Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Drug Chem Toxicol ; 45(1): 250-261, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31656103

RESUMEN

Long-term morphine use for therapeutic approaches may lead to serious side effects. Several studies have suggested opioid antagonist and antioxidant therapy for reducing adverse effects of morphine. Cinnamaldehyde has a potent anti-oxidant property. In this study, separate and combined effects of cinnamaldehyde and naloxone (an opioid receptor antagonist) on behavioral changes and cerebellar histological and biochemical outcomes were investigated after long-term morphine administration. Seventy-eight rats were divided into two major morphine-treated and morphine-untreated groups. Morphine-treated group was subdivided into seven subgroups for receiving vehicle, normal saline, cinnamaldehyde (1.25, 5, and 20 mg/kg), naloxone, and cinnamaldehyde plus naloxone before morphine. Morphine-untreated group was subdivided into six subgroups and treated with vehicle, cinnamaldehyde (1.25, 5, and 20 mg/kg), naloxone, and their combination. Chemical compounds were administered for 28 consecutive days. Behavioral tests including footprint, rotarod, and beam balance tests were employed. Histopathological and biochemical alterations of cerebellum were determined. Body and cerebellum weights, stride width, time spent on the rotarod, Purkinje cell number, thickness of molecular and granular layers, superoxide dismutase (SOD), and total antioxidant capacity (TAC) decreased as a result of administrating morphine. Morphine increased beam transverse time, malondealdehyde (MDA), tumor necrosis factor-α (TNF-α), and caspase-3 levels. Histopathological changes such as cellular vacuolation and loss were also produced as a result of treatment with morphine. Cinnamaldehyde, naloxone, and their combination treatments improved all the above-mentioned alterations induced by morphine. We concluded that cinnamaldehyde produced a neuroprotective effect through anti-oxidant, anti-inflammatory, apoptotic, and probably naloxone-sensitive opioid receptor interaction mechanisms.


Asunto(s)
Morfina , Naloxona , Acroleína/análogos & derivados , Animales , Cerebelo , Morfina/toxicidad , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Ratas
2.
Exp Neurol ; 343: 113787, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34153321

RESUMEN

Opioid addiction can produce severe side effects including physical dependence and withdrawal. Perturbations of the gut microbiome have recently been shown to alter opioid-induced side-effects such as addiction, tolerance and dependence. In the present study, we investigated the influence of the gut microbiome on opioid withdrawal by evaluating the effects of fecal microbiota transplantation (FMT), antibiotic and probiotic treatments, and pharmacological inhibition of gut permeability in a mouse model of opioid dependence. Repeated intraperitoneal (i.p.) morphine treatment produced physical dependence that was quantified by measuring somatic signs of withdrawal (i.e. number of jumps) precipitated using the opioid antagonist naloxone. Morphine-dependent mice that received FMT from morphine-treated donor mice exhibited fewer naloxone-precipitated jumps compared to morphine-dependent counterparts receiving FMT from saline-treated donor mice. Microbial contents in the mouse cecum were altered by morphine treatment but were not differentially impacted by FMT. A broad-spectrum antibiotic cocktail (ABX) regimen reduced the bacterial load and attenuated naloxone-precipitated morphine withdrawal in morphine-dependent mice, whereas commercially available probiotic strains did not reliably alter somatic signs of opioid withdrawal. ML-7, a pharmacological inhibitor of gut permeability, reduced the morphine-induced increase in gut permeability in vivo but did not reliably alter somatic signs of naloxone-precipitated opioid withdrawal. Our results suggest that the gut microbiome impacts the development of physical dependence induced by chronic morphine administration, and that therapeutic manipulations of the gut microbiome may reduce opioid withdrawal.


Asunto(s)
Antibacterianos/administración & dosificación , Trasplante de Microbiota Fecal/métodos , Dependencia de Morfina/terapia , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Síndrome de Abstinencia a Sustancias/terapia , Analgésicos Opioides/administración & dosificación , Animales , Terapia Combinada/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Dependencia de Morfina/tratamiento farmacológico , Dependencia de Morfina/metabolismo , Probióticos/administración & dosificación , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Síndrome de Abstinencia a Sustancias/metabolismo
3.
Cardiovasc Toxicol ; 21(3): 248-254, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33125619

RESUMEN

Several case reports suggest QT prolongation leading to ventricular arrhythmias with fatal outcome after intoxication with the µ-opioid receptor agonist and anti-diarrheal agent loperamide. The number of cases of loperamide misuse are growing due to its potential stimulating effects. Loperamide intoxications can be treated by naloxone. However, previous reports described a further QT prolongation associated with naloxone administration. Therefore, the aim of this study was to investigate the effects of loperamide and naloxone on the cardiac electrophysiology in a sensitive whole-heart model. Twenty-six hearts of New Zealand White rabbits were retrogradely perfused in a modified Langendorff apparatus. Monophasic action potentials were recorded by endo- and epicardially positioned catheters. Hearts were stimulated at different cycle lengths, thereby obtaining action potential duration at 90% of repolarization (APD90) and QT intervals. Programmed ventricular stimulation was used to assess ventricular vulnerability. Fourteen hearts were perfused with ascending concentrations of loperamide (0.2 µM, 0.35 µM, and 0.5 µM) after obtaining baseline data. Another 12 hearts were treated with naloxone (0.1 µM, 0.5 µM, 2 µM). Loperamide led to a significant increase in QT interval, APD90, and ventricular tachycardia (VT) episodes. In contrast, naloxone led to a decrease in QT interval and APD90. Accordingly, the number of VT episodes was unaltered. To the best of our knowledge, this is the first experimental study that investigated the effects of loperamide and naloxone in a whole-heart model. Loperamide led to a significant increase in action potential duration and QT interval. Simultaneously, the number of ventricular tachycardias was significantly increased. In contrast, naloxone led to a shortening of the action potential duration without altering arrhythmia susceptibility.


Asunto(s)
Analgésicos Opioides/toxicidad , Antidiarreicos/toxicidad , Corazón/efectos de los fármacos , Loperamida/toxicidad , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Taquicardia Ventricular/inducido químicamente , Potenciales de Acción/efectos de los fármacos , Animales , Estimulación Cardíaca Artificial , Cardiotoxicidad , Femenino , Corazón/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Preparación de Corazón Aislado , Conejos , Taquicardia Ventricular/fisiopatología , Factores de Tiempo
4.
Toxicol Lett ; 325: 34-42, 2020 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-32070766

RESUMEN

Carfentanil is an ultra-potent opioid with an analgesic potency 10,000 times that of morphine but has received little scientific investigation. Three experiments were conducted to evaluate the toxicity of carfentanil and the efficacy of naloxone in adult male African green monkeys. The first experiment determined the ED50 (found to be 0.71 µg/kg) of subcutaneous carfentanil for inducing bradypnea and/or loss of posture. Experiment 2 attempted to establish the ED50 of naloxone for rapidly reversing the bradypnea/loss of posture induced by carfentanil (1.15 µg/kg). Experiment 3 evaluated the effects of carfentanil (0.575 µg/kg) alone, the safety of naloxone (71-2841 µg/kg), and the efficacy of naloxone (71-710 µg/kg) administration at two time points following carfentanil (1.15 µg/kg) on operant choice reaction time. Collectively, these experiments characterized the temporal progression of carfentanil-induced toxic signs, determined the range of naloxone doses that restored respiratory and gross behavioral function, and determined the time course and range of naloxone doses that partially or completely reversed the effects of carfentanil on operant choice reaction time performance in African green monkeys. These results have practical relevance for the selection of opioid antagonists, initial doses, and expected functional outcomes following treatment of synthetic opioid overdose in a variety of operational/emergency response contexts.


Asunto(s)
Analgésicos Opioides/toxicidad , Fentanilo/análogos & derivados , Naloxona/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Analgésicos Opioides/antagonistas & inhibidores , Animales , Apnea/inducido químicamente , Apnea/prevención & control , Conducta Animal/efectos de los fármacos , Chlorocebus aethiops , Condicionamiento Operante/efectos de los fármacos , Sobredosis de Droga/tratamiento farmacológico , Fentanilo/antagonistas & inhibidores , Fentanilo/toxicidad , Masculino , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Tiempo de Reacción/efectos de los fármacos
5.
J Immunol Res ; 2020: 8874288, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33490291

RESUMEN

The aim of this study was to evaluate the efficacy of selenium nanoparticle (an immune booster) and naloxone (an opioid receptor antagonist) as a new adjuvant in increasing immune responses against killed whole-cell Vibrio cholerae in a mouse cholera model. The Se NPs were synthesized and characterized by UV-visible, DLS, and zeta potential analysis. The SEM image confirmed the uniformity of spherical morphology of nanoparticle shape with 34 nm in size. The concentration of the Se NPs was calculated as 0.654 µg/ml in the ICP method. The cytotoxic activity of Se NPs on Caco-2 cells was assessed by the MTT assay and revealed 82.05% viability of cells after 24 h exposure with 100 µg/ml of Se NPs. Female BALB/C mice were orally immunized three times on days 0, 14, and 28, and challenge experiments were performed on immunized neonates with toxigenic V. cholerae. Administration of Se NP diet led to significant increase in V. cholerae-specific IgG and IgA responses in serum and saliva and caused protective immunity and 83.3% survival in challenge experiment against 1 LD50 V. cholerae in a group receiving diet of Se NPs compared with other groups including Dukoral vaccine. The IL-4 and IL-5 were significantly increased in response to WC+daily diet of Se NPs with or without naloxone. Naloxone proved no effect on IL-4 and IL-5 increase and is proposed as null in the cytokine and antibody production process. These results reveal that daily diet of Se NPs could efficiently induce immune cell effectors in both humoral and mucosal levels.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra el Cólera/administración & dosificación , Cólera/prevención & control , Selenio/administración & dosificación , Adyuvantes Inmunológicos/toxicidad , Administración Oral , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Células CACO-2 , Cólera/sangre , Cólera/inmunología , Cólera/microbiología , Vacunas contra el Cólera/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunogenicidad Vacunal , Ratones , Naloxona/administración & dosificación , Naloxona/toxicidad , Nanopartículas/administración & dosificación , Nanopartículas/toxicidad , Selenio/toxicidad , Pruebas de Toxicidad Aguda , Vacunación/métodos , Vibrio cholerae/inmunología
6.
Metab Brain Dis ; 35(2): 305-313, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31630319

RESUMEN

Opioid-induced neuroinflammation plays a role in the development of opioid physical dependence. Moreover, nitric oxide (NO) has been implicated in several oxidative and inflammatory pathologies. Here, we sought to determine whether treatment with venlafaxine during the development of morphine dependence could inhibit naloxone-precipitated withdrawal symptoms. The involvement of neuro-inflammation related cytokines, oxidative stress, and L-arginine (L-arg)-NO pathway in these effects were also investigated. Mice received morphine (50 mg/kg/daily; s.c.), plus venlafaxine (5 and 40 mg/kg, i.p.) once a day for 3 consecutive days. In order to evaluate the possible role of L-arg-NO on the effects caused by venlafaxine, animals received L-arg, L-NAME or aminoguanidine with venlafaxine (40 mg/kg, i.p.) 30 min before each morphine injection for 3 consecutive days. On 4th day of experiment, behavioral signs of morphine-induced physical dependence were evaluated after i.p. naloxone injection. Then, brain levels of tissue necrosis factor-alpha (TNF-α), interleukin-1-beta (IL-1ß), interleukin-6 (IL-6), interleukin-10 (IL-10), brain-derived neurotrophic factor (BDNF), NO and oxidative stress factors including; total thiol, malondialdehyde (MDA) contents and glutathione peroxidase (GPx) activity were determined. Co-administration of venlafaxine (40 mg/kg) with morphine not only inhibited the naloxone-precipitated withdrawal signs including jumping and weight loss, but also reduced the up-regulation of TNF-α, IL-1ß, IL-6, NO and MDA contents in mice brain tissue. However, repeated administration of venlafaxine inhibited the decrease in the brain levels of BDNF, total thiol and GPx. Pre-administration of L-NAME and aminoguanidine improved, while L-arg antagonized the venlafaxine-induced effects. These results provide evidences that venlafaxine could be used as a candidate drug to inhibit morphine withdrawal through the involvement of inflammatory cytokines and l-arginine-NO in mice.


Asunto(s)
Citocinas/antagonistas & inhibidores , Morfina/efectos adversos , Naloxona/toxicidad , Óxido Nítrico/antagonistas & inhibidores , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Clorhidrato de Venlafaxina/uso terapéutico , Animales , Citocinas/metabolismo , Masculino , Ratones , Dependencia de Morfina/tratamiento farmacológico , Dependencia de Morfina/metabolismo , Óxido Nítrico/metabolismo , Inhibidores de Captación de Serotonina y Norepinefrina/farmacología , Inhibidores de Captación de Serotonina y Norepinefrina/uso terapéutico , Síndrome de Abstinencia a Sustancias/metabolismo , Clorhidrato de Venlafaxina/farmacología
7.
Drug Res (Stuttg) ; 70(1): 23-25, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31525817

RESUMEN

The experiment was conducted on 10 Wistar rats, male and female, with initial body weight 270-280 g (males) and 250-260 g (females). The drug was administered using a spray cap in 10 doses of 0.1 mg at 45 min intervals. The average cumulative dose of the drug per naloxone hydrochloride was 36.6 mg/kg for males and 39.4 mg/kg for females. The animals were monitored for 2 weeks after the exposure and then euthanized by a gentle decapitation.We noticed that after each drug administration the animals showed a decrease in motor activity. During the observation period there were no animal deaths or signs of abnormalities in their general state or behavior. Beginning on day 7 a significant increase in body weight of the animals was noted in comparison with the initial data. The relative mass of the internal organs of the treated rats remained within the physiological norm.We conclude that naloxone hydrochloride after an intranasal administration at 36.6 mg/kg for males and 39.4 mg/kg for females does not cause death of animals and or have a toxic effect on their general state, does not change their protein metabolism characteristics or the appearance of the internal organs and their mass.


Asunto(s)
Actividad Motora/efectos de los fármacos , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Pruebas de Toxicidad Aguda , Administración Intranasal , Administración Oral , Animales , Técnicas de Observación Conductual , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Modelos Animales , Naloxona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Ratas , Ratas Wistar , Factores Sexuales
8.
Psychopharmacology (Berl) ; 237(2): 375-384, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31712968

RESUMEN

RATIONALE: Acute naloxone-precipitated morphine withdrawal (MWD) produces a conditioned place aversion (CPA) in rats even after one or two exposures to high-dose (20 mg/kg, sc) morphine followed 24-h later by naloxone (1 mg/kg, sc). However, the somatic withdrawal reactions produced by acute naloxone-precipitated MWD in rats have not been investigated. A recently discovered fatty acid amide, N-oleoylglycine (OlGly), which has been suggested to act as a fatty acid amide hydrolase (FAAH) inhibitor and as a peroxisome proliferator-activated receptor alpha (PPARα) agonist, was previously shown to interfere with a naloxone-precipitated MWD-induced CPA in rats. OBJECTIVES: The aims of these studies were to examine the somatic withdrawal responses produced by acute naloxone-precipitated MWD and determine whether OlGly can also interfere with these responses. RESULTS: Here, we report that following two exposures to morphine (20 mg/kg, sc) each followed by naloxone (1 mg/kg, sc) 24 h later, rats display nausea-like somatic reactions of lying flattened on belly, abdominal contractions and diarrhea, and display increased mouthing movements and loss of body weight. OlGly (5 mg/kg, ip) interfered with naloxone-precipitated MWD-induced abdominal contractions, lying on belly, diarrhea and mouthing movements in male Sprague-Dawley rats, by both a cannabinoid 1 (CB1) and a PPARα mechanism of action. Since these withdrawal reactions are symptomatic of nausea, we evaluated the potential of OlGly to interfere with lithium chloride (LiCl)-induced and MWD-induced conditioned gaping in rats, a selective measure of nausea; the suppression of MWD-induced gaping reactions by OlGly was both CB1 and PPARα mediated. CONCLUSION: These results suggest that the aversive effects of acute naloxone-precipitated MWD reflect nausea, which is suppressed by OlGly.


Asunto(s)
Glicina/análogos & derivados , Morfina/efectos adversos , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Náusea/tratamiento farmacológico , Ácidos Oléicos/uso terapéutico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Animales , Femenino , Glicina/farmacología , Glicina/uso terapéutico , Masculino , Síntomas sin Explicación Médica , Dependencia de Morfina/tratamiento farmacológico , Dependencia de Morfina/fisiopatología , Náusea/inducido químicamente , Náusea/fisiopatología , Ácidos Oléicos/farmacología , Ratas , Ratas Sprague-Dawley , Musarañas , Síndrome de Abstinencia a Sustancias/etiología , Síndrome de Abstinencia a Sustancias/fisiopatología
9.
Nat Neurosci ; 22(7): 1053-1056, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31209376

RESUMEN

The lateral habenula encodes aversive stimuli contributing to negative emotional states during drug withdrawal. Here we report that morphine withdrawal in mice leads to microglia adaptations and diminishes glutamatergic transmission onto raphe-projecting lateral habenula neurons. Chemogenetic inhibition of this circuit promotes morphine withdrawal-like social deficits. Morphine withdrawal-driven synaptic plasticity and reduced sociability require tumor necrosis factor-α (TNF-α) release and neuronal TNF receptor 1 activation. Hence, habenular cytokines control synaptic and behavioral adaptations during drug withdrawal.


Asunto(s)
Citocinas/fisiología , Habénula/fisiología , Morfina/efectos adversos , Conducta Social , Síndrome de Abstinencia a Sustancias/fisiopatología , Transmisión Sináptica/fisiología , Adaptación Psicológica , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Naloxona/toxicidad , Plasticidad Neuronal , Distribución Aleatoria , Receptores de Glutamato/análisis , Receptores de N-Metil-D-Aspartato/análisis , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/fisiología , Síndrome de Abstinencia a Sustancias/psicología , Factor de Necrosis Tumoral alfa/fisiología
10.
Psychopharmacology (Berl) ; 236(9): 2623-2633, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30993360

RESUMEN

RATIONALE: Oleoyl glycine (OlGly), a recently discovered fatty acid amide that is structurally similar to N- acylethanolamines, which include the endocannabinoid, anandamide (AEA), as well as endogenous peroxisome proliferator-activated receptor alpha (PPARα) agonists oleoylethanolamide (OEA) and palmitoylethanolamide (PEA), has been shown to interfere with nicotine reward and dependence in mice. OBJECTIVES AND METHODS: Behavioral and molecular techniques were used to investigate the ability of OlGly to interfere with the affective properties of morphine and morphine withdrawal (MWD) in male Sprague-Dawley rats. RESULTS: Synthetic OlGly (1-30 mg/kg, intraperitoneal [ip]) produced neither a place preference nor aversion on its own; however, at doses of 1 and 5 mg/kg, ip, it blocked the aversive effects of MWD in a place aversion paradigm. This effect was reversed by the cannabinoid 1 (CB1) receptor antagonist, AM251 (1 mg/kg, ip), but not the PPARα antagonist, MK886 (1 mg/kg, ip). OlGly (5 or 30 mg/kg, ip) did not interfere with a morphine-induced place preference or reinstatement of a previously extinguished morphine-induced place preference. Ex vivo analysis of tissue (nucleus accumbens, amygdala, prefrontal cortex, and interoceptive insular cortex) collected from rats experiencing naloxone-precipitated MWD revealed that OlGly was selectively elevated in the nucleus accumbens. MWD did not modify levels of the endocannabinoids 2-AG and AEA, nor those of the PPARα ligands, OEA and PEA, in any region evaluated. CONCLUSION: Here, we show that OlGly interferes with the aversive properties of acute naloxone-precipitated morphine withdrawal in rats. These results suggest that OlGly may reduce the impact of MWD and may possess efficacy in treating opiate withdrawal.


Asunto(s)
Analgésicos Opioides/efectos adversos , Glicina/análogos & derivados , Morfina/efectos adversos , Naloxona/toxicidad , Ácidos Oléicos/administración & dosificación , Recompensa , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Glicina/administración & dosificación , Glicina/metabolismo , Masculino , Ratones , Antagonistas de Narcóticos/toxicidad , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Ácidos Oléicos/metabolismo , Ratas , Ratas Sprague-Dawley , Síndrome de Abstinencia a Sustancias/metabolismo , Síndrome de Abstinencia a Sustancias/psicología
11.
Pain ; 160(2): 307-321, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30412056

RESUMEN

Ocular pain is a core symptom of inflammatory or traumatic disorders affecting the anterior segment. To date, the management of chronic ocular pain remains a therapeutic challenge in ophthalmology. The main endogenous opioids (enkephalins) play a key role in pain control but exhibit only transient analgesic effects due to their rapid degradation. The aim of this study was to explore the antinociceptive and anti-inflammatory effects of topical administration of PL265 (a dual enkephalinase inhibitor) on murine models of corneal pain. On healthy corneas, chronic PL265 topical administration did not alter corneal integrity nor modify corneal mechanical and chemical sensitivity. Then, on murine models of corneal pain, we showed that repeated instillations of PL265 (10 mM) significantly reduced corneal mechanical and chemical hypersensitivity. PL265-induced corneal analgesia was completely antagonized by naloxone methiodide, demonstrating that PL265 antinociceptive effects were mediated by peripheral corneal opioid receptors. Moreover, flow cytometry (quantification of CD11b+ cells) and in vivo confocal microscopy analysis revealed that instillations of PL265 significantly decreased corneal inflammation in a corneal inflammatory pain model. Chronic PL265 topical administration also decreased Iba1 and neuronal injury marker (ATF3) staining in the nucleus of primary sensory neurons of ipsilateral trigeminal ganglion. These results open a new avenue for ocular pain treatment based on the enhancement of endogenous opioid peptides' analgesic effects in tissues of the anterior segment of the eye. Dual enkephalinase inhibitor PL265 seems to be a promising topical treatment for safe and effective alleviation of ocular pain and inflammation.


Asunto(s)
Córnea/patología , Inhibidores Enzimáticos/administración & dosificación , Inflamación/tratamiento farmacológico , Dolor/tratamiento farmacológico , Propionatos/administración & dosificación , Administración Tópica , Animales , Antiinfecciosos Locales/uso terapéutico , Compuestos de Benzalconio/uso terapéutico , Capsaicina/toxicidad , Córnea/efectos de los fármacos , Lesiones de la Cornea/inducido químicamente , Lesiones de la Cornea/complicaciones , Modelos Animales de Enfermedad , Hiperalgesia/fisiopatología , Inflamación/inducido químicamente , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Dolor/etiología , Umbral del Dolor/efectos de los fármacos , Fármacos del Sistema Sensorial/toxicidad , Ganglio del Trigémino/metabolismo , Ganglio del Trigémino/patología
12.
Cancer Chemother Pharmacol ; 79(2): 431-434, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28011980

RESUMEN

PURPOSE: Irinotecan-induced gut toxicity is mediated in part by Toll-Like receptor 4 (TLR4) signalling. The primary purpose of this preclinical study was to determine whether blocking TLR4 signalling by administering (-)-naloxone, a TLR4 antagonist, would improve irinotecan-induced gut toxicity. Our secondary aim was to determine the impact of (-)-naloxone on tumour growth. METHODS: Female Dark Agouti (DA) tumour-bearing rats were randomly assigned to four treatments (n = 6 in each); control, (-)-naloxone (100 mg/kg oral gavage at -2, 24, 48, and 72 h), irinotecan (175 mg/kg intraperitoneal at 0 h), and (-)-naloxone and irinotecan. Body weight and tumour growth were measured daily, and diarrhoea incidence and severity were recorded 4× per day up to 72 h post-treatment. RESULTS: At 72 h, all rats that received irinotecan lost weight compared to controls (p = 0.03). In addition, rats that received (-)-naloxone and irinotecan lost significantly more weight compared to controls (p < 0.005) than irinotecan only compared to controls (p = 0.001). (-)-Naloxone did not attenuate irinotecan-induced severe diarrhoea at 48 and 72 h. Finally, (-)-naloxone caused increased tumour growth compared to control at 72 h (p < 0.05) and significantly reduced the efficacy of irinotecan (p = 0.001). CONCLUSIONS: (-)-Naloxone in our preclinical model was unable to block irinotecan-induced gut toxicity and decreased the efficacy of irinotecan. As (-)-naloxone-oxycodone combination is used for cancer pain, this may present a potential safety concern for patients receiving (-)-naloxone-oxycodone and irinotecan concurrently and requires further investigation.


Asunto(s)
Camptotecina/análogos & derivados , Naloxona/toxicidad , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Camptotecina/toxicidad , Dolor en Cáncer/tratamiento farmacológico , Diarrea/inducido químicamente , Diarrea/prevención & control , Femenino , Irinotecán , Naloxona/farmacología , Ratas , Receptor Toll-Like 4/fisiología
13.
MMWR Morb Mortal Wkly Rep ; 65(41): 1148-1149, 2016 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-27764078

RESUMEN

Expanding access to office-based medication-assisted treatment with buprenorphine/naloxone for opioid dependence is a key part of the national strategy to address the opioid abuse epidemic (1). However, as buprenorphine/naloxone prescribing increased, emergency department (ED) visits and hospitalizations for unsupervised ingestions by young children began to increase, with buprenorphine/naloxone ingestions becoming the most common cause of hospitalization for medication ingestions by young children during 2010-2011 (2). Buprenorphine ingestions might be asymptomatic or can cause drowsiness, vomiting, or respiratory depression, which if untreated can result in death (3). Buprenorphine/naloxone was available only as tablets in multidose child-resistant bottles (Suboxone) until late 2010, when film strips packaged in unit-dose, child-resistant pouches were introduced. In 2013, tablets became available in unit-dose packaging (Zubsolv). Because unit-dose, child-resistant packaging encloses each dose until opened, it might limit unintended ingestions by young children compared with traditional child-resistant bottles that must be resecured after every use (4). This study compared ED visits for pediatric buprenorphine/naloxone ingestions before and after these product packaging/formulation changes.


Asunto(s)
Buprenorfina/toxicidad , Servicio de Urgencia en Hospital/estadística & datos numéricos , Naloxona/toxicidad , Pediatría , Combinación Buprenorfina y Naloxona , Preescolar , Embalaje de Medicamentos , Ingestión de Alimentos , Femenino , Hospitalización/estadística & datos numéricos , Humanos , Lactante , Masculino , Estados Unidos
14.
Pain ; 157(10): 2285-2296, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27306045

RESUMEN

Individuals with Alzheimer's disease (AD) are in susceptible patient groups in which pain is an important clinical issue that is often underdiagnosed. However, it is unclear whether decreased pain complaints in patients with AD result from elevated pain tolerance or an impaired ability to communicate sensations. Here, we explored if AD-related pathology is present in key regions of the pain pathway and assessed whether nociceptive thresholds to acute noxious stimulation are altered in the double-mutant APPswe × PS1.M146V (TASTPM) transgenic mouse model of AD. TASTPM mice exhibited an age-dependant cognitive deficit at the age of 6 months, but not at 4 months, a deficit that was accompanied by amyloid plaques in the cortex, hippocampus, and thalamus. In the spinal cord, ß-amyloid (APP/Aß) immunoreactivity was observed in dorsal and ventral horn neurons, and the expression of vesicular glutamate transporter 2 (VGLUT2) was significantly reduced, while the expression of the inhibitory peptides enkephalins was increased in TASTPM dorsal horn, consistent with an increased inhibitory tone. TASTPM mice displayed reduced sensitivity to acute noxious heat, which was reversed by naloxone, an opioid antagonist. This study suggests that increased inhibition and decreased excitation in the spinal cord may be responsible for the reduced thermal sensitivity associated with AD-related pathology.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Analgésicos Opioides/metabolismo , Trastornos del Conocimiento/etiología , Umbral Sensorial/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Carragenina/toxicidad , Modelos Animales de Enfermedad , Locomoción/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Proteínas del Tejido Nervioso/metabolismo , Dolor/inducido químicamente , Dimensión del Dolor , Presenilina-1/genética , Presenilina-1/metabolismo , Reconocimiento en Psicología/fisiología , Umbral Sensorial/efectos de los fármacos
15.
J Neurosci ; 36(11): 3391-7, 2016 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-26985045

RESUMEN

Mindfulness meditation, a cognitive practice premised on sustaining nonjudgmental awareness of arising sensory events, reliably attenuates pain. Mindfulness meditation activates multiple brain regions that contain a high expression of opioid receptors. However, it is unknown whether mindfulness-meditation-based analgesia is mediated by endogenous opioids. The present double-blind, randomized study examined behavioral pain responses in healthy human volunteers during mindfulness meditation and a nonmanipulation control condition in response to noxious heat and intravenous administration of the opioid antagonist naloxone (0.15 mg/kg bolus + 0.1 mg/kg/h infusion) or saline placebo. Meditation during saline infusion significantly reduced pain intensity and unpleasantness ratings when compared to the control + saline group. However, naloxone infusion failed to reverse meditation-induced analgesia. There were no significant differences in pain intensity or pain unpleasantness reductions between the meditation + naloxone and the meditation + saline groups. Furthermore, mindfulness meditation during naloxone produced significantly greater reductions in pain intensity and unpleasantness than the control groups. These findings demonstrate that mindfulness meditation does not rely on endogenous opioidergic mechanisms to reduce pain. SIGNIFICANCE STATEMENT: Endogenous opioids have been repeatedly shown to be involved in the cognitive inhibition of pain. Mindfulness meditation, a practice premised on directing nonjudgmental attention to arising sensory events, reduces pain by engaging mechanisms supporting the cognitive control of pain. However, it remains unknown if mindfulness-meditation-based analgesia is mediated by opioids, an important consideration for using meditation to treat chronic pain. To address this question, the present study examined pain reports during meditation in response to noxious heat and administration of the opioid antagonist naloxone and placebo saline. The results demonstrate that meditation-based pain relief does not require endogenous opioids. Therefore, the treatment of chronic pain may be more effective with meditation due to a lack of cross-tolerance with opiate-based medications.


Asunto(s)
Analgésicos Opioides/metabolismo , Meditación , Dolor/metabolismo , Dolor/rehabilitación , Resultado del Tratamiento , Adulto , Análisis de Varianza , Método Doble Ciego , Femenino , Voluntarios Sanos , Calor/efectos adversos , Humanos , Masculino , Meditación/psicología , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Dolor/inducido químicamente , Dimensión del Dolor , Psicofísica , Adulto Joven
16.
Can J Physiol Pharmacol ; 93(11): 935-44, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26440527

RESUMEN

Long-term use of opioids for pain management results in rapid development of tolerance and dependence leading to severe withdrawal symptoms. We have previously demonstrated that endothelin-A (ETA) receptor antagonists potentiate opioid analgesia and eliminate analgesic tolerance. This study was designed to investigate the involvement of central ET mechanisms in opioid withdrawal. The effect of intracerebroventricular administration of ETA receptor antagonist BQ123 on morphine and oxycodone withdrawal was determined in male Swiss Webster mice. Opioid tolerance was induced and withdrawal was precipitated by the opioid antagonist naloxone. Expression of ETA and ETB receptors, nerve growth factor (NGF), and vascular endothelial growth factor was determined in the brain using Western blotting. BQ123 pretreatment reversed hypothermia and weight loss during withdrawal. BQ123 also reduced wet shakes, rearing behavior, and jumping behavior. No changes in expression of vascular endothelial growth factor, ETA receptors, and ETB receptors were observed during withdrawal. NGF expression was unaffected in morphine withdrawal but significantly decreased during oxycodone withdrawal. A decrease in NGF expression in oxycodone- but not in morphine-treated mice could be due to mechanistic differences in oxycodone and morphine. It is concluded that ETA receptor antagonists attenuate opioid-induced withdrawal symptoms.


Asunto(s)
Antagonistas de los Receptores de la Endotelina A/uso terapéutico , Naloxona/toxicidad , Trastornos Relacionados con Opioides/tratamiento farmacológico , Péptidos Cíclicos/uso terapéutico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Animales , Antagonistas de los Receptores de la Endotelina A/farmacología , Masculino , Ratones , Antagonistas de Narcóticos/toxicidad , Trastornos Relacionados con Opioides/patología , Trastornos Relacionados con Opioides/fisiopatología , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Péptidos Cíclicos/farmacología , Síndrome de Abstinencia a Sustancias/patología , Síndrome de Abstinencia a Sustancias/fisiopatología
17.
Toxicol Lett ; 228(2): 75-84, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24769261

RESUMEN

Respiratory depression has been attributed to buprenorphine (BUP) misuse or combination with benzodiazepines. BUP/naloxone (NLX) has been marketed as maintenance treatment, aiming at preventing opiate addicts from self-injecting crushed pills. However, to date, BUP/NLX benefits in comparison to BUP alone remain debated. We investigated the plethysmography effects of BUP/NLX in comparison to BUP/solvent administered by intravenous route in naive and BUP-tolerant Sprague-Dawley rats, and in combination with diazepam (DZP) or its solvent. In naive rats, BUP/NLX in comparison to BUP significantly increased respiratory frequency (f, P<0.05) without altering minute volume (VE). In combination to DZP, BUP/NLX significantly increased expiratory time (P<0.01) and decreased f (P<0.01), tidal volume (VT, P<0.001), and VE (P<0.001) while BUP only decreased VT (P<0.5). In BUP-tolerant rats, no significant differences in respiratory effects were observed between BUP/NLX and BUP. In contrast, in combination to DZP, BUP/NLX did not significantly alter the plethysmography parameters, while BUP increased inspiratory time (P<0.001) and decreased f (P<0.01) and VE (P<0.001). In conclusion, differences in respiratory effects between BUP/NLX and BUP are only significant in combination with DZP, with increased depression in naive rats but reduced depression in BUP-tolerant rats. However, BUP/NLX benefits in humans remain to be determined.


Asunto(s)
Analgésicos Opioides/toxicidad , Buprenorfina/toxicidad , Diazepam/toxicidad , Hipnóticos y Sedantes/toxicidad , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Insuficiencia Respiratoria/inducido químicamente , Análisis de Varianza , Animales , Interacciones Farmacológicas , Tolerancia a Medicamentos , Masculino , Pletismografía Total , Ratas , Ratas Sprague-Dawley
18.
Brain Res ; 1510: 22-37, 2013 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-23399681

RESUMEN

Peroxizome proliferator-activated receptor gamma (PPARγ) is highly expressed in the central nervous system where it modulates numerous gene transcriptions. Nitric oxide synthase (NOS) expression could be modified by simulation of PPARγ which in turn activates nitric oxide (NO)/soluble guanylyl-cyclase (sGC)/cyclic guanosine mono phosphate (cGMP) pathway. It is well known that NO/cGMP pathway possesses pivotal role in the development of opioid dependence and this study is aimed to investigate the effect of PPARγ stimulation on opioid dependence in mice as well as human glioblastoma cell line. Pioglitazone potentiated naloxone-induced withdrawal syndrome in morphine dependent mice in vivo. While selective inhibition of PPARγ, neuronal NOS or GC could reverse the pioglitazone-induced potentiation of morphine withdrawal signs; sildenafil, a phosphodiesterase-5 inhibitor amplified its effect. We also showed that nitrite levels in the hippocampus were significantly elevated in pioglitazone-treated morphine dependent mice. In the human glioblastoma (U87) cell line, rendered dependent to morphine, cAMP levels did not show any alteration after chronic pioglitazone administration while cGMP measurement revealed a significant rise. We were unable to show a significant alteration in neuronal NOS mRNA expressions by pioglitazone in mice hippocampus or U87 cells. Our results suggest that pioglitazone has the ability to enhance morphine-dependence and to augment morphine withdrawal signs. The possible pathway underlying this effect is through activation of NO/GC/cGMP pathway.


Asunto(s)
GMP Cíclico/metabolismo , Hipoglucemiantes/efectos adversos , Dependencia de Morfina/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal/efectos de los fármacos , Tiazolidinedionas/efectos adversos , Animales , Línea Celular Tumoral , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Glioblastoma/patología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Masculino , Ratones , Dependencia de Morfina/complicaciones , Dependencia de Morfina/patología , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Pioglitazona , ARN Mensajero , Síndrome de Abstinencia a Sustancias/etiología , Síndrome de Abstinencia a Sustancias/metabolismo , Transfección
19.
Indian J Pharmacol ; 44(4): 443-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23087502

RESUMEN

OBJECTIVE: Role of nitric oxide (NO) in reversing morphine anti-nociception has been shown. However, the interaction between NO and naloxone-induced pain in the hippocampus is unknown. The present study aimed to investigate the involvement of molecule NO in naloxone-induced pain and its possible interaction with naloxone into cortical area 1 (CA1) of hippocampus. MATERIALS AND METHODS: Male Wistar rats (250-350 g), provided by Pasteur Institute of Iran, were housed two per cage with food and water ad libitum. The animals' skulls were cannulated bilaterally at coordinates adjusted for CA1 of hippocampus (AP: -3.8; L: ±1.8- 2.2: V: 3) by using stereotaxic apparatus. Each experimental group included 6-8 rats. To induce inflammation pain, the rats received subcutaneous (s.c.) injections of formalin (50 µL at 2.5%) once prior to testing. To evaluate the nociceptive effect of naloxone, the main narcotic antagonist of morphine (0.1-0.4 mg/kg) was injected intraperitoneally (i.p.) 10 min before injection of formalin. Injections of L-arginine, a precursor of NO, and N(G)-Nitro-L-arginine Methyl Ester (L-NAME), an inhibitor of NO synthase (NOS), intra-CA1, were conducted orderly prior to the administration of naloxone. The pain induction was analyzed by analysis of variance (ANOVA). RESULTS: Naloxone at the lower doses caused a significant (P<0.01) pain in the naloxone-treated animals. However, pre-administration (1-2 min) of L-arginine (0.04, 0.08, 0.15, 0.3, 1.0, and 3.0 µg/rat, intra-CA1) reversed the response to naloxone. But, the response to L-arginine was blocked by pre-microinjection (1-2 min) of L-NAME (0.15, 0.3, 1.0, and 3.0 µg/rat), whilst, L-arginine or L-NAME alone did not induce pain behavior. CONCLUSION: NO in the rat hippocampal CA1 area is involved in naloxone-induced nociception.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Naloxona/metabolismo , Naloxona/toxicidad , Óxido Nítrico/metabolismo , Dolor/inducido químicamente , Dolor/metabolismo , Animales , Región CA1 Hipocampal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Masculino , Microinyecciones/métodos , NG-Nitroarginina Metil Éster/administración & dosificación , Naloxona/administración & dosificación , Óxido Nítrico/antagonistas & inhibidores , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Ratas , Ratas Wistar
20.
Synapse ; 66(8): 694-704, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22407757

RESUMEN

Midbrain periaqueductal gray (PAG) and spinal cord dorsal horn are major action sites of opioid analgesics in the pain pathway. Our previous study has shown that opioid antagonists activate MORS196A-CSTA (a mutant of mu-opioid receptor) as full agonists in vitro cell models and naloxone showed antinociceptive effects after the expression of MORS196A-CSTA in the spinal cord in mice. The purpose of this study is to investigate the site-directed antinociceptive effects of naloxone in mice injected with dsAAV-MORS196A-CSTA-EGFP at spinal cord or at periaqueductal gray. MORS196A-CSTA-EGFP was administered to ICR mice using dsAAV as vector. We measured MORS196A-CSTA-EGFP expression by detecting the EGFP visualization with a fluorescence microscope. The antinociceptive effect of naloxone was determined by tail-flick test and hot plate test. Drug rewarding effect was evaluated by the conditioned place preference test. Naloxone (10 mg/kg, s.c.) elicited both supraspinal and spinal antinociceptive responses in mice injected with the virus at PAG while only spinal antinociceptive response was observed in mice injected with virus at dorsal horn region. Chronic naloxone treatment did not induce physical dependence or rewarding effect in mice injected with MORS196A-CSTA-EGFP in spinal cord or PAG. These data suggest that the observed naloxone-induced antinociceptive response is the consequence of the local expression of MORS196A-CSTA at specific sites of pain pathway. Injection of such MOR mutant and the systemic administration of naloxone can be a new strategy in the management of chronic pain without the various side effects associated with the use of morphine.


Asunto(s)
Analgésicos Opioides/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Receptores Opioides mu/agonistas , Analgésicos Opioides/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos ICR , Morfina/farmacología , Dependencia de Morfina/tratamiento farmacológico , Mutación , Naloxona/toxicidad , Antagonistas de Narcóticos/toxicidad , Narcóticos/farmacología , Nocicepción/efectos de los fármacos , Nocicepción/fisiología , Sustancia Gris Periacueductal/efectos de los fármacos , Pruebas Psicológicas , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Médula Espinal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...