Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 33
Filtrer
1.
Int J Mol Sci ; 24(9)2023 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-37175503

RÉSUMÉ

Epidural motor cortex stimulation (MCS) is an effective treatment for refractory neuropathic pain; however, some individuals are unresponsive. In this study, we correlated the effectiveness of MCS and refractoriness with the expression of cytokines, neurotrophins, and nociceptive mediators in the dorsal root ganglion (DRG), sciatic nerve, and plasma of rats with sciatic neuropathy. MCS inhibited hyperalgesia and allodynia in two-thirds of the animals (responsive group), and one-third did not respond (refractory group). Chronic constriction injury (CCI) increased IL-1ß in the nerve and DRG, inhibited IL-4, IL-10, and IL-17A in the nerve, decreased ß-endorphin, and enhanced substance P in the plasma, compared to the control. Responsive animals showed decreased NGF and increased IL-6 in the nerve, accompanied by restoration of local IL-10 and IL-17A and systemic ß-endorphin. Refractory animals showed increased TNF-α and decreased IFNγ in the nerve, along with decreased TNF-α and IL-17A in the DRG, maintaining low levels of systemic ß-endorphin. Our findings suggest that the effectiveness of MCS depends on local control of inflammatory and neurotrophic changes, accompanied by recovery of the opioidergic system observed in neuropathic conditions. So, understanding the refractoriness to MCS may guide an improvement in the efficacy of the technique, thus benefiting patients with persistent neuropathic pain.


Sujet(s)
Analgésie , Névralgie , Rats , Animaux , Interleukine-10/métabolisme , Interleukine-17/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , bêta-Endorphine/métabolisme , Névralgie/thérapie , Névralgie/métabolisme , Hyperalgésie/thérapie , Hyperalgésie/métabolisme , Nerf ischiatique/métabolisme , Ganglions sensitifs des nerfs spinaux/métabolisme
2.
Int J Mol Sci ; 23(19)2022 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-36232883

RÉSUMÉ

Pain is a worldwide public health problem and its treatment is still a challenge since clinically available drugs do not completely reverse chronic painful states or induce undesirable effects. Crotalphine is a 14 amino acids synthetic peptide that induces a potent and long-lasting analgesic effect on acute and chronic pain models, peripherally mediated by the endogenous release of dynorphin A and the desensitization of the transient receptor potential ankyrin 1 (TRPA1) receptor. However, the effects of crotalphine on the central nervous system (CNS) and the signaling pathway have not been investigated. Thus, the central effect of crotalphine was evaluated on the partial sciatic nerve ligation (PSNL)-induced chronic neuropathic pain model. Crotalphine (100 µg/kg, p.o.)-induced analgesia on the 14th day after surgery lasting up to 24 h after administration. This effect was prevented by intrathecal administration of CB1 (AM251) or CB2 (AM630) cannabinoid receptor antagonists. Besides that, crotalphine-induced analgesia was reversed by CTOP, nor-BNI, and naltrindole, antagonists of mu, kappa, and delta-opioid receptors, respectively, and also by the specific antibodies for ß-endorphin, dynorphin-A, and met-enkephalin. Likewise, the analgesic effect of crotalphine was blocked by the intrathecal administration of minocycline, an inhibitor of microglial activation and proliferation. Additionally, crotalphine decreased the PSNL-induced IL-6 release in the spinal cord. Importantly, in vitro, crotalphine inhibited LPS-induced CD86 expression and upregulated CD206 expression in BV-2 cells, demonstrating a polarization of microglial cells towards the M2 phenotype. These results demonstrated that crotalphine, besides activating opioid and cannabinoid analgesic systems, impairs central neuroinflammation, confirming the neuromodulatory mechanism involved in the crotalphine analgesic effect.


Sujet(s)
Analgésie , Cannabinoïdes , Névralgie , Acides aminés/métabolisme , Analgésiques/métabolisme , Analgésiques/pharmacologie , Analgésiques/usage thérapeutique , Analgésiques morphiniques/métabolisme , Ankyrines/métabolisme , Antagonistes des récepteurs de cannabinoïdes/usage thérapeutique , Cannabinoïdes/usage thérapeutique , Dynorphines/métabolisme , Méthionine-enképhaline/métabolisme , Humains , Interleukine-6/métabolisme , Lipopolysaccharides/métabolisme , Microglie/métabolisme , Minocycline/usage thérapeutique , Névralgie/métabolisme , Peptides , Phénotype , Récepteurs aux opioïdes/métabolisme , Moelle spinale , bêta-Endorphine/métabolisme
3.
Cells ; 11(2)2022 01 12.
Article de Anglais | MEDLINE | ID: mdl-35053363

RÉSUMÉ

Increased collagen-derived advanced glycation end-products (AGEs) are consistently related to painful diseases, including osteoarthritis, diabetic neuropathy, and neurodegenerative disorders. We have recently developed a model combining a two-dimensional glycated extracellular matrix (ECM-GC) and primary dorsal root ganglion (DRG) that mimicked a pro-nociceptive microenvironment. However, culturing primary cells is still a challenge for large-scale screening studies. Here, we characterized a new model using ECM-GC as a stimulus for human sensory-like neurons differentiated from SH-SY5Y cell lines to screen for analgesic compounds. First, we confirmed that the differentiation process induces the expression of neuron markers (MAP2, RBFOX3 (NeuN), and TUBB3 (ß-III tubulin), as well as sensory neuron markers critical for pain sensation (TRPV1, SCN9A (Nav1.7), SCN10A (Nav1.8), and SCN11A (Nav1.9). Next, we showed that ECM-GC increased c-Fos expression in human sensory-like neurons, which is suggestive of neuronal activation. In addition, ECM-GC upregulated the expression of critical genes involved in pain, including SCN9A and TACR1. Of interest, ECM-GC induced substance P release, a neuropeptide widely involved in neuroinflammation and pain. Finally, morphine, the prototype opiate, decreased ECM-GC-induced substance P release. Together, our results suggest that we established a functional model that can be useful as a platform for screening candidates for the management of painful conditions.


Sujet(s)
Analgésiques/analyse , Analgésiques/pharmacologie , Collagène/pharmacologie , Évaluation préclinique de médicament , Modèles biologiques , Cellules réceptrices sensorielles/cytologie , Animaux , Antigènes néoplasiques/métabolisme , Marqueurs biologiques/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Matrice extracellulaire/métabolisme , Galectine -3/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Glycosylation/effets des médicaments et des substances chimiques , Humains , Mitogen-Activated Protein Kinases/métabolisme , Canal sodique voltage-dépendant NAV1.7/génétique , Canal sodique voltage-dépendant NAV1.7/métabolisme , Neurites/effets des médicaments et des substances chimiques , Neurites/métabolisme , Neurones/cytologie , Neurones/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-fos/métabolisme , Rats , Récepteur de la neurokinine 1/génétique , Récepteur de la neurokinine 1/métabolisme , Cellules réceptrices sensorielles/effets des médicaments et des substances chimiques , Cellules réceptrices sensorielles/métabolisme , Substance P/métabolisme , bêta-Endorphine/métabolisme
4.
PLoS One ; 13(10): e0204830, 2018.
Article de Anglais | MEDLINE | ID: mdl-30286124

RÉSUMÉ

The objective of this study was to evaluate the effect of low-dose naltrexone (LDN) as a carboplatin chemotherapy-associated drug in female dogs with mammary carcinoma in benign mixed tumors (MC-BMT) after mastectomy and to assess its association with quality of life and survival rates. Sixty female dogs were included in this study, all of which had histopathological diagnosis of MC-BMT and were divided into three groups: G1 (control), consisting of animals submitted only to mastectomy with or without regional metastasis; G2, composed of treated animals that did not present with metastasis; and G3, treated dogs that presented with metastasis. G2 and G3 were also subdivided according to the treatment administered: chemotherapy alone (MC-BMT(-) C/MC-BMT(+) C) or LDN and chemotherapy (MC-BMT(-) C+LDN/MC-BMT(+) C+LDN). All animals were subjected to clinical evaluation, mastectomy, peripheral blood lymphocyte immunophenotyping, beta-endorphin and met-enkephalin quantification, and evaluation of survival rates and quality of life scores. The results showed higher serum concentrations of beta-endorphin and met-enkephalin, fewer chemotherapy-related side effects, and better quality of life and survival rates in the LDN-treated groups than in LDN-untreated groups (P < 0.05). Evaluation of clinical and pathological parameters indicated a significant association between the use of LDN and both prolonged survival and enhanced quality of life. These results indicate that LDN is a viable chemotherapy-associated treatment in female dogs with MC-BMT, maintaining their quality of life and prolonging survival rates.


Sujet(s)
Carboplatine/administration et posologie , Carboplatine/pharmacologie , Lymphocytes/métabolisme , Tumeurs mammaires de l'animal/traitement médicamenteux , Naltrexone/administration et posologie , Animaux , Chiens , Synergie des médicaments , Méthionine-enképhaline/métabolisme , Femelle , Immunophénotypage , Tumeurs mammaires de l'animal/immunologie , Tumeurs mammaires de l'animal/chirurgie , Mastectomie/médecine vétérinaire , Naltrexone/pharmacologie , Métastase tumorale , Qualité de vie , Analyse de survie , Résultat thérapeutique , bêta-Endorphine/métabolisme
5.
Physiol Behav ; 189: 107-115, 2018 05 15.
Article de Anglais | MEDLINE | ID: mdl-29563057

RÉSUMÉ

Sexual experience modifies brain functioning and copulatory efficiency. Sexual activity, ejaculation in particular, is a rewarding behavior associated with the release of endogenous opioids, which modulate the activity of the mesolimbic dopaminergic system (MLS). In sexually exhausted rats, repeated ejaculation produces µ (MOR) and δ opioid receptor (DOR) internalization in ventral tegmental area (VTA) neurons, as well as long-lasting behavioral changes suggestive of brain plasticity processes. We hypothesized that in sexually naïve rats the endogenous opioids released during sexual experience acquisition, might contribute to brain plasticity processes involved in the generation of the behavioral changes induced by sexual experience. To this aim, using double immunohistochemistry and confocal microscopy, we compared in vivo MOR, DOR and ß-arrestin2 densities and activation in the VTA of sexually naïve males, sexually experienced rats not executing sexual activity prior to sacrifice and sexually experienced animals that ejaculated once before sacrifice. Results showed that sexual experience acquisition improved male's copulatory ability and induced persistent changes in the density, cellular distribution and activation of MOR and ß-arrestin2 in VTA neurons. DOR density was not modified, but its cellular location changed after sexual experience, revealing that these two opioid receptors were differentially activated during sexual experience acquisition. It is concluded that the endogenous opioids released during sexual activity produce adjustments in VTA neurons of sexually naïve male rats that might contribute to the behavioral plasticity expressed as an improvement in male copulatory parameters, promoted by the acquisition of sexual experience.


Sujet(s)
Récepteur delta/métabolisme , Récepteur mu/métabolisme , Aire tegmentale ventrale/métabolisme , bêta-Arrestine 2/métabolisme , Animaux , Éjaculation/physiologie , Enképhalines/métabolisme , Mâle , Peptides opioïdes/métabolisme , Rats , Comportement sexuel chez les animaux/physiologie , bêta-Endorphine/métabolisme
6.
J Cosmet Dermatol ; 16(4): e59-e67, 2017 Dec.
Article de Anglais | MEDLINE | ID: mdl-28593645

RÉSUMÉ

BACKGROUND: Unwanted side effects such as dryness, hypersensitivity, and cutaneous photosensitivity are challenge for adherence and therapeutical success for patients using treatments for inflammatory and allergic skin response. AIMS: In this study, we compared the effects of two dermatological formulations, which are used in inflammatory and/or allergic skin conditions: dexchlorpheniramine maleate (DCP; 10 mg/g) and promethazine (PTZ; 20 mg/g). METHODS: We evaluated both formulations for phototoxicity potential, skin irritation, anti-inflammatory and antihistaminic abilities, and skin barrier repair in vitro and ex vivo using the standard OECD test guideline n° 432, the ECVAM protocol n° 78, and cultured skin explants from a healthy patient. Ultraviolet A was chosen as exogenous agent to induce allergic and inflammatory response. RESULTS: Both PTZ and DCP promoted increases in interleukin-1 (IL-1) synthesis in response to ultraviolet A (UVA) radiation compared to control. However, the increase observed with PTZ was significantly greater than the DCP, indicating that the latter has a lower irritant potential. DCP also demonstrated a protective effect on UVA-induced leukotriene B4 and nuclear factor kappa B (NF-κB) synthesis. Conversely, PTZ demonstrates more robust UVA antihistaminic activity. Likewise, PTZ promoted a significantly greater increase in the production of involucrin and keratin 14, both associated with protective skin barrier property. CONCLUSION: In conclusion, these data suggest possible diverging UVA response mechanisms of DCP and PTZ, which gives greater insight into the contrasting photosensitizing potential between DCP and PTZ observed in the patients.


Sujet(s)
Chlorphénamine/pharmacologie , Dermatite phototoxique/métabolisme , Antihistaminiques des récepteurs H1/pharmacologie , Prométhazine/pharmacologie , Cellules 3T3 , Animaux , Chlorphénamine/effets indésirables , Dermatite phototoxique/étiologie , Dermatite phototoxique/prévention et contrôle , Dinoprostone/métabolisme , Femelle , Histamine/métabolisme , Antihistaminiques des récepteurs H1/effets indésirables , Humains , Interleukine-1/métabolisme , Kératine-14/métabolisme , Leucotriène B4/métabolisme , Souris , Adulte d'âge moyen , Facteur de transcription NF-kappa B/métabolisme , Prométhazine/effets indésirables , Précurseurs de protéines/métabolisme , Peau/métabolisme , Crème pour la peau/effets indésirables , Phénomènes physiologiques de la peau/effets des médicaments et des substances chimiques , Canaux cationiques TRPV/métabolisme , Techniques de culture de tissus , Rayons ultraviolets/effets indésirables , bêta-Endorphine/métabolisme
7.
Lasers Med Sci ; 32(4): 865-872, 2017 May.
Article de Anglais | MEDLINE | ID: mdl-28283814

RÉSUMÉ

Neuropathic pain can be defined as the pain initiated or caused by a primary lesion or dysfunction of the central or peripheral nervous system. Photobiomodulation therapy (PBM) stands out among the physical therapy resources used for analgesia. However, application parameters, especially the energy density, remain controversial in the literature. Therefore, this study aimed to investigate the PBM effect, in different energy densities to control neuropathic pain in mice. Fifty (50) mice were induced to neuropathy by chronic constriction surgery of the sciatic nerve (CCI), treated with PBM (808 nm), and divided into five groups: GP (PBM simulation), GS (sham), GL10, GL20, GL40 (energy density of 10, 20, and 40 J/cm2, respectively). The evaluations were carried out using the hot plate test and Randall and Selitto test, before and after the CCI surgery, every 15 days during the 90 days experiment. ß-Endorphin blood dosage was also tested. For both the hot plate and Randall and Selitto tests, the GL20 and GL40 groups presented reduction of the nociceptive threshold from the 30th day of treatment, the GL10 group only after day 75, and the GP group did not show any improvement throughout the experiment. The ß-endorphin dosage was higher for all groups when compared to the GP group. However, only the GL20 group and GL40 presented a significant increase. This study demonstrates that PBM in higher energy density (20, 40 J/cm2) is more effective in the control of neuropathic pain.


Sujet(s)
Photothérapie de faible intensité , Névralgie/radiothérapie , Animaux , Constriction , Test ELISA , Hyperalgésie/radiothérapie , Mâle , Souris , Rat Sprague-Dawley , Nerf ischiatique/anatomopathologie , Nerf ischiatique/effets des radiations , bêta-Endorphine/métabolisme
8.
Int Immunopharmacol ; 44: 160-167, 2017 Mar.
Article de Anglais | MEDLINE | ID: mdl-28107753

RÉSUMÉ

This study aimed to investigate the effect of sulfated polysaccharide from red seaweed Solieria filiformis (Fraction F II) in the inflammatory hypernociception in the temporomandibular joint (TMJ) of rats. Male Wistar rats were pretreated (30min) with a subcutaneous injection (s.c.) of vehicle or FII (0.03, 0.3 or 3.0mg/kg) followed by intra-TMJ injection of 1.5% Formalin or 5-hydroxytryptamine (5-HT, 225µg/TMJ). In other set of experiments rats were pretreated (15min) with an intrathecal injection of the non-selective opioid receptors Naloxone, or µ-opioid receptor antagonist CTOP, or δ-opioid receptor Naltridole hydrochloride, or κ-opioid receptor antagonist Nor-Binaltorphimine (Nor-BNI) followed by injection of FII (s.c.). After 30min, the animals were treated with an intra-TMJ injection of 1.5% formalin. After TMJ treatment, behavioral nociception response was evaluated for a 45-min observation period, animals were terminally anesthetized and periarticular tissue, trigeminal ganglion and subnucleus caudalis (SC) were collected plasma extravasation and ELISA analysis. Pretreatment with F II significantly reduced formalin- and serotonin-induced TMJ nociception, inhibit the plasma extravasation and inflammatory cytokines release induced by 1.5% formalin in the TMJ. Pretreatment with intrathecal injection of Naloxone, CTOP, Naltridole or Nor-BNI blocked the antinociceptive effect of F II in the 1.5% formalin-induced TMJ nociception. In addition, F II was able to significantly increase the ß-endorphin release in the subnucleus caudalis. The results suggest that F II has a potential antinociceptive and anti-inflammatory effect in the TMJ mediated by activation of opioid receptors in the subnucleus caudalis and inhibition of the release of inflammatory mediators in the periarticular tissue.


Sujet(s)
Analgésiques morphiniques/administration et posologie , Analgésiques/usage thérapeutique , Anti-inflammatoires/usage thérapeutique , Noyau caudé/effets des médicaments et des substances chimiques , Douleur/traitement médicamenteux , Polyosides/usage thérapeutique , Articulation temporomandibulaire/effets des médicaments et des substances chimiques , Analgésiques morphiniques/effets indésirables , Animaux , Noyau caudé/métabolisme , Interactions médicamenteuses , Interleukine-1 bêta/métabolisme , Mâle , Douleur/induit chimiquement , Polyosides/composition chimique , Rats , Rat Wistar , Récepteurs aux opioïdes/métabolisme , Algue marine/immunologie , Sulfates/composition chimique , Articulation temporomandibulaire/anatomopathologie , Facteur de nécrose tumorale alpha/métabolisme , bêta-Endorphine/métabolisme
9.
Sci Rep ; 6: 29290, 2016 07 04.
Article de Anglais | MEDLINE | ID: mdl-27373214

RÉSUMÉ

Obesity is the result of a long-term positive energy balance in which caloric intake overrides energy expenditure. This anabolic state results from the defective activity of hypothalamic neurons involved in the sensing and response to adiposity. However, it is currently unknown what the earliest obesity-linked hypothalamic defect is and how it orchestrates the energy imbalance present in obesity. Using an outbred model of diet-induced obesity we show that defective regulation of hypothalamic POMC is the earliest marker distinguishing obesity-prone from obesity-resistant mice. The early inhibition of hypothalamic POMC was sufficient to transform obesity-resistant in obesity-prone mice. In addition, the post-prandial change in the blood level of ß-endorphin, a POMC-derived peptide, correlates with body mass gain in rodents and humans. Taken together, these results suggest that defective regulation of POMC expression, which leads to a change of ß-endorphin levels, is the earliest hypothalamic defect leading to obesity.


Sujet(s)
Hypothalamus/métabolisme , Inflammation/métabolisme , Obésité/métabolisme , Pro-opiomélanocortine/métabolisme , bêta-Endorphine/métabolisme , Adolescent , Adulte , Animaux , Régime alimentaire , Matières grasses alimentaires/métabolisme , Ration calorique , Humains , Hypothalamus/immunologie , Inflammation/immunologie , Mâle , Souris , Souris obèse , Obésité/immunologie , Pro-opiomélanocortine/immunologie , Rats , Rat Wistar , Jeune adulte
10.
Brain Res ; 1646: 366-376, 2016 09 01.
Article de Anglais | MEDLINE | ID: mdl-27321158

RÉSUMÉ

Neurons that express the prohormone proopiomelanocortin (POMC) in the arcuate hypothalamic nucleus (Arc) are engaged in the regulation of energy balance and glucose homeostasis. Additionally, POMC neurons are considered key first-order cells regulated by leptin. Interestingly, in the Arc, POMC cells that express the leptin receptor (POMC/LepR+ cells) are found side by side with POMC cells not directly responsive to leptin (POMC/LepR- cells). However, it remains unknown whether these distinct populations innervate different target regions. Therefore, the objective of the present study was to compare the projections of POMC/LepR+ and POMC/LepR- neurons. Using genetically modified LepR-reporter mice to identify leptin receptor-expressing cells and immunohistochemistry to stain POMC-derived peptides (α-MSH or ß-endorphin) we confirmed that approximately 80% of Arc ß-endorphin-positive neurons co-expressed leptin receptors. POMC/LepR+ and POMC/LepR- axons were intermingled in all of their target regions. As revealed by confocal microscopy, we found an elevated degree of co-localization between α-MSH+ axons and the reporter protein (tdTomato) in all brain regions analyzed, with co-localization coefficients ranging from 0.889 to 0.701. Thus, these two populations of POMC neurons seem to project to the same set of brain structures, although one of the two subtypes of POMC axons was sometimes found to be more abundant than the other in distinct subregions of the same nucleus. Therefore, POMC/LepR+ and POMC/LepR- cells may target separate neuronal populations and consequently activate distinct neuronal circuits within some target nuclei. These findings contribute to unravel the neuronal circuits involved in the regulation of energy balance and glucose homeostasis.


Sujet(s)
Noyau arqué de l'hypothalamus/cytologie , Encéphale/cytologie , Neurones/cytologie , Neurones/métabolisme , Pro-opiomélanocortine/métabolisme , Récepteurs à la leptine/métabolisme , Animaux , Noyau arqué de l'hypothalamus/métabolisme , Encéphale/métabolisme , Immunohistochimie , Souris , Souris de lignée C57BL , Souris transgéniques , Voies nerveuses/cytologie , Voies nerveuses/métabolisme , Techniques de traçage neuroanatomique , Terminaisons présynaptiques/métabolisme , Hormone mélanotrope alpha/métabolisme , bêta-Endorphine/métabolisme
11.
Article de Anglais | MEDLINE | ID: mdl-27255640

RÉSUMÉ

We have previously demonstrated that peripheral administration of 15d-PGJ2 in the Temporomandibular joint (TMJ) of rats can prevent nociceptor sensitization, mediated by peroxisome proliferator activated receptor-γ (PPAR-γ), and κ- and δ- opioid receptors. However, the mechanism that underlies the signaling of PPAR-γ (upon activation by 15d-PGJ2) to induce antinociception, and how the opioid receptors are activated via 15d-PGJ2 are not fully understood. This study demonstrates that peripheral antinociceptive effect of 15d-PGJ2 is mediated by PPAR-γ expressed in the inflammatory cells of TMJ tissues. Once activated by 15d-PGJ2, PPAR-γ induces the release of ß-endorphin and dynorphin, which activates κ- and δ-opioid receptors in primary sensory neurons to induce the antinociceptive effect.


Sujet(s)
Analgésiques/administration et posologie , Peptides opioïdes/métabolisme , Prostaglandine D2/analogues et dérivés , Articulation temporomandibulaire/métabolisme , Analgésiques/pharmacologie , Animaux , Dynorphines/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Mâle , Récepteur PPAR gamma/métabolisme , Prostaglandine D2/administration et posologie , Prostaglandine D2/pharmacologie , Rats , Rat Wistar , Transduction du signal/effets des médicaments et des substances chimiques , Articulation temporomandibulaire/effets des médicaments et des substances chimiques , bêta-Endorphine/métabolisme
12.
Mediators Inflamm ; 2015: 879126, 2015.
Article de Anglais | MEDLINE | ID: mdl-26538838

RÉSUMÉ

PURPOSE: This study quantified the expression of substance P (SP), calcitonin gene-related peptide (CGRP), ß-endorphins (ß-End), and methionine-enkephalin (Met-Enk) in human dental pulp following orthodontic intrusion. METHODS: Eight patients were selected according to preestablished inclusion criteria. From each patient, two premolars (indicated for extraction due to orthodontic reasons) were randomly assigned to two different groups: the asymptomatic inflammation group (EXPg), which would undergo controlled intrusive force for seven days, and the control group (CTRg), which was used to determine the basal levels of each substance. Once extracted, dental pulp tissue was prepared to determine the expression levels of both neuropeptides and endogenous opioids by radioimmunoassay (RIA). RESULTS: All samples from the CTRg exhibited basal levels of both neuropeptides and endogenous opioids. By day seven, all patients were asymptomatic, even when all orthodontic-intrusive devices were still active. In the EXPg, the SP and CGRP exhibited statistically significant different levels. Although none of the endogenous opioids showed statistically significant differences, they all expressed increasing trends in the EXPg. CONCLUSIONS: SP and CGRP were identified in dental pulp after seven days of controlled orthodontic intrusion movement, even in the absence of pain.


Sujet(s)
Analgésiques morphiniques/métabolisme , Pulpe dentaire/métabolisme , Neuropeptides/composition chimique , Adolescent , Peptide relié au gène de la calcitonine/métabolisme , Enfant , Enképhalines/métabolisme , Femelle , Humains , Inflammation , Mâle , Méthionine/métabolisme , Molaire , Orthodontie , Douleur , Projets pilotes , Dosage radioimmunologique , Substance P/métabolisme , bêta-Endorphine/métabolisme
13.
Respir Med ; 108(4): 609-20, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24418570

RÉSUMÉ

BACKGROUND: Neuromuscular electrical stimulation (NMES) improves muscle performance and exercise tolerance in chronic obstructive pulmonary disease (COPD) patients. In contrast, no study has assessed the effect of NMES on dynamic hyperinflation (DH) in COPD. This study investigated the effect of short-term, high-frequency NMES on DH in patients with COPD. METHODS: Twenty patients were randomly allocated to either a NMES applied bilaterally to the quadriceps muscles (n = 11: 8 weeks, 5 days/week, twice/day, 45 min/session) or a control group (n = 09). All patients received respiratory physical therapy and stretching exercises. Free fat mass, pulmonary function, time to exercise tolerance (Tlim), 6-min walk test distance (6-MWTD), tumor necrosis factor (TNF-α) and ß-endorphin levels, Borg dyspnea and leg score (BDS and BLS) and quality of life by the St. George's Respiratory Questionnaire score (SGRQ) were examined before and after the intervention. RESULTS: Compared with the control group, NMES increased FEV1 and FEV1/FVC, 6-MWD and Tlim (P < 0.01) and reduced BDS and SGRQ (P < 0.01). Additionally, changes in the Tlim were positively correlated with respiratory improvements in FEV1 (rho = 0.48, P < 0.01). Also, NMES reduced TNF-α and increased ß-endorphin levels, compared with the control group (P < 0.001). CONCLUSION: In summary, 8 weeks of NMES promotes reduction of the perceived sensation of dyspnea during exercise in patients with COPD. This finding is accompanied by improvements in FEV1, exercise tolerance and quality of life, and DH. Interestingly, these findings may be associated with enhanced vasodilatory function and a reduction in inflammatory responses. CLINICAL TRIAL REGISTRATION: NCT01695421.


Sujet(s)
Électrothérapie/méthodes , Broncho-pneumopathie chronique obstructive/thérapie , Adulte , Sujet âgé , Anthropométrie/méthodes , Composition corporelle/physiologie , Méthode en double aveugle , Épreuve d'effort/méthodes , Tolérance à l'effort/physiologie , Humains , Capacité inspiratoire/physiologie , Mâle , Adulte d'âge moyen , Force musculaire/physiologie , Projets pilotes , Broncho-pneumopathie chronique obstructive/sang , Broncho-pneumopathie chronique obstructive/physiopathologie , Muscle quadriceps fémoral/physiopathologie , Qualité de vie , Mécanique respiratoire/physiologie , Muscles respiratoires/physiopathologie , Facteur de nécrose tumorale alpha/métabolisme , bêta-Endorphine/métabolisme
14.
Pharmacology ; 89(1-2): 22-8, 2012.
Article de Anglais | MEDLINE | ID: mdl-22236644

RÉSUMÉ

The aim of the present study was to investigate the mechanisms underlying the endogenous control of nociception at a peripheral level during inflammation. Using a pharmacological approach and the rat paw pressure test, we assessed the effect of an intraplantar injection of naloxone, an opioid receptor antagonist, and bestatin, an aminopeptidase inhibitor, on hyperalgesia induced by carrageenan, which mimics an inflammatory process, or prostaglandin E(2) (PGE(2)), which directly sensitizes nociceptors. Naloxone induced a significant and dose-dependent (25, 50 or 100 µg) increase in carrageenan-induced hyperalgesia, but not PGE(2)-induced hyperalgesia. Bestatin (400 µg/paw) significantly counteracted carrageenan-induced hyperalgesia, inducing an increase in the nociceptive threshold compared to control, but it did not modify hyperalgesia induced by PGE(2) injection into the rat paw. Positive ß-endorphin immunoreactivity was increased in paw inflammation induced by carrageenan in comparison with the control group. However, PGE(2) did not significantly alter the immunostained area. These results provide evidence for activation of the endogenous opioidergic system during inflammation and indicate that this system regulates hyperalgesia through a negative feedback mechanism, modulating it at a peripheral level.


Sujet(s)
Inflammation/métabolisme , Peptides opioïdes/physiologie , Seuil nociceptif/physiologie , bêta-Endorphine/métabolisme , Animaux , Carragénane/effets indésirables , Carragénane/agonistes , Carragénane/antagonistes et inhibiteurs , Dinoprostone/effets indésirables , Relation dose-effet des médicaments , Hyperalgésie/induit chimiquement , Leucine/analogues et dérivés , Leucine/pharmacologie , Mâle , Naloxone/pharmacologie , Antagonistes narcotiques/pharmacologie , Seuil nociceptif/effets des médicaments et des substances chimiques , Inhibiteurs de protéases/pharmacologie , Rats , Rat Wistar
15.
Neuropeptides ; 44(1): 9-16, 2010 Feb.
Article de Anglais | MEDLINE | ID: mdl-20031207

RÉSUMÉ

The dopaminergic mesocorticolimbic system plays an important role in the reinforcing effects of ethanol. Opioid peptides modulate the activity of this system and have been suggested to mediate, at least in part, the reinforcing properties of ethanol. Thus, beta-endorphin (beta-END) could participate in the development of ethanol reinforcement and addiction. The aim of this work was to investigate the acute and chronic ethanol effects on beta-END content in regions of the mesolimbic system and to examine if chronic ethanol treatment alters ligand binding to mu opioid receptor (muOR). Male Wistar rats received a single acute ethanol dose of 2.5 g/kg or water by intra-gastric administration. For chronic ethanol treatment experiments, one group of rats was given ethanol (10% v/v solution) to drink, two groups were given equivalent volumes of sucrose (14.14% isocaloric solution) or water, respectively, and a fourth group had ad libitum access to food and water. Treatment was followed for 4 weeks. Beta-endorphin content in brain regions was quantified by radioimmunoassay and ligand binding studies to muOR were performed by quantitative autoradiography using 8 nM [(3)H]-DAMGO as radioligand. Acute ethanol decreased beta-END content in the hypothalamus (26%) 1h after administration. No ethanol effects were observed in the midbrain, ventral tegmental area, substantia nigra, nucleus accumbens, nucleus accumbens-septum and prefrontal cortex. Chronic ethanol treatment neither changed beta-END levels nor [(3)H]-DAMGO binding to mu opioid receptors in any of the regions studied. However, beta-END levels in the sucrose group were significantly increased in the nucleus accumbens and substantia nigra, in comparison to all other groups. These findings suggest that different neural mechanisms and specific brain regions may be involved in the reinforcing effects of ethanol and sucrose.


Sujet(s)
Chimie du cerveau/effets des médicaments et des substances chimiques , Dépresseurs du système nerveux central/pharmacologie , 2-Alanine-5-glycine-4-méthylphénylalanine-enképhaline/métabolisme , Éthanol/pharmacologie , bêta-Endorphine/métabolisme , Animaux , Cortex cérébral/effets des médicaments et des substances chimiques , Cortex cérébral/métabolisme , Dopamine/physiologie , Système limbique/effets des médicaments et des substances chimiques , Système limbique/métabolisme , Mâle , Protéines de tissu nerveux/métabolisme , Système neuroendocrinien/effets des médicaments et des substances chimiques , Système neuroendocrinien/métabolisme , Dosage radioimmunologique , Rats , Rat Wistar , Récepteur mu/effets des médicaments et des substances chimiques , Récepteur mu/métabolisme , Saccharose/pharmacologie
16.
Neuroscience ; 155(2): 350-8, 2008 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-18602454

RÉSUMÉ

Opioid mechanisms are involved in the control of water and NaCl intake and opioid receptors are present in the lateral parabrachial nucleus (LPBN), a site of important inhibitory mechanisms related to the control of sodium appetite. Therefore, in the present study we investigated the effects of opioid receptor activation in the LPBN on 0.3 M NaCl and water intake in rats. Male Holtzman rats with stainless steel cannulas implanted bilaterally in the LPBN were used. In normohydrated and satiated rats, bilateral injections of the opioid receptor agonist beta-endorphin (2 nmol/0.2 microl) into the LPBN induced 0.3 M NaCl (17.8+/-5.9 vs. saline: 0.9+/-0.5 ml/240 min) and water intake (11.4+/-3.0 vs. saline: 1.0+/-0.4 ml/240 min) in a two-bottle test. Bilateral injections of the opioid antagonist naloxone (100 nmol/0.2 microl) into the LPBN abolished sodium and water intake induced by beta-endorphin into the LPBN and also reduced 0.3 M NaCl intake (12.8+/-1.5 vs. vehicle: 22.4+/-3.1 ml/180 min) induced by 24 h of sodium depletion (produced by the treatment with the diuretic furosemide s.c.+sodium deficient food for 24 h). Bilateral injections of beta-endorphin into the LPBN in satiated rats produced no effect on water or 2% sucrose intake when water alone or simultaneously with 2% sucrose was offered to the animals. The results show that opioid receptor activation in the LPBN induces hypertonic sodium intake in satiated and normohydrated rats, an effect not due to general ingestive behavior facilitation. In addition, sodium depletion induced 0.3 M NaCl intake also partially depends on opioid receptor activation in the LPBN. The results suggest that deactivation of inhibitory mechanisms by opioid receptor activation in the LPBN releases sodium intake if excitatory signals were activated (sodium depletion) or not.


Sujet(s)
Pont/effets des médicaments et des substances chimiques , Pont/physiologie , Récepteur delta/métabolisme , Récepteur mu/métabolisme , Chlorure de sodium/pharmacocinétique , bêta-Endorphine/pharmacologie , Animaux , Consommation de boisson/effets des médicaments et des substances chimiques , Consommation de boisson/physiologie , Interactions médicamenteuses , 2-Alanine-5-glycine-4-méthylphénylalanine-enképhaline/pharmacologie , Mâle , Microinjections , Naloxone/pharmacologie , Antagonistes narcotiques/pharmacologie , Rats , Rat Sprague-Dawley , Récepteur delta/antagonistes et inhibiteurs , Récepteur mu/antagonistes et inhibiteurs , Sensation de satiété/effets des médicaments et des substances chimiques , Sensation de satiété/physiologie , Saccharose/pharmacologie , Eau/métabolisme , bêta-Endorphine/métabolisme
17.
J Cosmet Dermatol ; 7(2): 112-9, 2008 Jun.
Article de Anglais | MEDLINE | ID: mdl-18482014

RÉSUMÉ

BACKGROUND: The pathophysiology of sensitive skin consists of an inflammatory reaction resulting from the abnormal penetration in the skin of potentially irritating substances, which occurs due to skin barrier dysfunction and changes in the production of local neuromediators. AIMS: The therapeutic potential of L-carnosine and Rhodiola rosea, as antioxidant and neuromodulatory, respectively, leads us to investigate the effects of the R. rosea extract/L-carnosine-associated compound (RCAC) on sensitive skin alterations. METHODS: A double-blind comparative study was conducted on 124 volunteers with sensitive skin, who were selected by their reactivity to stinging test. Two randomized groups of 62 each received either a formulation containing 1% of RCAC or placebo, which was applied twice a day for 28 consecutive days. One perceptibility questionnaire was applied at the onset and at the end of the treatment to evaluate the subjective response to test product. Additionally, in vitro studies were performed to investigate RCAC neuroimmunomodulatory mechanisms. RESULTS: RCAC treatment produced in vivo protective effects in skin barrier function and a positive subjective response of sensitive skin volunteers. In vitro treatment promoted the release of proopiomelanocortin peptides and restored to normal the increased levels of neuropeptides and cytokines produced by keratinocytes exposed to ultraviolet radiation. Clinical effectiveness was measured by reduction of transepidermal water loss, positive perceptions of improvements in skin dryness and skin comfort sensation, and reduction of discomfort sensation after stinging test. CONCLUSIONS: The protective effect of RCAC in skin barrier function and the positive response produced in human subjects with sensitive skin could be partially explained by our in vitro results showing a significant increase in opioid peptides release, an inhibitory effect on neuropeptides production, and modulation of cytokines production by keratinocytes under ultraviolet stress.


Sujet(s)
Antioxydants/pharmacologie , Carnosine/pharmacologie , Kératinocytes/effets des médicaments et des substances chimiques , Agents du système nerveux périphérique/pharmacologie , Extraits de plantes/pharmacologie , Rhodiola , Peau/effets des médicaments et des substances chimiques , Administration par voie cutanée , Analyse de variance , Peptide relié au gène de la calcitonine/métabolisme , Cellules cultivées , Cytokines/métabolisme , Méthode en double aveugle , Enképhalines/métabolisme , Humains , Techniques immunoenzymatiques , Kératinocytes/métabolisme , Kératinocytes/effets des radiations , Adulte d'âge moyen , Peau/métabolisme , Substance P/métabolisme , Enquêtes et questionnaires , Rayons ultraviolets , Perte insensible en eau/effets des médicaments et des substances chimiques , bêta-Endorphine/métabolisme
18.
Fertil Steril ; 83 Suppl 1: 1119-24, 2005 Apr.
Article de Anglais | MEDLINE | ID: mdl-15831284

RÉSUMÉ

OBJECTIVE: To evaluate whether prolactin (PRL) is able to inhibit ovulation induced with exogenous gonadotropins in the rat and whether this effect could be mediated by the ovarian production of beta-endorphin, prostaglandin, and nitric oxide (NO). DESIGN: Controlled in vivo and in vitro experiments. SETTING: Academic research laboratories. ANIMAL(S): Immature female rats undergoing ovulation induction with equine gonadotropins and hCG. INTERVENTION(S): Prolactin (100 or 200 microg), PRL + the opioid antagonist naloxone (200 microg each), or placebo were injected SC 4 hours after hCG administration for ovulation induction. In the in vitro experiments, isolated preovulatory ovaries were incubated with or without PRL in a final concentration of 100 or 200 ng/mL. MAIN OUTCOME MEASURES(S): Number of oocytes ovulated in vivo, ovarian beta-endorphin, PGE(2) and NO(2)(-)/NO(3)(-) release, and NO synthase activity in vitro. RESULT(S): Prolactin reduced significantly the number of oocytes ovulated at the doses of 100 and 200 microg, and this effect was partially reversed by naloxone administration together with 200 mug PRL. PRL also induced a twofold increase in the ovarian release of beta-endorphin and a threefold decrease in the ovarian production of PGE(2). Ovarian NO synthase activity and the concentrations of NO(2)(-)/NO(3)(-) in the incubation medium were not modified by PRL. CONCLUSION(S): Prolactin is able to reduce the number of oocytes released and modulate ovarian beta-endorphin and PGE(2) release, which may account for its peripheral anovulatory effects. This local effect of PRL could interfere in the process of ovulation induction by exogenous gonadotropins.


Sujet(s)
Dinoprostone/métabolisme , Gonadotrophine équine/pharmacologie , Ovulation/effets des médicaments et des substances chimiques , Prolactine/pharmacologie , bêta-Endorphine/métabolisme , Animaux , Gonadotrophine chorionique/pharmacologie , Interactions médicamenteuses , Femelle , Naloxone/pharmacologie , Antagonistes narcotiques/pharmacologie , Monoxyde d'azote/métabolisme , Ovocytes/physiologie , Ovaire/cytologie , Ovaire/effets des médicaments et des substances chimiques , Ovaire/métabolisme , Induction d'ovulation , Rats , Rat Wistar
19.
J Mol Histol ; 35(8-9): 759-64, 2004 Nov.
Article de Anglais | MEDLINE | ID: mdl-15609088

RÉSUMÉ

Beta-endorphin and prolactin (PRL) are natural inhibitors of ovulation via central and peripheral mechanisms, but their possible interactions within the ovary are still unknown. The aims of the present study were to determine the gene expression and the topographic distribution of beta-endorphin, and the possible changes evoked by the pretreatment with PRL on the ovarian beta-endorphin localization in immature, superovulated rats. Prepuberal female Wistar rats weighing 60-70 g were superovulated with 20 IU equine gonadotrophins and, 48 h later, 20 IU human chorionic gonalotropin (hCG). Four hours after the hCG injection, the rats received either 200 microg rat PRL .i.p. (n = 12) or saline vehicle (n = 10). In the following morning the rats were killed and their ovaries were quickly removed. Beta-endorphin localization was assessed by immunohistochemistry and proopiomelanocortin (POMC) mRNA was detected by reverse transcription polymerase chain reaction (RT-PCR). Beta-endorphin was expressed mostly in the corpora lutea and perivascular stroma, but a weak to moderate immunostaining was also present in the theca cells and some granulosa cells of tertiary/antral and preovulatory follicles. The main differences observed in the distribution of ovarian beta-endorphin between the two groups were a more intense immunostaining in the granulosa cells of antral follicles, corpus luteum and stroma of PRL-treated rats. POMC gene transcripts were detected in 2/5 samples from the control group and in 3/7 samples from the PRL-treated group. Thus, the expression of beta-endorphin in tertiary/antral follicles is enhanced by PRL treatment in immature, superovulated rats, providing a putative mechanism by which PRL could inhibit the ovarian response to induced ovulation.


Sujet(s)
Ovaire/anatomie et histologie , Ovaire/effets des médicaments et des substances chimiques , Prolactine/pharmacologie , Superovulation , bêta-Endorphine/métabolisme , Animaux , Femelle , Régulation de l'expression des gènes , Humains , Prolactine/génétique , Prolactine/métabolisme , Puberté , Rats , Rat Wistar , bêta-Endorphine/génétique
20.
Reproduction ; 125(4): 469-77, 2003 Apr.
Article de Anglais | MEDLINE | ID: mdl-12683918

RÉSUMÉ

The aim of this study was to investigate the relationship between beta-endorphin and nitric oxide (NO) during the ovulatory process in rats. Immature rats were treated with equine chorionic gonadotrophin-hCG to induce ovulation. An intrabursal injection of beta-endorphin stimulated nitric oxide synthase (NOS) activity. This effect was completely reversed when naltrexone was co-injected with beta-endorphin. The stimulatory action of beta-endorphin on NOS activity was studied to determine whether it was exerted via prostaglandins. Treatment with prostaglandin E(2) (PGE(2)) completely reversed the beta-endorphin-induced stimulation of NOS activity. Moreover, intrabursal injection of meloxicam, an inhibitor of cyclooxygenase 2, increased NOS activity, but this effect was not altered by co-injection with beta-endorphin. The presence of both endothelial NOS (eNOS) and inducible NOS (iNOS) in the ovary at 10 h after hCG treatment was studied by western blot analysis. Local administration of beta-endorphin inhibited the expression of eNOS protein, whereas expression of iNOS protein was not detectable. Ovarian beta-endorphin content was diminished at 10 h after hCG injection. Treatment with prostaglandin synthesis inhibitors in vivo augmented the ovarian beta-endorphin content. In conclusion, these results indicate that beta-endorphin stimulates the activity of ovarian NOS indirectly by inhibiting prostaglandin production.


Sujet(s)
Dinoprostone/pharmacologie , Monoxyde d'azote/métabolisme , Ovaire/métabolisme , Ovulation/physiologie , bêta-Endorphine/pharmacologie , Animaux , Technique de Western/méthodes , Gonadotrophine chorionique/pharmacologie , Dinoprostone/métabolisme , Femelle , Indométacine/pharmacologie , Méloxicam , Modèles animaux , Naltrexone/pharmacologie , Nitric oxide synthase/analyse , Nitric oxide synthase/antagonistes et inhibiteurs , Nitric oxide synthase/métabolisme , Nitric oxide synthase type II , Nitric oxide synthase type III , Ovaire/composition chimique , Antagonistes des prostaglandines/pharmacologie , Rats , Rat Sprague-Dawley , Thiazines/pharmacologie , Thiazoles/pharmacologie , bêta-Endorphine/analyse , bêta-Endorphine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE