Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.665
Filtrer
1.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-39000048

RÉSUMÉ

Bisphenols are dangerous endocrine disruptors that pollute the environment. Due to their chemical properties, they are globally used to produce plastics. Structural similarities to oestrogen allow bisphenols to bind to oestrogen receptors and affect internal body systems. Most commonly used in the plastic industry is bisphenol A (BPA), which also has negative effects on the nervous, immune, endocrine, and cardiovascular systems. A popular analogue of BPA-bisphenol S (BPS) also seems to have harmful effects similar to BPA on living organisms. Therefore, with the use of double immunofluorescence labelling, this study aimed to compare the effect of BPA and BPS on the enteric nervous system (ENS) in mouse jejunum. The study showed that both studied toxins impact the number of nerve cells immunoreactive to substance P (SP), galanin (GAL), vasoactive intestinal polypeptide (VIP), the neuronal isoform of nitric oxide synthase (nNOS), and vesicular acetylcholine transporter (VAChT). The observed changes were similar in the case of both tested bisphenols. However, the influence of BPA showed stronger changes in neurochemical coding. The results also showed that long-term exposure to BPS significantly affects the ENS.


Sujet(s)
Composés benzhydryliques , Système nerveux entérique , Jéjunum , Phénols , Sulfones , Animaux , Phénols/toxicité , Composés benzhydryliques/toxicité , Souris , Jéjunum/effets des médicaments et des substances chimiques , Jéjunum/métabolisme , Système nerveux entérique/effets des médicaments et des substances chimiques , Système nerveux entérique/métabolisme , Sulfones/pharmacologie , Sulfones/toxicité , Substance P/métabolisme , Peptide vasoactif intestinal/métabolisme , Transporteurs vésiculaires de l'acétylcholine/métabolisme , Mâle , Galanine/métabolisme , Perturbateurs endocriniens/toxicité , Perturbateurs endocriniens/pharmacologie , Nitric oxide synthase type I/métabolisme
2.
Front Biosci (Landmark Ed) ; 29(5): 190, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38812321

RÉSUMÉ

Nitric oxide synthases (NOS) are essential regulators of vascular function, and their role in ocular blood vessels is of paramount importance for maintaining ocular homeostasis. Three isoforms of NOS-endothelial (eNOS), neuronal (nNOS), and inducible (iNOS)-contribute to nitric oxide production in ocular tissues, exerting multifaceted effects on vascular tone, blood flow, and overall ocular homeostasis. Endothelial NOS, primarily located in endothelial cells, is pivotal for mediating vasodilation and regulating blood flow. Neuronal NOS, abundantly found in nerve terminals, contributes to neurotransmitter release and vascular tone modulation in the ocular microvasculature. Inducible NOS, expressed under inflammatory conditions, plays a role in response to pathological stimuli. Understanding the distinctive contributions of these NOS isoforms in retinal blood vessels is vital to unravel the mechanisms underlying various ocular diseases, such diabetic retinopathy. This article delves into the unique contributions of NOS isoforms within the complex vascular network of the retina, elucidating their significance as potential therapeutic targets for addressing pathological conditions.


Sujet(s)
Nitric oxide synthase , Vaisseaux rétiniens , Humains , Vaisseaux rétiniens/métabolisme , Vaisseaux rétiniens/physiopathologie , Animaux , Nitric oxide synthase/métabolisme , Nitric oxide synthase type III/métabolisme , Nitric oxide synthase type I/métabolisme , Monoxyde d'azote/métabolisme , Rétinopathie diabétique/physiopathologie , Rétinopathie diabétique/enzymologie , Rétinopathie diabétique/métabolisme , Nitric oxide synthase type II/métabolisme
3.
Food Chem Toxicol ; 189: 114763, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38797315

RÉSUMÉ

Monosodium glutamate (MSG) administration has been shown to pronounce hypertension and oxidative status with increased renal blood flow (RBF), however, the precise mechanisms of action have never been demonstrated. This study aimed to investigate the MSG action by studying the alteration in renal architecture and specific protein expression in 2-kidney-1-clip hypertensive comparing to sham operative normotensive rats. The administered doses of MSG were 80, 160, or 320 mg/kg BW daily for 8 weeks. Using routine chemical staining, the congestion of glomerular capillaries, a lesser renal corpuscles and glomeruli size, a widen Bowman capsule's space, an increase in mesangial cell proliferation and mesangial matrix, renal interstitial fibrosis, focal cloudy swelling of renal tubular epithelial cells were observed. Immunological study revealed an increase in the expression of N-methyl-D-aspartate receptor (NMDA-R) and endothelial nitric oxide synthase (eNOS) but a decrease in neuronal NOS (nNOS). It is suggested that MSG may upregulate the NMDA-R levels which responsible for the oxidative stress, glomerular injury, and renal interstitial fibrosis. The NMDA-R may also stimulate eNOS overexpression which resulted in renal microvascular dilatation, a raise in RBF and GFR, and natriuresis and diuresis promotion. Long-term exposure of MSG may trigger adaptation of tubuloglomerular feedback through nNOS downregulation.


Sujet(s)
Hypertension artérielle , Rein , Nitric oxide synthase type III , Nitric oxide synthase type I , Récepteurs du N-méthyl-D-aspartate , Glutamate de sodium , Animaux , Nitric oxide synthase type III/métabolisme , Glutamate de sodium/toxicité , Récepteurs du N-méthyl-D-aspartate/métabolisme , Récepteurs du N-méthyl-D-aspartate/génétique , Nitric oxide synthase type I/métabolisme , Mâle , Rats , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Rein/anatomopathologie , Hypertension artérielle/induit chimiquement , Hypertension artérielle/physiopathologie , Hypertension artérielle/métabolisme , Rat Wistar
4.
Biochemistry ; 63(11): 1395-1411, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38747545

RÉSUMÉ

Nitric oxide synthase (NOS) in mammals is a family of multidomain proteins in which interdomain electron transfer (IET) is controlled by domain-domain interactions. Calmodulin (CaM) binds to the canonical CaM-binding site in the linker region between the FMN and heme domains of NOS and allows tethered FMN domain motions, enabling an intersubunit FMN-heme IET in the output state for NO production. Our previous cross-linking mass spectrometric (XL MS) results demonstrated site-specific protein dynamics in the CaM-responsive regions of rat neuronal NOS (nNOS) reductase construct, a monomeric protein [Jiang et al., Biochemistry, 2023, 62, 2232-2237]. In this work, we have extended our combined approach of XL MS structural mapping and AlphaFold structural prediction to examine the homodimeric nNOS oxygenase/FMN (oxyFMN) construct, an established model of the NOS output state. We employed parallel reaction monitoring (PRM) based quantitative XL MS (qXL MS) to assess the CaM-induced changes in interdomain dynamics and interactions. Intersubunit cross-links were identified by mapping the cross-links onto top AlphaFold structural models, which was complemented by comparing their relative abundances in the cross-linked dimeric and monomeric bands. Furthermore, contrasting the CaM-free and CaM-bound nNOS samples shows that CaM enables the formation of the intersubunit FMN-heme docking complex and that CaM binding induces extensive, allosteric conformational changes across the NOS regions. Moreover, the observed cross-links sites specifically respond to changes in ionic strength. This indicates that interdomain salt bridges are responsible for stabilizing and orienting the output state for efficient FMN-heme IET. Taken together, our targeted qXL MS results have revealed that CaM and ionic strength modulate specific dynamic changes in the CaM/FMN/heme complexes, particularly in the context of intersubunit interdomain FMN-heme interactions.


Sujet(s)
Calmoduline , Flavine mononucléotide , Hème , Spectrométrie de masse , Nitric oxide synthase type I , Nitric oxide synthase type I/métabolisme , Nitric oxide synthase type I/composition chimique , Flavine mononucléotide/métabolisme , Flavine mononucléotide/composition chimique , Hème/métabolisme , Hème/composition chimique , Animaux , Rats , Calmoduline/métabolisme , Calmoduline/composition chimique , Spectrométrie de masse/méthodes , Domaines protéiques , Modèles moléculaires , Sites de fixation , Réactifs réticulants/composition chimique , Liaison aux protéines
5.
Ann Anat ; 255: 152291, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38821428

RÉSUMÉ

BACKGROUND: What textbooks usually call the sublingual gland in humans is in reality a tissue mass of two types of salivary glands, the anteriorly located consisting of a cluster of minor sublingual glands and the posteriorly located major sublingual gland with its outlet via Bartholin's duct. Only recently, the adrenergic and cholinergic innervations of the major sublingual gland was reported, while information regarding the neuropeptidergic and nitrergic innervations is still lacking. METHODS: Bioptic and autoptic specimens of the human major sublingual gland were examined by means of immunohistochemistry for the presence of vasoactive intestinal peptide (VIP)-, neuropeptide Y (NPY)-, substance P (SP)-, calcitonin gene related-peptide (CGRP)-, and neuronal nitric oxide synthase (nNOS)-labeled neuronal structures. RESULTS: As to the neuropeptidergic innervation of secretory cells (here in the form of mucous tubular and seromucous cells), the findings showed many VIP-containing nerves, few NPY- and SP-containing nerves and a lack of CGRP-labeled nerves. As to the neuropeptidergic innervation of vessels, the number of VIP-containing nerves was modest, while, of the other neuropeptide-containing nerves under study, only few (SP and CGRP) to very few (NPY) nerves were observed. As to the nitrergic innervation, nNOS-containing nerves were very few close to secretory cells and even absent around vessels. CONCLUSION: The various innervation patterns may suggest potential transmission mechanisms involved in secretory and vascular responses of the major sublingual gland.


Sujet(s)
Neuropeptides , Glande sublinguale , Substance P , Humains , Glande sublinguale/innervation , Glande sublinguale/métabolisme , Mâle , Neuropeptides/métabolisme , Femelle , Substance P/métabolisme , Neuropeptide Y/métabolisme , Peptide relié au gène de la calcitonine/métabolisme , Peptide vasoactif intestinal/métabolisme , Immunohistochimie , Adulte d'âge moyen , Nitric oxide synthase type I/métabolisme , Sujet âgé , Adulte , Sujet âgé de 80 ans ou plus
6.
Vet Immunol Immunopathol ; 271: 110752, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38579442

RÉSUMÉ

Nitric oxide (NO) is gaseous bioactive molecule that is synthesized by NO synthase (NOS). Inducible NOS (iNOS) expression occurs in response to pathogenic challenges, resulting in the production of large amounts of NO. However, there is a lack of knowledge regarding neuronal NOS (nNOS) and endothelial NOS (eNOS) in birds during pathogenic challenge. Therefore, the present study was conducted to determine the influence of intraperitoneal (IP) injection of zymosan (cell wall component of yeast) and lipopolysaccharide (LPS, a cell wall component of gram-negative bacteria) on NOS expression in chicks (Gallus gallus). Furthermore, the effect of NOS inhibitors on the corresponding behavioral and physiological parameters was investigated. Zymosan and LPS injections induced iNOS mRNA expression in several organs. Zymosan had no effect on eNOS mRNA expression in the organs investigated, whereas LPS increased its expression in the pancreas. Zymosan and LPS decreased nNOS mRNA expression in the lung, heart, kidney, and pancreas. The decreased nNOS mRNA expression in pancreas was probably associated with the NO from iNOS provided that such effect was reproduced by IP injection of sodium nitroprusside, which is a NO donor. Furthermore, pancreatic nNOS mRNA expression decreased following subcutaneous injection of corticosterone. Furthermore, IP injections of a nonspecific NOS inhibitor, NG-nitro-L-arginine methyl ester, and an nNOS-specific inhibitor, 7-nitroindazole, resulted in the significant decreases in food intake, cloacal temperature, and feed passage via the digestive tract in chicks. Collectively, the current findings imply the decreased nNOS expression because of fungal and bacterial infections, which affects food intake, body temperature, and the digestive function in birds.


Sujet(s)
Poulets , Lipopolysaccharides , Nitric oxide synthase type I , Zymosan , Animaux , Zymosan/pharmacologie , Lipopolysaccharides/pharmacologie , Poulets/immunologie , Nitric oxide synthase type I/génétique , Nitric oxide synthase type I/métabolisme , Mâle , Indazoles/pharmacologie , Nitric oxide synthase type II/génétique , Nitric oxide synthase type II/métabolisme , Nitric oxide synthase type III/génétique , Nitric oxide synthase type III/métabolisme
7.
Biochem J ; 481(9): 601-613, 2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38592741

RÉSUMÉ

Knowledge of the primary structure of neuronal NO synthase (nNOS) in skeletal muscle is still conflicting and needs further clarification. To elucidate the expression patterns of nNOS isoforms at both mRNA and protein level, systematic reverse transcription (RT)-PCR and epitope mapping by qualitative immunoblot analysis on skeletal muscle of C57/BL6 mice were performed. The ability of the nNOS isoforms to form aggregates was characterized by native low-temperature polyacrylamide electrophoresis (LT-PAGE). The molecular analysis was focused on the rectus femoris (RF) muscle, a skeletal muscle with a nearly balanced ratio of nNOS α- and ß-isoforms. RT-PCR amplificates from RF muscles showed exclusive exon-1d mRNA expression, either with or without exon-µ. Epitope mapping demonstrated the simultaneous expression of the nNOS splice variants α/µ, α/non-µ, ß/µ and ß/non-µ. Furthermore, immunoblotting suggests that the transition between nNOS α- and ß-isoforms lies within exon-3. In LT-PAGE, three protein nNOS associated aggregates were detected in homogenates of RF muscle and tibialis anterior muscle: a 320 kDa band containing nNOS α-isoforms, while 250 and 300 kDa bands consist of nNOS ß-isoforms that form homodimers or heterodimers with non-nNOS proteins.


Sujet(s)
Muscles squelettiques , Nitric oxide synthase type I , Animaux , Mâle , Souris , Exons , Isoenzymes/métabolisme , Isoenzymes/génétique , Souris de lignée C57BL , Muscles squelettiques/enzymologie , Nitric oxide synthase type I/métabolisme , Nitric oxide synthase type I/génétique , ARN messager/génétique , ARN messager/métabolisme
9.
Nat Commun ; 15(1): 3610, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38688927

RÉSUMÉ

Puberty is a crucial phase for the development of female sexual behavior. Growing evidence suggests that stress during this period may interfere with the development of sexual behavior. However, the neural circuits involved in this alteration remain elusive. Here, we demonstrated in mice that pubertal stress permanently disrupted sexual performance without affecting sexual preference. This was associated with a reduced expression and activation of neuronal nitric oxide synthase (nNOS) in the ventrolateral part of the ventromedial hypothalamus (VMHvl). Fiber photometry revealed that VMHvl nNOS neurons are strongly responsive to male olfactory cues with this activation being substantially reduced in pubertally stressed females. Finally, treatment with a NO donor partially restored sexual performance in pubertally stressed females. This study provides insights into the involvement of VMHvl nNOS in the processing of olfactory cues important for the expression of female sexual behavior. In addition, exposure to stress during puberty disrupts the integration of male olfactory cues leading to reduced sexual behavior.


Sujet(s)
Nitric oxide synthase type I , Comportement sexuel chez les animaux , Maturation sexuelle , Stress psychologique , Animaux , Femelle , Mâle , Comportement sexuel chez les animaux/physiologie , Nitric oxide synthase type I/métabolisme , Nitric oxide synthase type I/génétique , Souris , Stress psychologique/physiopathologie , Neurones/métabolisme , Noyau ventromédial de l'hypothalamus/métabolisme , Signaux , Souris de lignée C57BL , Odorat/physiologie , Donneur d'oxyde nitrique/pharmacologie
10.
Nitric Oxide ; 146: 1-9, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38428514

RÉSUMÉ

BACKGROUND: Cannabidiol (CBD) is the second most abundant pharmacologically active component present in Cannabis sp. Unlike Δ-9-tetrahydrocannabinol (THC), it has no psychotomimetic effects and has recently received significant interest from the scientific community due to its potential to treat anxiety and epilepsy. CBD has excellent anti-inflammatory potential and can be used to treat some types of inflammatory and neuropathic pain. In this context, the present study aimed to evaluate the analgesic mechanism of cannabidiol administered systemically for the treatment of neuropathic pain and determine the endogenous mechanisms involved with this analgesia. METHODS: Neuropathic pain was induced by sciatic nerve constriction surgery, and the nociceptive threshold was measured using the paw compression test in mice. RESULTS: CBD produced dose-dependent antinociception after intraperitoneal injection. Selective inhibition of PI3Kγ dose-dependently reversed CBD-induced antinociception. Selective inhibition of nNOS enzymes reversed the antinociception induced by CBD, while selective inhibition of iNOS and eNOS did not alter this antinociception. However, the inhibition of cGMP production by guanylyl cyclase did not alter CBD-mediated antinociception, but selective blockade of ATP-sensitive K+ channels dose-dependently reversed CBD-induced antinociception. Inhibition of S-nitrosylation dose-dependently and completely reversed CBD-mediated antinociception. CONCLUSION: Cannabidiol has an antinociceptive effect when administered systemically and this effect is mediated by the activation of PI3Kγ as well as by nitric oxide and subsequent direct S-nitrosylation of KATP channels on peripheral nociceptors.


Sujet(s)
Analgésiques , Cannabidiol , Phosphatidylinositol 3-kinases de classe Ib , Canaux KATP , Névralgie , Nitric oxide synthase type I , Monoxyde d'azote , Transduction du signal , Animaux , Cannabidiol/pharmacologie , Canaux KATP/métabolisme , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Névralgie/traitement médicamenteux , Névralgie/métabolisme , Souris , Monoxyde d'azote/métabolisme , Phosphatidylinositol 3-kinases de classe Ib/métabolisme , Nitric oxide synthase type I/métabolisme , Analgésiques/pharmacologie , Analgésie
11.
Pharmacol Rep ; 76(2): 338-347, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38480667

RÉSUMÉ

BACKGROUND: Cocaine use disorder (CUD) remains a severe health problem with no effective pharmacological therapy. One of the potential pharmacological strategies for CUD pharmacotherapy includes manipulations of the brain glutamatergic (Glu) system which is particularly involved in drug withdrawal and relapse. Previous research indicated a pivotal role of ionotropic N-methyl-D-aspartate (NMDA) receptors or metabotropic receptors' type 5 (mGlu5) receptors in controlling the reinstatement of cocaine. Stimulation of the above molecules results in the activation of the downstream signaling targets such as neuronal nitric oxide synthase (nNOS) and the release of nitric oxide. METHODS: In this paper, we investigated the molecular changes in nNOS in the prefrontal cortex and nucleus accumbens following 3 and 10 days of cocaine abstinence as well as the effectiveness of nNOS blockade with the selective enzyme inhibitor N-ω-propyl-L-arginine hydrochloride (L-NPA) on cocaine seeking in male rats. The effect of L-NPA on locomotor activity in drug-naïve animals was investigated. RESULTS: Ten-day (but not 3-day) cocaine abstinence from cocaine self-administration increased nNOS gene and protein expression in the nucleus accumbens, but not in the prefrontal cortex. L-NPA (0.5-5 mg/kg) administered peripherally did not change locomotor activity but attenuated the reinstatement induced with cocaine priming or the drug-associated conditioned cue. CONCLUSIONS: Our findings support accumbal nNOS as an important molecular player for cocaine seeking while its inhibitors could be considered as anti-cocaine pharmacological tools in male rats.


Sujet(s)
Cocaïne , Comportement de recherche de substances , Animaux , Mâle , Rats , Encéphale/métabolisme , Cocaïne/pharmacologie , Nitric oxide synthase type I/métabolisme , Noyau accumbens/métabolisme , Autoadministration
12.
Cell Rep ; 43(4): 113970, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38512868

RÉSUMÉ

To meet the high energy demands of brain function, cerebral blood flow (CBF) parallels changes in neuronal activity by a mechanism known as neurovascular coupling (NVC). However, which neurons play a role in mediating NVC is not well understood. Here, we identify in mice and humans a specific population of cortical GABAergic neurons that co-express neuronal nitric oxide synthase and tachykinin receptor 1 (Tacr1). Through whole-tissue clearing, we demonstrate that Tacr1 neurons extend local and long-range projections across functionally connected cortical areas. We show that whisker stimulation elicited Tacr1 neuron activity in the barrel cortex through feedforward excitatory pathways. Additionally, through optogenetic experiments, we demonstrate that Tacr1 neurons are instrumental in mediating CBF through the relaxation of mural cells in a similar fashion to whisker stimulation. Finally, by electron microscopy, we observe that Tacr1 processes contact astrocytic endfeet. These findings suggest that Tacr1 neurons integrate cortical activity to mediate NVC.


Sujet(s)
Couplage neurovasculaire , Animaux , Souris , Couplage neurovasculaire/physiologie , Humains , Neurones/métabolisme , Neurones/physiologie , Vibrisses/physiologie , Souris de lignée C57BL , Neurones GABAergiques/métabolisme , Neurones GABAergiques/physiologie , Mâle , Cortex cérébral/physiologie , Cortex cérébral/vascularisation , Circulation cérébrovasculaire/physiologie , Nitric oxide synthase type I/métabolisme
13.
J Neurochem ; 168(7): 1402-1419, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38445395

RÉSUMÉ

The role of nitrergic system in modulating the action of psychostimulants on reward processing is well established. However, the relevant anatomical underpinnings and scope of the involved interactions with mesolimbic dopaminergic system have not been clarified. Using immunohistochemistry, we track the changes in neuronal nitric oxide synthase (nNOS) containing cell groups in the animals conditioned to intracranial self-stimulation (ICSS) via an electrode implanted in the lateral hypothalamus-medial forebrain bundle (LH-MFB) area. An increase in the nNOS immunoreactivity was noticed in the cells and fibers in the ventral tegmental area (VTA) and nucleus accumbens shell (AcbSh), the primary loci of the reward system. In addition, nNOS was up-regulated in the nucleus accumbens core (AcbC), vertical limb of diagonal band (VDB), locus coeruleus (LC), lateral hypothalamus (LH), superficial gray layer (SuG) of the superior colliculus, and periaqueductal gray (PAG). The brain tissue fragments drawn from these areas showed a change in nNOS mRNA expression, but in opposite direction. Intracerebroventricular (icv) administration of nNOS inhibitor, 7-nitroindazole (7-NI) showed decreased lever press activity in a dose-dependent manner in ICSS task. While an increase in the dopamine (DA) and 3, 4-dihydroxyphenylacetic acid (DOPAC) efflux was noted in the microdialysates collected from the AcbSh of ICSS rats, pre-administration of 7-NI (icv route) attenuated the response. The study identifies nitrergic centers that probably mediate sensory, cognitive, and motor components of the goal-directed behavior.


Sujet(s)
Nitric oxide synthase type I , Autostimulation , Animaux , Mâle , Rats , Nitric oxide synthase type I/métabolisme , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Indazoles/pharmacologie , Antienzymes/pharmacologie
14.
J Neural Transm (Vienna) ; 131(3): 275-280, 2024 03.
Article de Anglais | MEDLINE | ID: mdl-38253928

RÉSUMÉ

Nitric oxide (NO) has been thought to be a novel factor involved in the mechanisms of mental disorders pathogenesis for quite some time. However, little is known about potential crosstalk between neuronal NO signaling and neuroleptics action. The present work was, therefore, focused on gene expression of neuronal NO synthase (nNOS) in the brains of rats chronically treated with olanzapine, an atypical antipsychotic drug. Studies were carried out on adult, male Sprague-Dawley rats that were divided into 2 groups: control and experimental animals treated with olanzapine (28-day-long intraperitoneal injection, at dose 5 mg/kg daily). All individuals were killed under anesthesia and the whole brains excised. Immunohistochemical procedure was used for histological assessment of the whole brain, and for both descriptive and quantitative analysis of nNOS protein distribution in selected brain structures. Long-term treatment with olanzapine is reflected in different changes in the number of enzyme-expressing cells in the rat brain. Olanzapine decreased the number of nNOS-expressing cells and possibly reduced NO synthesis in the rat striatum. Olanzapine can be taken into account as a potential inhibitor of NO synthesis in the rat striatum.


Sujet(s)
Neuroleptiques , Corps strié , Animaux , Mâle , Rats , Neuroleptiques/pharmacologie , Corps strié/métabolisme , Monoxyde d'azote/métabolisme , Nitric oxide synthase type I/métabolisme , Olanzapine/pharmacologie , Rat Sprague-Dawley
15.
J Am Heart Assoc ; 13(3): e033279, 2024 Feb 06.
Article de Anglais | MEDLINE | ID: mdl-38258657

RÉSUMÉ

BACKGROUND: Gut dysmotility is common after ischemic stroke, but the mechanism underlying this response is unknown. Under homeostasis, gut motility is regulated by the neurons of the enteric nervous system that control contractile/relaxation activity of muscle cells in the gut wall. More recently, studies of gut inflammation revealed interactions of macrophages with enteric neurons are also involved in modulating gut motility. However, whether poststroke gut dysmotility is mediated by direct signaling to the enteric nervous system or indirectly via inflammatory macrophages is unknown. METHODS AND RESULTS: We examined these hypotheses by using a clinically relevant permanent intraluminal midcerebral artery occlusion experimental model of stroke. At 24 hours after stroke, we performed in vivo and ex vivo gut motility assays, flow cytometry, immunofluorescence, and transcriptomic analysis. Stroke-induced gut dysmotility was associated with recruitment of muscularis macrophages into the gastrointestinal tract and redistribution of muscularis macrophages away from myenteric ganglia. The permanent intraluminal midcerebral artery occlusion model caused changes in gene expression in muscularis macrophages consistent with an altered phenotype. While the size of myenteric ganglia after stroke was not altered, myenteric neurons from post-permanent intraluminal midcerebral artery occlusion mice showed a reduction in neuronal nitric oxide synthase expression, and this response was associated with enhanced intestinal smooth muscle contraction ex vivo. Finally, chemical sympathectomy with 6-hydroxydopamine prevented the loss of myenteric neuronal nitric oxide synthase expression and stroke-induced slowed gut transit. CONCLUSIONS: Our findings demonstrate that activation of the sympathetic nervous system after stroke is associated with reduced neuronal nitric oxide synthase expression in myenteric neurons, resulting in impaired smooth muscle relaxation and dysregulation of gut transit.


Sujet(s)
Système nerveux entérique , Accident vasculaire cérébral , Souris , Animaux , Nitric oxide synthase type I/génétique , Nitric oxide synthase type I/métabolisme , Système nerveux entérique/métabolisme , Neurones/physiologie , Relâchement musculaire , Accident vasculaire cérébral/métabolisme
16.
Cardiovasc Res ; 120(5): 519-530, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38270932

RÉSUMÉ

AIMS: A reduction in both dystrophin and neuronal nitric oxide synthase (NOS1) secondary to microRNA-31 (miR-31) up-regulation contributes to the atrial electrical remodelling that underpins human and experimental atrial fibrillation (AF). In contrast, patients with Duchenne muscular dystrophy (DMD), who lack dystrophin and NOS1 and, at least in the skeletal muscle, have raised miR-31 expression, do not have increase susceptibility to AF in the absence of left ventricular (LV) dysfunction. Here, we investigated whether dystrophin deficiency is also associated with atrial up-regulation of miR-31, loss of NOS1 protein, and increased AF susceptibility in young mdx mice. METHODS AND RESULTS: Echocardiography showed normal cardiac structure and function in 12-13 weeks mdx mice, with no indication by assay of hydroxyproline that atrial fibrosis had developed. The absence of dystrophin in mdx mice was accompanied by an overall reduction in syntrophin and a lower NOS1 protein content in the skeletal muscle and in the left atrial and ventricular myocardium, with the latter occurring alongside reduced Nos1 transcript levels (exons 1-2 by quantitative polymerase chain reaction) and an increase in NOS1 polyubiquitination [assessed using tandem polyubiquitination pulldowns; P < 0.05 vs. wild type (WT)]. Neither the up-regulation of miR-31 nor the substantial reduction in NOS activity observed in the skeletal muscle was present in the atrial tissue of mdx mice. At difference with the skeletal muscle, the mdx atrial myocardium showed a reduction in the constitutive NOS inhibitor, caveolin-1, coupled with an increase in NOS3 serine1177 phosphorylation, in the absence of differences in the protein content of other NOS isoforms or in the relative expression NOS1 splice variants. In line with these findings, transoesophageal atrial burst pacing revealed no difference in AF susceptibility between mdx mice and their WT littermates. CONCLUSION: Dystrophin depletion is not associated with atrial miR-31 up-regulation, reduced NOS activity, or increased AF susceptibility in the mdx mouse. Compared with the skeletal muscle, the milder atrial biochemical phenotype may explain why patients with DMD do not exhibit a higher prevalence of atrial arrhythmias despite a reduction in NOS1 content.


Sujet(s)
Fibrillation auriculaire , Modèles animaux de maladie humaine , Dystrophine , Souris de lignée mdx , microARN , Myopathie de Duchenne , Nitric oxide synthase type I , Animaux , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/complications , Fibrillation auriculaire/métabolisme , Fibrillation auriculaire/génétique , Fibrillation auriculaire/physiopathologie , Fibrillation auriculaire/étiologie , Fibrillation auriculaire/anatomopathologie , Nitric oxide synthase type I/métabolisme , Nitric oxide synthase type I/génétique , microARN/métabolisme , microARN/génétique , Dystrophine/génétique , Dystrophine/métabolisme , Humains , Mâle , Souris de lignée C57BL , Muscles squelettiques/métabolisme , Atrium du coeur/métabolisme , Atrium du coeur/physiopathologie , Atrium du coeur/anatomopathologie , Remodelage auriculaire , Souris
17.
J Biol Chem ; 300(1): 105464, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37979917

RÉSUMÉ

Neuronal nitric oxide synthase (nNOS) is a homodimeric cytochrome P450-like enzyme that catalyzes the conversion of L-arginine to nitric oxide in the presence of NADPH and molecular oxygen. The binding of calmodulin (CaM) to a linker region between the FAD/FMN-containing reductase domain, and the heme-containing oxygenase domain is needed for electron transfer reactions, reduction of the heme, and NO synthesis. Due to the dynamic nature of the reductase domain and low resolution of available full-length structures, the exact conformation of the CaM-bound active complex during heme reduction is still unresolved. Interestingly, hydrogen-deuterium exchange and mass spectrometry studies revealed interactions of the FMN domain and CaM with the oxygenase domain for iNOS, but not nNOS. This finding prompted us to utilize covalent crosslinking and mass spectrometry to clarify interactions of CaM with nNOS. Specifically, MS-cleavable bifunctional crosslinker disuccinimidyl dibutyric urea was used to identify thirteen unique crosslinks between CaM and nNOS as well as 61 crosslinks within the nNOS. The crosslinks provided evidence for CaM interaction with the oxygenase and reductase domain residues as well as interactions of the FMN domain with the oxygenase dimer. Cryo-EM studies, which gave a high-resolution model of the oxygenase domain, along with crosslink-guided docking provided a model of nNOS that brings the FMN within 15 Å of the heme in support for a more compact conformation than previously observed. These studies also point to the utility of covalent crosslinking and mass spectrometry in capturing transient dynamic conformations that may not be captured by hydrogen-deuterium exchange and mass spectrometry experiments.


Sujet(s)
Calmoduline , Réactifs réticulants , Modèles moléculaires , Nitric oxide synthase type I , Calmoduline/métabolisme , Hème/métabolisme , Spectrométrie de masse , Nitric oxide synthase type I/métabolisme , Oxygénases/métabolisme , Réactifs réticulants/composition chimique , Calcium/composition chimique , Structure quaternaire des protéines , Liaison aux protéines , Cryomicroscopie électronique
18.
Cell Mol Biol (Noisy-le-grand) ; 69(13): 128-133, 2023 Dec 10.
Article de Anglais | MEDLINE | ID: mdl-38158677

RÉSUMÉ

The neuronal nitric oxide synthase (nNOS; encoded by NOS1)-derived nitric oxide (NO) plays an important role in maintaining skeletal muscle mass. In adult skeletal muscle, nNOS localizes to the cell membrane, cytosol, and nucleus, and regulates muscle hypertrophy and atrophy in various subcellular fractions. However, its role in muscle stem cells (also known as muscle satellite cells), which provide myonuclei for postnatal muscle growth, maintenance, and regeneration, remains unclear. The present study aimed to determine nNOS expression in muscle satellite cell-derived primary myoblasts during differentiation and its DNA methylation levels, an epigenetic modification that controls gene expression. Undifferentiated and differentiated satellite cell-derived primary myoblasts were found to express nNOS. Immunohistochemical analysis revealed that nNOS colocalized with Pax7 (satellite cell marker) only in the undifferentiated myoblasts. Furthermore, nNOS immunoreactivity spread to the cytosol of Pax7-negative differentiated myotube-like cells. The level of Nos1µ mRNA, the main isoform of skeletal muscle nNOS, was increased in differentiated satellite cell-derived primary myoblasts compared to that in the undifferentiated cells. However, Nos1 methylation levels remained unchanged during differentiation. These findings suggest that nNOS induction and the appropriate transition of its subcellular localization may contribute to muscle differentiation.


Sujet(s)
Nitric oxide synthase type I , Cellules satellites du muscle squelettique , Humains , Différenciation cellulaire/physiologie , Fibres musculaires squelettiques/métabolisme , Muscles squelettiques/métabolisme , Nitric oxide synthase type I/génétique , Nitric oxide synthase type I/métabolisme , Cellules satellites du muscle squelettique/métabolisme
19.
Int J Mol Sci ; 24(21)2023 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-37958792

RÉSUMÉ

In this review, the structure, isoform, and physiological role of the carboxy-terminal PDZ ligand of neuronal nitric oxide synthase (CAPON) are summarized. There are three isoforms of CAPON in humans, including long CAPON protein (CAPON-L), short CAPON protein (CAPON-S), and CAPON-S' protein. CAPON-L includes three functional regions: a C-terminal PDZ-binding motif, carboxypeptidase (CPE)-binding region, and N-terminal phosphotyrosine (PTB) structural domain. Both CAPON-S and CAPON-S' only contain the C-terminal PDZ-binding motif. The C-terminal PDZ-binding motif of CAPON can bind with neuronal nitric oxide synthase (nNOS) and participates in regulating NO production and neuronal development. An overview is given on the relationship between CAPON and heart diseases, diabetes, psychiatric disorders, and tumors. This review will clarify future research directions on the signal pathways related to CAPON, which will be helpful for studying the regulatory mechanism of CAPON. CAPON may be used as a drug target, which will provide new ideas and solutions for treating human diseases.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Transduction du signal , Humains , Nitric oxide synthase type I/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme
20.
J Mol Neurosci ; 73(11-12): 875-883, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37843719

RÉSUMÉ

Glioblastoma multiforme (GBM) is a prevalent and aggressive primary brain tumor, presenting substantial treatment challenges and high relapse rates. GBM is characterized by alterations in molecular signaling and enzyme expression within malignant cells. This tumor exhibits elevated nitric oxide (NO.) levels. NO. is a crucial signaling molecule involved in the regulation of neuronal functions, synaptic transmission, and cell proliferation. It is primarily synthesized from L-arginine by nitric oxide synthase (NOS) enzymes. The increased levels of NO. in GBM stem from dysregulated activity and expression of clinically relevant NOS isoforms, particularly inducible NOS (iNOS) and neuronal NOS (nNOS). Based on this knowledge, we hypothesize that targeted pharmacological intervention with N6-(1-iminoethyl)-L-lysine (L-NIL), an iNOS inhibitor, and 7-Nitroindazole (7-NI), an nNOS inhibitor, may suggest a promising therapeutic strategy for the treatment of GBM. To test our hypothesis, we utilized the U87-MG cell line as an in vitro model of GBM. Our results showed that treatment with L-NIL and 7-NI led to a reduction in NO. levels, NOS activity, and clonogenic proliferation in U87-MG cells. These findings suggest that NO. and NOS enzymes might be prospective therapeutic targets for GBM.


Sujet(s)
Glioblastome , Humains , Glioblastome/traitement médicamenteux , Antienzymes/pharmacologie , Récidive tumorale locale , Nitric oxide synthase/métabolisme , Nitric oxide synthase type I/métabolisme , Nitric oxide synthase type II/génétique , Nitric oxide synthase type II/métabolisme , Monoxyde d'azote/métabolisme , Prolifération cellulaire
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE