Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell Neurosci ; 61: 23-33, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24844606

ABSTRACT

BACKGROUND & AIMS: In the central nervous system (CNS), reelin coordinates migration and lamination of neurons and regulates synaptic plasticity, whereas its role in the enteric nervous system (ENS) remains enigmatic. Thus we determined the expression pattern and localization of reelin in the human ENS and monitored the time course of mRNA expression of the reelin signaling system in the rat intestine as well as in GDNF treated ENS cultures. RESULTS: Reelin, its receptors and Dab1 were expressed in all intestinal layers as well as in isolated myenteric ganglia. Enteric ganglia and nerve fibers were immunoreactive for reelin which co-localized with PGP 9.5 and synaptophysin. In the rat small intestine, highest expression levels of reelin were detected at early postnatal stages. Enteric nerve cell cultures treated with GDNF showed marked up-regulation of reelin and its receptors. CONCLUSIONS: Reelin and its receptors are strongly expressed in the human ENS. Reelin is specifically localized in enteric neurons with highest expression levels during early postnatal life as well as in neuronal network forming enteric nerve cell cultures pointing to putative functions in the differentiation and maintenance of the ENS. EXPERIMENTAL METHODS: Gene expression of reelin, its receptors and Dab1 were analyzed in the human colon and isolated enteric ganglia. Co-localization of reelin with the pan-neuronal marker PGP 9.5 and the synaptic vesicle marker synaptophysin was studied by dual-label-immunocytochemistry. The time course of reelin expression was monitored in an ontogenetic study of rat intestines as well as in GDNF-treated cultures of enteric neurons.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation/physiology , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, Cell Surface/metabolism , Serine Endopeptidases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Age Factors , Animals , Animals, Newborn , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Extracellular Matrix Proteins/genetics , Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Humans , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Muscle, Smooth/metabolism , Myenteric Plexus/metabolism , Nerve Fibers/metabolism , Nerve Tissue Proteins/genetics , Rats , Rats, Wistar , Receptors, Cell Surface/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Reelin Protein , Serine Endopeptidases/genetics , Submucous Plexus/metabolism , Synaptophysin/metabolism , Ubiquitin Thiolesterase/metabolism
2.
J Gastroenterol ; 49(8): 1241-52, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24113817

ABSTRACT

BACKGROUND: The pathogenesis of diverticular disease (DD) is considered to be multifactorial and involves intestinal motor disturbances and an underlying enteric neuromuscular pathology. While an enteric neuropathy has been well documented, actual studies on concomitant alterations of the enteric musculature are limited. This study is aimed at reassessing the smooth muscle tissue by histological, ultrastructural and molecular-biological approaches. METHODS: Full-thickness sigmoid specimens were obtained from patients with DD (n = 20) and controls (n = 19). Morphometric analysis was performed to evaluate the thickness and connective tissue index of the circular and longitudinal muscle layers as well as the myenteric plexus. Structural alterations were determined by light and transmission electron microscopy. mRNA profiles of components of the contractile smooth muscle apparatus including smooth muscle α-actin, smoothelin, histone deacetylase 8, and smooth muscle myosin heavy chain (SMMHC) were assessed by qPCR. Altered gene expression levels were confirmed at protein level by immunohistochemistry. RESULTS: Compared to controls, patients with DD showed (1) increased thickness of the circular and longitudinal muscle layers, (2) architectural alterations of smooth muscle cells, (3) increased connective tissue index of the longitudinal muscle layer, (4) focally reduced density of myofilaments at ultrastructural level, (5) specific down-regulation of SMMHC mRNA levels, (6) decreased immunoreactivity of SMMHC, (7) oligo-neuronal hypoganglionosis. CONCLUSIONS: DD is associated with distinct structural and functional alterations of the enteric musculature. The enteric myopathy is characterized by disturbed muscular architecture, connective tissue replacement and loss of specific myofilaments and thus may contribute to the pathogenesis and progression of DD.


Subject(s)
Diverticulitis, Colonic/pathology , Muscle, Smooth/pathology , Myenteric Plexus/pathology , Sigmoid Diseases/pathology , Adult , Aged , Aged, 80 and over , Case-Control Studies , Diverticulitis, Colonic/genetics , Down-Regulation , Female , Gene Expression Regulation , Humans , Immunohistochemistry , Male , Microscopy, Electron, Transmission , Middle Aged , Muscle, Smooth/cytology , Myosin Heavy Chains/genetics , Polymerase Chain Reaction/methods , RNA, Messenger/metabolism , Sigmoid Diseases/genetics
3.
PLoS One ; 8(6): e66290, 2013.
Article in English | MEDLINE | ID: mdl-23805210

ABSTRACT

BACKGROUND & AIMS: Absence of glial cell line-derived neurotrophic factor (GDNF) leads to intestinal aganglionosis. We recently demonstrated that patients with diverticular disease (DD) exhibit hypoganglionosis suggesting neurotrophic factor deprivation. Thus, we screened mRNA expression pattern of the GDNF system in DD and examined the effects of GDNF on cultured enteric neurons. METHODS: Colonic specimens obtained from patients with DD (n = 21) and controls (n = 20) were assessed for mRNA expression levels of the GDNF system (GDNF, GDNF receptors GFRα1 and RET). To identify the tissue source of GDNF and its receptors, laser-microdissected (LMD) samples of human myenteric ganglia and intestinal muscle layers were analyzed separately by qPCR. Furthermore, the effects of GDNF treatment on cultured enteric neurons (receptor expression, neuronal differentiation and plasticity) were monitored. RESULTS: mRNA expression of GDNF and its receptors was significantly down-regulated in the muscularis propria of patients with DD. LMD samples revealed high expression of GDNF in circular and longitudinal muscle layers, whereas GDNF receptors were also expressed in myenteric ganglia. GDNF treatment of cultured enteric neurons increased mRNA expression of its receptors and promoted neuronal differentiation and plasticity revealed by synaptophysin mRNA and protein expression. CONCLUSIONS: Our results suggest that the GDNF system is compromised in DD. In vitro studies demonstrate that GDNF enhances expression of its receptors and promotes enteric neuronal differentiation and plasticity. Since patients with DD exhibit hypoganglionosis, we propose that the observed enteric neuronal loss in DD may be due to lacking neurotrophic support mediated by the GDNF system.


Subject(s)
Diverticulum/physiopathology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Aged , Cell Differentiation/drug effects , Cells, Cultured , Colon/cytology , Colon/drug effects , Colon/metabolism , Diverticulum/metabolism , Diverticulum/pathology , Down-Regulation/drug effects , Female , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Laser Capture Microdissection , Male , Middle Aged , Neuronal Plasticity/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism , Transcriptome/drug effects
4.
Gut ; 62(12): 1753-62, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23144076

ABSTRACT

OBJECTIVE: Disturbances of the enteric serotonergic system have been implicated in several intestinal motility disorders. Patients with diverticular disease (DD) have been reported to exhibit abnormal intestinal motility and innervation patterns. Gene expression profiles of the serotonergic system and distribution of the serotonin type 4 receptor (5HT-4R) were thus studied in patients with DD. DESIGN: Colonic specimens from patients with DD and controls were subjected to quantitative PCR for serotonin receptors 2B, 3A, 4, serotonin transporter and synthesising enzyme tryptophan hydroxylase. Localisation of 5HT-4R was determined by dual-label immunocytochemistry using smooth muscle actin (α-SMA) and pan-neuronal markers (PGP 9.5) and quantitative analysis was carried out. Site-specific gene expression analysis of 5HT-4R was assessed within myenteric ganglia and muscle layers. Correlation of 5HT-4R with muscarinic receptors 2 and 3 (M2R, M3R) messenger RNA expression was determined. RESULTS: 5HT-4R mRNA expression was downregulated in the tunica muscularis and upregulated in the mucosa of patients with DD, whereas the other components of the serotonergic system remained unchanged. 5HT-4R was detected in ganglia and muscle layers, but was decreased in the circular muscle layer and myenteric ganglia of patients with DD. 5HT-4R mRNA expression correlated with M2R/M3R mRNA expression in controls, but not in patients with DD. CONCLUSIONS: The serotonergic system is compromised in DD. Altered expression of 5HT-4R at mRNA and protein levels may contribute to intestinal motor disturbances reported in patients with DD. The findings support the hypothesis that DD is associated and possibly promoted by an enteric neuromuscular pathology.


Subject(s)
Diverticulum, Colon/physiopathology , Enteric Nervous System/physiopathology , Serotonergic Neurons/physiology , Aged , Case-Control Studies , Colon, Sigmoid/metabolism , Colon, Sigmoid/physiopathology , Diverticulum, Colon/metabolism , Enteric Nervous System/metabolism , Female , Humans , Male , Middle Aged , Polymerase Chain Reaction , Receptors, Serotonin, 5-HT2/metabolism , Receptors, Serotonin, 5-HT2/physiology , Receptors, Serotonin, 5-HT3/metabolism , Receptors, Serotonin, 5-HT3/physiology , Receptors, Serotonin, 5-HT4/metabolism , Receptors, Serotonin, 5-HT4/physiology , Serotonergic Neurons/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/physiology , Transcriptome/physiology , Tryptophan Hydroxylase/metabolism , Tryptophan Hydroxylase/physiology
5.
Neurobiol Dis ; 48(3): 474-80, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22850485

ABSTRACT

BACKGROUND: Alpha-synuclein (α-syn) is abundantly expressed in the central nervous system and involved in the regulation of neurotransmission. Insoluble fibrils of phosphorylated α-synuclein (p-α-syn) have been implicated in several neurodegenerative diseases (e.g. Parkinson's disease, Alzheimer's disease). The aim of the study was to determine the gene expression pattern and localization of α-syn/p-α-syn in the human enteric nervous system (ENS). METHODS: Human colonic specimens (n=13, 15-83 years) were processed for α-syn and p-α-syn immunohistochemistry. Colocalization of α-syn was assessed by dual-labeling with pan-neuronal markers (PGP 9.5, HuC/D). For qPCR studies, tissue was obtained from full-thickness sections, tunica muscularis, submucosa, mucosa, and laser-microdissected (LMD) enteric ganglia. RESULTS: Highest α-syn levels were detectable within the tunica muscularis and submucosa. Ganglia isolated by LMD showed high expression of α-syn mRNA. All myenteric and submucosal ganglia and nerve fibers were immunoreactive for α-syn. Dual-labeling revealed colocalization of α-syn with both pan-neuronal markers. p-α-syn immunoreactivity was consistently observed in specimens from adults with increasing age. CONCLUSIONS: α-syn is abundantly expressed in all nerve plexus of the human ENS including both neuronal somata and processes. The presence of p-α-syn within the ENS is a regular finding in adults with increasing age and may not be regarded as pathological correlate. The data provide a basis to unravel the functions of α-syn and to evaluate altered α-syn in enteric neuropathies and α-synucleinopathies of the CNS with gastrointestinal manifestations.


Subject(s)
Enteric Nervous System/metabolism , alpha-Synuclein/analysis , alpha-Synuclein/biosynthesis , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry , Male , Microdissection , Middle Aged , Neurons/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome , Young Adult
6.
Ger Med Sci ; 10: Doc06, 2012.
Article in English | MEDLINE | ID: mdl-22557939

ABSTRACT

Uni- or bilateral ejaculatory duct obstruction (EDO) is a rare but correctable cause of infertility, chronic pelvic pain and postejaculatory pain. EDO is a congenital or acquired condition, it is the underlying cause of infertility in approximately 5% of infertile men. If acquired, the etiology often remains unresolved, but prostatitis or urethritis with post-inflammatory adhesion of the duct walls seems to be a common underlying pathomechanism.Although a certain constellation of physicochemical semen parameters may lead to correct diagnosis, EDO often resembles a diagnosis by exclusion. Imaging of acquired EDO remains a challenge and the established surgical therapy, transurethral resection of the ejaculatory ducts (TURED), leads to a low rate of natural conception and a high rate of complications such as reflux of urine and epididymitis. We present a case of a male with suspected EDO who underwent a combined approach to both, semi-invasive diagnosis and therapy by transrectal puncture of the seminal vesicles and antegrade balloon-dilation of the ejaculatory ducts. Possibilities and pitfalls of this procedure are described and the literature is reviewed.Furthermore, we suggest a CT- or MRI-guided, percutaneous intervention for treatment of ejaculatory duct obstruction by balloon dilation and demonstrate initial steps of this procedure with a body donor. We call this new procedure PTED (percutaneous transgluteal ejaculatory ductoplasty).


Subject(s)
Catheterization/methods , Ejaculatory Ducts/pathology , Genital Diseases, Male/therapy , Adult , Aged , Cadaver , Constriction, Pathologic/therapy , Humans , Magnetic Resonance Imaging, Interventional , Male , Radiography, Interventional , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...