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











Publication year range
1.
Acta Pharmacol Sin ; 42(6): 898-908, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33154555

ABSTRACT

Hydrogen sulfide (H2S), which is closely related to various cardiovascular disorders, lowers blood pressure (BP), but whether this action is mediated via the modification of baroreflex afferent function has not been elucidated. Therefore, the current study aimed to investigate the role of the baroreflex afferent pathway in H2S-mediated autonomic control of BP regulation. The results showed that baroreflex sensitivity (BRS) was increased by acute intravenous NaHS (a H2S donor) administration to renovascular hypertensive (RVH) and control rats. Molecular expression data also showed that the expression levels of critical enzymes related to H2S were aberrantly downregulated in the nodose ganglion (NG) and nucleus tractus solitarius (NTS) in RVH rats. A clear reduction in BP by the microinjection of NaHS or L-cysteine into the NG was confirmed in both RVH and control rats, and a less dramatic effect was observed in model rats. Furthermore, the beneficial effects of NaHS administered by chronic intraperitoneal infusion on dysregulated systolic blood pressure (SBP), cardiac parameters, and BRS were verified in RVH rats. Moreover, the increase in BRS was attributed to activation and upregulation of the ATP-sensitive potassium (KATP) channels Kir6.2 and SUR1, which are functionally expressed in the NG and NTS. In summary, H2S plays a crucial role in the autonomic control of BP regulation by improving baroreflex afferent function due at least in part to increased KATP channel expression in the baroreflex afferent pathway under physiological and hypertensive conditions.


Subject(s)
Afferent Pathways/metabolism , Baroreflex/physiology , Blood Pressure/physiology , Hydrogen Sulfide/metabolism , Hypertension/physiopathology , Animals , Antihypertensive Agents/pharmacology , Baroreflex/drug effects , Blood Pressure/drug effects , Cardiotonic Agents/pharmacology , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/pharmacology , Hypertension/drug therapy , Male , Nodose Ganglion/drug effects , Nodose Ganglion/enzymology , Nodose Ganglion/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Rats, Sprague-Dawley , Solitary Nucleus/drug effects , Solitary Nucleus/enzymology , Solitary Nucleus/metabolism , Sulfides/pharmacology , Sulfonylurea Receptors/metabolism , Sulfurtransferases/metabolism
2.
J Mol Neurosci ; 57(3): 435-45, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26076739

ABSTRACT

The aim of this study is to investigate whether systemic 5-hydroxytryptamine (5-HT) can promote long-lasting form of respiratory plasticity in vivo via 5-HT2AR-activated protein kinase C (PKC) mechanism. The frequency and peak amplitude of hypoglossal nerve discharges in anesthetized rats were compared before and after intravenous injections of different treatments, including saline, 5-HT, ketanserin tartrate, or staurosporine. The administration of 5-HT at a systemic bolus imposed an initial ephemeral inhibition subsequently followed by striking facilitation, which demonstrates a biphasic manner of hypoglossal nerve output in anesthetized adult rats. The facilitatory stage conformed to the reinforced hypoglossal activity that lasted for more than 60 min after drug administration. The 5-HT evoked biphasic manner of hypoglossal output and hypoglossal nerve activity LTF (hLTF) were 5-HT2A receptor-dependent and coupled to PKC activation. The initial inhibition of hypoglossal activity was associated with nodose ganglion, and the subsequent facilitation was associated with carotid body. The reactive oxygen species (ROS) formation was triggered in the systemic 5-HT2-dependent hLTF model in vivo. The expressions of immunofluorescent histochemistry provide morphological evidence of a 5-HT/5-HT2A receptor coupled to PKC mechanism. In conclusion, systemic 5-HT challenge contributes to long-lasting form of respiratory plasticity and to elicit hLTF or elevated hLTF in animals, which with decreased or even with inhibited peripheral inhibitory activations. The effect of systemic 5-HT was regulated by a 5-HT2AR-activated PKC mechanism.


Subject(s)
Hypoglossal Nerve/drug effects , Respiration/drug effects , Serotonin/pharmacology , Action Potentials/drug effects , Anesthesia, General , Animals , Carotid Body/enzymology , Carotid Body/physiology , Hypoglossal Nerve/physiology , Injections, Intravenous , Ketanserin/pharmacology , Long-Term Potentiation/drug effects , Male , Nerve Tissue Proteins/physiology , Nodose Ganglion/enzymology , Nodose Ganglion/physiology , Oxidative Stress/drug effects , Phosphorylation , Protein Isoforms/metabolism , Protein Kinase C/physiology , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptor, Serotonin, 5-HT2A/physiology , Serotonin/administration & dosage , Serotonin 5-HT2 Receptor Agonists/pharmacology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Staurosporine/administration & dosage , Staurosporine/pharmacology
3.
Cells Tissues Organs ; 195(3): 272-86, 2012.
Article in English | MEDLINE | ID: mdl-21625066

ABSTRACT

In this study, we investigated the expression of neuronal nitric oxide synthase (nNOS) and nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d), two specific enzymes for nitric oxide (NO) synthesis, in the development of liver fibrosis induced by chronic bile duct ligation (BDL) in the rabbit. We specifically studied the liver-innervated nitroxidergic neurons that originate in the nodose ganglion (NG), nucleus of the solitary tract (NTS) and dorsal motor vagal nucleus (DMV). Our data showed that BDL resulted in overexpression of NADPH-d/nNOS in the NG, NTS and DMV neurons. Using densitometric analysis, we found a significant increase in NADPH-d expression as a result of BDL in the NG, NTS and DMV (72.6, 79.4 and 57.4% increase, respectively). These findings were corroborated by serum biochemistry and hepatic histopathological examination, which were influenced by NADPH-d/nNOS-generated NO in the liver following BDL. Upregulation of NADPH-d/nNOS expression may have important implications, including (1) facilitation of extrahepatic biliary parasympathetic tone that promotes gallbladder emptying of excess stagnant bile; (2) relaxation of smooth muscles of bile canaliculi thus participating in the pathogenesis of cholestasis; (3) dilation of hepatic sinusoids to counter BDL-induced intrahepatic portal hypertension in which endothelia may be damaged, and (4) alterations in hepatic metabolism, such as glycogenesis, bile formation and secretion, and bilirubin clearance.


Subject(s)
Biliary Tract/physiology , Jaundice, Obstructive/pathology , NADPH Dehydrogenase/metabolism , Nitrergic Neurons/pathology , Nitric Oxide Synthase Type I/metabolism , Vagus Nerve/pathology , Animals , Jaundice, Obstructive/metabolism , Nitrergic Neurons/enzymology , Nodose Ganglion/enzymology , Nodose Ganglion/pathology , Rabbits , Vagus Nerve/enzymology
4.
Auton Neurosci ; 164(1-2): 20-6, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21664195

ABSTRACT

Cholecystokinin (CCK) directly activates vagal afferent neurons resulting in coordinated gastrointestinal functions and satiation. In vitro, the effects of CCK on dissociated vagal afferent neurons are mediated via activation of the vanilloid family of transient receptor potential (TRPV) cation channels leading to membrane depolarization and an increase in cytosolic calcium. However, the cellular transduction pathway(s) involved in this process between CCK receptors and channel opening have not been identified. To address this question, we monitored CCK-induced cytosolic calcium responses in dissociated nodose neurons from rat in the presence or absence of reagents that interact with various intracellular signaling pathways. We found that the phospholipase C (PLC) inhibitor U-73122 significantly attenuated CCK-induced responses, whereas the inactive analog U-73433 had no effect. Responses to CCK were also cross-desensitized by a brief pretreatment with m-3M3FBS, a PLC stimulator. Together these observations strongly support the participation of PLC in the effects of CCK on vagal afferent neurons. In contrast, pharmacological antagonism of phospholipase A(2), protein kinase A, and phosphatidylinositol 3-kinase revealed that they are not critical in the CCK-induced calcium response in nodose neurons. Further investigations of the cellular pathways downstream of PLC showed that neither protein kinase C (PKC) nor generation of diacylglycerol (DAG) or release of calcium from intracellular stores participates in the response to CCK. These results suggest that alteration of membrane phosphatidylinositol 4,5-bisphosphate (PIP(2)) content by PLC activity mediates CCK-induced calcium response and that this pathway may underlie the vagally-mediated actions of CCK to induce satiation and alter gastrointestinal functions.


Subject(s)
Cholecystokinin/physiology , Gastrointestinal Tract/innervation , Nodose Ganglion/enzymology , Nodose Ganglion/physiology , Signal Transduction/physiology , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Cholecystokinin/pharmacology , Gastrointestinal Tract/cytology , Gastrointestinal Tract/physiology , Male , Nodose Ganglion/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Cholecystokinin/physiology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/enzymology , Sensory Receptor Cells/physiology , Signal Transduction/drug effects , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/physiology
5.
Cell Mol Neurobiol ; 30(3): 339-46, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19777342

ABSTRACT

Gene transfer has been used to examine the role of putative neurotransmitters in the nucleus tractus solitarii (NTS). Most such studies used adenovirus vector-mediated gene transfer although adenovirus vector transfects both neuronal and non-neuronal cells. Successful transfection in the NTS has also been reported with lentivirus as the vector. Feline immunodeficiency virus (FIV), a lentivirus, may preferentially transfect neurons and could be a powerful tool to delineate physiological effects produced by altered synthesis of transmitters in neurons. However, it has not been studied in NTS. Therefore, we sought to determine whether FIV transfects rat NTS cells and to define the type of cell transfected. We found that injection of FIV encoding LacZ gene (FIVLacZ) into the NTS led to transfection of numerous NTS cells. Injection of FIVLacZ did not alter immunoreactivity (IR) for neuronal nitric oxide synthase, which we have shown resides in NTS neurons. A majority (91.7 +/- 3.9%) of transfected cells contained IR for neuronal nuclear antigen, a neuronal marker; 2.1 +/- 3.8% of transfected cells contained IR for glial fibrillary acidic protein, a glial marker. No transfected neurons or fibers were observed in the nodose ganglion, which sends afferents to the NTS. We conclude that FIV almost exclusively transfects neurons in the rat NTS from which it is not retrogradely transported. The cell-type specificity of FIV in the NTS may provide a molecular method to study local physiological functions mediated by potential neurotransmitters in the NTS.


Subject(s)
Genetic Vectors/genetics , Immunodeficiency Virus, Feline/genetics , Neurotransmitter Agents/biosynthesis , Solitary Nucleus/metabolism , Transfection/methods , Afferent Pathways/cytology , Afferent Pathways/enzymology , Animals , Antigens, Nuclear/genetics , Axonal Transport/physiology , Brain Mapping/methods , Genes, Reporter , Glial Fibrillary Acidic Protein/genetics , Male , Nerve Tissue Proteins/genetics , Neuroanatomical Tract-Tracing Techniques/methods , Neurons/cytology , Neurons/enzymology , Neurons/virology , Nodose Ganglion/cytology , Nodose Ganglion/enzymology , Rats , Rats, Sprague-Dawley , Solitary Nucleus/cytology , Solitary Nucleus/enzymology , Staining and Labeling/methods , beta-Galactosidase/genetics
6.
Gastroenterology ; 134(4): 1094-103, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18395090

ABSTRACT

BACKGROUND & AIMS: Changes in the properties of visceral sensory neurons contribute to the development of gastrointestinal pain. However, little is known about the molecules involved in mechanosensation from the gastrointestinal tract. We investigated the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), a member of the mitogen-activated protein kinase cascade, in dorsal root ganglion (DRG) and nodose ganglion (NG) neurons by noxious gastric distention (GD) and its involvement in acute visceral pain in rats. METHODS: Electromyographic responses to gastric balloon distention through gastrostomy were recorded from the acromiotrapezius muscle in rats after splanchnic nerve resection or vagotomy and in control rats. We then examined the phosphorylated-ERK1/2 (p-ERK1/2) labeling in the DRG and NG after GD using immunohistochemistry. RESULTS: Gastric distention induced p-ERK1/2 in DRG and NG neurons with a peak at 2 minutes after stimulation. We found a stimulus intensity-dependent increase in the number of activated neurons, and this activation corresponded well with the incidence of the visceromotor response. Most of these p-ERK1/2-labeled neurons were small- and medium-sized neurons that coexpressed transient receptor potential vanilloid 1 ion channel and acid-sensing ion channel 3. Splanchnic nerve resection, but not vagotomy, affected the visceromotor response, and attenuated the ERK1/2 activation in DRG neurons produced by GD. Furthermore, intrathecal administration of the mitogen-activated protein kinase kinase 1/2 inhibitor, U0126, altered the response to noxious GD. CONCLUSIONS: The activation of ERK1/2 pathways in DRG neurons by noxious GD may be correlated with functional activity, and may be involved in acute visceral pain.


Subject(s)
Abdominal Pain/enzymology , Catheterization/adverse effects , Mitogen-Activated Protein Kinase 3/metabolism , Neurons, Afferent/enzymology , Stomach/innervation , Abdominal Pain/etiology , Abdominal Pain/physiopathology , Acid Sensing Ion Channels , Acute Disease , Animals , Butadienes/pharmacology , Disease Models, Animal , Electromyography , Enzyme Activation , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/enzymology , Ganglia, Spinal/physiopathology , Gastric Emptying/drug effects , Gastric Emptying/physiology , Immunohistochemistry , Male , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurofilament Proteins/metabolism , Nitriles/pharmacology , Nodose Ganglion/enzymology , Nodose Ganglion/physiopathology , Physical Stimulation , Rats , Rats, Sprague-Dawley , Sodium Channels/metabolism , Stomach/enzymology , Stomach/physiopathology , TRPV Cation Channels/metabolism
7.
J Anat ; 209(2): 239-50, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16879602

ABSTRACT

Sleep disorders are a form of stress associated with increased sympathetic activity, and they are a risk factor for the occurrence of cardiovascular disease. Given that nitric oxide (NO) may play an inhibitory role in the regulation of sympathetic tone, this study set out to determine the NO synthase (NOS) reactivity in the primary cardiovascular afferent neurons (i.e. nodose neurons) following total sleep deprivation (TSD). TSD was performed by the disc-on-water method. Following 5 days of TSD, all experimental animals were investigated for quantitative nicotinamine adenine dinucleotide phosphate-diaphorase (NADPH-d, a co-factor of NOS) histochemistry, neuronal NOS immunohistochemistry and neuronal NOS activity assay. In order to evaluate the endogenous metabolic activity of nodose neurons, cytochrome oxidase (COX) reactivity was further tested. All the above-mentioned reactivities were objectively assessed by computerized image analysis. The clinical significance of the reported changes was demonstrated by alterations of mean arterial blood pressure (MAP). The results indicated that in normal untreated rats, numerous NADPH-d/NOS- and COX-reactive neurons were found in the nodose ganglion (NG). Following TSD, however, both the labelling and staining intensity of NADPH-d/NOS as well as COX reactivity were drastically reduced in the NG compared with normal untreated ganglions. MAP was significantly higher in TSD rats (136+/-4 mmHg) than in normal untreated rats (123+/-2 mmHg). NO may serve as an important sympathoinhibition messenger released by the NG neurons, and decrease of NOS immunoexpression following TSD may account for the decrease in NOS content. In association with the reduction of NOS activity, a defect in NOS expression in the primary cardiovascular afferent neurons would enhance clinical hypertension, which might serve as a potential risk factor in the development of TSD-relevant cardiovascular disturbances.


Subject(s)
Electron Transport Complex IV/metabolism , Nitric Oxide Synthase/metabolism , Nodose Ganglion/enzymology , Sleep Deprivation/metabolism , Animals , Blood Pressure/physiology , Cell Count , Hypertension/etiology , Male , NADPH Dehydrogenase/metabolism , Nodose Ganglion/cytology , Rats , Rats, Wistar , Sleep Deprivation/physiopathology
8.
J Pineal Res ; 39(2): 206-14, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16098100

ABSTRACT

This study aimed to elucidate whether melatonin would exert beneficial effects on the neuronal functions of the nodose ganglion (NG) following acute hypoxic insult. The cytochrome oxidase (COX) and the nicotinamine adenine dinucleotide phosphate diaphorase (NADPH-d) histochemistry along with the nitric oxide synthase (NOS) immunofluorescence were used to examine the metabolic stage and nitric oxide production in nodose neurons respectively. Adult rats were injected intraperitoneally with melatonin at 5 or 100 mg/kg. Hypoxia was achieved by placing the rats into an altitude chamber (PO2 = 43 torr) for 4 hr. The results show that in normal untreated rats, nearly all and about 43% of the NG neurons displayed COX and NOS/NADPH-d reactivities with various staining intensities respectively. However, COX reactivity was drastically decreased while NOS/NADPH-d reactivity was significantly upregulated following hypoxia treatment. In melatonin pretreated rats, the hypoxia-induced reduction of COX reactivity was obviously prevented and the augmentation of NOS/NADPH-d reactivity was successfully suppressed. The deficit in the metabolic stage and the over-activation of NOS would contribute to the generation of oxidative stress. By effectively preventing the metabolic disruption, melatonin may have potential utility in therapeutic treatment of neuronal dysfunctions where oxidative stress is a participant.


Subject(s)
Antioxidants/pharmacology , Electron Transport Complex IV/metabolism , Hypoxia/enzymology , Melatonin/pharmacology , Nodose Ganglion/enzymology , Acute Disease , Animals , Dose-Response Relationship, Drug , Electron Transport Complex IV/drug effects , Fluorescent Antibody Technique , Hypoxia/drug therapy , Male , NADP/metabolism , Nodose Ganglion/drug effects , Rats , Rats, Wistar
9.
J Chem Neuroanat ; 29(2): 149-56, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15652701

ABSTRACT

This study aimed to test the hypothesis that mild hypoxic preconditioning (MHPC)-induced NOS expression would attenuate the neuropathological changes in the nodose ganglion (NG) of severe hypoxic exposure (SHE) rats. Thus, the young adult rats were caged in the altitude chamber for 4 weeks prior to SHE for 4 h to gain hypoxic preconditioning. The altitude chamber was used to set the height at the level from 5500 m (0.50 atm; pO2=79 Torr) to 10,000 m (0.27 atm; pO2=43 Torr) for MHPC and SHE, respectively. The experimental animals were allowed to survive for 0, 7, 14, 30 and 60 successive days, respectively. Nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) histochemistry and neuronal nitric oxide synthase (nNOS) immunohistochemistry were used to detect NADPH-d/nNOS reactivity in the NG at various time points following hypoxic exposure. The present results showed that about 38% of the neurons in the NG displayed NADPH-d/nNOS positive [NADPH-d/nNOS(+)] in normoxic rats. In SHE rats, a peak in the percentage (71%) and staining intensity (230%) of NADPH-d/nNOS(+) nodose neurons at 0 day, which then gradually decreased at 7-60 days. About 25% of the nodose neurons died 60 days after SHE. However, in MHPC rats subjected to SHE, NADPH-d/nNOS(+) neurons peaked in the percentage (51%) and staining intensity (171%) at 0 day, which then decreased at 7-60 days. In addition, neuronal survival was markedly increased by MHPC. These results suggested that MHPC might have a neuroprotective effect that reduces the susceptibility of the nodose neurons to NOS mediated neuropathy subsequent to SHE.


Subject(s)
Altitude Sickness/enzymology , Hypoxia/enzymology , NADPH Dehydrogenase/metabolism , Nerve Tissue Proteins/metabolism , Neurons, Afferent/enzymology , Nitric Oxide Synthase/metabolism , Nodose Ganglion/enzymology , Altitude Sickness/physiopathology , Altitude Sickness/prevention & control , Animals , Apoptosis/physiology , Atmosphere Exposure Chambers , Cell Count , Disease Models, Animal , Hypoxia/physiopathology , Immunohistochemistry , Male , Neurons, Afferent/cytology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I , Nodose Ganglion/cytology , Rats , Rats, Wistar , Time Factors
10.
Mol Cell Neurosci ; 27(3): 267-79, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15519242

ABSTRACT

The role of c-Jun activation for survival and regeneration of sensory neurons is unclear. Here we report that c-Jun N-terminal kinase (JNK)-mediated c-Jun activation is important for axonal outgrowth of sensory neurons in rat nodose and dorsal root ganglia (DRG). Peripheral severance of the vagus or the sciatic nerve resulted in a massive and rapid, but transient increase of the activated JNK (p-JNK) in neuronal nuclei, followed by c-Jun phosphorylation and activating transcription factor-3 (ATF3) induction. JNK inhibition by the selective JNK inhibitors SP600125 and (D)-JNKI1 did not affect neuronal survival in explanted or dissociated ganglia, but dramatically reduced axonal outgrowth, c-Jun activation, and ATF3 induction. Using retrograde labeling, we demonstrated that activated c-Jun (p-c-Jun) and ATF3 were associated with regenerative neurons. Taken together, our results suggest that JNK-mediated c-Jun activation is one of the first cell body reactions in response to nerve injury and that this activation and subsequent ATF3 induction are associated with axonal outgrowth.


Subject(s)
Axons/enzymology , Ganglia, Spinal/enzymology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurons, Afferent/enzymology , Nodose Ganglion/enzymology , Animals , Anthracenes/pharmacology , Axons/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons, Afferent/cytology , Neurons, Afferent/drug effects , Nodose Ganglion/cytology , Nodose Ganglion/drug effects , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
12.
Brain Res ; 999(1): 73-80, 2004 Feb 27.
Article in English | MEDLINE | ID: mdl-14746923

ABSTRACT

Recent studies have shown that (-)-epigallocatechin gallate (EGCG), one of the green tea polyphenols, has a potent antioxidant property. Nitric oxide (NO) plays an important role in the neuropathogenesis induced by brain ischemia/reperfusion and hypoxia. This study aimed to explore the potential neuroprotective effect of EGCG on the ganglionic neurons of the nodose ganglion (NG) in acute hypoxic rats. Thus, the young adult rats were pretreated with EGCG (10, 25, or 50 mg/kg, i.p.) 30 min before they were exposed to the altitude chamber at 10,000 m with the partial pressure of oxygen set at the level of 0.27 atm (pO2=43 Torr) for 4 h. All the animals examined were allowed to survive for 3, 7, and 14 successive days, respectively, except for those animals sacrificed immediately following hypoxic exposure. Nicotinamide adenine dinucleotide phosphate diaphorase (NADPH-d) histochemistry and neuronal nitric oxide synthase (nNOS) immunohistochemistry were carried out to detect the neuronal NADPH-d/nNOS expression in the NG. The present results show a significant increase in the expression of NADPH-d/nNOS reactivity in neurons of the NG at various time intervals following hypoxia. However, the hypoxia-induced increase in NADPH-d/nNOS expression was significantly depressed only in the hypoxic rats treated with high dosages of EGCG (25 or 50 mg/kg). These data suggest that EGCG may attenuate the oxidative stress following acute hypoxia.


Subject(s)
Catechin/analogs & derivatives , Catechin/pharmacology , Flavonoids/pharmacology , Hypoxia, Brain/drug therapy , NADPH Dehydrogenase/drug effects , Neuroprotective Agents/pharmacology , Nodose Ganglion/drug effects , Phenols/pharmacology , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Catechin/therapeutic use , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Hypoxia, Brain/enzymology , Hypoxia, Brain/physiopathology , Immunohistochemistry , Male , NADPH Dehydrogenase/metabolism , Neuroprotective Agents/therapeutic use , Nitric Oxide/metabolism , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Nodose Ganglion/enzymology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Polyphenols , Rats , Rats, Wistar , Time Factors
13.
Auton Neurosci ; 108(1-2): 22-31, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-14614961

ABSTRACT

Intraganglionic laminar endings (IGLEs) represent major vagal afferent structures throughout the gastrointestinal tract. Both morphological and functional data suggested a mechanosensory role. Elucidation of their functional significance in a particular organ would be facilitated by the availability of animal models with significantly altered numbers of IGLEs. The present study was aimed at searching for mouse strains fulfilling this criterion in the esophagus. Anterograde wheat germ agglutinin-horseradish peroxidase tracing (WGA-HRP) from nodose ganglion was used in order to label esophageal IGLEs in mice deficient for neurotrophin-3 (NT-3) or tyrosine kinase C-receptor (TrkC) and in control littermates. This approach was feasible only in heterozygous mutants which are viable. IGLEs were counted in tetramethylbenzidine (TMB) processed wholemounts using a standardised protocol. Quantification of myenteric neurons was done in cuprolinic blue-stained specimens. Nodose neuron counts were performed in cryostat sections stained with cresyl violet. Numbers of IGLEs in the esophagus were significantly reduced in both heterozygous NT-3 (NT-3+/-) and heterozygous TrkC (TrkC+/-) mutants (65% and 40% reduction, respectively). Numbers of nodose neurons were also significantly reduced in NT-3+/- mice (48% reduction), while their reduction in TrkC+/- mutants was insignificant (11% reduction). There was no reduction of myenteric neurons in the esophagus of either mutant strain. The numeric deficiency of IGLEs was unlikely to be secondary to reduction of myenteric neurons. Although only heterozygous mutants could be studied, these results suggest that esophageal IGLEs share neurotrophin dependence on NT-3/TrkC with spinal proprioceptors and some cutaneous mechanosensors. This concurs with their proposed function as vagal mechanosensors crucial for reflex peristalsis.


Subject(s)
Down-Regulation/genetics , Esophagus/metabolism , Mechanoreceptors/metabolism , Neurotrophin 3/deficiency , Neurotrophin 3/genetics , Nodose Ganglion/physiology , Receptor, trkC/deficiency , Receptor, trkC/genetics , Animals , Down-Regulation/physiology , Esophagus/enzymology , Female , Male , Mechanoreceptors/enzymology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Neurotrophin 3/biosynthesis , Nodose Ganglion/enzymology , Receptor, trkC/biosynthesis , Vagus Nerve/enzymology , Vagus Nerve/physiology
14.
J Comp Neurol ; 450(3): 283-91, 2002 Aug 26.
Article in English | MEDLINE | ID: mdl-12209856

ABSTRACT

Tachykinins depolarize guinea pig intracardiac neurons by activating nonselective cationic channels. Recently, members of the transient receptor potential family of membrane channels (TRPC) have been implicated in the generation of G protein-coupled receptor-activated nonselective cationic currents. We have investigated whether guinea pig cardiac neurons exhibit immunoreactivity to TRPC. Our results showed that nerve fibers within guinea pig intrinsic cardiac ganglia exhibited immunoreactivity to TRPC6. After culture of cardiac ganglia whole-mount explants for 72 hours, the TRPC6-IR fiber networks were absent. Therefore, the TRPC6-IR fibers were derived from sources extrinsic to the heart. A small percentage ( approximately 3%) of intracardiac neurons also exhibited TRPC6 immunoreactivity in control preparations, and the percentage of cells exhibiting TRPC6 immunoreactivity was not changed following explant culture for 72 hours. The few intrinsic TRPC6-IR neurons also exhibited nitric oxide synthase (NOS) immunoreactivity, indicating that they were nitrergic as well. We compared the immunohistochemical staining patterns of TRPC6-IR fibers with the staining patterns of a number of other neurotransmitters or neurotransmitter synthetic enzymes that mark specific extrinsic inputs to the intrinsic cardiac ganglia. The TRPC6-IR fibers were not immunoreactive for choline acetyltransferase, tyrosine hydroxylase, or substance P. However, the TRPC6-IR fibers exhibited immunoreactivity to neuronal NOS. Therefore, we propose that the TRPC6-IR fibers within the guinea pig intrinsic cardiac ganglia are vagal sensory fibers that also contain NOS. We found, in support of this conclusion, that TRPC6-IR cells were also present in sections of nodose ganglia.


Subject(s)
Calcium Channels/metabolism , Ganglia, Parasympathetic/enzymology , Guinea Pigs/metabolism , Heart/innervation , Myocardium/enzymology , Nitrergic Neurons/enzymology , Nitric Oxide Synthase/metabolism , Vagus Nerve/enzymology , Visceral Afferents/enzymology , Animals , Axons/enzymology , Axons/ultrastructure , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Female , Ganglia, Parasympathetic/cytology , Immunohistochemistry , Male , Myocardium/cytology , Neurons, Afferent/enzymology , Neurons, Afferent/ultrastructure , Nitrergic Neurons/cytology , Nitric Oxide/metabolism , Nodose Ganglion/cytology , Nodose Ganglion/enzymology , Sympathetic Fibers, Postganglionic/cytology , Sympathetic Fibers, Postganglionic/enzymology , Tyrosine 3-Monooxygenase/metabolism , Vagus Nerve/cytology , Visceral Afferents/cytology
15.
Morfologiia ; 122(4): 32-6, 2002.
Article in Russian | MEDLINE | ID: mdl-12596548

ABSTRACT

The expression NADPH-diaphorase and inducible NO-synthase (NOS) was studied in vagal nucleus dorsalis and ganglion nodosum neurons following acethylcholine inhalation in healthy rats and rats with ovalbumin-induced experimental bronchial asthma (BA). It was found that NOS activity regulation is mediated by cholinoreceptors; functioning of this mechanism is disturbed in hypoxic state. It is shown that both in conditions of physiological norm and in experimental BA, changes in NOS activity are determined by its constitutive, neuronal isoform.


Subject(s)
Acetylcholine/pharmacology , Asthma/enzymology , Nitric Oxide Synthase/metabolism , Olivary Nucleus/enzymology , Vagus Nerve/enzymology , Acetylcholine/administration & dosage , Administration, Inhalation , Animals , Asthma/pathology , Immunohistochemistry , Male , NADPH Dehydrogenase/metabolism , Neurons/enzymology , Nitric Oxide Synthase Type II , Nodose Ganglion/enzymology , Nodose Ganglion/pathology , Olivary Nucleus/pathology , Rats , Vagus Nerve/pathology
16.
Brain Res ; 920(1-2): 84-96, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11716814

ABSTRACT

Pre-clinical and clinical studies are currently underway to evaluate the potential of phosphodiesterase-4 (PDE4) inhibitors for the treatment of chronic obstructive pulmonary disease and other inflammatory conditions of the airways. The most common side effect associated with this class of compounds is emesis. The squirrel monkey provides a model for evaluating the efficacy of PDE4 inhibitors and their emetic potential. The distribution of three PDE4 isoforms (A, C and D) has been investigated in the squirrel monkey medulla and nodose ganglion to determine which isoform(s) could be responsible for the emetic adverse effects. The distribution of PDE4 isoforms was delineated using immunohistochemistry with antibodies specific for PDE4A, PDE4C and PDE4D and by in situ hybridization with isoform-selective riboprobes. PDE4A was present in the medulla where expression was mostly restricted to glial cells and the vasculature. PDE4C was not detected in either the medulla or nodose ganglion. Finally, the PDE4D isoform was localized to neurons in the nodose ganglion and found through many structures of medulla including the area postrema, neurons of the nucleus tractus solitarius and locus coeruleus. These data are consistent with a role for PDE4D in the emetic response.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Medulla Oblongata/enzymology , Nodose Ganglion/enzymology , Animals , Base Sequence , Blotting, Western , Cyclic Nucleotide Phosphodiesterases, Type 4 , Female , Immunohistochemistry , In Situ Hybridization , Isoenzymes/metabolism , Male , Microscopy, Fluorescence , Molecular Sequence Data , RNA Probes , Reflex/physiology , Saimiri , Substance P/metabolism , Vomiting/physiopathology
18.
J Comp Neurol ; 398(2): 289-307, 1998 Aug 24.
Article in English | MEDLINE | ID: mdl-9700572

ABSTRACT

Vagal afferent neurons contain a variety of neurochemical markers and neuroactive substances, most of which are present also in dorsal root ganglion cells. To test for the suitability of the calcium-binding protein calretinin as a specific marker for vagal afferent fibers in the periphery, immunocytochemistry for this protein was combined with retrograde tracing. Nerve fibers in the rat esophagus, as well as vagal and spinal sensory neurons innervating the esophagus, were investigated for co-localization of calretinin with calbindin, calcitonin gene-related peptide, and NADPH diaphorase. The results indicated that calretinin immunocytochemistry demonstrates neuronal structures known as vagal afferent from other studies, in particular intraganglionic laminar endings. A few enteric neurons whose distribution was unrelated to intraganglionic laminar endings also stained for calretinin. Strikingly, calretinin immunoreactivity was absent from spinal afferent neurons innervating the rat esophagus. In intraganglionic laminar endings and nodose ganglion cells calretinin was highly co-localized with calbindin but not with calcitonin gene-related peptide. On the other hand, calbindin was also found in spinal afferents to the esophagus where it was co-localized with calcitonin gene-related peptide. Vagal afferent neurons innervating the esophagus were never positive for NADPH diaphorase. Thus, calretinin appears to be a more specific marker for vagal afferent structures in the esophagus than calbindin, which is expressed by both vagal and spinal sensory neurons. Calretinin immunocytochemistry may be utilized as a valuable tool for investigations of subpopulations of vagal afferents in certain viscera.


Subject(s)
Calcium-Binding Proteins/analysis , Esophagus/innervation , Rats, Sprague-Dawley/anatomy & histology , Spinal Cord/cytology , Stilbamidines , Vagus Nerve/cytology , Animals , Brain Stem/chemistry , Brain Stem/cytology , Brain Stem/enzymology , Calbindin 2 , Calbindins , Calcitonin Gene-Related Peptide/analysis , Female , Fluorescent Dyes , Ganglia, Spinal/chemistry , Ganglia, Spinal/cytology , Ganglia, Spinal/enzymology , Male , NADPH Dehydrogenase/analysis , Nodose Ganglion/chemistry , Nodose Ganglion/cytology , Nodose Ganglion/enzymology , Rats , Rats, Wistar , S100 Calcium Binding Protein G/analysis , Spinal Cord/chemistry , Spinal Cord/enzymology , Vagotomy , Vagus Nerve/chemistry , Vagus Nerve/enzymology
20.
Eur J Pharmacol ; 315(2): 127-33, 1996 Nov 14.
Article in English | MEDLINE | ID: mdl-8960875

ABSTRACT

The present study has investigated whether nitric oxide (NO) is involved in neurotransmission of rat vagal afferent neurons. The diethylamine-NO complex (diethylamine-NO, 10-100 microM) and S-nitroso-N-acetylpenicillamine (3-100 microM) both elicited a concentration-dependent depolarisation of the isolated rat nodose ganglion preparation. Pre-treatment with 1 H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, 300 nM), 6-(phenylamino)-5,8-quinolinedione (LY83,583, 30 microM) and Methylene blue (100 microM) all caused a significant shift to the right in the concentration-response curve to diethylamine-NO. Incubation of rat nodose ganglion sections with a 35S-labeled antisense oligonucleotide to neuronal NO synthase resulted in visualisation of the mRNA encoding NO synthase over vagal afferent perikarya. The anatomical findings, therefore, suggest that a number of rat vagal afferent perikarya possess the ability to produce the enzyme required for the biosynthesis of NO. Collectively, these data suggest that NO may be functionally important as a neuromodulator of rat vagal afferent neurons.


Subject(s)
Nitric Oxide Synthase/genetics , Nitric Oxide/physiology , Nodose Ganglion/physiology , Aminoquinolines/pharmacology , Animals , Electrophysiology , Enzyme Inhibitors/pharmacology , In Situ Hybridization , Male , Nodose Ganglion/drug effects , Nodose Ganglion/enzymology , Oligonucleotides, Antisense , RNA, Messenger/physiology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL