Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 986
Filter
1.
Int J Mol Sci ; 22(10)2021 May 12.
Article in English | MEDLINE | ID: mdl-34065933

ABSTRACT

Neonicotinoid insecticides are nicotine-derived molecules which exert acute neurotoxic effects over the insect central nervous system by activating nicotinic acetylcholine receptors (nAChRs). However, these receptors are also present in the mammalian central and peripheral nervous system, where the effects of neonicotinoids are faintly known. In mammals, cholinergic synapses are crucial for the control of vascular tone, blood pressure and skeletal muscle contraction. We therefore hypothesized that neonicotinoids could affect cholinergic networks in mammals and sought to highlight functional consequences of acute intoxication in rats with sub-lethal concentrations of the highly used acetamiprid (ACE) and clothianidin (CLO). In this view, we characterized their electrophysiological effects on rat α3ß4 nAChRs, knowing that it is predominantly expressed in ganglia of the vegetative nervous system and the adrenal medulla, which initiates catecholamine secretion. Both molecules exhibited a weak agonist effect on α3ß4 receptors. Accordingly, their influence on epinephrine secretion from rat adrenal glands was also weak at 100 µM, but it was stronger at 500 µM. Challenging ACE or CLO together with nicotine (NIC) ended up with paradoxical effects on secretion. In addition, we measured the rat arterial blood pressure (ABP) in vivo by arterial catheterization. As expected, NIC induced a significant increase in ABP. ACE and CLO did not affect the ABP in the same conditions. However, simultaneous exposure of rats to both NIC and ACE/CLO promoted an increase of ABP and induced a biphasic response. Modeling the interaction of ACE or CLO on α3ß4 nAChR is consistent with a binding site located in the agonist pocket of the receptor. We present a transversal experimental approach of mammal intoxication with neonicotinoids at different scales, including in vitro, ex vivo, in vivo and in silico. It paves the way of the acute and chronic toxicity for this class of insecticides on mammalian organisms.


Subject(s)
Epinephrine/metabolism , Insecticides/toxicity , Neonicotinoids/toxicity , Nicotine/toxicity , Receptors, Nicotinic/metabolism , Adrenal Medulla/drug effects , Adrenal Medulla/metabolism , Animals , Arterial Pressure/drug effects , Disease Models, Animal , Drug Partial Agonism , Ganglia/drug effects , Ganglia/metabolism , Gene Expression Regulation/drug effects , Guanidines/toxicity , Male , Rats , Thiazoles/toxicity , Toxicity Tests, Subacute
2.
Sci Rep ; 11(1): 7934, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846426

ABSTRACT

Parkinson's disease (PD) is associated with neuronal damage in the brain and gut. This work compares changes in the enteric nervous system (ENS) of commonly used mouse models of PD that exhibit central neuropathy and a gut phenotype. Enteric neuropathy was assessed in five mouse models: peripheral injection of MPTP; intracerebral injection of 6-OHDA; oral rotenone; and mice transgenic for A53T variant human α-synuclein with and without rotenone. Changes in the ENS of the colon were quantified using pan-neuronal marker, Hu, and neuronal nitric oxide synthase (nNOS) and were correlated with GI function. MPTP had no effect on the number of Hu+ neurons but was associated with an increase in Hu+ nuclear translocation (P < 0.04). 6-OHDA lesioned mice had significantly fewer Hu+ neurons/ganglion (P < 0.02) and a reduced proportion of nNOS+ neurons in colon (P < 0.001). A53T mice had significantly fewer Hu+ neurons/area (P < 0.001) and exhibited larger soma size (P < 0.03). Treatment with rotenone reduced the number of Hu+ cells/mm2 in WT mice (P < 0.006) and increased the proportion of Hu+ translocated cells in both WT (P < 0.02) and A53T mice (P < 0.04). All PD models exhibited a degree of enteric neuropathy, the extent and type of damage to the ENS, however, was dependent on the model.


Subject(s)
Gastrointestinal Tract/pathology , Intestinal Pseudo-Obstruction/pathology , Parkinson Disease/pathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Acute Disease , Animals , Cell Count , Chronic Disease , Colon/drug effects , Colon/pathology , Disease Models, Animal , Feces , Ganglia/drug effects , Ganglia/pathology , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/pathology , Nitric Oxide Synthase Type I/metabolism , Oxidopamine , Phenotype , Rotenone/pharmacology
3.
Eur J Pharmacol ; 886: 173536, 2020 Nov 05.
Article in English | MEDLINE | ID: mdl-32896550

ABSTRACT

The cardiac plexus, which contains parasympathetic ganglia, plays an important role in regulating cardiac function. Histamine is known to excite intracardiac ganglion neurons, but the underlying mechanism is obscure. In the present study, therefore, the effect of histamine on rat intracardiac ganglion neurons was investigated using perforated patch-clamp recordings. Histamine depolarized acutely isolated neurons with a half-maximal effective concentration of 4.5 µM. This depolarization was markedly inhibited by the H1 receptor antagonist triprolidine and mimicked by the H1 receptor agonist 2-pyridylethylamine, thus implicating histamine H1 receptors. Consistently, reverse transcription-PCR (RT-PCR) and Western blot analyses confirmed H1 receptor expression in the intracardiac ganglia. Under voltage-clamp conditions, histamine evoked an inward current that was potentiated by extracellular Ca2+ removal and attenuated by extracellular Na+ replacement with N-methyl-D-glucamine. This implicated the involvement of non-selective cation channels, which given the link between H1 receptors and Gq/11-protein-phospholipase C signalling, were suspected to be transient receptor potential canonical (TRPC) channels. This was confirmed by the marked inhibition of the inward current through the pharmacological disruption of either Gq/11 signalling or intracellular Ca2+ release and by the application of the TRPC blockers Pyr3, Gd3+ and ML204. Consistently, RT-PCR analysis revealed the expression of several TRPC subtypes in the intracardiac ganglia. Whilst histamine was also separately found to inhibit the M-current, the histamine-induced depolarization was only significantly inhibited by the TRPC blockers Gd3+ and ML204, and not by the M-current blocker XE991. These results suggest that TRPC channels serve as the predominant mediator of neuronal excitation by histamine.


Subject(s)
Ganglia/cytology , Ganglia/drug effects , Heart/drug effects , Heart/innervation , Histamine/pharmacology , Ion Channels/drug effects , Neurons/drug effects , TRPC Cation Channels/drug effects , Animals , Calcium Signaling/drug effects , Female , Histamine Agonists/pharmacology , Histamine H1 Antagonists/pharmacology , Male , Meglumine/pharmacology , Patch-Clamp Techniques , Potassium Channels/drug effects , Pyridines/pharmacology , Rats , Rats, Wistar , Triprolidine/pharmacology , Type C Phospholipases/drug effects
4.
Aquat Toxicol ; 223: 105482, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32371337

ABSTRACT

The use of online remote control for 24/7 behavioural monitoring can play a key role in estimating the environmental status of aquatic ecosystems. Recording the valve activity of bivalve molluscs is a relevant approach in this context. However, a clear understanding of the underlying disturbances associated with behaviour is a key step. In this work, we studied freshwater Asian clams after exposure to crude oil (measured concentration, 167 ± 28 µg·L-1) for three days in a semi-natural environment using outdoor artificial streams. Three complementary approaches to assess and explore disturbances were used: behaviour by high frequency non-invasive (HFNI) valvometry, tissue contamination with polycyclic aromatic hydrocarbons (PAH), and proteomic analysis. Two tissues were targeted: the pool adductor muscles - retractor pedal muscle - cerebral and visceral ganglia, which is the effector of any valve movement and the gills, which are on the frontline during contamination. The behavioural response was marked by an increase in valve closure-duration, a decrease in valve opening-amplitude and an increase in valve agitation index during opening periods. There was no significant PAH accumulation in the muscle plus nervous ganglia pool, contrary to the situation in the gills, although the latter remained in the low range of data available in literature. Major proteomic changes included (i) a slowdown in metabolic and/or cellular processes in muscles plus ganglia pool associated with minor toxicological effect and (ii) an increase of metabolic and/or cellular processes in gills associated with a greater toxicological effect. The nature of the proteomic changes is discussed in terms of unequal PAH distribution and allows to propose a set of explanatory mechanisms to associate behaviour to underlying physiological changes following oil exposure. First, the first tissues facing contaminated water are the inhalant siphon, the mantle edge and the gills. The routine nervous activity in the visceral ganglia should be modified by nervous information originating from these tissues. Second, the nervous activity in the visceral ganglia could be modified by its own specific contamination. Third, a decrease in nervous activity of the cerebral ganglia close to the mouth, including some kind of narcosis, could contribute to a decrease in visceral ganglia activity via a decrease or blockage of the downward neuromodulation by the cerebro-visceral connective. This whole set of events can explain the decrease of metabolic activity in the adductor muscles, contribute to initiate the catch mechanism and then deeply modify the valve behaviour.


Subject(s)
Behavior, Animal/drug effects , Corbicula/drug effects , Corbicula/metabolism , Petroleum/toxicity , Polycyclic Aromatic Hydrocarbons/toxicity , Proteome/metabolism , Water Pollutants, Chemical/toxicity , Animals , Ecosystem , Fresh Water/chemistry , Ganglia/drug effects , Ganglia/metabolism , Gills/drug effects , Gills/metabolism , Muscles/drug effects , Muscles/metabolism , Proteomics
5.
Brain Res Bull ; 157: 90-99, 2020 04.
Article in English | MEDLINE | ID: mdl-32017970

ABSTRACT

Diabetic cardiac autonomic neuropathy (DCAN) is a serious complication of diabetes mellitus, which often leads to cardiac dysfunction and even threatens patients' life. Osthole, a natural coumarin derivative, has anti-inflammatory, anti-oxidant and antihypertensive effects. The P2X3 receptor is related to DCAN. The objective of this study will investigate whether osthole relieves DCAN associated with the P2X3 receptor in the stellate ganglia of diabetic rats. A type 2 diabetes mellitus rat model was induced by a combination of diet and streptozotocin. Our results showed that osthole improved the abnormal changes of blood pressure, heart rate, and heart rate variability in diabetic rats and significantly reduced the up-regulated expression levels of the P2X3 receptor, tumor necrosis factor-α and interleukin-1ß in stellate ganglia of diabetic rats. Meanwhile, osthole significantly decreased the elevated serum adrenaline concentration and phosphorylation level of extracellular regulated protein kinase 1/2. In addition, the molecular docking result indicated that osthole was a perfect fit for interacting with the P2X3 receptor. Overall, osthole alleviates the sympathetic relative excitation via inhibiting the expression of P2X3 receptors in the stellate ganglia, to achieve a balance between sympathetic and parasympathetic nerves, relieves the DCAN.


Subject(s)
Coumarins/pharmacology , Diabetic Neuropathies/drug therapy , Ganglia/drug effects , Receptors, Purinergic P2X3/drug effects , Animals , Blood Pressure/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Diabetic Neuropathies/pathology , Ganglia/pathology , Male , Molecular Docking Simulation , Rats, Sprague-Dawley , Receptors, Purinergic P2X3/metabolism , Streptozocin/pharmacology
6.
Environ Sci Pollut Res Int ; 26(14): 14461-14472, 2019 May.
Article in English | MEDLINE | ID: mdl-30868461

ABSTRACT

Rotenone (ROT) was shown to affect cerebral ganglions (CGs) of Lumbricus terrestris as a pioneering observation in our earlier investigation. Though ROT is a well-known neurotoxin causing neurodegeneration (ND), the precipitation of movement dysfunction remains largely unknown. We have designed the current study to analyze motor abnormalities in worms by exposing them to different concentrations (0.0-0.4 ppm) of ROT for 7 days. GABA, cholinergic receptor, serotonin transporter (SERT), acetylcholine esterase (AchE), and dopamine-ß-hydroxylase that are well known for their involvement in neuromuscular junctions were investigated by qRT-PCR. Further, neuronal mitochondrial genes (cytochrome C oxidase-2, NADH deydrogenase-1, cytochrome-b) and actin-1 that are essential for regeneration and calreticulin (phagocytosis) were investigated. The levels of neurotransmitters, lipids, ATPase, neuronal behavior analyses, and fluorescence analysis (lipid droplets) were performed in CGs which showed significant variations at 0.3 ppm. Ultrastructural changes in lipid droplet and neuromelatonin were prominent in 0.3 ppm. Dose-dependent effect of ROT on behavior alteration and expression of m-RNAs studied suggested that at 0.3 ppm, it could deteriorate motor and cognitive functions. We predict that perhaps, by virtue of its effect on cerebral ganglionic genes and their neurotransmitting potential, ROT may cause morbidities that resemble features characteristic of hemiparkinsonic degeneration.


Subject(s)
Neurons/drug effects , Neurotoxins/toxicity , Oligochaeta/physiology , Rotenone/toxicity , Animals , Cognition/drug effects , Ganglia/drug effects , Motor Activity/drug effects , Neurotransmitter Agents/metabolism , Oligochaeta/drug effects , RNA, Messenger/metabolism , Soil Pollutants/toxicity , Toxicity Tests
7.
Antioxid Redox Signal ; 30(11): 1389-1410, 2019 04 10.
Article in English | MEDLINE | ID: mdl-29587485

ABSTRACT

AIMS: Cisplatin can damage spiral ganglion neurons (SGNs) and cause sensorineural hearing loss. Wnt activation protects against neomycin-induced hair cell damage in the mouse cochlea, but the role of Wnt signaling in protecting SGNs from cisplatin treatment has not yet been elucidated. This study was designed to investigate the neuroprotective effects of Wnt signaling against cisplatin-induced SGN damage. RESULTS: First, we found that Wnt signaling was activated in SGNs after cisplatin treatment. Next, we discovered that overexpression (OE) of Wnt signaling in SGNs reduced cisplatin-induced SGN loss by inhibiting caspase-associated apoptosis, thus preventing the loss of SGN function after cisplatin treatment. In contrast, inhibition of Wnt signaling increased apoptosis, made SGNs more vulnerable to cisplatin treatment, and exacerbated hearing loss. TP53-induced glycolysis and apoptosis regulator (TIGAR), which scavenges intracellular reactive oxygen species (ROS), was upregulated in SGNs in response to cisplatin administration. Wnt/ß-catenin activation increased TIGAR expression and reduced ROS level, while inhibition of Wnt/ß-catenin in SGNs reduced TIGAR expression and increased the ROS level. Moreover, OE of TIGAR reduced ROS and decreased caspase 3 expression, as well as increased the survival of SGNs in Wnt-inhibited SGNs. Finally, antioxidant treatment rescued the more severe SGN loss induced by ß-catenin deficiency after cisplatin treatment. Innovation and Conclusion: This study is the first to indicate that Wnt signaling activates TIGAR and protects SGNs against cisplatin-induced damage through the inhibition of oxidative stress and apoptosis in SGNs, and this might offer novel therapeutic targets for the prevention of SGN injury. Antioxid. Redox Signal. 00, 000-000.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cisplatin/adverse effects , Cochlea/cytology , Cochlea/metabolism , Necrosis/chemically induced , Phosphoric Monoester Hydrolases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Cochlea/drug effects , Ganglia/cytology , Ganglia/drug effects , Ganglia/metabolism , Glycolysis/drug effects , Glycolysis/genetics , Glycolysis/physiology , Hearing Loss/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Phosphoric Monoester Hydrolases/genetics , Spiral Ganglion/cytology , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology
8.
Behav Brain Res ; 360: 341-353, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30528940

ABSTRACT

Although it is well documented that exposure to aversive stimuli induces modulation of neural circuits and subsequent behavioral changes, the means by which an aversive stimulus concomitantly alters behaviors of different natures (e.g., defensive and appetitive) remains unclear. Here, we addressed this issue by using the learning-induced concurrent modulation of defensive and appetitive behaviors that occurs when the mollusk Aplysia is exposed to aversive stimuli. In Aplysia, aversive stimuli concomitantly enhance withdrawal reflexes (i.e., sensitization) and suppress feeding. Sensitization and feeding suppression, which are expressed in the short term and long term, depending on the training protocol, are accompanied by increased excitability of the tail sensory neurons (TSNs) controlling the withdrawal reflexes, and by decreased excitability of feeding decision-making neuron B51, respectively. Serotonin (5-HT) has been shown to mediate sensitization, but not feeding suppression. In this study, we examined which other neurotransmitter might be responsible for feeding suppression and its underlying cellular changes. Our results indicate that nitric oxide (NO) contributes to both short-term and long-term feeding suppression, as well as to the underlying decreased B51 excitability. NO was also necessary for the induction of long-term sensitization and for the expression of short-term increased TSN excitability in vitro, revealing a previously undocumented interaction between 5-HT and NO signaling cascades in sensitization. Overall, these results revealed a scenario in which multiple modulators contribute to the widespread changes induced by sensitizing stimuli in Aplysia.


Subject(s)
Avoidance Learning/physiology , Feeding Behavior/physiology , Neurons/physiology , Nitric Oxide/metabolism , Reflex/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Aplysia , Avoidance Learning/drug effects , Electric Stimulation/adverse effects , Enzyme Inhibitors/pharmacology , Feeding Behavior/drug effects , Ganglia/cytology , Ganglia/drug effects , Ganglia/physiology , In Vitro Techniques , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Donors/pharmacology , Patch-Clamp Techniques , S-Nitroso-N-Acetylpenicillamine/pharmacology , Serotonin/pharmacology , Statistics, Nonparametric
9.
Toxins (Basel) ; 10(12)2018 12 08.
Article in English | MEDLINE | ID: mdl-30544796

ABSTRACT

The non-proteinogenic amino acid beta-methyl-amino-l-alanine (BMAA) is a neurotoxin produced by cyanobacteria. BMAA accumulation in the brain of animals via biomagnification along the food web can contribute to the development of neurodegenerative diseases such as Amyotrophic lateral sclerosis/Parkinsonism dementia complex (ALS/PDC), the latter being associated with a loss of dopaminergic neurons. Daphnia magna is an important microcrustacean zooplankton species that plays a key role in aquatic food webs, and BMAA-producing cyanobacteria often form part of their diet. Here, we tested the effects of BMAA on putative neurodegeneration of newly identified specific dopaminergic neurons in the optic ganglia/brain complex of D. magna using quantitative tyrosine-hydroxylase immunohistochemistry and fluorescence cytometry. The dopaminergic system was analysed in fed and starved isogenic D. magna adults incubated under different BMAA concentrations over 4 days. Increased BMAA concentration showed significant decrease in the stainability of dopaminergic neurons of D. magna, with fed animals showing a more extreme loss. Furthermore, higher BMAA concentrations tended to increase offspring mortality during incubation. These results are indicative of ingested BMAA causing neurodegeneration of dopaminergic neurons in D. magna and adversely affecting reproduction. This may imply similar effects of BMAA on known human neurodegenerative diseases involving dopaminergic neurons.


Subject(s)
Amino Acids, Diamino/toxicity , Bacterial Toxins/toxicity , Dopaminergic Neurons/drug effects , Neurotoxins/toxicity , Animals , Brain/cytology , Brain/drug effects , Cyanobacteria , Cyanobacteria Toxins , Daphnia , Female , Ganglia/drug effects , Reproduction/drug effects
10.
Elife ; 72018 10 16.
Article in English | MEDLINE | ID: mdl-30325308

ABSTRACT

The Large Cell (LC) motor neurons of the crab cardiac ganglion have variable membrane conductance magnitudes even within the same individual, yet produce identical synchronized activity in the intact network. In a previous study we blocked a subset of K+ conductances across LCs, resulting in loss of synchronous activity (Lane et al., 2016). In this study, we hypothesized that this same variability of conductances makes LCs vulnerable to desynchronization during neuromodulation. We exposed the LCs to serotonin (5HT) and dopamine (DA) while recording simultaneously from multiple LCs. Both amines had distinct excitatory effects on LC output, but only 5HT caused desynchronized output. We further determined that DA rapidly increased gap junctional conductance. Co-application of both amines induced 5HT-like output, but waveforms remained synchronized. Furthermore, DA prevented desynchronization induced by the K+ channel blocker tetraethylammonium (TEA), suggesting that dopaminergic modulation of electrical coupling plays a protective role in maintaining network synchrony.


Subject(s)
Crustacea/physiology , Dopamine/metabolism , Ganglia/physiology , Gap Junctions/metabolism , Motor Neurons/physiology , Action Potentials , Animals , Ganglia/drug effects , Motor Neurons/drug effects , Patch-Clamp Techniques , Serotonin/metabolism
11.
PLoS One ; 12(12): e0189297, 2017.
Article in English | MEDLINE | ID: mdl-29220373

ABSTRACT

This study aimed to determine whether nerve regeneration by means of an artificial nerve conduit is promoted by ethanol-induced cervical sympathetic ganglion block (CSGB) in a canine model. This study involved two experiments-in part I, the authors examined the effect of CSGB by ethanol injection on long-term blood flow to the orofacial region; part II involved evaluation of the effect of CSGB by ethanol injection on inferior alveolar nerve (IAN) repair using polyglycolic acid-collagen tubes. In part I, seven Beagles were administered left CSGB by injection of 99.5% ethanol under direct visualization by means of thoracotomy, and changes in oral mucosal blood flow in the mental region and nasal skin temperature were evaluated. The increase in blood flow on the left side lasted for 7 weeks, while the increase in average skin temperature lasted 10 weeks on the left side and 3 weeks on the right. In part II, fourteen Beagles were each implanted with a polyglycolic acid-collagen tube across a 10-mm gap in the left IAN. A week after surgery, seven of these dogs were administered CSGB by injection of ethanol. Electrophysiological findings at 3 months after surgery revealed significantly higher sensory nerve conduction velocity and recovery index (ratio of left and right IAN peak amplitudes) after nerve regeneration in the reconstruction+CSGB group than in the reconstruction-only group. Myelinated axons in the reconstruction+CSGB group were greater in diameter than those in the reconstruction-only group. Administration of CSGB with ethanol resulted in improved nerve regeneration in some IAN defects. However, CSGB has several physiological effects, one of which could possibly be the long-term increase in adjacent blood flow.


Subject(s)
Ethanol/pharmacology , Ganglia/drug effects , Nerve Regeneration , Spine/drug effects , Sympathetic Nervous System/drug effects , Animals , Dogs , Male , Microscopy, Electron, Transmission
12.
BJU Int ; 120(6): 861-872, 2017 12.
Article in English | MEDLINE | ID: mdl-28782252

ABSTRACT

OBJECTIVES: To evaluate neuronal nitric oxide (NO) synthase (nNOS) phosphorylation, nNOS uncoupling, and oxidative stress in the penis and major pelvic ganglia (MPG), before and after the administration of the cAMP-dependent protein kinase A (PKA) agonist colforsin in a rat model of bilateral cavernous nerve injury (BCNI),which mimics nerve injury after prostatectomy. MATERIALS AND METHODS: Adult male Sprague-Dawley rats were divided into BCNI and sham-operated groups. Each group included two subgroups: vehicle and colforsin (0.1 mg/kg/day i.p.). After 3 days, erectile function (intracavernosal pressure) was measured and penis and MPG were collected for molecular analyses of phospho (P)-nNOS (Ser-1412 and Ser-847), total nNOS, nNOS uncoupling, binding of protein inhibitor of nNOS (PIN) to nNOS, gp91phox subunit of NADPH oxidase, active caspase 3, PKA catalytic subunit α (PKA-Cα; by Western blot) and oxidative stress (hydrogen peroxide [H2 O2 ] and superoxide by Western blot and microdialysis method). RESULTS: Erectile function was decreased 3 days after BCNI and normalized by colforsin. nNOS phosphorylation on both positive (Ser-1412) and negative (Ser-847) regulatory sites, and nNOS uncoupling, were increased after BCNI in the penis and MPG, and normalized by colforsin. H2 O2 and total reactive oxygen species production were increased in the penis after BCNI and normalized by colforsin. Protein expression of gp91phox was increased in the MPG after BCNI and was normalized by colforsin treatment. Binding of PIN to nNOS was increased in the penis after BCNI and was normalized by colforsin treatment. Protein expression of active Caspase 3 was increased in the MPG after BCNI and was normalized by colforsin treatment. Protein expression of PKA-Cα was decreased in the penis after BCNI and normalized by colforsin. CONCLUSION: Collectively, BCNI impairs nNOS function in the penis and MPG by mechanisms involving its phosphorylation and uncoupling in association with increased oxidative stress, resulting in erectile dysfunction. PKA activation by colforsin reverses these molecular changes and preserves penile erection in the face of BCNI.


Subject(s)
Erectile Dysfunction/physiopathology , Neuroprotective Agents , Nitric Oxide Synthase Type I , Penile Erection/drug effects , Protein Processing, Post-Translational , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Ganglia/drug effects , Male , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type I/pharmacology , Oxidative Stress , Pelvis/innervation , Phosphorylation , Rats , Rats, Sprague-Dawley
13.
Pol J Vet Sci ; 20(1): 173-183, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28525332

ABSTRACT

Cocaine- and amphetamine-regulated transcript (CART) peptides are widely expressed not only in the brain but also in numerous endocrine/neuroendocrine cells as well as in neurons of the peripheral nervous system. The present study investigated the distribution patterns of CART-like immunoreactivity in the pelvic plexus (PP) of the female pig. The co-expression of CART with principal neurotransmitter markers: choline acetyltransferase (ChAT), tyrosine hydroxylase (TH), serotonin (5-HT) or biologically active neuropeptides: pituitary adenylate cyclase-activating polypeptide (PACAP), substance P (SP), calbindin was analyzed using double immunohistochemical stainings. Amongst neurons immunopositive to Hu C/D panneuronal marker as many as 4.1 ± 1.2% in right and 4.4 ± 1.6% in left pelvic ganglia were found to express CART. The vast majority of CART-IR ganglionic neurons were predominantly small in size and were evenly scattered throughout particular ganglia. Immunoreactivity to CART was also detected in numerous nerve terminals (which frequently formed pericellular formations around CART-negative perikarya) as well as in numerous nerve fibres within nerve branches interconnecting the unilateral pelvic ganglia. Immunohistochemistry revealed that virtually all CART-IR neurons were cholinergic in nature and CART-IR basket-like formations frequently encircled TH-positive/CART-negative perikarya. None of CART-IR ganglionic neurons showed immunoreactivity to SP, PACAP, 5-HT or calbindin. CART-IR nerve fibres ran in a close vicinity to serotonin-containing cells or faintly labelled SP-expressing neurons. On the other hand, PACAP-IR, SP-IR (but not 5-HT-positive) nerve terminals were found to run in close proximity to CART-IR neurons. Our results indicate that: 1) CART present in PP may influence the activity of pelvic ganglionic neurons/SIF cells, 2) PP should be considered as a potential source of CART-like supply to pelvic viscera and 3) functional interactions between CART and SP or PACAP are possible at the periphery.


Subject(s)
Ganglia/drug effects , Gene Expression Regulation/physiology , Nerve Tissue Proteins/metabolism , Swine/physiology , Animals , Female , Ganglia/physiology , Immunohistochemistry , Nerve Tissue Proteins/genetics , Protein Transport
14.
J Physiol Biochem ; 73(1): 77-87, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27738973

ABSTRACT

Urinary bladder function consists in the storage and controlled voiding of urine. Translational studies require animal models that match human characteristics, such as Octodon degus, a diurnal rodent. This study aims to characterize the contractility of the detrusor muscle and the morphology and code of the vesical plexus from O. degus. Body temperature was measured by an intra-abdominal sensor, the contractility of detrusor strips was evaluated by isometric tension recording, and the vesical plexus was studied by electrical field stimulation (EFS) and immunofluorescence. The animals showed a diurnal chronotype as judged from core temperature. The myogenic contractile response of the detrusor muscle to increasing doses of KCl reached its maximum (31.04 mN/mm2) at 60 mM. In the case of cumulative dose-response of bethanecol, the maximum response (37.42 mN/mm2) was reached at 3.2 × 10-4 M. The response to ATP was clearly smaller (3.8 mN/mm2). The pharmacological dissection of the EFS-induced contraction identified ACh and sensory fibers as the main contributors to this response. The neurons of the vesical plexus were located mainly in the trigone area, grouped in big and small ganglia. Out of them, 48.1 % of the neurons were nitrergic and 62.7 % cholinergic. Our results show functional and morphological similarities between the urinary bladder of O. degus and that of humans.


Subject(s)
Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/innervation , Octodon/physiology , Urinary Bladder/drug effects , Urinary Bladder/innervation , Adenosine Triphosphate/metabolism , Animals , Bethanechol/pharmacology , Body Temperature , Cholinergic Neurons/cytology , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , Electric Stimulation , Fluorescent Antibody Technique, Indirect , Ganglia/anatomy & histology , Ganglia/drug effects , Ganglia/metabolism , Ganglia/physiology , Humans , In Vitro Techniques , Male , Muscarinic Agonists/pharmacology , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Natriuretic Agents/pharmacology , Nitrergic Neurons/cytology , Nitrergic Neurons/drug effects , Nitrergic Neurons/metabolism , Nitrergic Neurons/physiology , Octodon/anatomy & histology , Potassium Chloride/pharmacology , Species Specificity , Urinary Bladder/metabolism , Urinary Bladder/physiology
15.
Cell Mol Neurobiol ; 37(4): 729-742, 2017 May.
Article in English | MEDLINE | ID: mdl-27488854

ABSTRACT

Neurons of the Grueneberg ganglion respond to cool temperatures as well as to distinct odorants and extend axonal processes to the olfactory bulb of the brain. Analyses of transgenic mice, in which Grueneberg ganglion neurons and their axons are labeled, revealed that these axons innervated nine distinct glomeruli distributed in a characteristic topographical pattern in dorsal, lateral, ventral, and medial regions of rather posterior areas in the bulb. To assess activation of these glomeruli (hereinafter designated as Grueneberg glomeruli) upon stimulation of Grueneberg ganglion neurons, mice were exposed to the odorant 2,3-dimethylpyrazine (2,3-DMP) and the expression of the activity-dependent marker c-Fos in juxtaglomerular cells of the relevant glomeruli was monitored. It was found that all of these glomeruli were activated, irrespective of their localization in the bulb. To verify that the activation of juxtaglomerular cells in Grueneberg glomeruli was indeed based on stimulation of Grueneberg ganglion neurons, the 2,3-DMP-induced responses in these glomeruli were investigated in mice lacking the cyclic nucleotide-gated channel CNGA3 which is critical for chemo- and thermosensory signal transduction in Grueneberg ganglion neurons. This approach revealed that elimination of CNGA3 led to a reduction of the odorant-induced activity in Grueneberg glomeruli, indicating that the activation of these glomeruli is based on a preceding stimulation of the Grueneberg ganglion. Analyzing whether Grueneberg glomeruli in the bulb might also process thermosensory information, it was found that upon exposure to coolness, Grueneberg glomeruli were activated. Investigating mice lacking CNGA3, the activation of these glomeruli by cool temperatures was attenuated.


Subject(s)
Olfactory Bulb/drug effects , Pyrazines/pharmacology , Animals , Axons/drug effects , Axons/metabolism , Cold Temperature , Ganglia/drug effects , Ganglia/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Olfactory Bulb/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Signal Transduction
16.
Toxicon ; 120: 61-8, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27452932

ABSTRACT

Chinese tarantula Ornithoctonus huwena is one of the most venomous spiders distributing in the hilly areas of southern China. In this study, using whole-cell patch-clamp technique we investigated electrophysiological and pharmacological properties of ion channels from tarantula subesophageal ganglion neurons. It was found that the neurons express multiple kinds of ion channels at least including voltage-gated calcium channels, TTX-sensitive sodium channels and two types of potassium channels. They exhibit pharmacological properties similar to mammalian subtypes. Spider calcium channels were sensitive to ω-conotoxin GVIA and diltiazem, two well-known inhibitors of mammalian neuronal high-voltage-activated (HVA) subtypes. 4-Aminopyridine and tetraethylammonium could inhibit spider outward transient and delayed-rectifier potassium channels, respectively. Huwentoxin-I and huwentoxin-IV are two abundant toxic components in the venom of Ornithoctonus huwena. Interestingly, although in our previous work they inhibit HVA calcium channels and TTX-sensitive sodium channels from mammalian sensory neurons, respectively, they fail to affect the subtypes from spider neurons. Moreover, the crude venom has no effect on delayed-rectifier potassium channels and only slightly reduces transient outward potassium channels with an IC50 value of ∼51.3 mg/L. Therefore, our findings provide important evidence for ion channels from spiders having an evolution as self-defense and prey mechanism.


Subject(s)
Esophagus/drug effects , Ganglia/drug effects , Ion Channels/physiology , Neurons/drug effects , Spider Venoms/toxicity , Spiders/drug effects , Animals , Esophagus/cytology , Female , Ganglia/cytology , Ion Channel Gating
17.
Exp Neurol ; 283(Pt A): 224-34, 2016 09.
Article in English | MEDLINE | ID: mdl-27343802

ABSTRACT

Experimental allergic encephalomyelitis (EAE), an animal model of multiple sclerosis produced by immunization with myelin oligodendrocyte glycoprotein (MOG) and adjuvants, results from profound T-cell mediated CNS demyelination. EAE is characterized by progressive, ascending motor dysfunction and symptoms of ongoing pain and hypersensitivity, in some cases preceding or concomitant with the motor deficits. In this regard, the EAE model mimics major features of multiple sclerosis, where a central neuropathic pain state is common. Although the latter condition is presumed to arise from a CNS loss of inhibitory controls secondary to the demyelination, dysfunction of sensory neurons may also contribute. Based on our previous studies that demonstrated the utility of monitoring expression of activating transcription factor 3 (ATF3), a sensitive marker of injured sensory neurons, here we followed both ATF3 and CD4+ T cells invasion of sensory ganglia (as well as the CNS) at different stages of the EAE model. We found that ATF3 is induced in peripheral sensory ganglia and brainstem well before the appearance of motor deficits. Unexpectedly, the ATF3 induction always preceded T cell infiltration, typically in adjacent, but non-overlapping regions. Surprisingly, control administration of the pertussis toxin and/or Complete Freund's adjuvants, without MOG, induced ATF3 in sensory neurons. In contrast, T cell infiltration only occurred with MOG. Taken together, our results suggest that the clinical manifestations in the EAE result not only from central demyelination but also from neuronal stress and subsequent pathophysiology of sensory neurons.


Subject(s)
Activating Transcription Factor 3/metabolism , Brain/pathology , CD4-Positive T-Lymphocytes/physiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Neurons/metabolism , Neutrophil Infiltration/physiology , Animals , Brain/metabolism , Disease Models, Animal , Disease Progression , Female , Freund's Adjuvant/toxicity , Ganglia/drug effects , Ganglia/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/toxicity , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurotransmitter Agents/metabolism , Neutrophil Infiltration/drug effects , Peptide Fragments/toxicity , Pertussis Toxin/pharmacology , TRPV Cation Channels/metabolism
18.
Chemosphere ; 152: 468-80, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27003369

ABSTRACT

Rotenone is well-documented to cause neurodegenerative condition such as Parkinson's, in the exposed systems. However, its detrimental effect on particular sites of neuronal pathway is still under investigation. We aimed at elucidating the impact of rotenone on cerebral ganglions (CG) of Lumbricus terrestris which control movement and behaviour of the worms. Worms were exposed to 0-0.4 ppm/mL of rotenone. Mitochondrial and lysosomal integrities were found to be affected beyond 0.2 ppm/mL of rotenone. Activities of cholinergic enzymes and the expression of tyrosine hydroxylase showed an impaired neuronal transmission in CGs at the dose of 0.2 ppm/mL of rotenone. Histopathological and immunoflourescent analyses showed neuronal apoptosis, reduced nucleic acid content and inhibited of neurosecretion at 0.3 ppm/mL. Electron microscopy showed that the neurons and neuromuscular junctions were affected at 0.2 ppm/mL. Dose-dependent changes were also observed in the motor function such as burrowing behaviours and locomotion. Conduction velocity (CV) and locomotion assessment showed that the CV of lateral giant fiber (LGF) was reduced while that of MGF remains unaffected at 0.2 ppm, the dose at which the burrowing behaviour was also not affected. LGF, cholinergic enzymes and tyrosine hydroxylase are primarily targeted by rotenone affecting locomotion at 0.2 ppm/mL while MGF, neuropile and the burrowing behaviour were affected at 0.3 ppm/mL. We demonstrate, in addition to dose-dependent effects, that the bioaccumulation factors range 0.28-0.32 ppm/µg of rotenone cause degenerative impact on giant fibers affecting neuronal behaviors/locomotion of worms. We also propose worms for studying mechanisms of neuronal pathology caused by chemicals prevailing in earth's atmosphere.


Subject(s)
Ganglia/drug effects , Lysosomes/drug effects , Mitochondria/drug effects , Neurotoxins/toxicity , Oligochaeta/drug effects , Rotenone/toxicity , Animals , Apoptosis/drug effects , Female , Ganglia/physiology , Locomotion/drug effects , Lysosomes/metabolism , Male , Mitochondria/metabolism , Neuromuscular Junction/drug effects , Neuromuscular Junction/ultrastructure , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Oligochaeta/metabolism , Oligochaeta/physiology , Synaptic Transmission/drug effects , Tyrosine 3-Monooxygenase/metabolism
19.
Acta Biomater ; 32: 89-99, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26776147

ABSTRACT

Erectile dysfunction (ED) is a serious medical condition in which current treatments are ineffective in prostatectomy and diabetic patients, due to injury to the cavernous nerve (CN), which causes irreversible remodeling of the penis (decreased smooth muscle and increased collagen), through a largely undefined mechanism. We propose that sonic hedgehog (SHH) and neural innervation, are indispensable regulators of collagen in the penis, with decreased SHH protein being an integral component of the fibrotic response to loss of innervation. We examined collagen abundance and morphology in control (Peyronie's), prostatectomy and diabetic patients, and in rat models of penile development, CN injury, SHH inhibition and under regenerative conditions, utilizing self-assembling peptide amphiphile (PA) nanofiber hydrogels for SHH delivery. Collagen abundance increased in penis of ED patients. In rats, collagen increased with CN injury in a defined time frame independent of injury severity. An inverse relationship between SHH and collagen abundance was identified; SHH inhibition increased and SHH treatment decreased penile collagen. SHH signaling in the pelvic ganglia (PG)/CN is important to maintain CN integrity and when inhibited, downstream collagen induction occurs. Collagen increased throughout penile development and with age, which is important when considering how to treat fibrosis clinically. These studies show that SHH PA treatment reduces collagen under regenerative post-prostatectomy conditions, indicating broad application for ED prevention in prostatectomy, diabetic and aging patients and in other peripheral nerve injuries. The PA nanofiber protein vehicle may be widely applicable as an in vivo delivery tool. STATEMENT OF SIGNIFICANCE: We use self-assembling peptide amphiphiles (PA) as biological delivery vehicles to prevent cavernous nerve (CN) injury induced erectile dysfunction (ED). These versatile hydrogels were molecularly pre-programmed for sonic hedgehog (SHH) protein delivery, either from an injectable solution with fast, in situ assembly into a soft hydrogel, or by highly aligned monodomain nanofiber bundles. We used PAs to examine a novel neuronal component to collagen regulation and the role of SHH in the fibrotic response to CN injury. SHH perturbation in the penis or the CN, selectively impacts collagen, with SHH inhibition increasing and SHH treatment suppressing collagen. These results suggest that SHH treatment by PA has translational potential to suppress collagen induction and remodelling, an irreversible component of ED development.


Subject(s)
Drug Delivery Systems , Erectile Dysfunction/drug therapy , Erectile Dysfunction/pathology , Hedgehog Proteins/administration & dosage , Hedgehog Proteins/therapeutic use , Hydrogels/chemistry , Nanofibers/chemistry , Animals , Collagen/metabolism , Diabetes Mellitus/physiopathology , Disease Models, Animal , Erectile Dysfunction/physiopathology , Fibrosis , Ganglia/drug effects , Humans , Hydroxyproline/metabolism , Immunohistochemistry , Male , Penis/drug effects , Penis/innervation , Penis/pathology , Penis/physiopathology , Peptides/pharmacology , Prostatectomy , Rats, Sprague-Dawley , Staining and Labeling , Time Factors
20.
Toxicol Appl Pharmacol ; 290: 107-15, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26524655

ABSTRACT

Prenatal nicotinic exposure (PNE) prolongs bronchopulmonary C-fiber (PCF)-mediated apneic response to intra-atrial bolus injection of capsaicin in rat pups. The relevant mechanisms remain unclear. Pulmonary substance P and adenosine and their receptors (neurokinin-A receptor, NK1R and ADA1 receptor, ADA1R) and transient receptor potential cation channel subfamily V member 1 (TRPV1) expressed on PCFs are critical for PCF sensitization and/or activation. Here, we compared substance P and adenosine in BALF and NK1R, ADA1R, and TRPV1 expression in the nodose/jugular (N/J) ganglia (vagal pulmonary C-neurons retrogradely labeled) between Ctrl and PNE pups. We found that PNE failed to change BALF substance P and adenosine content, but significantly upregulated both mRNA and protein TRPV1 and NK1R in the N/J ganglia and only NK1R mRNA in pulmonary C-neurons. To define the role of NK1R in the PNE-induced PCF sensitization, the apneic response to capsaicin (i.v.) without or with pretreatment of SR140333 (a peripheral and selective NK1R antagonist) was compared and the prolonged apnea by PNE significantly shortened by SR140333. To clarify if the PNE-evoked responses depended on action of nicotinic acetylcholine receptors (nAChRs), particularly α7nAChR, mecamylamine or methyllycaconitine (a general nAChR or a selective α7nAChR antagonist) was administrated via another mini-pump over the PNE period. Mecamylamine or methyllycaconitine eliminated the PNE-evoked mRNA and protein responses. Our data suggest that PNE is able to elevate PCF NK1R expression via activation of nAChRs, especially α7nAChR, which likely contributes to sensitize PCFs and prolong the PCF-mediated apneic response to capsaicin.


Subject(s)
Lung/drug effects , Nerve Fibers, Unmyelinated/drug effects , Receptors, Neurokinin-1/metabolism , Receptors, Neurokinin-2/metabolism , Up-Regulation , Adenosine/pharmacology , Animals , Animals, Newborn , Apnea/drug therapy , Bronchoalveolar Lavage Fluid , Capsaicin/pharmacology , Female , Ganglia/drug effects , Ganglia/metabolism , Male , Nerve Fibers, Unmyelinated/metabolism , Nicotine/blood , Nicotine/toxicity , Pregnancy , Prenatal Exposure Delayed Effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/genetics , Receptors, Neurokinin-2/genetics , Substance P/pharmacology , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Vagus Nerve/drug effects , Vagus Nerve/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...