Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Cell Mol Neurobiol ; 42(5): 1569-1583, 2022 Jul.
Article in English | MEDLINE | ID: mdl-33544273

ABSTRACT

The neurotrophin GDNF acts through its co-receptor RET to direct embryonic development of the intestinal nervous system. Since this continues in the post-natal intestine, co-cultures of rat enteric neurons and intestinal smooth muscle cells were used to examine how receptor activation mediates neuronal survival or axonal extension. GDNF-mediated activation of SRC was essential for neuronal survival and axon outgrowth and activated the major downstream signaling pathways. Selective inhibition of individual pathways had little effect on survival but JNK activation was required for axonal maintenance, extension or regeneration. This was localized to axonal endings and retrograde transport was needed for central JUN activation and subsequent axon extension. Collectively, GDNF signaling supports neuronal survival via SRC activation with multiple downstream events, with JNK signaling mediating structural plasticity. These pathways may limit neuron death and drive subsequent regeneration during challenges in vivo such as intestinal inflammation, where supportive strategies could preserve intestinal function.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor , MAP Kinase Kinase 4/metabolism , Neurons , src-Family Kinases/metabolism , Animals , Cells, Cultured , Enzyme Activation , Female , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Intestines , Neuronal Outgrowth , Neurons/metabolism , Pregnancy , Rats
2.
Mol Cell Neurosci ; 115: 103655, 2021 09.
Article in English | MEDLINE | ID: mdl-34273501

ABSTRACT

Intestinal inflammation challenges both function and structure of the enteric nervous system (ENS). In the animal model of TNBS-induced colitis, an influx of immune cells causes early neuron death in the neuromuscular layers, followed by axonal outgrowth from surviving neurons associated with upregulation of the neurotrophin GDNF (glial cell line-derived neurotrophic factor). Inflammation could involve ischemia and metabolic inhibition leading to neuronal damage, which might be countered by a protective action of GDNF. This was examined in a primary co-culture model of rat myenteric neurons and smooth muscle, where metabolic challenge was caused by dinitrophenol (DNP), O-methyl glucose (OMG) or hypoxia. These caused the specific loss of 50% of neurons by 24 h that was blocked by GDNF both in vitro and in whole mounts. Neuroprotection was lost with RET inhibition by vandetanib or GSK3179106, which also caused neuron loss in untreated controls. Thus, both basal and upregulated GDNF levels signal via RET for neuronal survival. This includes a key role for upregulation of HIF-1α, which was detected in neurons in colitis, since the inhibitor chetomin blocked rescue by GDNF or ischemic pre-conditioning in vitro. In DNP-treated co-cultures, neuron death was not inhibited by zVAD, necrosulfonamide or GSK872, and cleaved caspase-3 or - 8 were undetectable. However, combinations of inhibitors or the RIP1kinase inhibitor Nec-1 prevented neuronal death, evidence for RIPK1-dependent necroptosis. Therefore, inflammation challenges enteric neurons via ischemia, while GDNF is neuroprotective, activating RET and HIF-1α to limit programmed cell death. This may support novel strategies to address recurrent inflammation in IBD.


Subject(s)
Enteric Nervous System , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , Apoptosis , Cell Survival , Glial Cell Line-Derived Neurotrophic Factor Receptors , Neurons , Proto-Oncogene Proteins c-ret , Rats
3.
Neurogastroenterol Motil ; 29(10): 1-12, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28466581

ABSTRACT

BACKGROUND: Mouse models of inflammatory bowel disease (IBD) identify an impact on the enteric nervous system (ENS) but do not distinguish between Crohn's disease and ulcerative colitis phenotypes. In these models, analgesia is required, but its influence on different strains and disease outcomes is unknown. Therefore, changes to the ENS and intestinal smooth muscle were studied in trinitrobenzene sulfonic acid (TNBS) and dextran sodium sulfate (DSS) induced colitis to identify the effects of analgesia, and compared between two mouse strains. METHODS: Colitis was induced in CD1 or BALB/c mice receiving analgesia with either buprenorphine or tramadol. Euthanasia was on Day 8 (DSS) or Day 4 (TNBS). Outcomes were Disease Activity Index and cytokine assay, and quantitative histology and immunocytochemistry were used to evaluate effects of inflammation on neurons and smooth muscle. KEY RESULTS: In BALB/c mice, both models of colitis caused >2-fold increase in smooth muscle cell number. DSS caused axon proliferation without neuron loss while TNBS caused significant neuron loss and axonal damage. Buprenorphine (but not tramadol) was generally anti-inflammatory in both strains, but correlated with lethal outcomes to TNBS in BALB/c mice. CONCLUSIONS AND INFERENCES: Smooth muscle growth is common to both models of colitis. In contrast, ENS damage in TNBS is correlated with the severe response of a Crohn's disease-like phenotype, while DSS correlates with a milder, ulcerative colitis-like outcome in the deeper tissues. Analgesia with tramadol over buprenorphine is supported for mouse studies of IBD.


Subject(s)
Analgesics, Opioid/pharmacology , Colitis/chemically induced , Colitis/pathology , Intestines/drug effects , Analgesia/methods , Animals , Buprenorphine/pharmacology , Dextran Sulfate/toxicity , Disease Models, Animal , Inflammation/pathology , Intestines/pathology , Mice , Mice, Inbred BALB C , Tramadol/pharmacology , Trinitrobenzenesulfonic Acid/toxicity
4.
Neurogastroenterol Motil ; 28(4): 569-80, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26744175

ABSTRACT

BACKGROUND: A relationship between stress and the symptoms of irritable bowel syndrome (IBS) has been well established but the cellular mechanisms are poorly understood. Therefore, we investigated effects of stress and stress hormones on colonic descending inhibition and transit in mouse models and human tissues. METHODS: Stress was applied using water avoidance stress (WAS) in the animal model or mimicked using stress hormones, adrenaline (5 nM), and corticosterone (1 µM). Intracellular recordings were obtained from colonic circular smooth muscle cells in isolated smooth muscle/myenteric plexus preparations and the inhibitory junction potential (IJP) was elicited by nerve stimulation or balloon distension oral to the site of recording. KEY RESULTS: Water avoidance stress increased the number of fecal pellets compared to control (p < 0.05). WAS also caused a significant increase in IJP amplitude following balloon distension. Stress hormones also increased the IJP amplitude following nerve stimulation and balloon distension (p < 0.05) in control mice but had no effect in colons from stressed mice. No differences were observed with application of ATP between stress and control tissues, suggesting the actions of stress hormones were presynaptic. Stress hormones had a large effect in the nerve stimulated IJP in human colon (increased >50%). Immunohistochemical studies identified alpha and beta adrenergic receptor immunoreactivity on myenteric neurons in human colon. CONCLUSIONS & INFERENCES: These studies suggest that WAS and stress hormones can signal via myenteric neurons to increase inhibitory neuromuscular transmission. This could lead to greater descending relaxation, decreased transit time, and subsequent diarrhea.


Subject(s)
Colon/physiopathology , Gastrointestinal Motility/physiology , Irritable Bowel Syndrome/physiopathology , Stress, Psychological/physiopathology , Adult , Aged , Aged, 80 and over , Animals , Electrophysiology , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Middle Aged , Muscle, Smooth/physiopathology , Myenteric Plexus/physiopathology , Neural Inhibition/physiology , Stress, Psychological/complications , Synaptic Transmission/physiology
5.
Neuroscience ; 290: 357-68, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25655216

ABSTRACT

Intestinal inflammation causes initial axonal degeneration and neuronal death, as well as the proliferation of intestinal smooth muscle cells (ISMC), but subsequent axonal outgrowth leads to re-innervation. We recently showed that expression of glial cell-derived neurotrophic factor (GDNF), the critical neurotrophin for the post-natal enteric nervous system (ENS) is upregulated in ISMC by inflammatory cytokines, leading us to explore the relationship between ISMC growth and GDNF expression. In co-cultures of myenteric neurons and ISMC, GDNF or fetal calf serum (FCS) was equally effective in supporting neuronal survival, with neurons forming extensive axonal networks among the ISMC. However, only GDNF was effective in low-density cultures where neurons lacked contact with ISMC. In early-passage cultures of colonic circular smooth muscle cells (CSMC), polymerase chain reaction (PCR) and western blotting showed that proliferation was associated with expression of GDNF, and the successful survival of neonatal neurons co-cultured on CSMC was blocked by vandetanib or siGDNF. In tri-nitrobenzene sulfonic acid (TNBS)-induced colitis, immunocytochemistry showed the selective expression of GDNF in proliferating CSMC, suggesting that smooth muscle proliferation supports the ENS in vivo as well as in vitro. However, high-passage CSMC expressed significantly less GDNF and failed to support neuronal survival, while expressing reduced amounts of smooth muscle marker proteins. We conclude that in the inflamed intestine, smooth muscle proliferation supports the ENS, and thus its own re-innervation, by expression of GDNF. In chronic inflammation, a compromised smooth muscle phenotype may lead to progressive neural damage. Intestinal stricture formation in human disease, such as inflammatory bowel disease (IBD), may be an endpoint of failure of this homeostatic mechanism.


Subject(s)
Cell Survival/physiology , Enteric Nervous System/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Intestines/physiology , Muscle, Smooth/physiology , Neurons/physiology , Animals , Axons/drug effects , Axons/physiology , Cattle , Cell Proliferation/physiology , Cell Survival/drug effects , Coculture Techniques , Colitis/physiopathology , Enteric Nervous System/drug effects , Glial Cell Line-Derived Neurotrophic Factor/antagonists & inhibitors , Intestines/drug effects , Intestines/immunology , Male , Mice, Inbred BALB C , Muscle, Smooth/drug effects , Neurons/drug effects , Rats, Sprague-Dawley , Trinitrobenzenesulfonic Acid
6.
Neurobiol Dis ; 75: 40-52, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25562655

ABSTRACT

Intestinal inflammation affects the enteric nervous system (ENS) that lies adjacent to the smooth muscle layers. Previously, we showed that the loss of ENS neurons in animal models such as tri-nitrobenzene sulphonic acid (TNBS)-induced colitis was a limited and early event despite progressive worsening of inflammation. Here, we demonstrated that the rapid appearance of activated immune cells in the intestinal wall is selectively neurotoxic via iNOS-derived NO, using TNBS-induced colitis in both rats and mice, and a co-culture model of ENS neurons and smooth muscle. An influx of neutrophils and macrophages occurred within hours of initiation of rat colitis, correlating with iNOS expression, acutely elevated NO and neuronal death. In vitro, chemical donors of NO selectively caused axonal damage and neuronal death. These outcomes were similar to those seen with combined culture with either activated peritoneal immune cells or the immune cell lines RAW-264 and RBL-2H3. Immune cell-mediated neurotoxicity was blocked by the iNOS inhibitor L-NIL, and neuronal death was inhibited by the RIP-1 kinase inhibitor necrostatin. In a mouse model, the stereotypic loss of myenteric neurons by Day 4 post-TNBS was abrogated by the selective iNOS inhibitors L-NIL or 1400W without effect on other parameters of intestinal inflammation. Preservation of ENS neurons also ameliorated the hyperplasia of smooth muscle that is characteristic of intestinal inflammation, in line with prior work showing neural regulation of smooth muscle phenotype. This identifies a predominant pathway of immune cell damage to the ENS, where early, acute elevation of NO from iNOS can be cytotoxic to myenteric neurons.


Subject(s)
Colitis/enzymology , Enteric Nervous System/enzymology , Neurons/enzymology , Nitric Oxide Synthase Type II/metabolism , Animals , Cell Line , Coculture Techniques , Colitis/pathology , Disease Models, Animal , Enteric Nervous System/drug effects , Enteric Nervous System/immunology , Enteric Nervous System/pathology , Female , Hyperplasia/drug therapy , Hyperplasia/pathology , Hyperplasia/physiopathology , Macrophages/drug effects , Macrophages/pathology , Macrophages/physiology , Male , Mice, Inbred BALB C , Mice, Inbred C3H , Muscle, Smooth/drug effects , Muscle, Smooth/enzymology , Muscle, Smooth/immunology , Muscle, Smooth/pathology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Neutrophils/drug effects , Neutrophils/pathology , Neutrophils/physiology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Rats, Sprague-Dawley , Trinitrobenzenesulfonic Acid
7.
Neurogastroenterol Motil ; 23(2): e44-56, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21087354

ABSTRACT

BACKGROUND: The enteric nervous system (ENS) continues its structural and functional growth after birth, with formation of ganglia and the innervation of growing smooth muscle. However, little is known about factors in the postnatal intestine that influence these processes. METHODS: We examined the presence and potential role of glial cell line-derived nerve growth factor (GDNF) in the rat postnatal ENS using neonatal tissue, primary co-cultures of the myenteric plexus, smooth muscle, and glial cells as well as cell lines of smooth muscle or glial cells. KEY RESULTS: Western blot analysis showed that GDNF and its co-receptors rearranged during transfection (RET) and GDNF family receptor alpha-1 were expressed in the muscle layer of the neonatal and adult rat intestine. Immunohistochemistry localized the receptors for GDNF to myenteric neurons, while GDNF was localized to smooth muscle cells. In a co-culture model, GDNF but not nerve growth factor, brain derived neurotrophic factor or neurotrophin-3 significantly increased neuronal survival and more than doubled the numbers of neurites in vitro. RT-PCR, qPCR, Western blotting, ELISA, and immunocytochemistry as well as bioassays of neuronal survival and of RET phosphorylation all identified intestinal smooth muscle as the source of GDNF in vitro. GDNF also induced morphological changes in the structure and organization of neurons and axons, causing marked aggregation of neuronal cell bodies and collinear development of axons. As well, GDNF (50-150 ng mL(-1)) significantly increased [(3)H]-choline uptake and stimulated [(3)H]-acetylcholine release. CONCLUSIONS & INFERENCES: We conclude that GDNF derived from intestinal smooth muscle cells is a key factor influencing the structural and functional development of postnatal myenteric neurons.


Subject(s)
Animals, Newborn/metabolism , Enteric Nervous System/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Nerve Growth Factors/metabolism , Animals , Cells, Cultured , Coculture Techniques , Enteric Nervous System/cytology , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Models, Animal , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Myenteric Plexus/cytology , Myenteric Plexus/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Rats , Rats, Sprague-Dawley
8.
Neurogastroenterol Motil ; 22(2): 210-6, e67, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19740117

ABSTRACT

BACKGROUND: Intraluminal acid evokes sustained oesophageal longitudinal smooth muscle (LSM) contraction and oesophageal shortening, which may play a role in oesophageal pain and the aetiology of hiatus hernia. In the opossum model, this reflex has been shown to involve mast cell activation and release of neurokinins from capsaicin-sensitive neurons. The aim of this study was to determine whether proteinase-activated receptor-2 (PAR-2) activation evokes reflex LSM contraction via similar mechanisms. METHODS: Tension recording studies were performed using opossum oesophageal LSM strips in the presence and absence of pharmacological agents. In addition, the effect of trypsin on single isolated LSM cells was determined using videomicroscopy, and the expression of PAR-2 in oesophageal tissue was examined using immunohistochemistry. KEY RESULTS: The PAR-2 agonist trypsin evoked sustained, concentration-dependent contraction of LSM muscle strips, but had no effect on isolated LSM cells. The trypsin-induced contraction was blocked by capsaicin desensitization, substance P (SP) desensitization or application of the selective neurokinin-2 (NK-2) receptor antagonist MEN 10376. Immunohistochemistry revealed co-localization of SP, calcitonin gene-related peptide and PAR-2 in axons of opossum oesophageal LSM. CONCLUSIONS & INFERENCES: Longitudinal smooth muscle contraction induced by trypsin involves capsaicin-sensitive neurons and subsequent activation of NK-2, which is identical to the pathway involved in acid-induced LSM contraction and oesophageal shortening. This suggests that acid-induced LSM contraction may involve mast cell-derived mediators that activate capsaicin-sensitive neurons via PAR-2.


Subject(s)
Esophagus/metabolism , Muscle Contraction/physiology , Muscle, Smooth/metabolism , Receptor, PAR-2/metabolism , Receptors, Neurokinin-2/metabolism , Animals , Axons/metabolism , Calcitonin Gene-Related Peptide/metabolism , Capsaicin/pharmacology , Dose-Response Relationship, Drug , Esophagus/drug effects , Female , Immunohistochemistry , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Neurokinin A/analogs & derivatives , Neurokinin A/pharmacology , Opossums , Peptide Fragments/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Substance P/metabolism , Substance P/pharmacology , Trypsin/pharmacology
9.
Gut ; 54(12): 1699-706, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16014744

ABSTRACT

BACKGROUND: The altered motility of the inflamed intestine derives in part from changes to the contractility of the intestinal smooth muscle cell. While modifications to the muscarinic receptor system are identified, changes to 5-hydroxytryptamine (5-HT; serotonin) receptors that also mediate contraction are less well studied. METHODS: In the trinitrobenzene sulphonic acid model of rat colitis, we used receptor antagonists to identify changes in receptor utilisation that accompany the selective reversal of the impaired contractile response to acetylcholine (ACh) and 5-HT during colitis (day 4 (D4)) and following resolution of inflammation (day 36 (D36)). RESULTS: In isolated circular smooth muscle cells, challenged with ACh, the muscarinic 3 receptor (M3R) antagonists 4-DAMP and pF-HSD each showed a 50% decrease in antagonism on D4 while the M2R antagonist methoctramine more than doubled its potency, showing a decreased role of M3R and an increased role of M2R, respectively. These changes were fully reversed by D36. In contrast, the 5-HT2 receptor (5-HT2R) antagonist ketanserin was sharply decreased in effectiveness on D4, with a further decrease by D36, when the contribution of 5-HT(2A)R was only 22% of control. There were no changes in response to the 5-HT4R antagonist SDZ-205-557 at any time. Western blotting identified decreased expression of 5-HT(2A)R on D36 versus controls, further supporting the conclusion that the persistence of the impaired response to 5-HT was due to decreased expression of the excitatory 5-HT(2A)R. CONCLUSIONS: Thus the lasting decrease in receptor expression and resulting impairment of the contractile response will compromise the capacity for an appropriate response to 5-HT, which may contribute to the intestinal dysfunction seen in post-enteritis syndromes.


Subject(s)
Colitis/physiopathology , Receptors, Muscarinic/physiology , Receptors, Serotonin/physiology , Acetylcholine/pharmacology , Animals , Cells, Cultured , Colitis/chemically induced , Colitis/pathology , Male , Muscle Contraction/drug effects , Muscle, Smooth/physiopathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Rats , Rats, Sprague-Dawley , Serotonin/pharmacology , Signal Transduction , Trinitrobenzenesulfonic Acid
10.
Exp Neurol ; 191(2): 337-43, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15649490

ABSTRACT

Nerve growth factor (NGF) enhances neuronal survival during injury to the mature central and peripheral nervous systems, but its potential as a neuroprotective factor in the enteric nervous system (ENS) has not been examined. We used the trinitrobenzene sulfonic acid (TNBS)-induced model of colitis to examine if NGF-sensitive neurons were selectively spared from inflammation-induced cell loss. Immunocytochemistry of whole mounts of the rat colon showed that total myenteric neuronal number decreased by 32.9% +/- 1.4% by 35 days after inflammation. At this time, the proportion of neurons expressing both the p75 and trkA receptor decreased to 38.4% from a control value of 62.0%. The distribution of expression of neural phenotypes among the NGF receptor-expressing population was differentially affected by inflammation, with selective decrease among cholinergic excitatory neurons and calbindin-expressing neurons, and a trend to increase among inhibitory nitrergic neurons. This is evidence of a novel mechanism whereby intestinal inflammation can give rise to a permanent imbalance between excitatory and inhibitory neural pathways, thus tending to compromise intestinal function.


Subject(s)
Colitis/pathology , Colon/pathology , Enteric Nervous System/pathology , Nerve Growth Factor/physiology , Neurons/pathology , Animals , Biomarkers/analysis , Calbindins , Cell Count , Colitis/chemically induced , Colon/drug effects , Colon/innervation , Disease Models, Animal , Enteric Nervous System/drug effects , Male , Neurons/classification , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor/biosynthesis , Receptor, trkA/biosynthesis , S100 Calcium Binding Protein G/biosynthesis , Trinitrobenzenesulfonic Acid
11.
Pflugers Arch ; 448(5): 515-24, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15107998

ABSTRACT

Intestinal inflammation affects smooth muscle contractility contributing to altered motility, but changes to the individual smooth muscle cells are not well described. We used video microscopy to study the contractility of circular smooth muscle cells (CSMC) isolated from the rat mid-descending colon throughout the course of TNBS-induced colitis, measuring their shortening response to carbachol (CCh), 5-HT, histamine or high K(+). In control CSMC, CCh caused a maximal shortening response of 28 (2%), similar to that for 5-HT of 27 (1%), but by day 4 of colitis, these responses were decreased by 35% and 37%, respectively. By day 36, all aspects of cholinergic contraction returned to control levels, while 5-HT-induced contraction remained significantly attenuated. In contrast, the contractile responses to histamine remained similar at all time points. K(+)-induced contraction was impaired only on day 4, and the maximal response remained substantially greater than CCh or 5-HT. Colitis caused a 121% increase in CSMC length by day 2 that persisted through day 36, independent evidence for phenotypic change. We conclude that impaired CSMC contractility at both the receptor and non-receptor levels contribute to altered smooth muscle function during colitis. Persistent changes in contractile response remained detectable after resolution of inflammation, and similar events may occur in post-enteritis syndromes seen in humans.


Subject(s)
Colitis/physiopathology , Inflammation/physiopathology , Muscle Contraction/physiology , Myocytes, Smooth Muscle/physiology , Animals , Carbachol/pharmacology , Cells, Cultured , Cholinergic Agonists/pharmacology , Colitis/chemically induced , Dose-Response Relationship, Drug , Histamine/pharmacology , Inflammation/chemically induced , Male , Microscopy, Video , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Potassium/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin/pharmacology , Trinitrobenzenesulfonic Acid/adverse effects
12.
Can J Physiol Pharmacol ; 81(5): 451-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12774851

ABSTRACT

Acid-induced esophagitis is associated with sustained longitudinal smooth muscle (LSM) contraction and consequent esophageal shortening. In addition, LSM strips from opossums with esophagitis are hyper-responsive, while the circular smooth muscle (CSM) contractility is impaired. To determine the origin of these changes, studies were performed on esophageal smooth muscle cells isolated from opossum esophagi perfused intraluminally on 3 consecutive days with either saline (control; n = 8) or HCl (n = 9). CSM and LSM cells, obtained by enzymatic digestion, were exposed to various concentrations of carbachol (CCh) and fixed. CCh induced concentration-dependent contraction of both LSM and CSM cells. CCh-induced LSM cell contraction was not different between control and esophagitis animals; however, there was marked attenuation in the CCh-induced contraction of CSM cells from esophagitis animals. Morphological studies revealed significant hypertrophy of the CSM cells. These findings suggest that impaired CSM contractility can be attributed at least in part to alterations to the CSM cell itself. In contrast, hyper-contractility demonstrated in LSM strips is likely related to factors in the surrounding tissue.


Subject(s)
Esophagitis, Peptic/chemically induced , Esophagus , Muscle, Smooth , Animals , Carbachol/pharmacology , Cell Separation , Esophagitis, Peptic/pathology , Esophagitis, Peptic/physiopathology , Esophagus/pathology , Esophagus/physiopathology , Hydrochloric Acid , Hypertrophy , In Vitro Techniques , Manometry , Muscle Contraction/drug effects , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Myocytes, Smooth Muscle/pathology , Opossums
13.
Am J Physiol Gastrointest Liver Physiol ; 282(6): G1097-104, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12016136

ABSTRACT

The calcium-binding protein neuronal calcium sensor 1 (NCS-1) is involved in modulation of neurotransmitter release in the peripheral and central nervous systems. Since intestinal inflammation impairs neurotransmitter release, we evaluated the expression of NCS-1 in the normal rat colon and in dinitrobenzene sulfonic acid (DNBS)-induced colitis. Immunocytochemistry and Western blots showed high levels of NCS-1 in the myenteric plexus and in axons in the smooth muscle layers; 23 +/- 2% of myenteric neurons were NCS-1 positive, with staining restricted to the largest neurons. NCS-1-positive axons decreased to 13.3 +/- 0.4% of total axons by day 2 and dropped further to 7.0 +/- 0.1% by day 4, returning to control levels by day 16. Dual-label Western blot analysis showed that the expression of NCS-1 relative to PGP 9.5 decreased by 50% on day 4 but returned to control by day 16. The selective loss of NCS-1 during colitis may underlie the altered neural function seen in the inflamed intestine.


Subject(s)
Calcium-Binding Proteins/biosynthesis , Colitis/metabolism , Colon/innervation , Neuropeptides/biosynthesis , Synaptic Vesicles/metabolism , Animals , Benzenesulfonates , Blotting, Western , Calcium-Binding Proteins/analysis , Colitis/chemically induced , Colitis/immunology , Colon/immunology , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Immunohistochemistry , Male , Neuronal Calcium-Sensor Proteins , Neuropeptides/analysis , Rats , Rats, Sprague-Dawley , Synaptic Vesicles/chemistry
14.
Am J Physiol Gastrointest Liver Physiol ; 279(3): G511-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10960349

ABSTRACT

The loss of intrinsic neurons is an early event in inflammation of the rat intestine that precedes the growth of intestinal smooth muscle cells (ISMC). To study this relationship, we cocultured ISMC and myenteric plexus neurons from the rat small intestine and examined the effect of scorpion venom, a selective neurotoxin, on ISMC growth. By 5 days after neuronal ablation, ISMC number increased to 141+/-13% (n = 6) and the uptake of [(3)H]thymidine in response to mitogenic stimulation was nearly doubled. Atropine caused a dose-dependent increase in [(3)H]thymidine uptake in cocultures, suggesting the involvement of neural stimulation of cholinergic receptors in regulation of ISMC growth. In contrast, coculture of ISMC with sympathetic neurons increased [(3)H]thymidine uptake by 45-80%, which was sensitive to propranolol (30 microM) and was lost when the neurons were separated from ISMC by a permeable filter. Western blotting showed that coculture with myenteric neurons increased alpha-smooth muscle-specific actin nearly threefold to a level close to ISMC in vivo. Therefore, factors derived from enteric neurons maintain the phenotype of ISMC through suppression of the growth response, whereas catecholamines released by neurons extrinsic to the intestine may stimulate their growth. Thus inflammation-induced damage to intestinal innervation may initiate or modulate ISMC hyperplasia.


Subject(s)
Jejunum/cytology , Jejunum/innervation , Muscle, Smooth/cytology , Muscle, Smooth/innervation , Myenteric Plexus/physiology , Actins/analysis , Adrenergic beta-Antagonists/pharmacology , Animals , Atropine/pharmacology , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Cholinergic Fibers/drug effects , Cholinergic Fibers/physiology , Colon/cytology , Colon/growth & development , Colon/innervation , Ganglionic Blockers/pharmacology , Hexamethonium/pharmacology , In Vitro Techniques , Jejunum/growth & development , Male , Muscle Development , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/cytology , Muscle, Smooth/growth & development , Myenteric Plexus/cytology , Parasympatholytics/pharmacology , Phenotype , Propranolol/pharmacology , Rats , Rats, Sprague-Dawley , Scorpion Venoms/pharmacology , Sympathetic Nervous System/cytology , Sympathetic Nervous System/physiology
15.
Neuroreport ; 11(1): 177-81, 2000 Jan 17.
Article in English | MEDLINE | ID: mdl-10683853

ABSTRACT

Since activated immune cells may damage peripheral nerves during inflammation, we developed a co-culture model that permits the direct study of macrophage-induced neuronal damage. Sympathetic neurons were enzymatically isolated from neonatal mice and co-cultured with increasing numbers of peritoneal macrophages for 24 h. This caused rapid neuronal cell death, reducing neuronal number by 24.1 +/- 4% with the addition of 11.5 x 10(3) macrophages, representing a ratio of 8 macrophages per neuron. Nuclear analysis showed that cell death occurred by both apoptosis and necrosis. These effects were not mimicked by addition of macrophage-conditioned medium, and were prevented by 10 microM dexamethasone. Although no appreciable neuronal death occurred beyond 24 h, the density of neurites was decreased between 1 and 2 days of co-culture (p < 0.05). There is, therefore, a rapid induction of cytotoxicity by macrophages after their addition to the neuronal cultures, followed by axonal damage without neuronal cell death.


Subject(s)
Inflammation/pathology , Macrophages, Peritoneal/physiology , Neurons/physiology , Peripheral Nerve Injuries , Sympathetic Nervous System/cytology , Animals , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Coculture Techniques , Dexamethasone/pharmacology , Electrophysiology , Fluorescent Dyes , In Situ Nick-End Labeling , Mice , Superior Cervical Ganglion/cytology
16.
Neurosci Lett ; 281(1): 25-8, 2000 Mar 03.
Article in English | MEDLINE | ID: mdl-10686407

ABSTRACT

Over-activation of glutamate receptors is implicated in neurodegeneration. Using mice with a deletion in the GluR2 gene, we studied the sensitivity of sympathetic neurons to reduced levels of nerve growth factor (NGF), which can cause neuronal cell death. Under standard culture conditions of 50 ng/ml NGF, neurons from the superior cervical ganglion survived and grew equally well compared with wild type controls. However, the subsequent reduction of NGF levels caused significantly poorer survival among mutant neurons by 48 h, at 44+/-13% of control at 10 ng/ml NGF, and dropping further to 14+/-6% at 0.05 ng/ml NGF. These results suggest that the absence of GluR2 impairs the ability of these NGF-sensitive neurons to survive under limiting amounts of this neurotrophic factor.


Subject(s)
Nerve Growth Factor/pharmacology , Neurons/drug effects , Receptors, AMPA/physiology , Sympathetic Nervous System/drug effects , Animals , Animals, Newborn , Cell Count/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Dose-Response Relationship, Drug , Mice , Mice, Mutant Strains , Mutation , Neurites/drug effects , Neurons/cytology , Receptors, AMPA/genetics , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/drug effects , Sympathetic Nervous System/cytology
17.
Am J Pathol ; 155(4): 1051-7, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10514387

ABSTRACT

Inflammation of the intestine causes pain and altered motility, at least in part through effects on the enteric nervous system. While these changes may be reversed with healing, permanent damage may contribute to inflammatory bowel disease (IBD) and post-enteritis irritable bowel syndrome. Since little information exists, we induced colitis in male Sprague-Dawley rats with dinitrobenzene sulfonic acid and used immunocytochemistry to examine the number and distribution of enteric neurons at times up to 35 days later. Inflammation caused significant neuronal loss in the inflamed region by 24 hours, with only 49% of neurons remaining by days 4 to 6 and thereafter, when inflammation had subsided. Eosinophils were found within the myenteric plexus at only at the earliest time points, despite a general infiltration of neutrophils into the muscle wall. While the number of myenteric ganglia remained constant, there was significant decrease in the number of ganglia in the submucosal plexus. Despite reduced neuronal number and hyperplasia of smooth muscle, the density of axons among the smooth muscle cells remained unchanged during and after inflammation. Intracolonic application of the topical steroid budesonide caused a dose-dependent prevention of neuronal loss, suggesting that evaluation of anti-inflammatory therapy in inflammatory bowel disease should include quantitative assessment of neural components.


Subject(s)
Colitis/pathology , Enteric Nervous System/pathology , Animals , Anti-Inflammatory Agents/therapeutic use , Axons/drug effects , Benzenesulfonates , Budesonide/therapeutic use , Cell Count/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colon/drug effects , Colon/enzymology , Colon/innervation , Colon/pathology , Dose-Response Relationship, Drug , Enteric Nervous System/drug effects , Immunohistochemistry , Inflammation/chemically induced , Inflammation/enzymology , Inflammation/pathology , Male , Myenteric Plexus/cytology , Myenteric Plexus/drug effects , Neurons/cytology , Neurons/metabolism , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Submucous Plexus/cytology , Submucous Plexus/drug effects , Thiolester Hydrolases/metabolism , Time Factors , Ubiquitin Thiolesterase
18.
Dig Dis Sci ; 44(7): 1265-72, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10489904

ABSTRACT

Inflammation of the human intestine causes thickening of the smooth muscle layers, and studies in rats infected with Trichinella spiralis (Tsp) have shown hyperplasia of the intestinal smooth muscle cells (ISMC). We have shown that Tsp-induced inflammation caused a fivefold increase in total protein per ISMC over control, while ISMC from the noninflamed distal ileum also showed a threefold increase. The amount of alpha-smooth muscle (SM) actin per ISMC increased nearly 500% over control by postinfection (PI) day 6. The proportion of alpha-SM actin in the total cellular protein increased 200% by day 6 PI, indicating a higher density of alpha-SM actin in the hypertrophied ISMC. Gamma-SM actin mRNA increased sharply and was matched by an increased fractional content of gamma-SM actin protein. These increases in the smooth muscle-specific actins may affect force production and further demonstrate the plasticity of smooth muscle in the inflamed intestine.


Subject(s)
Enteritis/pathology , Intestinal Diseases, Parasitic/pathology , Muscle, Smooth/pathology , Trichinella spiralis , Trichinellosis/pathology , Actins/genetics , Animals , Gene Expression/physiology , Humans , Hypertrophy , Intestinal Mucosa/pathology , Jejunum/pathology , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
19.
J Immunol ; 162(7): 4271-6, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10201958

ABSTRACT

Nerve growth factor (NGF) is well recognized to have a number of potent effects on mast cells, including increasing mast cell numbers in vivo and inducing mast cell degranulation in vitro. More recently, NGF has been demonstrated to induce PGD2 production by mast cells through the induction of mast cell cyclooxygenase expression. We have observed that NGF at doses as low as 10 ng/ml will induce IL-6 production and inhibit TNF-alpha release from rat peritoneal mast cells in the presence of lysophosphatidylserine as a cofactor. NGF synergizes with LPS treatment of peritoneal mast cells (PMC) for the induction of IL-6. Examination of the mechanism of this phenomenon has revealed that NGF can induce both rat PMC and mouse bone marrow-derived cultured mast cells to produce substantial levels of PGE2. This response is maximal at later time points 18-24 h after NGF activation. The ability of NGF to induce PGE2 is not dependent on mast cell degranulation. Other stimuli capable of inducing IL-6, such as LPS, do not induce production of this prostanoid. Inhibition of cyclooxygenase activity by PMC using either flurbiprofen or indomethacin inhibited both the NGF-induced PGE2 synthesis and the NGF-induced alterations in TNF-alpha and IL-6 production. These results suggest a role for mast cell-derived prostanoids in the regulation of local inflammatory responses and neuronal degeneration after tissue injury involving induction of NGF production.


Subject(s)
Dinoprostone/physiology , Interleukin-6/biosynthesis , Mast Cells/metabolism , Nerve Growth Factors/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Degranulation/immunology , Cells, Cultured , Cytotoxicity Tests, Immunologic , Histamine Release/immunology , Hybridomas , Lymphocyte Activation , Male , Mast Cells/drug effects , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred Lew
20.
Exp Neurol ; 152(2): 251-8, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9710525

ABSTRACT

Acetylcholine (ACh) is a major neurotransmitter in the enteric nervous system. Since increasing evidence suggests that inflammation alters neural regulation of intestinal function, we examined the synthesis and breakdown of ACh in smooth muscle/myenteric plexus (SM/MP) preparations from the jejunum of the rat during inflammation caused by infection with the nematode parasite Trichinella spiralis. Both total and neuron-specific uptake of the ACh precursor [3H]choline into SM/MP preparations was increased by over twofold on Day 6 postinfection. Further, a radiochemical assay of choline acetyltransferase activity showed significant increase by Day 1, with peak values reached by Day 3 and maintained without reversal thereafter. Despite the enhancement of these steps, measurement of the conversion of [3H]choline into [3H]ACh in SM/MP preparations in vitro showed a nearly fourfold decrease by Day 6, implying a large decrease in ACh production in the inflamed jejunum. Examination of acetylcholinesterase in the rat jejunum showed decreased histochemical staining intensity in the muscle wall, and quantitative evaluation showed significantly decreased (>50%) acetylcholinesterase activity in SM/MP preparations. These results show that cholinergic innervation of the intestine can undergo rapid and long-lasting alterations during inflammation. Upregulation of major steps in the synthetic pathway for ACh was not matched by increased ACh production, suggesting that defects in ACh packaging, storage, and granule exocytosis may also be present.


Subject(s)
Acetylcholine/metabolism , Inflammation/metabolism , Jejunal Diseases/metabolism , Myenteric Plexus/metabolism , Trichinella spiralis/isolation & purification , Trichinellosis/metabolism , Animals , Choline/metabolism , Choline O-Acetyltransferase/metabolism , Inflammation/parasitology , Jejunal Diseases/parasitology , Male , Muscle, Smooth/metabolism , Rats , Rats, Sprague-Dawley , Trichinellosis/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...