Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Neuropharmacology ; 206: 108934, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34933049

ABSTRACT

Genetic differences in cerebellar sensitivity to alcohol (EtOH) influence EtOH consumption phenotype in animal models and contribute to risk for developing an alcohol use disorder in humans. We previously determined that EtOH enhances cerebellar granule cell (GC) tonic GABAAR currents in low EtOH consuming rodent genotypes, but suppresses it in high EtOH consuming rodent genotypes. Moreover, pharmacologically counteracting EtOH suppression of GC tonic GABAAR currents reduces EtOH consumption in high alcohol consuming C57BL/6J (B6J) mice, suggesting a causative role. In the low EtOH consuming rodent models tested to date, EtOH enhancement of GC tonic GABAAR currents is mediated by inhibition of neuronal nitric oxide synthase (nNOS) which drives increased vesicular GABA release onto GCs and a consequent enhancement of tonic GABAAR currents. Consequently, genetic variation in nNOS expression across rodent genotypes is a key determinant of whether EtOH enhances or suppresses tonic GABAAR currents, and thus EtOH consumption. We used behavioral, electrophysiological, and immunocytochemical techniques to further explore the relationship between EtOH consumption and GC GABAAR current responses in C57BL/6N (B6N) mice. B6N mice consume significantly less EtOH and achieve significantly lower blood EtOH concentrations than B6J mice, an outcome not mediated by differences in taste. In voltage-clamped GCs, EtOH enhanced the GC tonic current in B6N mice but suppressed it in B6J mice. Immunohistochemical and electrophysiological studies revealed significantly higher nNOS expression and function in the GC layer of B6N mice compared to B6Js. Collectively, our data demonstrate that despite being genetically similar, B6N mice consume significantly less EtOH than B6J mice, a behavioral difference paralleled by increased cerebellar nNOS expression and opposite EtOH action on GC tonic GABAAR currents in each genotype.


Subject(s)
Alcohol Drinking/physiopathology , Alcoholism/physiopathology , Central Nervous System Depressants/pharmacology , Cerebellar Cortex , Electrophysiological Phenomena , Ethanol/pharmacology , Nitric Oxide Synthase Type I , Receptors, GABA-A , Animals , Behavior, Animal/physiology , Central Nervous System Depressants/administration & dosage , Cerebellar Cortex/drug effects , Cerebellar Cortex/metabolism , Disease Models, Animal , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Ethanol/administration & dosage , Male , Mice , Mice, Inbred C57BL/genetics , Nitric Oxide Synthase Type I/drug effects , Nitric Oxide Synthase Type I/metabolism , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Species Specificity
2.
Neuroreport ; 32(3): 228-237, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33470757

ABSTRACT

Memantine has demonstrated beneficial effects on several types of brain insults via therapeutic mechanisms mainly related to its activity as a receptor antagonist of N-methyl-d-aspartate. However, the influences of memantine on intracerebral hemorrhage (ICH) remain obscure. This research probed into the neurovascular protective mechanisms of memantine after ICH and its impacts on neuronal nitric oxide synthase (nNOS) ser1412 phosphorylation. ICH model was established by employing intrastriatal collagenase injection in rats. After modeling, rats were then allocated randomly into sham-operated (sham), vehicle-treated (ICH+V), and memantine-administrated (ICH+M) groups. Memantine (20 mg/kg/day) was intraperitoneally administered 30 min after ICH and thenceforth once daily. Rats were dedicated at 0.25, 6, 12, 24 h, 3 and 7 d post-ICH for measurement of corresponding indexes. Behavioral changes, brain edema, levels of nNOS ser1412 phosphorylation, peroxynitrite, matrix metalloproteinase (MMP)-9, NLRP3, IL-1ß and numbers of dying neurons, as well as the cellular localization of gelatinolytic activity, were detected among the groups. Memantine improved the neurologic deficits and mitigated brain water content, levels of MMP-9, NLRP3, IL-1ß and dying neurons. Additionally, treatment with memantine also reduced nNOS ser1412 phosphorylation and peroxynitrite formation compared with the ICH+V group at 24 h after ICH. In situ zymography simultaneously revealed that gelatinase activity was primarily colocalized with vessel walls and neurons. We concluded that memantine ameliorated blood-brain barrier disruption and neurologic dysfunction in an ICH rat model. The underlying mechanism might involve repression of nNOS ser1412 phosphorylation, as well as peroxynitrite-related MMP-9 and NLRP3 inflammasome activation.


Subject(s)
Blood-Brain Barrier/drug effects , Cerebral Hemorrhage/metabolism , Matrix Metalloproteinase 9/drug effects , Memantine/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type I/drug effects , Peroxynitrous Acid/metabolism , Animals , Brain Edema , Collagenases/toxicity , Disease Models, Animal , Gelatinases/metabolism , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/metabolism , Matrix Metalloproteinase 9/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nitric Oxide Synthase Type I/metabolism , Phosphorylation/drug effects , Rats
3.
Cells ; 9(11)2020 11 23.
Article in English | MEDLINE | ID: mdl-33238404

ABSTRACT

Perlecan is an extracellular matrix molecule anchored to the sarcolemma by a dystrophin-glycoprotein complex. Perlecan-deficient mice are tolerant to muscle atrophy, suggesting that perlecan negatively regulates mechanical stress-dependent skeletal muscle mass. Delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma to the cytosol triggers protein degradation, thereby initiating skeletal muscle atrophy. We hypothesized that perlecan regulates nNOS delocalization and activates protein degradation during this process. To determine the role of perlecan in nNOS-mediated mechanotransduction, we used sciatic nerve transection as a denervation model of gastrocnemius muscles. Gastrocnemius muscle atrophy was significantly lower in perinatal lethality-rescued perlecan-knockout (Hspg2-/--Tg) mice than controls (WT-Tg) on days 4 and 14 following surgery. Immunofluorescence microscopy showed that cell membrane nNOS expression was reduced by denervation in WT-Tg mice, with marginal effects in Hspg2-/--Tg mice. Moreover, levels of atrophy-related proteins-i.e., FoxO1a, FoxO3a, atrogin-1, and Lys48-polyubiquitinated proteins-increased in the denervated muscles of WT-Tg mice but not in Hspg2-/--Tg mice. These findings suggest that during denervation, perlecan promotes nNOS delocalization from the membrane and stimulates protein degradation and muscle atrophy by activating FoxO signaling and the ubiquitin-proteasome system.


Subject(s)
Heparan Sulfate Proteoglycans/therapeutic use , Muscular Atrophy/chemically induced , Nitric Oxide Synthase Type I/drug effects , Animals , Heparan Sulfate Proteoglycans/pharmacology , Humans , Mice , Mice, Knockout
4.
Am J Physiol Gastrointest Liver Physiol ; 319(4): G481-G493, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32812777

ABSTRACT

Diabetic gastroparesis (DG) is a clinical syndrome characterized by delayed gastric emptying (DGE). Loss of nuclear factor erythroid 2-related factor 2 (Nrf2) is associated with reduced neuronal nitric oxide synthase-α (nNOSα)-mediated gastric motility and DGE. Previous studies have shown that nuclear exclusion and inactivation of Nrf2 is partly regulated by glycogen synthase kinase 3ß (GSK-3ß). In the current study, the molecular signaling of GSK-3ß-mediated Nrf2 activation and its mechanistic role on DG were investigated in high-fat diet (HFD)-induced obese/Type 2 diabetes (T2D) female mice. Adult female C57BL/6J mice were fed with HFD or normal diet (ND) with or without GSK-3ß inhibitor (SB 216763, 10 mg/kg body wt ip) start from the 14th wk and continued feeding mice for an additional 3-wk time period. Our results show that treatment with GSK-3ß inhibitor SB attenuated DGE in obese/T2D mice. Treatment with SB restored impaired gastric 1) Nrf2 and phase II antioxidant enzymes through PI3K/ERK/AKT-mediated pathway, 2) tetrahydrobiopterin (BH4, cofactor of nNOS) biosynthesis enzyme dihydrofolate reductase, and 3) nNOSα dimerization in obese/T2 diabetic female mice. SB treatment normalized caspase 3 activity and downstream GSK-3ß signaling in the gastric tissues of the obese/T2 diabetic female mice. In addition, GSK-3ß inhibitor restored impaired nitrergic relaxation in hyperglycemic conditions. Finally, SB treatment reduced GSK3 marker, pTau in adult primary enteric neuronal cells. These findings emphasize the importance of GSK-3ß on regulating gastric Nrf2 and nitrergic mediated gastric emptying in obese/diabetic rodents.NEW & NOTEWORTHY Inhibition of glycogen synthase kinase 3ß (GSK-3ß) with SB 216763 attenuates delayed gastric emptying through gastric nuclear factor erythroid 2-related factor 2 (Nrf2)-phase II enzymes in high-fat diet-fed female mice. SB 216763 restored impaired gastric PI3K/AKT/ ß-catenin/caspase 3 expression. Inhibition of GSK-3ß normalized gastric dihydrofolate reductase, neuronal nitric oxide synthase-α expression, dimerization and nitrergic relaxation. SB 216763 normalized both serum estrogen and nitrate levels in female obese/Type 2 diabetes mice. SB 216763 reduced downstream signaling of GSK-3ß in enteric neuronal cells in vitro.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Gastric Emptying/drug effects , Gastroparesis/drug therapy , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Indoles/pharmacology , Maleimides/pharmacology , Obesity/complications , Animals , Antioxidants/physiology , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 2/complications , Diet, High-Fat , Female , Gastric Emptying/physiology , Gastroparesis/etiology , Glucose/metabolism , Glycogen Synthase Kinase 3 beta/physiology , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/drug effects , Nitric Oxide Synthase Type I/drug effects , Obesity/etiology
5.
Auton Neurosci ; 227: 102675, 2020 09.
Article in English | MEDLINE | ID: mdl-32474374

ABSTRACT

Considering the antioxidant, neuroprotective, inflammatory and nitric oxide modulatory actions of quercetin, the aim of this study was to test the effect of quercetin administration in drinking water (40 mg/day/rat) on neuronal nitric oxide synthase (nNOS), vasoactive intestinal peptide (VIP), overall population of myenteric neurons (HuC/D) and nitric oxide (NO) levels in the jejunal samples from diabetic rats. Male Wistar rats were distributed into four groups (8 rats per group): euglycemic (E), euglycemic administered with quercetin (E+Q), diabetic (D) and diabetic administered with quercetin (D+Q). Rats were induced to diabetes with streptozotocin (35mg/kg/iv) and, after 120 days, the proximal jejunum were collected and processed for immunohistochemical (VIP, nNOS and HuC/D) and chemiluminescence (quantification of tissue NO levels) techniques. Diabetes mellitus reduced the number of nNOS-IR (immunoreactive) (p <0.05) and HuC/D-IR (p <0.001) neurons, however, promoted an increased morphometric area of nNOS-IR neurons (p <0.001) and VIP-IR varicosities (p <0.05). In D+Q group, neuroplasticity effects were observed on HuC/D-IR neurons, accompanied by a reduction of cell body area of neurons nNOS- and VIP-IR varicosities (p <0.05). The NO levels were increased in the E+Q (p <0.05) and D+Q group (p <0.001) compared to the control group. In conclusion, the results showed that quercetin supplementation increased the bioavailability of NO in the jejunum in euglycemic and mitigate the effects of diabetes on nNOS-IR neurons and VIP-IR varicosities in the myenteric plexus of diabetic rats.


Subject(s)
Antioxidants/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Jejunum/drug effects , Myenteric Plexus/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , Nitric Oxide Synthase Type I/drug effects , Nitric Oxide/metabolism , Quercetin/pharmacology , Vasoactive Intestinal Peptide/drug effects , Animals , Antioxidants/administration & dosage , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Male , Myenteric Plexus/pathology , Quercetin/administration & dosage , Rats , Rats, Wistar
6.
Behav Brain Res ; 390: 112671, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32437889

ABSTRACT

Although recent studies have shown that angiotensin (1-7) (Ang [1-7]) exerts anti-stress and anxiolytic-like effects, the underlying mechanisms remain elusive. The ventral hippocampus (VH) is proposed to be a critical brain region for mood and stress management through the N-methyl-d-aspartate receptor (NMDAR) signaling pathway. However, the role of VH NMDAR signaling in the effects of Ang (1-7) remains unclear. In the present study, Ang (1-7) was injected into the bilateral VH of stressed rats, or in combination with a Fyn kinase inhibitor, NMDAR antagonist, neuronal nitric oxide synthase (nNOS) inhibitor, or nitric oxide (NO) scavenger. Anxiety-like behaviors were assessed using the open field test and elevated plus maze test, while alterations in NMDAR-nNOS-NO signaling and serotonergic metabolism were examined in the VH. After 21 days of chronic restraint stress, anxiety-like behaviors were evident. Levels of phosphorylated NR2B (a key NMDAR subunit), its upstream kinase Fyn, as well as activity of nNOS and monoamine oxidase (MAO) were markedly reduced. In contrast, levels of serotonin were increased. Bilateral VH infusion of Ang (1-7) recovered NMDAR-nNOS-NO signaling and MAO-mediated serotonin metabolism, as well as reducing anxiety-like behaviors in stressed rats. These effects were diminished by blockade of MasR (Ang [1-7]-specific receptor), Fyn kinase, NMDAR, nNOS, or NO production. Altogether, these findings indicate that Ang (1-7) exerts anxiolytic effects through modulation of the NMDAR-nNOS-NO pathway and serotonergic metabolism. Future translational research should focus on the relationship between Ang (1-7), glutamatergic neurotransmission, and serotonergic neurotransmission in the VH.


Subject(s)
Angiotensin I/pharmacology , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Anxiety/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Monoamine Oxidase/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Peptide Fragments/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Serotonin/metabolism , Signal Transduction/drug effects , Angiotensin I/administration & dosage , Animals , Anti-Anxiety Agents/administration & dosage , Behavior, Animal/drug effects , Disease Models, Animal , Male , Monoamine Oxidase/drug effects , Nitric Oxide Synthase Type I/drug effects , Peptide Fragments/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects
7.
J Psychopharmacol ; 34(4): 429-440, 2020 04.
Article in English | MEDLINE | ID: mdl-31913077

ABSTRACT

BACKGROUND: Endocannabinoid neurotransmission in the bed nucleus of the stria terminalis is involved in the control of cardiovascular responses to stress. However, the local mechanisms involved is this regulation are not known. AIMS: The purpose of this study was to assess an interaction of bed nucleus of the stria terminalis endocannabinoid neurotransmission with local nitrergic signaling, as well as to investigate the involvement of local N-methyl-D-aspartate glutamate receptor and nitric oxide signaling in the control of cardiovascular responses to acute restraint stress by bed nucleus of the stria terminalis endocannabinoid neurotransmission in rats. METHODS: The first protocol evaluated the effect of intra-bed nucleus of the stria terminalis microinjection of the selective cannabinoid receptor type 1 receptor antagonist AM251 in nitrite/nitrate content in the bed nucleus of the stria terminalis following restraint stress. The other protocols evaluated the impact of local pretreatment with the selective N-methyl-D-aspartate glutamate receptor antagonist LY235959, the selective neuronal nitric oxide synthase inhibitor Nω-propyl-L-arginine, the soluble guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, or the protein kinase G inhibitor KT5823 in restraint-evoked cardiovascular changes following bed nucleus of the stria terminalis treatment with AM251. RESULTS: Bilateral microinjection of AM251 into the bed nucleus of the stria terminalis increased local nitric oxide release during restraint stress. Bed nucleus of the stria terminalis treatment with the cannabinoid receptor type 1 receptor antagonist also enhanced the tachycardia caused by restraint stress, but without affecting arterial pressure increase and sympathetic-mediated cutaneous vasoconstriction. The facilitation of restraint-evoked tachycardia following bed nucleus of the stria terminalis treatment with the cannabinoid receptor type 1 receptor antagonist was completely inhibited by local pretreatment with LY235959, Nω-propyl-L-arginine, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, or KT5823. CONCLUSIONS: Our results provide evidence that bed nucleus of the stria terminalis endocannabinoid neurotransmission inhibits local N-methyl-D-aspartate/neuronal nitric oxide synthase/soluble guanylate cyclase/protein kinase G signaling, and this mechanism is involved in the control of the cardiovascular responses to stress.


Subject(s)
Hemodynamics/drug effects , Receptor, Cannabinoid, CB1/drug effects , Septal Nuclei/drug effects , Signal Transduction/drug effects , Stress, Psychological/complications , Stress, Psychological/drug therapy , Animals , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/drug effects , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/drug effects , Male , Microinjections , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/drug effects , Piperidines/administration & dosage , Piperidines/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects , Restraint, Physical , Synaptic Transmission/drug effects
8.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 54(3): 188-193, 2019 Mar 09.
Article in Chinese | MEDLINE | ID: mdl-30856697

ABSTRACT

Objective: To investigate the effects of estrogen and remifemin on the expression of neuronal nitric oxide synthase (nNOS), transient receptor potential vanilloid 1 (TRPV1), muscarinic acetylcholine receptor, member 1 and 3 (M1 and M3 receptor) and acetylcholinesterase (AChE) in the submandibular gland of rats. Methods: Forty SD female adult rats were divided into SHAM group (sham operation), OVX group (ovarian removal), OVX+E group (ovarian removal + estrogen treatment) and OVX+ICR group (ovarian removal + remifemin treatment), 10 per group. The rats were recovered for 2 weeks after operation. The SHAM group and the OVX group were treated with distilled water, the OVX+E group and the OVX+ICR group were treated with ß-estradiol and remifemin respectively. After 4 weeks, the location and expression of nNOS, TRPV1, M1 and M3 receptors in the submandibular gland were evaluated by immunohistochemistry. The changes of AChE expression in rat submandibular gland were observed by AChE staining. Results: Compared with SHAM group (0.23±0.02, 0.28±0.01, 0.25±0.03, 0.19±0.03), the expression of nNOS, TRPV1, M1 and M3 receptors in OVX group (0.16±0.01, 0.21±0.01, 0.15±0.02, 0.09±0.02) were significantly lower (P<0.05); there were no significant difference between OVX+E group (0.23±0.01, 0.28±0.02, 0.23±0.03, 0.19±0.01) and SHAM group (P>0.05). But compared with OVX group, the expression of nNOS, TRPV1 and M3 receptors in OVX+ICR group were no significantly changed (P>0.05), and only M1 receptor expression (0.22±0.03) was significantly increased (P<0.05). The expression of AChE in OVX group (0.14±0.01) was significantly higher than that in SHAM group (0.10±0.01) (P<0.05). The expression of AChE in OVX+E group (0.15±0.01) was significantly higher than that in SHAM group (P<0.05). The expression of AChE in OVX+ICR group (0.09±0.01) was not significantly different from that in SHAM group (P>0.05). Conclusions: Estrogen can significantly increase the expression of nNOS and TRPV1 in the submandibular gland of rats, suggesting that estrogen may regulate the salivary secretion function of the submandibular gland through nNOS and TRPV1. The mechanism of remifemin is different from that of estrogen, and remifemin does not play a regulatory role by nNOS and TRPV1.


Subject(s)
Cimicifuga , Estrogens , Nitric Oxide Synthase Type I , Plant Extracts , Submandibular Gland , Animals , Estradiol , Estrogens/pharmacology , Female , Nitric Oxide , Nitric Oxide Synthase Type I/drug effects , Nitric Oxide Synthase Type I/metabolism , Ovariectomy , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley , Submandibular Gland/drug effects , Submandibular Gland/metabolism , TRPV Cation Channels/drug effects , TRPV Cation Channels/metabolism
9.
Neurosci Bull ; 35(1): 79-90, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30617765

ABSTRACT

Chronic intermittent hypobaric hypoxia (CIHH) is known to have an anti-hypertensive effect, which might be related to modulation of the baroreflex in rats with renal vascular hypertension (RVH). In this study, RVH was induced by the 2-kidney-1-clip method (2K1C) in adult male Sprague-Dawley rats. The rats were then treated with hypobaric hypoxia simulating 5000 m altitude for 6 h/day for 28 days. The arterial blood pressure (ABP), heart rate (HR), and renal sympathetic nerve activity (RSNA) were measured before and after microinjection of L-arginine into the nucleus tractus solitarii (NTS) in anesthetized rats. Evoked excitatory postsynaptic currents (eEPSCs) and spontaneous EPSCs (sEPSCs) were recorded in anterogradely-labeled NTS neurons receiving baroreceptor afferents. We measured the protein expression of neuronal nitric oxide synthase (nNOS) and endothelial NOS (eNOS) in the NTS. The results showed that the ABP in RVH rats was significantly lower after CIHH treatment. The inhibition of ABP, HR, and RSNA induced by L-arginine was less in RVH rats than in sham rats, and greater in the CIHH-treated RVH rats than the untreated RVH rats. The eEPSC amplitude in NTS neurons receiving baroreceptor afferents was lower in the RVH rats than in the sham rats and recovered after CIHH. The protein expression of nNOS and eNOS in the NTS was lower in the RVH rats than in the sham rats and this decrease was reversed by CIHH. In short, CIHH treatment decreases ABP in RVH rats via up-regulating NOS expression in the NTS.


Subject(s)
Hypertension/metabolism , Hypoxia/chemically induced , Nitric Oxide Synthase Type I/metabolism , Solitary Nucleus/metabolism , Animals , Baroreflex/physiology , Blood Pressure/drug effects , Kidney/drug effects , Kidney/metabolism , Male , Nitric Oxide Synthase Type I/drug effects , Rats, Sprague-Dawley
10.
Neurochem Res ; 44(2): 421-427, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30523577

ABSTRACT

Under pathological conditions, nitric oxide can become a mediator of oxidative cellular damage, generating an unbalance between oxidant and antioxidant systems. The participation of neuronal nitric oxide synthase (nNOS) in the neurodegeneration mechanism has been reported; the activation of N-methyl-D-aspartate (NMDA) receptors by agonist quinolinic acid (QUIN) triggers an increase in nNOS function and promotes oxidative stress. The aim of the present work was to elucidate the participation of nNOS in QUIN-induced oxidative stress in knock-out mice (nNOS-/-). To do so, we microinjected saline solution or QUIN in the striatum of wild-type (nNOS +/+), heterozygote (nNOS+/-), and knock-out (nNOS-/-) mice, and measured circling behavior, GABA content levels, oxidative stress, and NOS expression and activity. We found that the absence of nNOS provides a protection against striatal oxidative damage induced by QUIN, resulting in decreased circling behavior, oxidative stress, and a partial protection reflected in GABA depletion. We have shown that nNOS-derived NO is involved in neurological damage induced by oxidative stress in a QUIN-excitotoxic model.


Subject(s)
Corpus Striatum/drug effects , Nitric Oxide Synthase Type I/drug effects , Oxidative Stress/drug effects , Quinolinic Acid/pharmacology , Animals , Antioxidants/pharmacology , Corpus Striatum/metabolism , Lipid Peroxidation/drug effects , Male , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I/metabolism , Quinolinic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism
11.
BMC Neurosci ; 19(1): 50, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30103682

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) is one of the leading causes of disability and chronic pain. In SCI-induced pathology, homeostasis of the nitric oxide (NO) metabolome is lost. Major NO metabolites such as S-nitrosoglutathione (GSNO) and peroxynitrite are reported to play pivotal roles in regulating the activities of key cysteine proteases, calpains. While peroxynitrite (a metabolite of NO and superoxide) up regulates the activities of calpains leading to neurodegeneration, GSNO (a metabolite of NO and glutathione) down regulates the activities of calpains leading to neuroprotection. In this study, effect of GSNO on locomotor function and pain threshold and their relationship with the levels of peroxynitrite and the activity of calpain in the injured spinal cord were investigated using a 2-week rat model of contusion SCI. RESULTS: SCI animals were initially treated with GSNO at 2 h after the injury followed by a once daily dose of GSNO for 14 days. Locomotor function was evaluated by "Basso Beattie and Bresnahan (BBB) locomotor rating scale" and pain by mechanical allodynia. Peroxynitrite level, as expression of 3-nitrotyrosine (3-NT), calpain activity, as the degradation products of calpain substrate alpha II spectrin, and nNOS activity, as the expression phospho nNOS, were measured by western blot analysis. Treatment with GSNO improved locomotor function and mitigated pain. The treatment also reduced the levels of peroxynitrite (3-NT) and decreased activity of calpains. Reduced levels of peroxynitrite resulted from the GSNO-mediated inhibition of aberrant activity of neuronal nitric oxide synthase (nNOS). CONCLUSIONS: The data indicates that higher levels of 3-NT and aberrant activities of nNOS and calpains correlated with SCI pathology and functional deficits. Treatment with GSNO improved locomotor function and mitigated mechanical allodynia acutely post-injury. Because GSNO shows potential to ameliorate experimental SCI, we discuss implications for GSNO therapy in clinical SCI research.


Subject(s)
Calpain/metabolism , Nitrosoguanidines/pharmacology , Peroxynitrous Acid/metabolism , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Animals , Brain/drug effects , Disease Models, Animal , Male , Neurons/drug effects , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type I/drug effects , Rats, Sprague-Dawley
12.
Neurochem Res ; 43(11): 2025-2037, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30145742

ABSTRACT

Epilepsy is the third most common chronic brain disorder. Modafinil is an awakening agent approved for narcolepsy. In addition to its clinical uses some reports revealed that modafinil was associated with some alterations in seizure threshold. The purpose of this study was to clarify the effect of acute administration of modafinil in clonic seizure threshold (CST) induced by pentylenetetrazole in mice and the involvement of glutamate, nitric oxide, gamma amino butyric acid (GABA), and serotonin systems in this feature. Modafinil at 80 and 150 mg/kg showed anti- and pro-convulsant effects respectively and expressed maximum anti- and pro-convulsant activities at 30 min after injection. Both modulatory effects were blunted by pretreatment of L-NAME [nonspecific nitric oxide synthase (NOS) inhibitor; 10 mg/kg, i.p.], 7-nitroindazole (a neuronal NOS inhibitor; 40 mg/kg, i.p.), and aminoguanidine (an inducible NOS inhibitor; 50 mg/kg, i.p.). Injection of the NOS precursor L-arginine (60 mg/kg, i.p.) before modafinil did not change the anti-convulsant effect, while thoroughly reversed the pro-convulsant one. Our experiments displayed that administration of diazepam (a GABAA receptor agonist; 0.02 mg/kg, i.p.) and MK-801 (a NMDA receptor antagonist; 0.05 mg/kg, i.p.) before different doses of modafinil significantly increased CST. Finally, pretreatment of citalopram (a selective serotonin reuptake inhibitor; 0.1 mg/kg, i.p.) did not modify the convulsant activities of modafinil. Therefore, nitric oxide system may mediate anti-convulsant activity, while glutamate, nitric oxide, and GABA pathways may involve in pro-convulsant property. Serotonin receptors have no role on convulsant effects of modafinil.


Subject(s)
Glutamic Acid/metabolism , Modafinil/pharmacology , Nitric Oxide Synthase Type I/drug effects , Nitric Oxide/metabolism , Pentylenetetrazole/pharmacology , Serotonin/metabolism , Animals , Anticonvulsants/pharmacology , Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Male , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/metabolism , Seizures/chemically induced
13.
Am J Obstet Gynecol ; 219(1): 113.e1-113.e9, 2018 07.
Article in English | MEDLINE | ID: mdl-29709511

ABSTRACT

BACKGROUND: Maternal inflammation is a risk factor for neonatal brain injury and future neurological deficits. Pomegranates have been shown to exhibit anti-inflammatory, anti-apoptotic and anti-oxidant activities. OBJECTIVE: We hypothesized that pomegranate juice (POM) may attenuate fetal brain injury in a rat model of maternal inflammation. STUDY DESIGN: Pregnant rats (24 total) were randomized for intraperitoneal lipopolysaccharide (100 µg/kg) or saline at time 0 at 18 days of gestation. From day 11 of gestation, 12 dams were provided ad libitum access to drinking water, and 12 dams were provided ad libitum access to drinking water with pomegranate juice (5 mL per day), resulting in 4 groups of 6 dams (saline/saline, pomegranate juice/saline, saline/lipopolysaccharide, pomegranate juice/lipopolysaccharide). All dams were sacrificed 4 hours following the injection and maternal blood and fetal brains were collected from the 4 treatment groups. Maternal interleukin-6 serum levels and fetal brain caspase 3 active form, nuclear factor-κB p65, neuronal nitric oxide synthase (phosphoneuronal nitric oxide synthase), and proinflammatory cytokine levels were determined by enzyme-linked immunosorbent assay and Western blot. RESULTS: Maternal lipopolysaccharide significantly increased maternal serum interleukin-6 levels (6039 ± 1039 vs 66 ± 46 pg/mL; P < .05) and fetal brain caspase 3 active form, nuclear factor-κB p65, phosphoneuronal nitric oxide synthase, and the proinflammatory cytokines compared to the control group (caspase 3 active form 0.26 ± 0.01 vs 0.20 ± 0.01 U; nuclear factor-κB p65 0.24 ± 0.01 vs 0.1 ± 0.01 U; phosphoneuronal nitric oxide synthase 0.23 ± 0.01 vs 0.11 ± 0.01 U; interleukin-6 0.25 ± 0.01 vs 0.09 ± 0.01 U; tumor necrosis factor-α 0.26 ± 0.01 vs 0.12 ± 0.01 U; chemokine (C-C motif) ligand 2 0.23 ± 0.01 vs 0.1 ± 0.01 U). Maternal supplementation of pomegranate juice to lipopolysaccharide-exposed dams (pomegranate juice/lipopolysaccharide) significantly reduced maternal serum interleukin-6 levels (3059 ± 1121 pg/mL, fetal brain: caspase 3 active form (0.2 ± 0.01 U), nuclear factor-κB p65 (0.22 ± 0.01 U), phosphoneuronal nitric oxide synthase (0.19 ± 0.01 U) as well as the brain proinflammatory cytokines (interleukin-6, tumor necrosis factor-α and chemokine [C-C motif] ligand 2) compared to lipopolysaccharide group. CONCLUSION: Maternal pomegranate juice supplementation may attenuate maternal inflammation-induced fetal brain injury. Pomegranate juice neuroprotective effects might be secondary to the suppression of both the maternal inflammatory response and inhibition of fetal brain apoptosis, neuronal nitric oxide synthase, and nuclear factor-κB activation.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Fetus/drug effects , Fruit and Vegetable Juices , Lipopolysaccharides/pharmacology , Lythraceae , Nitric Oxide Synthase Type I/drug effects , Transcription Factor RelA/drug effects , Animals , Antioxidants , Apoptosis/immunology , Brain/immunology , Brain/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Chemokine CCL2/drug effects , Chemokine CCL2/immunology , Cytokines/drug effects , Cytokines/immunology , Dietary Supplements , Female , Fetus/immunology , Fetus/metabolism , Inflammation , Interleukin-6/immunology , NF-kappa B/drug effects , NF-kappa B/immunology , NF-kappa B/metabolism , Nitric Oxide Synthase Type I/immunology , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Pregnancy , Rats , Transcription Factor RelA/immunology , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/immunology
14.
Transl Psychiatry ; 7(4): e1078, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28375208

ABSTRACT

The aim of this study was to identify molecular pathways related to antidepressant response. We administered paroxetine to the DBA/2J mice for 28 days. Following the treatment, the mice were grouped into responders or non-responders depending on the time they spent immobile in the forced swim test. Hippocampal metabolomics and proteomics analyses revealed that chronic paroxetine treatment affects glutamate-related metabolite and protein levels differentially in the two groups. We found significant differences in the expression of N-methyl-d-aspartate receptor and neuronal nitric oxide synthase proteins between the two groups, without any significant alterations in the respective transcript levels. In addition, we found that chronic paroxetine treatment altered the levels of proteins associated with the ubiquitin-proteasome system (UPS). The soluble guanylate cyclase-ß1, proteasome subunit α type-2 and ubiquitination levels were also affected in peripheral blood mononuclear cells from antidepressant responder and non-responder patients suffering from major depressive disorder. We submit that the glutamatergic system and UPS have a crucial role in the antidepressant treatment response in both mice and humans.


Subject(s)
Antidepressive Agents/metabolism , Depressive Disorder, Major/metabolism , Glutamic Acid/metabolism , Nitric Oxide Synthase Type I/drug effects , Paroxetine/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Hippocampus/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Male , Metabolomics , Mice , Mice, Inbred DBA , Nitric Oxide Synthase Type I/metabolism , Paroxetine/administration & dosage , Paroxetine/pharmacology , Proteomics , Receptors, N-Methyl-D-Aspartate/metabolism , Swimming/physiology , Ubiquitin
15.
Neuroscience ; 349: 303-317, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28285942

ABSTRACT

Excessive activation of NMDA receptor (NMDAR) signaling within the spinal dorsal horn contributes to central sensitization and the induction and maintenance of pathological pain states. However, direct antagonism of NMDARs produces undesirable side effects which limit their clinical use. NMDAR activation produces central sensitization, in part, by initiating a signaling cascade that activates the enzyme neuronal nitric oxide synthase (nNOS) and generates the signaling molecule nitric oxide. NMDAR-mediated activation of nNOS requires a scaffolding protein, postsynaptic density protein 95kDa (PSD95), which tethers nNOS to NMDARs. Thus, disrupting the protein-protein interaction between PSD95 and nNOS may inhibit pro-nociceptive signaling mechanisms downstream of NMDARs and suppress central sensitization while sparing unwanted side effects associated with NMDAR antagonists. We examined the impact of small molecule PSD95-nNOS protein-protein interaction inhibitors (ZL006, IC87201) on both nociceptive behavior and formalin-evoked Fos protein expression within the lumbar spinal cord of rats. Comparisons were made with ZL007, an inactive analog of ZL006, and the NMDAR antagonist MK-801. IC87201 and ZL006, but not ZL007, suppressed phase 2 of formalin-evoked pain behavior and decreased the number of formalin-induced Fos-like immunoreactive cells in spinal dorsal horn regions associated with nociceptive processing. MK-801 suppressed Fos protein expression in both dorsal and ventral horns. MK-801 produced motor ataxia in the rotarod test whereas IC87201 and ZL006 failed to do so. ZL006 but not ZL007 suppressed paclitaxel-induced mechanical and cold allodynia in a model of chemotherapy-induced neuropathic pain. Co-immunoprecipitation experiments revealed the presence of the PSD95-nNOS complex in lumbar spinal cord of paclitaxel-treated rats, although ZL006 did not reliably disrupt the complex in all subjects. The present findings validate use of putative small molecule PSD95-nNOS protein-protein interaction inhibitors as novel analgesics and demonstrate, for the first time, that these inhibitors suppress inflammation-evoked neuronal activation at the level of the spinal dorsal horn.


Subject(s)
Behavior, Animal/drug effects , Disks Large Homolog 4 Protein/antagonists & inhibitors , Dizocilpine Maleate/pharmacology , Nitric Oxide Synthase Type I/drug effects , Nociceptors/drug effects , Spinal Cord/drug effects , Animals , Formaldehyde/pharmacology , Male , Nitric Oxide Synthase Type I/metabolism , Nociceptors/metabolism , Rats, Sprague-Dawley , Spinal Cord/metabolism
16.
J Headache Pain ; 17(1): 99, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27778243

ABSTRACT

BACKGROUND: Antioxidants have been proven to weaken hyperalgesia in neuropathic pain. Endogenous antioxidant defense system may have a role in the prevention of hyperalgesia in migraine. In this study, we aimed to evaluate the role of nuclear factor E2-related factor 2/antioxidant response element (Nrf2/ARE) pathway in regulating the activation of the trigeminovascular system (TGVS) and hypersensitivity in nitroglycerin (NTG)-induced hyperalgesia rats. METHODS: The expression levels of Nrf2, HO, HO1, and NQO1 in the trigeminal nucleus caudalis (TNC) were detected by western blot. Immunofluorescence was used to demonstrate the cell-specific localization of Nrf2 in TNC. Sulforaphane, a Nrf2 activator, was administered to NTG-induced rats. Then, the number of c-Fos- and nNOS-immunoreactive neurons in TNC was evaluated using immunofluorescence, and c-Fos and nNOS protein levels were quantified using western blot. Von Frey hair testing was used to evaluate the tactile thresholds of rats at different time points in different groups. RESULTS: Total cellular and nuclear levels of the proteins Nrf2, HO1, and NQO1 were elevated in TNC after NTG injection, and Nrf2 was found to be located in the nucleus and cytoplasm of the neurons. Sulforaphane pretreatment significantly increased the nuclear Nrf2, HO1, and NQO1 levels in TNC. In addition, sulforaphane exposure effectively inhibited the expression of nNOS and c-Fos, reduced the number of nNOS and c-Fos immunoreactive neurons in TNC, and attenuated the tactile thresholds induced by NTG injection. CONCLUSION: Oxidative stress was involved in nitroglycerin-induced hyperalgesia. Activation of the Nrf2/ARE pathway inhibited the activation of TGVS and prevented the induction of hyperalgesia. Sulforaphane might therefore be an effective agent for hyperalgesia. Further studies are needed to discover the underlying mechanisms of the process.


Subject(s)
Antioxidant Response Elements/drug effects , Hyperalgesia/metabolism , NF-E2-Related Factor 2/drug effects , Neurons/drug effects , Proto-Oncogene Proteins c-fos/drug effects , Trigeminal Caudal Nucleus/drug effects , Animals , Anticarcinogenic Agents/pharmacology , Heme Oxygenase (Decyclizing)/drug effects , Heme Oxygenase (Decyclizing)/metabolism , Hyperalgesia/chemically induced , Isothiocyanates/pharmacology , Male , Migraine Disorders/metabolism , NAD(P)H Dehydrogenase (Quinone)/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Neurons/metabolism , Nitric Oxide Synthase Type I/drug effects , Nitric Oxide Synthase Type I/metabolism , Nitroglycerin/pharmacology , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Sulfoxides , Trigeminal Caudal Nucleus/metabolism , Vasodilator Agents/pharmacology
17.
Am J Physiol Endocrinol Metab ; 311(2): E335-45, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27221119

ABSTRACT

It has been argued whether insulin accelerates or prevents atherosclerosis. Although results from in vitro studies have been conflicting, recent in vivo mice studies demonstrated antiatherogenic effects of insulin. Insulin is a known activator of endothelial nitric oxide synthase (NOS), leading to increased production of NO, which has potent antiatherogenic effects. We aimed to examine the role of NOS in the protective effects of insulin against atherosclerosis. Male apolipoprotein E-null mice (8 wk old) fed a high-cholesterol diet (1.25% cholesterol) were assigned to the following 12-wk treatments: control, insulin (0.05 U/day via subcutaneous pellet), N(ω)-nitro-l-arginine methyl ester hydrochloride (l-NAME, via drinking water at 100 mg/l), and insulin plus l-NAME. Insulin reduced atherosclerotic plaque burden in the descending aorta by 42% compared with control (plaque area/aorta lumen area: control, 16.5 ± 1.9%; insulin, 9.6 ± 1.3%, P < 0.05). Although insulin did not decrease plaque burden in the aortic sinus, macrophage accumulation in the plaque was decreased by insulin. Furthermore, insulin increased smooth muscle actin and collagen content and decreased plaque necrosis, consistent with increased plaque stability. In addition, insulin treatment increased plasma NO levels, decreased inducible NOS staining, and tended to increase phosphorylated vasodilator-stimulated phosphoprotein staining in the plaques of the aortic sinus. All these effects of insulin were abolished by coadministration of l-NAME, whereas l-NAME alone showed no effect. Insulin also tended to increase phosphorylated endothelial NOS and total neuronal NOS staining, effects not modified by l-NAME. In conclusion, we demonstrate that insulin treatment decreases atherosclerotic plaque burden and increases plaque stability through NOS-dependent mechanisms.


Subject(s)
Aorta/drug effects , Enzyme Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/drug effects , Plaque, Atherosclerotic/metabolism , Actins/drug effects , Actins/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Collagen/drug effects , Collagen/metabolism , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Knockout , Necrosis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I/drug effects , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/drug effects , Nitric Oxide Synthase Type III/metabolism , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Plaque, Atherosclerotic/pathology , Sinus of Valsalva/drug effects , Sinus of Valsalva/metabolism , Sinus of Valsalva/pathology
18.
PLoS One ; 11(1): e0147634, 2016.
Article in English | MEDLINE | ID: mdl-26799743

ABSTRACT

UNLABELLED: Altered neuronal nitric oxide synthase function in Duchenne muscular dystrophy leads to impaired mitochondrial function which is thought to be one cause of muscle damage in this disease. The study tested if increased intramuscular nitric oxide concentration can improve mitochondrial energy metabolism in Duchenne muscular dystrophy using a novel therapeutic approach through the combination of L-arginine with metformin. Five ambulatory, genetically confirmed Duchenne muscular dystrophy patients aged between 7­10 years were treated with L-arginine (3 x 2.5 g/d) and metformin (2 x 250 mg/d) for 16 weeks. Treatment effects were assessed using mitochondrial protein expression analysis in muscular biopsies, indirect calorimetry, Dual-Energy X-Ray Absorptiometry, quantitative thigh muscle MRI, and clinical scores of muscle performance. There were no serious side effects and no patient dropped out. Muscle biopsy results showed pre-treatment a significantly reduced mitochondrial protein expression and increased oxidative stress in Duchenne muscular dystrophy patients compared to controls. Post-treatment a significant elevation of proteins of the mitochondrial electron transport chain was observed as well as a reduction in oxidative stress. Treatment also decreased resting energy expenditure rates and energy substrate use shifted from carbohydrates to fatty acids. These changes were associated with improved clinical scores. In conclusion pharmacological stimulation of the nitric oxide pathway leads to improved mitochondria function and clinically a slowing of disease progression in Duchenne muscular dystrophy. This study shall lead to further development of this novel therapeutic approach into a real alternative for Duchenne muscular dystrophy patients. TRIAL REGISTRATION: ClinicalTrials.gov NCT02516085.


Subject(s)
Arginine/therapeutic use , Metformin/therapeutic use , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Arginine/administration & dosage , Biopsy , Child , Drug Therapy, Combination , Humans , Magnetic Resonance Imaging , Metformin/administration & dosage , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Nitric Oxide Synthase Type I/drug effects , Pilot Projects
19.
Neurogastroenterol Motil ; 28(1): 101-15, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26526599

ABSTRACT

BACKGROUND: Cachexia is a significant problem in patients with cancer. The effect of cancer on interstitial cells of Cajal (ICC) and neurons of the gastrointestinal tract have not been studied previously. Although supplementation with L-glutamine 2% may have beneficial effects in cancer-related cachexia, and be protective of ICC in models of oxidative stress such as diabetes, its effects on ICC in cancer have also not been studied. METHODS: Twenty-eight male Wistar rats were divided into four groups: control (C), control supplemented with L-glutamine (CG), Walker 256 tumor (WT), and Walker 256 tumor supplemented with L-glutamine (WTG). Rats were implanted with tumor cells or injected with saline in the right flank. After 14 days, the jejunal tissues were collected and processed for immunohistochemical techniques including whole mounts and cryosections and Western blot analysis. KEY RESULTS: Tumor-bearing rats demonstrate reduced numbers of Myenteric ICC and deep muscular plexus ICC and yet increased Ano1 protein expression and enhanced ICC networks. In addition, there is more nNOS protein expressed in tumor-bearing rats compared to controls. L-glutamine treatment had a variety of effects on ICC that may be related to the disease state and the interaction of ICC and nNOS neurons. Regardless, L-glutamine reduced the size of tumors and also tumor-induced cachexia that was not due to altered food intake. CONCLUSIONS & INFERENCES: There are significant effects on ICC in the Walker 256 tumor model. Although supplementation with L-glutamine has differential and complex effects of ICC, it reduces tumor size and tumor-associated cachexia, which supports its beneficial therapeutic role in cancer.


Subject(s)
Cachexia/metabolism , Carcinoma 256, Walker/metabolism , Chloride Channels/drug effects , Glutamine/pharmacology , Interstitial Cells of Cajal/drug effects , Jejunum/drug effects , Myenteric Plexus/drug effects , Nitric Oxide Synthase Type I/drug effects , Oxidative Stress/drug effects , Animals , Anoctamin-1 , Blotting, Western , Carcinoma 256, Walker/pathology , Chloride Channels/metabolism , Immunohistochemistry , Interstitial Cells of Cajal/metabolism , Male , Myenteric Plexus/cytology , Nitric Oxide Synthase Type I/metabolism , Rats , Rats, Wistar , Tumor Burden
20.
BMC Neurosci ; 16: 61, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26443997

ABSTRACT

BACKGROUND: It is well documented that the nitric oxide (NO) might be directly involved in brain response to hypobaric hypoxia, and could contribute to memory deficiencies. Recent studies have shown that melatonin could attenuate hypoxia or ischemia-induced nerve injuries by decreasing the production of free radicals. The present study, using immunohistochemical and immunoblot methods, aimed to explore whether melatonin treatment may affect the expression of nitric oxide system and protein nitration, and provide neuroprotection in the rat hippocampus injured by hypobaric hypoxia. Prior to hypoxic treatment, adult rats were pretreated with melatonin (100 mg/kg, i.p.) before they were exposed to the altitude chamber with 48 Torr of the partial oxygen concentration (pO2) for 7 h to mimic the ambience of being at 9000 m in height. They were then sacrificed after 0 h, 1, and 3 days of reoxygenation. RESULTS: The results obtained from the immunohistochemical and immunoblotting analyses showed that the expressions of neuronal nitric oxide synthase (nNOS), endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS), nitrotyrosine (Ntyr) and Caspase 3 in the hypoxic hippocampus were increased from 0 h to 3 days of reoxygenation. Interestingly, the hypoxia-induced increase of nNOS, eNOS, iNOS, Ntyr and Caspase 3 protein expression was significantly depressed in the hypoxic rats treated with melatonin. CONCLUSIONS: Activation of the nitric oxide system and protein nitration constitutes a hippocampal response to hypobaric hypoxia and administration of melatonin could provide new therapeutic avenues to prevent and/or treat the symptoms produced by hypobaric hypoxia.


Subject(s)
Altitude Sickness/drug therapy , Antioxidants/pharmacology , Caspase 3/metabolism , Hippocampus/metabolism , Hypoxia/drug therapy , Melatonin/pharmacology , Neurons/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Tyrosine/analogs & derivatives , Animals , Antioxidants/administration & dosage , Caspase 3/drug effects , Disease Models, Animal , Hippocampus/drug effects , Hypoxia/etiology , Male , Melatonin/administration & dosage , Neurons/drug effects , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase Type I/drug effects , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/drug effects , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/drug effects , Nitric Oxide Synthase Type III/metabolism , Rats , Rats, Wistar , Tyrosine/drug effects , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL