Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 660
Filtrar
Más filtros

Publication year range
1.
Toxicol Appl Pharmacol ; 483: 116802, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38184280

RESUMEN

The incidence of postoperative myocardial injury remains high as the underlying pathogenesis is still unknown. The dorsal root ganglion (DRG) neurons express transient receptor potential vanilloid 1 (TRPV1) and its downstream effector, calcitonin gene-related peptide (CGRP) participating in transmitting pain signals and cardiac protection. Opioids remain a mainstay therapeutic option for moderate-to-severe pain relief clinically, as a critical component of multimodal postoperative analgesia via intravenous and epidural delivery. Evidence indicates the interaction of opioids and TRPV1 activities in DRG neurons. Here, we verify the potential impairment of myocardial viability by epidural usage of opioids in postoperative analgesia. We found that large dose of epidural morphine (50 µg) significantly worsened the cardiac performance (+dP/dtmax reduction by 11% and -dP/dtmax elevation by 24%, all P < 0.001), the myocardial infarct size (morphine vs Control, 0.54 ± 0.09 IS/AAR vs. 0.23 ± 0.06 IS/AAR, P < 0.001) and reduced CGRP in the myocardium (morphine vs. Control, 9.34 ± 2.24 pg/mg vs. 21.23 ± 4.32 pg/mg, P < 0.001), while induced definite suppression of nociception in the postoperative animals. It was demonstrated that activation of µ-opioid receptor (µ-OPR) induced desensitization of TRPV1 by attenuating phosphorylation of the channel in the dorsal root ganglion neurons, via inhibiting the accumulation of cAMP. CGRP may attenuated the buildup of ROS and the reduction of mitochondrial membrane potential in cardiomyocytes induced by hypoxia/reoxygenation. The findings of this study indicate that epidurally giving large dose of µ-OPR agonist may aggravate myocardial injury by inhibiting the activity of TRPV1/CGRP pathway.


Asunto(s)
Analgésicos Opioides , Péptido Relacionado con Gen de Calcitonina , Animales , Analgésicos Opioides/toxicidad , Péptido Relacionado con Gen de Calcitonina/farmacología , Receptores Opioides mu/agonistas , Morfina/toxicidad , Miocardio/patología , Dolor/tratamiento farmacológico , Dolor/metabolismo , Dolor/patología , Miocitos Cardíacos/metabolismo , Canales Catiónicos TRPV/metabolismo , Ganglios Espinales
2.
J Neuroinflammation ; 19(1): 87, 2022 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-35414088

RESUMEN

BACKGROUND: Chronic morphine exposure induces immunosuppression in the peripheral and central nervous system, resulting in susceptibility of patients to invading pathogens. Mitophagy is a crucial regulator of inflammation, and dysregulated mitophagy may cause immunosuppression, but whether mitophagy is linked with morphine-induced immunosuppression in the brain remains unknown. NLRX1 is the only mitochondrially localized NOD family receptor protein which serves as a critical regulator in immunity and mitophagy activation, but it remains an enigma how NLRX1 functions in the crosstalk between microglial inflammatory defense and mitophagy in the presence of morphine. METHODS: Primary microglia and astrocytes, BV2 and MA cell lines were utilized. Mice were stimulated with repeated morphine treatment to mimic chronic morphine exposure, and activation of mitophagy, lysosomal functions, and inflammation were assayed in specific brain regions and immune organs with or without NLRX1-silencing. RESULTS: Morphine induced microglial mitophagy in a LC3 (microtubule-associated proteins light chain 3)-dependent manner, which was mediated by NLRX1. Contrastingly, morphine impaired lysosomal functions, including generation, acidification and mitophagosome-lysosome fusion, thus leading to insufficient mitophagy activation in microglia. NLRX1-silencing inhibited mitophagy activity and rescued lysosomal functions including generation and acidification in microglia. The NLRX1-mediated incomplete mitophagy in microglial cells contributed to immunosuppression and vulnerability towards pathogenic challenge after morphine treatment. In vivo, NLRX1-mediated microglial mitophagy activation by morphine was mainly located in the murine brain cortex, striatum, and cerebellum, where NLRX1 functioned as a negative immune regulator and facilitated septic shock. Collectively, microglial immune responses to septic shock were amenable to NLRX1 silencing in the brain with morphine treatment. CONCLUSION: Morphine activated insufficient mitophagy in microglia which was regulated by NLRX1, ultimately leading to host immunosuppression and susceptible conditions in the brain.


Asunto(s)
Mitofagia , Choque Séptico , Animales , Humanos , Terapia de Inmunosupresión , Ratones , Ratones Endogámicos NOD , Microglía/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Morfina/toxicidad , Choque Séptico/metabolismo , Choque Séptico/patología
3.
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
4.
J Cell Physiol ; 236(6): 4445-4454, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33184860

RESUMEN

Morphine, a mu-opioid receptor (MOR) agonist, has been extensively used to treat advanced cancer pain. In particular, in patients with cancer metastasis, both morphine and anticancer drugs are given simultaneously. However, evidence showed that morphine might be a risk factor in promoting the tumor's malignant potential. In this study, we report that treatment with morphine could activate MOR and lead to the promotion of proliferation, migration, and invasion in HCT116 and DLD1 colorectal cancer (CRC) cells with time-concentration dependence. Moreover, morphine can also contribute to cetuximab's drug resistance, a targeted drug widely used to treat advanced CRC by inducing the activation of epidermal growth factor receptor (EGFR). The cell phenotype includes proliferation, migration, invasion, and drug resistance, which may be reversed by MOR knockdown or adding nalmefene, the MOR receptor antagonist. Receptor tyrosine kinase array analysis revealed that morphine selectively induced the transactivation of EGFR. EGFR transactivation resulted in the activation of ERK1/2 and AKT. In conclusion, morphine induces the transactivation of EGFR via MOR. It activates the downstream signal pathway AKT-MTOR and RAS-MAPK, increases proliferation, migration, and invasion, and promotes resistance to EGFR inhibitors in a CRC cell line. Furthermore, we verified that EGFR inhibition by cetuximab strongly reversed the protumoral effects of morphine in vitro and in vivo. Collectively, we provide evidence that morphine-EGFR signaling might be a promising therapeutic target for CRC patients, especially for cetuximab-resistant CRC patients.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Cetuximab/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Resistencia a Antineoplásicos , Morfina/toxicidad , Receptores Opioides mu/agonistas , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HCT116 , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas ras/metabolismo
5.
J Vasc Res ; 58(6): 392-402, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34521095

RESUMEN

Recent studies have shown that chronic use of prescription or illicit opioids leads to an increased risk of cardiovascular events and pulmonary arterial hypertension. Indices of vascular age and arterial stiffness are also shown to be increased in opioid-dependent patients, with the effects being more marked in women. There are currently no studies investigating sex-specific vascular dysfunction in opioid use, and the mechanisms leading to opioid-induced vascular damage remain unknown. We hypothesized that exposure to exogenous opioids causes sex-specific vascular remodeling that will be more pronounced in female. Acknowledging the emerging roles of cofilins and extracellular signal-regulated kinases (ERKs) in mediating actin dynamics, we investigated the effects of morphine on these molecules. Twenty-four hour exposure to morphine increased inactivated cofilin and activated ERKs in resistance arteries from female mice, which may promote stress fiber over-assembly. We also performed continuous intraluminal infusion of morphine in pressurized resistance arteries from male and female mice using culture pressure myographs. We observed that morphine reduced the vascular diameter in resistance arteries from female, but not male mice. These results have significant implications for the previously unexplored role of exogenous opioids as a modifiable cardiovascular risk factor, especially in women.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Analgésicos Opioides/toxicidad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hemodinámica/efectos de los fármacos , Arterias Mesentéricas/efectos de los fármacos , Morfina/toxicidad , Remodelación Vascular/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Masculino , Arterias Mesentéricas/enzimología , Arterias Mesentéricas/patología , Arterias Mesentéricas/fisiopatología , Ratones Endogámicos C57BL , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fosforilación , Ratas Sprague-Dawley , Factores Sexuales , Transducción de Señal
6.
Toxicol Appl Pharmacol ; 416: 115458, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33607128

RESUMEN

Morphine is an opioid agonist and a nonselective mu, kappa and delta receptor agonist. It is a commonly used analgesic drug for the treatment of acute and chronic pain as well as cancer pain. Morphine is particularly important to address the problem of morphine tolerance. Tcf7l2, known as a risk gene for schizophrenia and autism, encodes a member of the LEF1/TCF transcription factor family. TCF7L2 is an important transcription factor that is upregulated in neuropathic pain models. However, the relationship between TCF7L2 and morphine tolerance has not been reported. In this study, we found that morphine tolerance led to the upregulation of TCF7L2 in the spinal cord, and also led to the upregulation of TCF7L2 expression in glial cells, which promoted inflammation related signal, and activated TLR4 / NF-κB/NLRP3 pathway. In addition, TCF7L2 regulated microglial cell activation induced by chronic morphine treatment. Mechanically, we found that TCF7L2 transcriptionally regulated TLR4 expression, and the depletion of TCF7L2 alleviated morphine tolerance induced by chronic morphine treatment, and further alleviated pain hypersensitivity induced by chronic morphine treatment. We therefore suggested that TCF7L2 regulates the activation of TLR4/ NF-κB/NLRP3 pathway in microglia, and is involved in the formation of morphine tolerance. Our results provide a new idea for the regulation mechanism of morphine tolerance.


Asunto(s)
Analgésicos Opioides/toxicidad , Tolerancia a Medicamentos , Hiperalgesia/inducido químicamente , Microglía/efectos de los fármacos , Morfina/toxicidad , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Dolor Nociceptivo/prevención & control , Receptor Toll-Like 4/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Masculino , Ratones , Microglía/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Dolor Nociceptivo/metabolismo , Dolor Nociceptivo/fisiopatología , Umbral del Dolor/efectos de los fármacos , Transducción de Señal , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Receptor Toll-Like 4/genética , Proteína 2 Similar al Factor de Transcripción 7/genética , Regulación hacia Arriba
7.
Int J Toxicol ; 40(6): 530-541, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34610777

RESUMEN

Respiratory monitoring, using impedance with implanted telemetry in socially housed animals, was not possible until the recent development of digital signal transmission. The objective of this study was to evaluate digital telemetry monitoring of cardiopulmonary parameters (respiratory rate, tidal volume, minute volume, electrocardiography (DII), systemic arterial blood pressure, physical activity, and body temperature) in conscious, single-housed, non-rodent species commonly used in toxicology studies following administration of positive/negative controls (saline, dexmedetomidine, morphine, amphetamine, and doxapram), and also, the effects of various social housing arrangements in untreated female and/or male cynomolgus monkeys, Beagle dogs, and Göttingen minipigs (n = 4 per species). Aggressions were observed in socially housed male minipigs, however, which prevented pair-housed assessments in this species. All tested pharmacological agents significantly altered more than one organ system, highlighting important inter-organ dependencies when analyzing functional endpoints. Stress-related physiological changes were observed with single-housing or pair-housing with a new cage mate in cynomolgus monkeys and Beagle dogs, suggesting that stable social structures are preferable to limit variability, especially around dosing. Concomitant monitoring of cardiovascular and respiratory parameters from the same animals may help reduce the number of animals (3 Rs) needed to fulfill the S7A guidelines and allows for identification of organ system functional correlations. Globally, the data support the use of social housing in non-rodents for safety pharmacology multi-organ system (heart and lungs) monitoring investigations.


Asunto(s)
Anfetamina/toxicidad , Analgésicos Opioides/toxicidad , Sistema Cardiovascular/efectos de los fármacos , Dexmedetomidina/toxicidad , Doxapram/toxicidad , Electrocardiografía/efectos de los fármacos , Morfina/toxicidad , Animales , Estimulantes del Sistema Nervioso Central/toxicidad , Perros , Impedancia Eléctrica , Macaca fascicularis , Porcinos , Porcinos Enanos
8.
Mol Biol Rep ; 47(8): 6073-6081, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32705505

RESUMEN

Morphine is the most common analgesic drug that is widely used in post-operative interventions. This drug causes free radical accumulation leading to spermatogenesis failure. Antioxidant agents like Sumach (Rhus coriaria) neutralize cellular free radicals. In this study, the properties of antioxidative, modulative of inflammatory cytokines, and apoptotic genes following Sumach extract administration on morphine-induced fertility destruction in male Wistar rats was evaluated. Sixty-four animals were grouped (n = 8) including; 1: control, 2: morphine, 3-5: Sumach (200, 400, 800 mg/kg), and 6-8: morphine + Sumach. Hydroalcoholic extract of Sumach seeds was prepared. Treatments with Sumach extract were applied orally and intraperitoneally daily for 8 weeks. The P53, Bcl2 and caspase-3 genes expression were measured by real-time PCR. Cytokines involved in inflammation were evaluated by ELISA. Sperm parameters, total antioxidant capacity (TAC), testosterone, and germinal layer height (GLH) were assessed. All parameters (investigated in this study) in Morphine group reduced significantly than the control group (P ˂ 0.01) (except P53 and caspase-3 genes expression and inflammatory cytokine which were improved). All factors in Sumach and Sumach + Morphine groups were significantly enhanced compared to the Morphine group (P ˂ 0.01) (except P53 and caspase-3 genes expression and inflammatory cytokine which were declined). Morphine disrupted the physiological function of male fertility system. Besides, all doses of Sumach showed no therapeutic changes compared to the control group. Sumach with anti-infertility features compensates the toxic effect of Morphine administration.


Asunto(s)
Infertilidad Masculina/tratamiento farmacológico , Morfina/toxicidad , Fitoterapia , Extractos Vegetales/uso terapéutico , Rhus/química , Administración Oral , Animales , Antioxidantes/análisis , Caspasa 3/biosíntesis , Caspasa 3/genética , Citocinas/sangre , Regulación de la Expresión Génica/efectos de los fármacos , Infertilidad Masculina/sangre , Infertilidad Masculina/inducido químicamente , Inyecciones Intraperitoneales , Masculino , Extractos Vegetales/administración & dosificación , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/genética , Distribución Aleatoria , Ratas , Ratas Wistar , Semillas/química , Túbulos Seminíferos/efectos de los fármacos , Túbulos Seminíferos/ultraestructura , Espermatozoides/efectos de los fármacos , Espermatozoides/ultraestructura , Testosterona/sangre , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
9.
Anesth Analg ; 128(1): 161-167, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29596097

RESUMEN

BACKGROUND: Opioids are effective postoperative analgesics. Disturbingly, we have previously reported that opioids such as morphine can worsen inflammatory pain and peripheral and central neuropathic pain. These deleterious effects are mediated by immune mediators that promote neuronal hyperexcitability in the spinal dorsal horn. Herein, we tested whether perioperative morphine could similarly prolong postoperative pain in male rats. METHODS: Rats were treated with morphine for 7 days, beginning immediately after laparotomy, while the morphine was tapered in a second group. Expression of genes for inflammatory mediators was quantified in the spinal dorsal horn. In the final experiment, morphine was administered before laparotomy for 7 days. RESULTS: We found that morphine treatment after laparotomy extended postoperative pain by more than 3 weeks (time × treatment: P < .001; time: P < .001; treatment: P < .05). Extension of postoperative pain was not related to morphine withdrawal, as it was not prevented by dose tapering (time × treatment: P = .8; time: P < .001; treatment: P = .9). Prolonged postsurgical pain was associated with increased expression of inflammatory genes, including those encoding Toll-like receptor 4, NOD like receptor protein 3 (NLRP3), nuclear factor kappa B (NFκB), caspase-1, interleukin-1ß, and tumor necrosis factor (P < .05). Finally, we showed that of preoperative morphine, concluding immediately before laparotomy, similarly prolonged postoperative pain (time × treatment: P < .001; time: P < .001; treatment: P < .001). There is a critical window for morphine potentiation of pain, as a 7-day course of morphine that concluded 1 week before laparotomy did not prolong postsurgical pain. CONCLUSIONS: These studies indicate the morphine can have a deleterious effect on postoperative pain. These studies further suggest that longitudinal studies could be performed to test whether opioids similarly prolong postoperative pain in the clinic.


Asunto(s)
Analgésicos Opioides/toxicidad , Hiperalgesia/inducido químicamente , Morfina/toxicidad , Umbral del Dolor/efectos de los fármacos , Dolor Postoperatorio/inducido químicamente , Células del Asta Posterior/efectos de los fármacos , Analgésicos Opioides/administración & dosificación , Animales , Modelos Animales de Enfermedad , Esquema de Medicación , Hiperalgesia/diagnóstico por imagen , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Mediadores de Inflamación/metabolismo , Laparotomía , Masculino , Morfina/administración & dosificación , Dimensión del Dolor , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/metabolismo , Dolor Postoperatorio/fisiopatología , Células del Asta Posterior/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
10.
Bull Environ Contam Toxicol ; 103(5): 710-716, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31482305

RESUMEN

The manufacturing and consumption of drugs of addiction has increased globally and their widespread occurrence in the environment is an emerging concern. This study evaluated the phytotoxicity of three compounds: methamphetamine, codeine and morphine; commonly reported in Australian urban water, to the aquatic plant Lemna minor under controlled conditions. L. minor was sensitive to lower drug concentrations when administered in multi-compound mixtures (100-500 µg L-1) than when applied individually (range 600-2500 µg L-1), while no adverse effects were observed at environmentally-relevant concentrations (1-5 µg L-1) detected in wastewater effluent. In conclusion, the results show that the concentrations of these compounds discharged into the environment are unlikely to pose adverse phytotoxic effects. These three compounds are known to be the most stable of their group under such conditions indicating that with this respect it is safe to use recycled water for existing regulated reclaimed purposes including agricultural or parklands irrigation or replenishing surface and groundwater. However, more research on the analysis of methamphetamines and opiates in municipal effluents is needed to reassure the likely environmental hazard of these neuroactive drug classes to aquatic organisms. Given the ever-growing production and aquatic disposal of discharge wastewater globally, this study provides timely and valuable insights into the likely drug-related impacts of effluent disposal on aquatic plants in receiving environments.


Asunto(s)
Araceae/efectos de los fármacos , Codeína/toxicidad , Drogas Ilícitas/toxicidad , Metanfetamina/toxicidad , Morfina/toxicidad , Contaminantes Químicos del Agua/toxicidad , Riego Agrícola , Australia , Codeína/análisis , Relación Dosis-Respuesta a Droga , Monitoreo del Ambiente , Drogas Ilícitas/análisis , Metanfetamina/análisis , Morfina/análisis , Reciclaje , Aguas Residuales/química , Contaminantes Químicos del Agua/análisis
11.
J Pharmacol Exp Ther ; 361(2): 280-291, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28336575

RESUMEN

Opioid-induced constipation (OIC) is a common side effect of opioid pharmacotherapy for the management of pain because opioid agonists bind to µ-opioid receptors in the enteric nervous system (ENS). Naloxegol, a polyethylene glycol derivative of naloxol, which is a derivative of naloxone and a peripherally acting µ-opioid receptor antagonist, targets the physiologic mechanisms that cause OIC. Pharmacologic measures of opioid activity and pharmacokinetic measures of central nervous system (CNS) penetration were employed to characterize the mechanism of action of naloxegol. At the human µ-opioid receptor in vitro, naloxegol was a potent inhibitor of binding (Ki = 7.42 nM) and a neutral competitive antagonist (pA2 - 7.95); agonist effects were <10% up to 30 µM and identical to those of naloxone. The oral doses achieving 50% of the maximal effect in the rat for antagonism of morphine-induced inhibition of gastrointestinal transit and morphine-induced antinociception in the hot plate assay were 23.1 and 55.4 mg/kg for naloxegol and 0.69 and 1.14 mg/kg by for naloxone, respectively. In the human colon adenocarcinoma cell transport assay, naloxegol was a substrate for the P-glycoprotein transporter, with low apparent permeability in the apical to basolateral direction, and penetrated the CNS 15-fold slower than naloxone in a rat brain perfusion model. Naloxegol-derived radioactivity was poorly distributed throughout the rat CNS and was eliminated from most tissues within 24 hours. These findings corroborate phase 3 clinical studies demonstrating that naloxegol relieves OIC-associated symptoms in patients with chronic noncancer pain by antagonizing the µ-opioid receptor in the ENS while preserving CNS-mediated analgesia.


Asunto(s)
Estreñimiento/tratamiento farmacológico , Morfinanos/farmacología , Alcaloides Opiáceos/toxicidad , Polietilenglicoles/farmacología , Receptores Opioides mu/antagonistas & inhibidores , Analgésicos Opioides/farmacología , Analgésicos Opioides/toxicidad , Animales , Encéfalo/metabolismo , Células CACO-2 , Permeabilidad de la Membrana Celular , Estreñimiento/inducido químicamente , Femenino , Tránsito Gastrointestinal/efectos de los fármacos , Células HEK293 , Humanos , Masculino , Morfinanos/farmacocinética , Morfina/farmacología , Morfina/toxicidad , Alcaloides Opiáceos/farmacología , Polietilenglicoles/farmacocinética , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/agonistas , Distribución Tisular
12.
Pak J Pharm Sci ; 30(6): 2067-2074, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29175775

RESUMEN

Opioid addiction is associated with oxidative cell injury in neuronal cells. In this study, Bacopa monnieri (L.), a reputed nootropic plant, was evaluated against morphine-induced histopathological changes in the cerebellum of rats. B. monnieri methanolic extract (mBME) (40 mg/kg, p.o) and ascorbic acid (50 mg/kg, i.p) were administered two hours before morphine (20 mg/kg, i.p) for 14 and 21 days. The in vitro antioxidant activity of mBME was determined by 2,2-diphenyl-1-picrylhydrazyl (DPPH) free-radical scavenging assay. Morphine produced vacuolization of basket and stellate cells and reduced the size of Purkinje cells in the cerebellum after 14 days. However, treatment for 21 days was associated with severe shrinkage of Purkinje cells with loss of their characteristic flask-shaped appearance as well as degeneration of basket, stellate and granule cells. Pretreatment with mBME and ascorbic acid for 14 and 21 days attenuated the morphine-induced histopathological changes in the cerebellum. The EC50 for the DPPH free-radical scavenging assay of mBME (39.06 µ/mL) as compared to ascorbic acid (30.25 µ/mL) and BHT (34.34 µ/mL) revealed that mBME strongly scavenged the free-radicals and thus possessed an efficient antioxidant propensity. These results concluded that B. monnieri having strong antioxidant activity exerted a protective effect against morphineinduced cerebellar toxicity.


Asunto(s)
Analgésicos Opioides/toxicidad , Bacopa , Cerebelo/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Morfina/toxicidad , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , Animales , Ácido Ascórbico/farmacología , Bacopa/química , Compuestos de Bifenilo/química , Hidroxitolueno Butilado/farmacología , Cerebelo/metabolismo , Cerebelo/patología , Citoprotección , Depuradores de Radicales Libres/aislamiento & purificación , Masculino , Fármacos Neuroprotectores/aislamiento & purificación , Estrés Oxidativo/efectos de los fármacos , Picratos/química , Extractos Vegetales/aislamiento & purificación , Células de Purkinje/efectos de los fármacos , Células de Purkinje/metabolismo , Células de Purkinje/patología , Ratas Sprague-Dawley
13.
Synapse ; 70(1): 15-23, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26418221

RESUMEN

The distribution pattern of calbindin (CB)-D28k-expressing neurons results to be altered in several brain regions of chronic morphine exposed adult mice. In this study, the influence of chronic maternal exposure to morphine on the distribution pattern of CB-D28k-expressing neurons in the brain of mouse offspring was investigated. Females of CD-1 mice were daily administered with saline or morphine for 7 days before mating, during the whole gestation period, and until 21 day post-partum. Their offspring were sacrificed on postnatal day 18, and the brains were examined by histology using cresyl violet and by immunohistochemistry using a rabbit polyclonal anti-CB-D28k antibody. Histology revealed no significant differences in the distribution pattern and the number of neurons between the offspring forebrain of the control group of mice and the two groups of mice treated with different doses of morphine. However, immunohistochemical analysis revealed that the number of CB-D28k-immunoreactive neurons remarkably decreased in the cingulate cortex, in the layers II-IV of the parietal cortex and in all regions of the hippocampus, while it increased in the layers V-VI of the parietal cortex and in the subicular region of the offspring brain of morphine treated mice. Overall, our findings demonstrate that maternal exposure to morphine alters the pattern of CB-D28k-expressing neuron pattern in specific regions of murine developing brain, in a layer- and dose-dependent way, thus suggesting that these alterations might represent a mechanism by which morphine modifies the functional aspects of developing brain.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Calbindina 1/metabolismo , Morfina/toxicidad , Narcóticos/toxicidad , Efectos Tardíos de la Exposición Prenatal , Animales , Benzoxazinas , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Relación Dosis-Respuesta a Droga , Femenino , Inmunohistoquímica , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Embarazo
14.
Behav Pharmacol ; 27(1): 37-43, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26274041

RESUMEN

Inhibitors of the endocannabinoid metabolic enzyme fatty acid amide hydrolase exert therapeutic effects, but might also be associated with some of the adverse effects of cannabis. However, at least one fatty acid amide hydrolase inhibitor, URB597, has beneficial effects without signs of abuse or dependence. Although previous investigations have evaluated URB597-morphine interactions, the effects of URB597 on morphine tolerance and cognition deficits have not been studied previously. Rats were rendered tolerant to or dependent on morphine by an injection of morphine (10 mg/kg, subcutaneous) twice daily, respectively, for 7 or 10 days. URB597 (1 mg/kg, intraperitoneal) was administered before morphine. The tail-flick and passive avoidance learning tests were used to evaluate tolerance and cognition. Chronic morphine injection led to significant tolerance to the antinociceptive effect on days 5 and 7. URB597 completely prevented the development of morphine tolerance. URB597 also enhanced memory acquisition in the passive avoidance learning test, and although morphine impaired memory, URB597 alleviated this effect. These data show that URB597 protects against tolerance and memory deficits in chronic usage of morphine and suggests URB597 as a promising candidate for the treatment of adverse effects of opioids.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Benzamidas/farmacología , Carbamatos/farmacología , Trastornos del Conocimiento/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Dependencia de Morfina/psicología , Psicotrópicos/farmacología , Amidohidrolasas/metabolismo , Analgésicos Opioides/farmacología , Analgésicos Opioides/toxicidad , Animales , Reacción de Prevención/efectos de los fármacos , Trastornos del Conocimiento/inducido químicamente , Tolerancia a Medicamentos , Masculino , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/tratamiento farmacológico , Morfina/farmacología , Morfina/toxicidad , Dependencia de Morfina/tratamiento farmacológico , Nocicepción/efectos de los fármacos , Distribución Aleatoria , Ratas Wistar
15.
Mol Cell Neurosci ; 65: 11-20, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25616162

RESUMEN

Infection of the CNS with HIV-1 occurs rapidly after primary peripheral infection. HIV-1 can induce a wide range of neurological deficits, collectively known as HIV-1-associated neurocognitive disorders. Our previous work has shown that the selected neurotoxic effects induced by individual viral proteins, Tat and gp120, and by HIV(+) supernatant are enhanced by co-exposure to morphine. This mimics co-morbid neurological effects observed in opiate-abusing HIV(+) patients. Although there is a correlation between opiate drug abuse and progression of HIV-1-associated neurocognitive disorders, the mechanisms underlying interactions between HIV-1 and opiates remain obscure. Previous studies have shown that HIV-1 induces neurotoxic effects through abnormal activation of GSK3ß. Interestingly, expression of GSK3ß has shown to be elevated in brains of young opiate abusers indicating that GSK3ß is also linked to neuropathology seen with opiate-abusing patients. Thus, we hypothesize that GSK3ß activation is a point of convergence for HIV- and opiate-mediated interactive neurotoxic effects. Neuronal cultures were treated with supernatant from HIV-1SF162-infected THP-1 cells, in the presence or absence of morphine and GSK3ß inhibitors. Our results show that GSK3ß inhibitors, including valproate and small molecule inhibitors, significantly reduce HIV-1-mediated neurotoxic outcomes, and also negate interactions with morphine that result in cell death, suggesting that GSK3ß-activation is an important point of convergence and a potential therapeutic target for HIV- and opiate-mediated neurocognitive deficits.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , VIH-1/patogenicidad , Morfina/toxicidad , Neuronas/efectos de los fármacos , Animales , Línea Celular Tumoral , Células Cultivadas , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Ratones , Neuronas/virología , Ácido Valproico/farmacología
16.
Eur J Anaesthesiol ; 33(11): 832-839, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27496317

RESUMEN

BACKGROUND: Morphine and other opioids are routinely used systemically and as wound infusions in the postoperative period. Their effect on wound and fracture healing remains unclear. OBJECTIVE: The primary outcome was to assess the potential cytotoxicity of clinically relevant concentrations of morphine on human fibroblasts. DESIGN: Laboratory in-vitro study. SETTING: Institute of Physiology, Zurich Center for Integrative Human Physiology, University of Zurich. MATERIALS: Monolayers of human fibroblasts. INTERVENTION(S): Exposure of human fibroblast monolayers to several concentrations of morphine, for different periods of time, with and without an artificially induced inflammatory process. MAIN OUTCOME MEASURES: Cell count, cell viability, cell proliferation and apoptosis. RESULTS: A concentration, time and exposure-dependent cytotoxic effect of morphine-mediated apoptosis was observed. Simulated inflammatory conditions seemed to lessen toxic effects. CONCLUSION: Cytotoxic effects of morphine are exposure, time and concentration dependent. Simulating aspects of inflammatory conditions seems to increase resistance to morphine cytotoxicity especially in the presence of higher concentration and longer exposure times.


Asunto(s)
Analgésicos Opioides/toxicidad , Proliferación Celular/efectos de los fármacos , Citotoxinas/toxicidad , Fibroblastos/efectos de los fármacos , Morfina/toxicidad , Recuento de Células/métodos , Proliferación Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Fibroblastos/fisiología , Humanos
17.
Drug Chem Toxicol ; 39(4): 418-23, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26821693

RESUMEN

Prolonged exposure to opiates induces a constellation of neuroadaptations, especially in the mesolimbic dopamine system (MLDS), which leads to alteration in the function of motivational circuitry. The neural cell adhesion molecule (NCAM) mediates cell-cell interactions and plays an important role in processes associated with neural plasticity. Moreover, it has been shown that NCAM were related to risk of alcoholism in human populations. Here, coimmunoprecipitation and western blotting were used to investigate whether morphine treatment induced alteration of the expression of NCAM or its signaling level in MLDS. The rats receiving escalating dose of morphine treatment were divided into three groups: morphine 1d, 3d and 5d group, which were injected subcutaneously with morphine hydrochloride for 1 day, 3 days and 5 days, respectively. Twelve hours after the last injection, animals were sacrificed and the tissues of ventral tegmental area (VTA), prefrontal cortex (PFC) and nucleus accumbens (NAc) were punched out to examine the expression of NCAM or its signaling level. The results showed that morphine treatment had no significant effect on the expression of NCAM, but downregulated the phosphorylation of NCAM-associated focal adhesion kinase (FAK) in the VTA and PFC of rats. In the NAc of rats, however, the expression of NCAM and its signaling were not altered significantly by morphine treatment. These results indicated that the downregulation of NCAM signaling in the VTA and PFC might be involved in the formation of morphine addiction.


Asunto(s)
Morfina/toxicidad , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Núcleo Accumbens/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Transducción de Señal , Área Tegmental Ventral/efectos de los fármacos , Animales , Western Blotting , Relación Dosis-Respuesta a Droga , Inmunoprecipitación , Inyecciones Subcutáneas , Masculino , Moléculas de Adhesión de Célula Nerviosa/genética , Núcleo Accumbens/metabolismo , Fosforilación , Corteza Prefrontal/metabolismo , Ratas Sprague-Dawley , Área Tegmental Ventral/metabolismo
18.
Environ Toxicol ; 31(1): 58-67, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25044278

RESUMEN

Opioids are considered as emerging contaminants in aquatic ecosystems, mainly due to their large illicit consume worldwide. Morphine (MOR) is the main opiate and it was commonly found at measurable concentrations in freshwaters. Even though its occurrence is well documented, just limited information is available regarding its hazard to nontarget organisms. The aim of this study was of the evaluation of sublethal effects induced by MOR to the freshwater bivalve Dreissena polymorpha. We exposed mussels to two MOR concentrations (0.05 µg/L and 0.5 µg/L) for 14 days and we investigated the sublethal effects by a suite of biomarkers. The Neutral Red Retention Assay (NRRA) was used as a test of cytotoxicity, while the oxidative stress was evaluated by the activity of antioxidant and detoxifying enzymes, namely catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx) and glutathione-S-transferase (GST), and by measuring the levels of lipid peroxidation (LPO) and protein carbonylation (PCC). The genetic damage was assessed by the Single Cell Gel Electrophoresis (SCGE) assay, the DNA diffusion assay and the micronucleus test (MN test). Finally, the filtration rate of D. polymorpha was evaluated in order to investigate possible physiological effects. Both tested concentrations reduced the lysosome membrane stability of bivalves, but only the highest MOR concentration induced significant changes in the activity of antioxidant enzymes (SOD, CAT, and GPx) and increase in lipid peroxidation levels. Slight increase in primary DNA fragmentation was noticed, while no fixed genetic damage and alterations of the filtering rate were found.


Asunto(s)
Dreissena/efectos de los fármacos , Morfina/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Antioxidantes/metabolismo , Biomarcadores/metabolismo , Catalasa/metabolismo , Ensayo Cometa , Fragmentación del ADN/efectos de los fármacos , Dreissena/enzimología , Dreissena/metabolismo , Glutatión Peroxidasa/metabolismo , Glutatión Transferasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Modelos Biológicos , Rojo Neutro/química , Estrés Oxidativo/efectos de los fármacos , Carbonilación Proteica/efectos de los fármacos , Superóxido Dismutasa/metabolismo
19.
J Neurosci ; 34(27): 9076-87, 2014 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-24990928

RESUMEN

Opioids are commonly used for pain relief, but their strong rewarding effects drive opioid misuse and abuse. How pain affects the liability of opioid abuse is unknown at present. In this study, we identified an epigenetic regulating cascade activated by both pain and the opioid morphine. Both persistent pain and repeated morphine upregulated the transcriptional regulator MeCP2 in mouse central nucleus of the amygdala (CeA). Chromatin immunoprecipitation analysis revealed that MeCP2 bound to and repressed the transcriptional repressor histone dimethyltransferase G9a, reducing G9a-catalyzed repressive mark H3K9me2 in CeA. Repression of G9a activity increased expression of brain-derived neurotrophic factor (BDNF). Behaviorally, persistent inflammatory pain increased the sensitivity to acquiring morphine-induced, reward-related behavior of conditioned place preference in mice. Local viral vector-mediated MeCP2 overexpression, Cre-induced G9a knockdown, and CeA application of BDNF mimicked, whereas MeCP2 knockdown inhibited, the pain effect. These results suggest that MeCP2 directly represses G9a as a shared mechanism in central amygdala for regulation of emotional responses to pain and opioid reward, and for their behavioral interaction.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Dolor Crónico/fisiopatología , N-Metiltransferasa de Histona-Lisina/fisiología , Proteína 2 de Unión a Metil-CpG/fisiología , Morfina/farmacología , Narcóticos/farmacología , Recompensa , Animales , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/farmacología , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/etiología , Condicionamiento Clásico/fisiología , Cruzamientos Genéticos , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Emociones/fisiología , Epigénesis Genética , Conducta Exploratoria/efectos de los fármacos , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Vectores Genéticos , Masculino , Ratones , Ratones Endogámicos C57BL , Morfina/uso terapéutico , Morfina/toxicidad , Dependencia de Morfina/etiología , Dependencia de Morfina/psicología , Narcóticos/uso terapéutico , Narcóticos/toxicidad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Refuerzo en Psicología , Transgenes
20.
J Neurovirol ; 21(5): 592-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26037114

RESUMEN

The abuse of opiates such as morphine in synergy with HIV infection accelerates neurocognitive impairments and neuropathology in the CNS of HIV-infected subjects, collectively referred to as HAND. To identify potential pathogenic markers associated with HIV and morphine in perturbing the synaptic architecture, we performed quantitative mass spectrometry proteomics on purified synaptosomes isolated from the caudate of two groups of rhesus macaques chronically infected with SIV differing by one regimen-morphine treatment. The upregulation of heat shock 70-kDa protein 5 in the SIV + morphine group points to increased cellular stress during SIV/morphine interaction thus leading to CNS dysfunction.


Asunto(s)
Analgésicos Opioides/toxicidad , Proteínas de Choque Térmico/biosíntesis , Morfina/toxicidad , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Sinapsis/metabolismo , Animales , Western Blotting , Línea Celular , Chaperón BiP del Retículo Endoplásmico , Humanos , Macaca mulatta , Neuronas/efectos de los fármacos , Proteómica/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Virus de la Inmunodeficiencia de los Simios , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Sinapsis/efectos de los fármacos , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda