostasis of myenteric neuronal circuitries. Isolation of pure myenteric plexus preparations from adult tissue permits to investigate changes in the ENS involving specific ECM, such as hyaluronan. This approach is based upon the possibility to isolate myenteric ganglia from the intestinal wall of either adult animals or humans, after microdissection and subsequent enzymatic digestion of the tissue. Enteric ganglia are free of connective tissue, extracellular collagen, and blood vessels, and thus treatment of intact intestinal segments with highly purified collagenases permits ganglia isolation from the surrounding smooth muscle cells. In this chapter, we describe methods for visualizing HA in isolated primary cultures of adult rat small intestine myenteric ganglia.


Subject(s)
Fluorescent Antibody Technique/methods , Ganglia/chemistry , Hyaluronic Acid/analysis , Intestine, Small/innervation , Microscopy, Confocal/methods , Myenteric Plexus/chemistry , Animals , Cells, Cultured , Ganglia/cytology , Ganglia/ultrastructure , Myenteric Plexus/cytology , Myenteric Plexus/ultrastructure , Rats , Tissue Culture Techniques/methods
8.
Article in English | MEDLINE | ID: mdl-28836741

ABSTRACT

BACKGROUND: It remains unclear whether regional variation exists in the human enteric nervous system (ENS) ie, whether intrinsic innervation varies along the gut. Recent classification of gastrointestinal neuropathies has highlighted inadequacies in the quantification of the human ENS. This study used paired wholemounts to accurately quantify and neurochemically code the hindgut myenteric plexus, comparing human distal colon and rectum. METHODS: Paired human descending colonic/rectal specimens were procured from 15 patients undergoing anterior resection. Wholemounts of myenteric plexi were triple-immunostained with anti-Hu/NOS/ChAT antibodies. Images were acquired by motorized epifluorescence microscopy, allowing assessment of ganglionic density/size, ganglionic area density, and neuronal density. 'Stretch-corrected' values were calculated using stretched/relaxed tissue dimensions. KEY RESULTS: Tile-stitching created a collage with average area 99 300 000 µm2 . Stretch-corrected ganglionic densities were similar (colon: median 510 ganglia/100 mm2 [range 386-1170], rectum: 585 [307-923]; P = .99), as were average ganglionic sizes (colon: 57 593 µm2 [40 301-126 579], rectum: 54 901 [38 701-90 211], P = .36). Ganglionic area density (colon: 11.92 mm2 per 100 mm2 [7.53-18.64], rectum: 9.84 [5.80-17.19], P = .10) and stretch-corrected neuronal densities (colon: 189 neurons/mm2 [117-388], rectum: 182 [89-361], P = .31) were also similar, as were the neurochemical profiles of myenteric ganglia, with comparable proportions of NOS+ and ChAT+ neurons (P > .10). CONCLUSIONS AND INFERENCES: This study has revealed similar neuronal and ganglionic densities and neurochemical profiles in human distal colon and rectum. Further investigation of other components of the ENS, incorporating additional immunohistochemical markers are required to confirm that there is no regional variation in the human hindgut ENS.


Subject(s)
Colon/chemistry , Myenteric Plexus/chemistry , Rectum/chemistry , Aged , Aged, 80 and over , Colon/innervation , Female , Ganglia/chemistry , Humans , Immunohistochemistry , Male , Middle Aged , Neurons/chemistry , Rectum/innervation
9.
Article in English | MEDLINE | ID: mdl-28256032

ABSTRACT

BACKGROUND: Serotonin plays a pivotal role in regulating gut motility, visceral sensitivity, and fluid secretion via specific receptors. Among these receptors, 5-HT4 exerts a prominent control on gut motor function. Although the prokinetic effect exerted by 5-HT4 agonists is well known, the cellular sites of 5-HT4 expression remain poorly understood in large mammals, e.g., horses. In this study, we evaluated the distribution of 5-HT4 in the horse intestine and in foals with enteric aganglionosis, reminiscent of human Hirschsprung's disease. METHODS: The intestine and spinal ganglia were obtained from three healthy horses and two foals with hereditary ileocolonic aganglionosis. Tissues were processed for immunohistochemistry using a specific antibody to 5-HT4 and a variety of neuronal markers. Myenteric and submucosal plexus 5-HT4 -immunoreactive (IR) neurons were quantified as relative percentage (mean±SD) to the total number of neurons counted. Furthermore, the density of 5-HT4 -IR nerve fibers was evaluated in the mucosa and tunica muscularis. KEY RESULTS: The 5-HT4 immunoreactivity was localized to large percentages of myenteric neurons ranging from 28±9% (descending colon) to 63±19% (ileum), and submucosal neurons ranging from 54±6% (ileum) to 68±14% (duodenum). The 5-HT4 -immunoreactivity was co-expressed by some substance P-IR (SP-IR) spinal ganglion neurons and extrinsic sensory fibers of aganglionic foals. CONCLUSIONS & INFERENCES: The presence of 5-HT4 in many enteric and extrinsic sensory neurons and nerve fibers provides solid morphological evidence of the cellular sites of 5-HT4 expression in horses. The evidence of SP-IR sensory neurons positive for 5-HT4 suggests its role in visceral sensitivity.


Subject(s)
Enteric Nervous System/chemistry , Gastrointestinal Tract/chemistry , Receptors, Serotonin, 5-HT4/analysis , Sensory Receptor Cells/chemistry , Animals , Enteric Nervous System/metabolism , Gastrointestinal Tract/metabolism , Horses , Male , Myenteric Plexus/chemistry , Myenteric Plexus/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Sensory Receptor Cells/metabolism
10.
J Endocrinol ; 191(2): 481-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17088418

ABSTRACT

Obestatin, a 23 amino acid peptide recently isolated from the rat stomach, is encoded by the same gene that encodes ghrelin. With the use of an antiserum directed against the mouse/rat obestatin, obestatin immunoreactivity (irOBS) was detected in cells of the gastric mucosa, myenteric plexus, and in Leydig cells of the testis in Sprague-Dawley rats. Double labeling the myenteric plexus with obestatin antiserum and choline acetyltransferase (ChAT) antiserum revealed that nearly all irOBS neurons were ChAT positive and vice versa. For comparative purposes, myenteric ganglion cells, cells in the gastric mucosa, and Leydig cells of the testis were shown to be immunoreactive to preproghrelin. The biological activity of obestatin on rat central neurons was assessed by the calcium microfluorimetric Fura-2 method. Obestatin (100 nM) administered to dissociated and cultured rat cerebral cortical neurons elevated cytosolic calcium concentrations [Ca2+]i in a population of cortical neurons. The result provides the first immunohistochemical evidence that obestatin is expressed in cells of the gastric mucosa and myenteric ganglion cells, and also in Leydig cells of the testis; the peptide is biologically active on central neurons.


Subject(s)
Gastric Mucosa/chemistry , Leydig Cells/chemistry , Myenteric Plexus/chemistry , Neurons/chemistry , Peptide Hormones/analysis , Animals , Calcium/metabolism , Cells, Cultured , Choline O-Acetyltransferase/analysis , Fluorometry , Gastric Mucosa/metabolism , Ghrelin , Growth Hormone/metabolism , Immunohistochemistry/methods , Leydig Cells/metabolism , Male , Microscopy, Confocal , Myenteric Plexus/metabolism , Neurons/metabolism , Peptide Hormones/metabolism , Rats , Rats, Sprague-Dawley
11.
Virchows Arch ; 448(3): 288-94, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16308708

ABSTRACT

Interstitial cell of Cajal (ICC) hyperplasia has been documented in conditions associated with multiple gastrointestinal stromal tumours (GISTs) (familial GIST syndromes, Carney's triad and von Recklinghausen's disease) and rarely in the vicinity of sporadic GISTs. The incidence of sporadic ICC hyperplasia and the so-called seedling leiomyoma (SLM) of the lower oesophagus has not been studied in the KIT era. In a retrospective review of 77 consecutive, routinely processed oesophagogastric resection specimens for distal oesophageal carcinoma, we found foci of ICC hyperplasia in 7 of 77 (9.1%) cases and foci of SLM in 17 of 77 (22%) cases. Two types of ICC hyperplasia were recognized: a non-circumscribed type and a nodular expansile type with peripherally compressed myenteric neural tissues. All cases of ICC hyperplasia were vimentin+/CD34+/CD117+. SLMs were desmin+/vimentin(-)/CD34(-)/CD117(-), similar to smooth muscles of the gut wall. In a prospective study of 32 non-carcinomatous specimens from age-matched patients (mostly autopsy cases), we found SLMs in only one case, but we were unable to detect ICC hyperplasia in any of the cases. We concluded that sporadic KIT-positive spindle-cell hyperplasia and SLMs were unexpectedly common in distal oesophageal specimens harbouring carcinomas. The possible mechanisms leading to the development of these putative precursor lesions will be discussed.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Myenteric Plexus/pathology , Precancerous Conditions/pathology , Adenocarcinoma/chemistry , Adenocarcinoma/surgery , Aged , Aged, 80 and over , Barrett Esophagus/metabolism , Barrett Esophagus/surgery , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/surgery , Esophageal Neoplasms/chemistry , Esophageal Neoplasms/surgery , Female , Humans , Hyperplasia , Leiomyoma/pathology , Male , Middle Aged , Myenteric Plexus/chemistry , Precancerous Conditions/chemistry , Retrospective Studies
12.
Life Sci ; 78(14): 1610-6, 2006 Feb 28.
Article in English | MEDLINE | ID: mdl-16289621

ABSTRACT

The cellular expression patterns of mu-, delta- and kappa-opioid receptors in the rat ileum were examined using fluorescence immunohistochemistry. Double-labelling was used to examine cellular receptor co-localisation as a pre-requisite for intracellular molecular interactions, such as heterodimerisation. Tissues were stained as whole-mount preparations. Strong, broadly distributed immunoreactivity (ir) was observed for each receptor in the myenteric and submucous plexuses. Although intracellular mu- and delta-ir patterns differed in ganglion neurons, mu/delta co-expression was extensive in these cells. mu/delta co-expression was also observed in interstitial cells, which were diffusely distributed in submucous plexus preparations but generally located adjacent to myenteric plexus structures. Punctate kappa-ir was seen broadly in nerve fibres in both plexuses, suggesting localisation in varicosities. Neuronal mu/kappa co-localisation was not apparent, although kappa-ir fibres were often apposed against mu-ir cells. mu/kappa co-localisation was detected in interstitial cells in submucous plexus preparations. Similarities in mu and delta expression patterns might reflect similar functional properties previously detected for these receptors. This study indicates that the rat gastrointestinal tract might provide a useful tool for the future study of molecular interactions between opioid receptor types.


Subject(s)
Ileum/chemistry , Myenteric Plexus/chemistry , Receptors, Opioid/analysis , Animals , Female , Ileum/innervation , Immunohistochemistry , Myenteric Plexus/cytology , Neurons/chemistry , Rats , Rats, Wistar , Tissue Distribution
13.
J Comp Neurol ; 524(13): 2577-603, 2016 09 01.
Article in English | MEDLINE | ID: mdl-26850701

ABSTRACT

A full description of the terminal architecture of sympathetic axons innervating the gastrointestinal (GI) tract has not been available. To label sympathetic fibers projecting to the gut muscle wall, dextran biotin was injected into the celiac and superior mesenteric ganglia (CSMG) of rats. Nine days postinjection, animals were euthanized and stomachs and small intestines were processed as whole mounts (submucosa and mucosa removed) to examine CSMG efferent terminals. Myenteric neurons were counterstained with Cuprolinic Blue; catecholaminergic axons were stained immunohistochemically for tyrosine hydroxylase. Essentially all dextran-labeled axons (135 of 136 sampled) were tyrosine hydroxylase-positive. Complete postganglionic arbors (n = 154) in the muscle wall were digitized and analyzed morphometrically. Individual sympathetic axons formed complex arbors of varicose neurites within myenteric ganglia/primary plexus and, concomitantly, long rectilinear arrays of neurites within circular muscle/secondary plexus or longitudinal muscle/tertiary plexus. Very few CSMG neurons projected exclusively (i.e., ∼100% of an arbor's varicose branches) to myenteric plexus (∼2%) or smooth muscle (∼14%). With less stringent inclusion criteria (i.e., ≥85% of an axon's varicose branches), larger minorities of neurons projected predominantly to either myenteric plexus (∼13%) or smooth muscle (∼27%). The majority (i.e., ∼60%) of all individual CSMG postganglionics formed mixed, heterotypic arbors that coinnervated extensively (>15% of their varicose branches per target) both myenteric ganglia and smooth muscle. The fact that ∼87% of all sympathetics projected either extensively or even predominantly to smooth muscle, while simultaneously contacting myenteric plexus, is consistent with the view that these neurons control GI muscle directly, if not exclusively. J. Comp. Neurol. 524:2577-2603, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Ganglia, Sympathetic/physiology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/physiology , Muscle, Smooth/innervation , Muscle, Smooth/physiology , Myenteric Plexus/physiology , Sympathetic Fibers, Postganglionic/physiology , Animals , Ganglia/chemistry , Ganglia/physiology , Ganglia, Sympathetic/chemistry , Gastrointestinal Tract/chemistry , Male , Muscle, Smooth/chemistry , Myenteric Plexus/chemistry , Neurons/chemistry , Neurons/physiology , Rats , Rats, Inbred F344 , Sympathetic Fibers, Postganglionic/chemistry
14.
J Neurosci ; 22(15): 6732-41, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12151552

ABSTRACT

The type 3 serotonin (5-HT3) receptor is the only ligand-gated ion channel receptor for serotonin in vertebrates. Two 5-HT3 receptor subunits have been cloned, subunit A (5-HT3A) and subunit B (5-HT3B). We used in situ hybridization histochemistry and reverse transcriptase-PCR amplification to demonstrate that 5-HT3A subunit transcripts are expressed in central and peripheral neurons. In contrast, 5-HT3B subunit transcripts are restricted to peripheral neurons. Thus, the prevalent form of 5-HT3 receptor synthesized within the CNS lacks the 5-HT3B subunit. Because coexpression of 5-HT3A and 5-HT3B subunits produces heteromeric 5-HT3A/3B receptors with properties that differ from those of 5-HT3A homomeric receptors, we investigated possible coexpression of both subunits at the cellular level. We found that near to 90% of all 5-HT3B expressing neurons coexpress the 5-HT3A subunit in superior cervical and nodose ganglia (NG). In addition, there is a cellular population that expresses only the 5-HT3A subunit. Therefore, peripheral neurons have the capacity to synthesize two different 5-HT3 receptors, 5-HT3A+/3B- and 5-HT3A+/3B+ receptors. We also determined that neurons of NG projecting to the nucleus tractus solitarium and those of dorsal root ganglia projecting to superficial layers of the spinal cord express 5-HT3A or 5-HT3A/3B subunits. Thus, presynaptic 5-HT3 receptors containing the 5-HT3B subunit might be present in these target brain areas. The compartmentalized structural composition of the 5-HT3 receptor may be the basis of functional diversity within this receptor. This raises the possibility that 5-HT3 receptors participating in sympathetic, parasympathetic and sensory functions may be functionally different from those involved in cognition and emotional behavior.


Subject(s)
Central Nervous System/metabolism , Peripheral Nervous System/metabolism , Receptors, Serotonin/biosynthesis , Stilbamidines , Animals , Autoradiography , Cell Compartmentation , Cell Count , Cell Size , Central Nervous System/chemistry , Fluorescent Dyes , In Situ Hybridization , Male , Myenteric Plexus/chemistry , Myenteric Plexus/metabolism , Neurons/cytology , Neurons/metabolism , Nodose Ganglion/chemistry , Nodose Ganglion/metabolism , Peripheral Nervous System/chemistry , Protein Subunits , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Serotonin/analysis , Receptors, Serotonin/genetics , Receptors, Serotonin, 5-HT3 , Reverse Transcriptase Polymerase Chain Reaction , Superior Cervical Ganglion/chemistry , Superior Cervical Ganglion/metabolism
15.
Peptides ; 26(7): 1196-206, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15949638

ABSTRACT

Urocortin 3 (Ucn 3) or stresscopin (SCP) is a new member of the corticotropin-releasing factor (CRF) neuropeptide family and is a specific ligand for CRF type 2 receptor (CRF2). CRF receptors are known to be expressed in the gastrointestinal tract and are considered to play pathophysiological roles, for example, in gastrointestinal motility under stress. We, therefore, examined Ucn 3 expression in the normal human large intestine obtained from surgery and autopsy in order to clarify this local response to stress in human intestine. Both immunohistochemistry and mRNA in situ hybridization demonstrated Ucn 3 expression in myenteric and submucosal nervous plexus, in vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs) of blood vessels in subserosa, in smooth muscle layers of the large intestine, and in enterochromaffin cells. In contrast to Urocortin 1 (Ucn 1), Ucn 3 was hardly detected in lamina propria (LP) inflammatory cells in colonic mucosa. In addition, immunohistochemistry demonstrated CRF2 expression in myenteric and submucosal nervous plexus, in smooth muscle layers, in VECs, in VSMCs and in lamina propria inflammatory cells. Immunoreactive Ucn 3 was also detected in the large intestine by RIA, with high concentrations detected in the rectum (15.4+/-9.5 pmol/g wet weight, mean+/-SEM, n=3) and sigmoid colon (6.5+/-3.5 pmol/g wet weight, n=5). Reverse-phase HPLC of the human large intestine disclosed peaks eluting in the position of synthetic Ucn 3 or SCP. These findings all suggest that Ucn 3 plays some physiological or pathological roles in the modulation of gastrointestinal functions during stressful conditions in different manners from Ucn 1.


Subject(s)
Colon/metabolism , Corticotropin-Releasing Hormone/metabolism , Stress, Physiological/metabolism , Colon/cytology , Colon/innervation , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/physiology , Humans , Immunohistochemistry , In Situ Hybridization , Intestinal Mucosa/metabolism , Myenteric Plexus/chemistry , Myenteric Plexus/immunology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Urocortins
16.
World J Gastroenterol ; 21(23): 7172-80, 2015 Jun 21.
Article in English | MEDLINE | ID: mdl-26109803

ABSTRACT

AIM: To investigate the abundance and potential diagnostic significance of neuroligin-1 and glutamate (Glu) in Hirschsprung's disease (HSCR). METHODS: Ninety children with HSCR and 50 children without HSCR matched for similar nutritional status, age and basal metabolic index were studied. The expression and localization of neuroligin-1 and Glu were assessed using double-labeling immunofluorescence staining of longitudinal muscles with adherent myenteric plexus from the surgically excised colon of children with HSCR. Western blot analysis, quantitative real-time PCR (qRT-PCR) and immunohistochemistry were performed to evaluate the abundance of neuroligin-1 and Glu in different HSCR-affected segments (ganglionic, transitional, and aganglionic segments). Enzyme-linked immunosorbent assay (ELISA) was used to detect and compare serum Glu levels in the long-segment HSCR, short-segment HSCR and non-HSCR samples. RESULTS: Neuroligin-1 and Glu were co-expressed highest to lowest in the ganglionic, transitional and aganglionic segments based on Western blot (neuroligin-1: 0.177 ± 0.008 vs 0.101 ± 0.006, 0.177 ± 0.008 vs 0.035 ± 0.005, and 0.101 ± 0.006 vs 0.035 ± 0.005, P < 0.005; Glu: 0.198 ± 0.006 vs 0.115 ± 0.008, 0.198 ± 0.006 vs 0.040 ± 0.003, and 0.115 ± 0.008 vs 0.040 ± 0.003, P < 0.005) and qRT-PCR (neuroligin-1: 9.58 × 10(-5) ± 9.94 × 10(-6) vs 2.49 × 10(-5) ± 1.38 × 10(-6), 9.58 × 10(-5) ± 9.94 × 10(-6) vs 7.17 × 10(-6 ±) 1.12 × 10(-6), and 2.49 × 10(-5) ± 1.38 × 10(-6) vs 7.17 × 10(-6) ± 1.12 × 10(-6), P < 0.005). Serum Glu level was the highest to lowest in the non-HSCR, short-type HSCR and long-type HSCR samples based on ELISA (in nmol/µL, 0.93 ± 0.31 vs 0.57 ± 0.25, 0.93 ± 0.31 vs 0.23 ± 0.16, and 0.57 ± 0.25 vs 0.23 ± 0.16, P < 0.005). CONCLUSION: Neuroligin-1 and Glu may represent new markers of ganglion cells, whose expression may correlate with the pathogenesis, diagnosis, differential diagnosis or classification of HSCR.


Subject(s)
Cell Adhesion Molecules, Neuronal/analysis , Colon/innervation , Glutamic Acid/analysis , Hirschsprung Disease/metabolism , Myenteric Plexus/chemistry , Biomarkers/analysis , Blotting, Western , Case-Control Studies , Cell Adhesion Molecules, Neuronal/genetics , Child , Child, Preschool , Diagnosis, Differential , Enzyme-Linked Immunosorbent Assay , Genetic Markers , Hirschsprung Disease/classification , Hirschsprung Disease/diagnosis , Hirschsprung Disease/genetics , Humans , Immunohistochemistry , Predictive Value of Tests , Real-Time Polymerase Chain Reaction
17.
J Comp Neurol ; 353(2): 161-78, 1995 Mar 06.
Article in English | MEDLINE | ID: mdl-7538152

ABSTRACT

The aim of this study was to investigate the neurochemical coding of myenteric neurons in the guinea pig gastric corpus by using immunohistochemical methods. Antibodies and antisera against calbindin (CALB), calretinin (CALRET), choline acetyltransferase (ChAT), calcitonin gene-related peptide (CGRP), dopamine beta-hydroxylase (DBH), beta-endorphin (ENK), neuropeptide Y (NPY), neuron-specific enolase (NSE), nitric oxide synthase (NOS), protein gene product 9.5 (PGP), parvalbumin (PARV), serotonin (5-HT), somatostatin (SOM), substance P (SP), tyrosine hydroxylase (TH), and vasoactive intestinal peptide (VIP) were used. Double- and triple-labeling studies revealed colocalization of certain transmitters and enabled the identification of distinct subpopulations of gastric enteric neurons. NPY/VIP/NOS/ENK were present in 28% of all neurons, whereas 11% had NPY/VIP/DBH/ChAT; NOS-only neurons made up 2% of the population. The combination SP/ChAT/ENK occurred in 21% of the population, whereas SP/ChAT/ENK/CALRET and SP/CHAT/SOM/ +/- CALRET was identified in 5% and 6% of all cells, respectively. 5-HT-containing neurons comprised 2% of all cells and could be further classified by the presence of additional antigens as 5-HT/SP/(ChAT) or 5-HT/VIP/(ChAT). Approximately 21% of all neurons contained only ChAT with no additional antigen present and are referred to as ChAT/-. Gastric myenteric ganglion cells were not immunoreactive for CALB, PARV, CGRP, or TH. The results of this study indicate that gastric myenteric neurons can be characterized on the basis of different chemical coding. Neurochemical coding of corpus myenteric neurons revealed some similarities and significant differences in comparison with other regions of the gut. These differences might reflect adaptation of enteric nerves according to regional specialization and the distinct functions of the proximal stomach as a gastric reservoir.


Subject(s)
Guinea Pigs/metabolism , Myenteric Plexus/chemistry , Neurons/chemistry , Stomach/innervation , Animals , Epitopes , Female , Guinea Pigs/anatomy & histology , Immunohistochemistry , Intestine, Small/innervation , Male , Myenteric Plexus/cytology , Neurotransmitter Agents/analysis
18.
J Comp Neurol ; 396(4): 429-41, 1998 Jul 13.
Article in English | MEDLINE | ID: mdl-9651003

ABSTRACT

In recent years, considerable progress has been made in characterising the neural circuitry of the pelvic plexus, particularly in the male rat. However, the small ganglia on the adventitial surface of the rectum remain largely unstudied. We have used immunohistochemistry and retrograde tracing techniques to determine the content and projections of these neurons. The adventitial ganglia contain 600-1,000 neurons. All of these are immunoreactive for choline acetyltransferase, 44% are immunoreactive for calbindin, and 35% are immunoreactive for vasoactive intestinal peptide. Very few (1-5%) adventitial neurons contain tyrosine hydroxylase or neuropeptide Y. In contrast, most adventitial neurons are surrounded by varicose axons that do contain tyrosine hydroxylase or neuropeptide Y. Retrograde tracing studies showed that the primary targets of adventitial neurons within the bowel are the internal anal sphincter and the circular muscle directly adjacent to the sphincter. However, more adventitial neurons project out of the gut wall than to targets within the bowel. These are most likely to be viscerofugal and rectospinal neurons. Combining the immunohistochemical and tracing observations, these studies suggest that the rat adventitial ganglia do not represent an additional source of pelvic (autonomic postganglionic) neurons but, instead, that they are comprised primarily of viscerofugal and rectospinal neurons. This is very different from the adventitial rectal ganglia of the cat, which represent merely an extension of the pelvic plexus.


Subject(s)
Cats/metabolism , Ganglia, Autonomic/chemistry , Hypogastric Plexus/chemistry , Rats, Wistar/metabolism , Rectum/innervation , Animals , Ganglia, Autonomic/physiology , Hypogastric Plexus/physiology , Immunohistochemistry , Intestines/chemistry , Male , Myenteric Plexus/chemistry , Nerve Endings/chemistry , Neural Pathways/physiology , Neurons/chemistry , Neurons/ultrastructure , Organ Culture Techniques , Rats , Species Specificity , Sympathetic Nervous System/chemistry
19.
J Comp Neurol ; 437(4): 423-37, 2001 Sep 03.
Article in English | MEDLINE | ID: mdl-11503144

ABSTRACT

The morphological and physiological characteristics of myenteric neurons in the guinea pig distal colon were determined using Lucifer yellow- or N-(2-aminoethyl) biotinamide-containing microelectrodes and intracellular recording and staining methods. The neurons in this study (n = 204) were classified on the basis of the shapes of their cell bodies and short processes or dendrites and the number of long processes or axons as Dogiel type I (n = 75 neurons; 36.8%), filamentous (n = 31 neurons; 15.2%), Dogiel type II (n = 38 neurons; 18.6%), and unclassified (n = 60 neurons; 29.4%). All Dogiel type II neurons had action potentials followed by an after-spike hyperpolarization (AH), and most of them (84%) had large, smooth somata and filamentous, short processes in addition to multiple, long processes or axons. Most of Dogiel type I, filamentous, and unclassified neurons (98%) had a single, long process, but four Dogiel type I neurons and one unclassified neuron had two long processes terminating as varicosities within other ganglia or on the surface of longitudinal muscle. The projections of monoaxonal neurons were distributed equally between oral and aboral directions, and most of them received fast excitatory postsynaptic potentials (EPSPs). All of the Dogiel type II neurons and seven Dogiel type I neurons were positive for calbindin immunoreactivity, but three filamentous neurons received fEPSPs, had spikes followed by AH, and were negative for calbindin. The presence of calbindin-immunoreactive(-IR) neurons was quite variable among the ganglia. These results confirm that neither the presence of calbindin immunoreactivity nor the absence of fEPSPs can be used as a predictor of cellular morphology or electrophysiological properties of myenteric neurons in the distal colon.


Subject(s)
Colon/innervation , Myenteric Plexus/cytology , Nerve Tissue Proteins/analysis , Neurons/cytology , S100 Calcium Binding Protein G/analysis , Action Potentials , Animals , Axons/ultrastructure , Biomarkers , Calbindins , Cell Size , Dendrites/ultrastructure , Excitatory Postsynaptic Potentials , Guinea Pigs , Male , Myenteric Plexus/chemistry , Neurons/chemistry , Neurons/classification , Neurons/physiology
20.
J Comp Neurol ; 414(2): 193-211, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10516591

ABSTRACT

The formation of the enteric nervous system (ENS) from neural crest-derived cell precursors requires the growth factor glial cell line-derived neurotrophic factor (GDNF) and the receptors Ret and GDNF family receptor alpha 1 (GFRalpha1). We investigated the location(s), the timing, and the extent to which these GDNF receptors appear in the population of crest-derived precursors that form the avian ENS using immunohistochemistry and in situ hybridization. Sections and whole mounts of embryonic chick gastrointestinal tract were costained with antibodies to the receptors and to HNK-1, a marker for crest-derived cells. Neural crest-derived precursors migrate through the primitive esophagus to colonize the gizzard where an extensive cellular network forms. Ret-immunoreactivity (ir) was found in a network of cells in the gizzard at embryonic day (E)3.5. As development proceeded, Ret-immunoreactive cells appeared at progressively more caudal positions and were present in the colon at E7.5. Costaining with Ret and HNK-1 was performed to determine the number of Ret-immunoreactive cells in the crest-derived population. Ret appeared in some HNK-1 cells in the esophagus and gizzard at embryonic day (E)3.5. During development, the number of crest cells with Ret increased in the ganglia of the gizzard and small intestine. GFRalpha1-ir was also found in HNK-1 cells in the esophagus at E3.5 but did not appear in the gizzard until E4.5. Surprisingly, the colonizing vanguard of crest-derived cells lacked both Ret- and GFRalpha-ir. Between E4.5 and E6.5, the fraction of HNK-1-positive cells expressing GFRalpha1 increased considerably in the foregut. Ret and GFRalpha1 were coexpressed in many cells at E6.5, and the number of such cells increased as development progressed. In the adult, GFRalpha1 and Ret were found in the neuropil of enteric ganglia. We conclude that the population of cells expressing the receptors increases during development and persists in the adult, findings that support a neurotrophic role for GDNF in the formation and maintenance of the avian ENS.


Subject(s)
Chick Embryo/embryology , Drosophila Proteins , Myenteric Plexus/chemistry , Myenteric Plexus/embryology , Nerve Tissue Proteins , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Submucous Plexus/chemistry , Submucous Plexus/embryology , Age Factors , Animals , Antibodies , Blotting, Western , CD57 Antigens/analysis , Chickens , Duodenum/innervation , ELAV Proteins , Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor Receptors , In Situ Hybridization , Neural Crest/chemistry , Neural Crest/embryology , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-ret , Quail , RNA, Messenger/analysis , RNA-Binding Proteins/analysis , Receptor Protein-Tyrosine Kinases/analysis , Substance P/analysis , Vagus Nerve/chemistry , Vagus Nerve/embryology , Vasoactive Intestinal Peptide/analysis
SELECTION OF CITATIONS
SEARCH DETAIL