Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 7(3)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34914639

RESUMEN

Ca2+/calmodulin-stimulated group I adenylyl cyclase (AC) isoforms AC1 and AC8 have been involved in nociceptive processing and morphine responses. However, whether AC3, another member of group I ACs, is involved in nociceptive transmission and regulates opioid receptor signaling remains elusive. Here, we report that conditional KO of AC3 (AC3 CKO) in L3 and L4 DRGs robustly facilitated the mouse nociceptive responses, decreased voltage-gated potassium (Kv) channel currents, and increased neuronal excitability. Furthermore, we report AC3 CKO eliminated the analgesic effect of κ-opioid receptor (KOR) agonist and its inhibition on Kv channel by classical Gαi/o signaling or nonclassical direct interaction of KOR and AC3 proteins. Interestingly, significantly upregulated AC1 level and cAMP concentration were detected in AC3-deficient DRGs. Inhibition of AC1 completely reversed cAMP upregulation, neuronal excitability enhancement, and nociceptive behavioral hypersensitivity in AC3-CKO mice. Our findings suggest a crucial role of peripheral AC3 in nociceptive modulation and KOR opioid analgesia.


Asunto(s)
Técnicas de Ablación/métodos , Adenilil Ciclasas/genética , Analgesia/métodos , Regulación de la Expresión Génica , Hiperalgesia/genética , Receptores Opioides/genética , Adenilil Ciclasas/biosíntesis , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Hiperalgesia/metabolismo , Hiperalgesia/terapia , Ratones , ARN/genética , ARN/metabolismo , Receptores Opioides/biosíntesis , Receptor de Nociceptina
2.
Exp Neurol ; 337: 113572, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33340498

RESUMEN

Neuropathic pain is the most common clinical disorder destroying the quality of patient life and leading to a marked economic and social burden. Opioids are still last option for pharmacological treatment of this disorder, but their antinociceptive effects are limited in part due to the downregulation of opioid receptors in the primary afferent neurons after peripheral nerve trauma. How this downregulation occurs is not completely understood, but recent studies have demonstrated that peripheral nerve trauma drives the alterations in epigenetic modifications (including DNA methylation, histone methylation and mciroRNAs), expression of transcription factors, post-transcriptional modifications (e.g., RNA methylation) and protein translation initiation in the neurons of nerve trauma-related dorsal root ganglion (DRG) and that these alternations may be associated with nerve trauma-caused downregulation of DRG opioid receptors. This review presents how opioid receptors are downregulated in the DRG after peripheral nerve trauma, specifically focusing on distinct molecular mechanisms underlying transcriptional and translational processes. This review also discusses how this downregulation contributes to the induction and maintenance of neuropathic pain. A deeper understanding of these molecular mechanisms likely provides a novel avenue for prevention and/or treatment of neuropathic pain.


Asunto(s)
Neuronas Aferentes/metabolismo , Traumatismos de los Nervios Periféricos/genética , Receptores Opioides/genética , Animales , Regulación hacia Abajo/genética , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Humanos , Traumatismos de los Nervios Periféricos/metabolismo , Traumatismos de los Nervios Periféricos/patología , Receptores Opioides/biosíntesis
3.
Int Immunopharmacol ; 86: 106718, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32585612

RESUMEN

The metastasis of cervical cancer has always been a clinical challenge. We investigated the effects of low-dose naltrexone (LDN) on the epithelial mesenchymal transition of cervical cancer cells in vitro as well as its influence on macrophage polarization and associated cytokines in vivo. The results suggested that LDN supressed the proliferation, migration and invasion abilities and promote their apoptosis in Hela cells, whereas the opioid growth factor receptor (OGFr) silenced significantly reversed these effects in vitro. Knockdown the expression of OGFr, the inhibitory of LDN on EMT was weakened. LDN could inhibit cervical cancer progression in nude mice. In additon, LDN indirectly reduced the number of tumor-associated macrophages (TAMs), mainly M2 macrophages, and decreased expression of anti-inflammatory factor IL-10 in the serum of nude mice. These findings demonstrate that LDN could be a potential treatment for cervical cancer.


Asunto(s)
Antineoplásicos/farmacología , Naltrexona/farmacología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/sangre , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Activación de Macrófagos/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Desnudos , Naltrexona/uso terapéutico , Receptores Opioides/biosíntesis , Receptores Opioides/efectos de los fármacos , Receptores Opioides/genética , Factores de Transcripción de la Familia Snail/metabolismo , Macrófagos Asociados a Tumores/efectos de los fármacos , Neoplasias del Cuello Uterino/patología , Vimentina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Artículo en Inglés | MEDLINE | ID: mdl-31904442

RESUMEN

Potent opioid-based therapies are often unsuccessful in promoting satisfactory analgesia in neuropathic pain. Moreover, the side effects associated with opioid therapy are still manifested in neuropathy-like diseases, including tolerance, abuse, addiction and hyperalgesia, although the mechanisms underlying these effects remain unclear. Studies in the spinal cord and periphery indicate that neuropathy alters the expression of mu-[MOP], delta-[DOP] or kappa-[KOP] opioid receptors, interfering with their activity. However, there is no consensus as to the supraspinal opioidergic modulation provoked by neuropathy, the structures where the sensory and affective-related pain components are processed. In this study we explored the effect of chronic constriction of the sciatic nerve (CCI) over 7 and 30 days (CCI-7d and CCI-30d, respectively) on MOP, DOP and KOP mRNAs expression, using in situ hybridization, and the efficacy of G-protein stimulation by DAMGO, DPDPE and U-69593 (MOP, DOP and KOP specific agonists, respectively), using [35S]GTPγS binding, within opioid-sensitive brain structures. After CCI-7d, CCI-30d or both, opioid receptor mRNAs expression was altered throughout the brain: MOP - in the paracentral/centrolateral thalamic nuclei, ventral posteromedial thalamic nuclei, superior olivary complex, parabrachial nucleus [PB] and posterodorsal tegmental nucleus; DOP - in the somatosensory cortex [SSC], ventral tegmental area, caudate putamen [CPu], nucleus accumbens [NAcc], raphe magnus [RMg] and PB; and KOP - in the locus coeruleus. Agonist-stimulated [35S]GTPγS binding was altered following CCI: MOP - CPu and RMg; DOP - prefrontal cortex [PFC], SSC, RMg and NAcc; and KOP - PFC and SSC. Thus, this study shows that several opioidergic circuits in the brain are recruited and modified following neuropathy.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Encéfalo/metabolismo , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides/biosíntesis , Analgésicos Opioides/farmacología , Animales , Encéfalo/efectos de los fármacos , Expresión Génica , Masculino , Ratas , Ratas Sprague-Dawley , Receptores Opioides/genética , Neuropatía Ciática/tratamiento farmacológico , Neuropatía Ciática/metabolismo
5.
Front Immunol ; 11: 615327, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33408720

RESUMEN

Clinical management of neuropathic pain is unsatisfactory, mainly due to its resistance to the effects of available analgesics, including opioids. Converging evidence indicates the functional interactions between chemokine and opioid receptors and their influence on nociceptive processes. Recent studies highlight that the CC chemokine receptors type 2 (CCR2) and 5 (CCR5) seem to be of particular interest. Therefore, in this study, we investigated the effects of the dual CCR2/CCR5 antagonist, cenicriviroc, on pain-related behaviors, neuroimmune processes, and the efficacy of opioids in rats after chronic constriction injury (CCI) of the sciatic nerve. To define the mechanisms of action of cenicriviroc, we studied changes in the activation/influx of glial and immune cells and, simultaneously, the expression level of CCR2, CCR5, and important pronociceptive cytokines in the spinal cord and dorsal root ganglia (DRG). We demonstrated that repeated intrathecal injections of cenicriviroc, in a dose-dependent manner, alleviated hypersensitivity to mechanical and thermal stimuli in rats after sciatic nerve injury, as measured by von Frey and cold plate tests. Behavioral effects were associated with the beneficial impact of cenicriviroc on the activation/influx level of C1q/IBA-1-positive cells in the spinal cord and/or DRG and GFAP-positive cells in DRG. In parallel, administration of cenicriviroc decreased the expression of CCR2 in the spinal cord and CCR5 in DRG. Concomitantly, we observed that the level of important pronociceptive factors (e.g., IL-1beta, IL-6, IL-18, and CCL3) were increased in the lumbar spinal cord and/or DRG 7 days following injury, and cenicriviroc was able to prevent these changes. Additionally, repeated administration of this dual CCR2/CCR5 antagonist enhanced the analgesic effects of morphine and buprenorphine in neuropathic rats, which can be associated with the ability of cenicriviroc to prevent nerve injury-induced downregulation of all opioid receptors at the DRG level. Overall, our results suggest that pharmacological modulation based on the simultaneous blockade of CCR2 and CCR5 may serve as an innovative strategy for the treatment of neuropathic pain, as well as in combination with opioids.


Asunto(s)
Analgésicos/uso terapéutico , Antagonistas de los Receptores CCR5/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Imidazoles/uso terapéutico , Neuralgia/tratamiento farmacológico , Receptores CCR2/antagonistas & inhibidores , Neuropatía Ciática/complicaciones , Sulfóxidos/uso terapéutico , Analgésicos/administración & dosificación , Analgésicos/farmacología , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Animales , Buprenorfina/farmacología , Buprenorfina/uso terapéutico , Antagonistas de los Receptores CCR5/administración & dosificación , Antagonistas de los Receptores CCR5/farmacología , Citocinas/biosíntesis , Citocinas/genética , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Imidazoles/administración & dosificación , Imidazoles/farmacología , Inyecciones Espinales , Masculino , Morfina/farmacología , Morfina/uso terapéutico , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuralgia/etiología , Neuralgia/fisiopatología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar , Receptores CCR2/biosíntesis , Receptores CCR2/genética , Receptores CCR5/biosíntesis , Receptores CCR5/genética , Receptores Opioides/biosíntesis , Receptores Opioides/genética , Médula Espinal/metabolismo , Sulfóxidos/administración & dosificación , Sulfóxidos/farmacología
6.
J Neuroendocrinol ; 31(12): e12810, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31715027

RESUMEN

The onset of puberty is the result of an increase in secretion of hypothalamic gonadotrophin-releasing hormone (GnRH). This action is a result of not only the development of stimulatory inputs to its release, but also the gradual decrease in inhibitory inputs that restrain release of the peptide prior to pubertal onset. Dynorphin (DYN) is one of the inhibitory inputs produced in the medial basal hypothalamus (MBH); however, little is known about what substance(s) control its prepubertal synthesis and release. Because neurokinin B (NKB) increases in the hypothalamus as puberty approaches, we considered it a candidate for such a role. An initial study investigated the acute effects of an NKB agonist, senktide, on the secretion of DYN from MBH tissues incubated in vitro. In other experiments, central injections of senktide were administered to animals for 4 days then MBHs were collected for assessment of DYN synthesis or for the in vitro secretion of both DYN and GnRH. Because insulin-like growth factor (IGF)-1 has been shown to play an important role at puberty, additional animals received central injections of this peptide for 4 days to assess NKB and DYN synthesis or the in vitro secretion of NKB. The results obtained show that senktide administration up-regulates the NKB receptor protein, at the same time as suppressing the DYN and its receptor. Senktide consistently suppressed DYN and elevated GnRH secretion in the same tissue incubates from both the acute and chronic studies. IGF-1 administration caused an increase in NKB protein, at the same time as decreasing DYN protein. Furthermore, the central administration of IGF-1 caused an increase in NKB release, an action blocked by the IGF-1 receptor blocker, JB-1. These results indicate that the IGF-1/NKB pathway contributes to suppressing the DYN inhibitory tone on prepubertal GnRH secretion and thus facilitates the puberty-related increase in the release of GnRH to accelerate the onset of puberty.


Asunto(s)
Dinorfinas/metabolismo , Hipotálamo Medio/metabolismo , Animales , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo Medio/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/farmacología , Microinyecciones , Neuroquinina B/metabolismo , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Ratas , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptores de Neuroquinina-3/biosíntesis , Receptores Opioides/biosíntesis , Maduración Sexual , Sustancia P/análogos & derivados , Sustancia P/farmacología , Regulación hacia Arriba
7.
Neuroscience ; 363: 50-61, 2017 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-28673713

RESUMEN

Widespread pain and anxiety are commonly reported in cancer patients. We hypothesize that cancer is accompanied by attenuation of endogenous opioid-mediated inhibition, which subsequently causes widespread pain and anxiety. To test this hypothesis we used a mouse model of oral squamous cell carcinoma (SCC) in the tongue. We found that mice with tongue SCC exhibited widespread nociceptive behaviors in addition to behaviors associated with local nociception that we reported previously. Tongue SCC mice exhibited a pattern of reduced opioid receptor expression in the spinal cord; intrathecal administration of respective mu (MOR), delta (DOR), and kappa (KOR) opioid receptor agonists reduced widespread nociception in mice, except for the fail flick assay following administration of the MOR agonist. We infer from these findings that opioid receptors contribute to widespread nociception in oral cancer mice. Despite significant nociception, mice with tongue SCC did not differ from sham mice in anxiety-like behaviors as measured by the open field assay and elevated maze. No significant differences in c-Fos staining were found in anxiety-associated brain regions in cancer relative to control mice. No correlation was found between nociceptive and anxiety-like behaviors. Moreover, opioid receptor agonists did not yield a statistically significant effect on behaviors measured in the open field and elevated maze in cancer mice. Lastly, we used an acute cancer pain model (injection of cancer supernatant into the mouse tongue) to test whether adaptation to chronic pain is responsible for the absence of greater anxiety-like behavior in cancer mice. No changes in anxiety-like behavior were observed in mice with acute cancer pain.


Asunto(s)
Ansiedad/metabolismo , Dolor en Cáncer/metabolismo , Carcinoma de Células Escamosas/complicaciones , Neoplasias de Cabeza y Cuello/complicaciones , Dolor Nociceptivo/metabolismo , Receptores Opioides/biosíntesis , Neoplasias de la Lengua/complicaciones , Animales , Ansiedad/etiología , Dolor en Cáncer/etiología , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Femenino , Neoplasias de Cabeza y Cuello/metabolismo , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Médula Espinal/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello , Neoplasias de la Lengua/metabolismo
8.
Alcohol Alcohol ; 52(2): 172-179, 2017 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-28043969

RESUMEN

AIMS: Nuclear factor kappa light chain enhancer of activated B cells (NFkB) is a ubiquitous transcription factor well known for its role in the innate immune response. As such, NFkB is a transcriptional activator of inflammatory mediators such as cytokines. It has recently been demonstrated that alcohol and other drugs of abuse can induce NFkB activity and cytokine expression in the brain. A number of reviews have been published highlighting this effect of alcohol, and have linked increased NFkB function to neuroimmune-stimulated toxicity. However, in this review we focus on the potentially non-immune functions of NFkB as possible links between NFkB and addiction. METHODS: An extensive review of the literature via Pubmed searches was used to assess the current state of the field. RESULTS: NFkB can induce the expression of a diverse set of gene targets besides inflammatory mediators, some of which are involved in addictive processes, such as opioid receptors and neuropeptides. NFkB mediates complex behaviors including learning and memory, stress responses, anhedonia and drug reward, processes that may lie outside the role of NFkB in the classic neuroimmune response. CONCLUSIONS: Future studies should focus on these non-immune functions of NFkB signaling and their association with addiction-related processes.


Asunto(s)
Regulación de la Expresión Génica , FN-kappa B/fisiología , Trastornos Relacionados con Sustancias/genética , Trastornos Relacionados con Sustancias/fisiopatología , Anhedonia/fisiología , Conducta Adictiva/genética , Conducta Adictiva/fisiopatología , Humanos , Inflamación/genética , Inflamación/inmunología , Aprendizaje/fisiología , Memoria/fisiología , FN-kappa B/inmunología , Neuropéptidos/biosíntesis , Receptores Opioides/biosíntesis
9.
Biopolymers ; 106(4): 460-9, 2016 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-27271345

RESUMEN

We previously showed that an antagonist-based peptide ligand, H-Cys(Npys)-Arg-Tyr-Tyr-Arg- Ile-Lys-NH2 , captures the free thiol groups in the ligand-binding site of the nociceptin receptor ORL1. However, the exact receptor sites of this thiol-disulfide exchange reaction have not been uncovered, although such identification would help to clarify the ligand recognition site. Since the Cys→Ala substitution prevents the reaction, we performed the so-called Ala scanning for all the Cys residues in the transmembrane (TM) domains of the ORL1 receptor. Seven different mutant receptors were soundly expressed in the COS-7 cells and examined for their specific affinity labeling by a competitive binding assay using nociceptin and [(3) H]nociceptin. The results of in vitro Ala scanning analyses revealed that the labeled residues were Cys59 in TM1, Cys215 and Cys231 in TM5, and Cys310 in TM7. The present study has provided a novel method of Cys(Npys)-affinity labeling for identification of the ligand-binding sites in the ORL1 receptor. © 2016 Wiley Periodicals, Inc. Biopolymers (Pept Sci) 106: 460-469, 2016.


Asunto(s)
Péptidos/química , Receptores Opioides , Coloración y Etiquetado/métodos , Sustitución de Aminoácidos , Animales , Sitios de Unión , Células COS , Chlorocebus aethiops , Humanos , Ligandos , Mutación Missense , Receptores Opioides/biosíntesis , Receptores Opioides/química , Receptores Opioides/genética , Receptor de Nociceptina
10.
Brain Res ; 1632: 34-41, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26657743

RESUMEN

Growing evidence indicates that targeting nociceptin receptor (NOP) signaling may have therapeutic efficacy in treating alcohol and opioid addiction. However, little is known about the therapeutic value of selective NOP agonists for the treatment of cocaine dependence. Recently, we identified a highly selective, brain-penetrant NOP small molecule agonist (SR-8993), and using this compound, we previously showed that nociceptin receptor activation attenuated consolidation of fear-related memories. Here, we sought to determine whether SR-8993 also affects the rewarding properties of cocaine. Using a conditioned place preference (CPP) procedure, we show that SR-8993 (3 or 10 mg/kg) failed to disrupt acquisition or expression of cocaine CPP (7.5 or 15 mg/kg) in C57BL/6 mice. Additionally, SR-8993 did not affect rate of extinction or reinstatement (yohimbine- and cocaine-induced) of cocaine CPP. These studies indicate that selective activation of NOP may not be sufficient in reducing behavioral responses to cocaine.


Asunto(s)
Conducta Adictiva/metabolismo , Cocaína/administración & dosificación , Condicionamiento Psicológico/fisiología , Extinción Psicológica/fisiología , Receptores Opioides/agonistas , Receptores Opioides/biosíntesis , Animales , Conducta Adictiva/tratamiento farmacológico , Condicionamiento Psicológico/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Yohimbina/farmacología , Receptor de Nociceptina
11.
PLoS One ; 10(4): e0123407, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25853862

RESUMEN

The dormancy of tumor cells is a major problem in chemotherapy, since it limits the therapeutic efficacy of anti-tumor drugs that only target dividing cells. One potential way to overcome chemo-resistance is to "wake up" these dormant cells. Here we show that the opioid antagonist methylnaltrexone (MNTX) enhances the effect of docetaxel (Doc) by blocking a cell growth-suppressive pathway. We found that PENK, which encodes opioid growth factor (OGF) and suppresses cell growth, is predominantly expressed in diffuse-type gastric cancers (GCs). The blockade of OGF signaling by MNTX releases cells from their arrest and boosts the effect of Doc. In comparison with the use of Doc alone, the combined use of Doc and MNTX significantly prolongs survival, alleviates abdominal pain, and diminishes Doc-resistant spheroids on the peritoneal membrane in model mice. These results suggest that blockade of the pathways that suppress cell growth may enhance the effects of anti-tumor drugs.


Asunto(s)
Naltrexona/análogos & derivados , Receptores Opioides/biosíntesis , Neoplasias Gástricas/tratamiento farmacológico , Taxoides/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Docetaxel , Sinergismo Farmacológico , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/genética , Humanos , Ratones , Naltrexona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Compuestos de Amonio Cuaternario/administración & dosificación , Receptores Opioides/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología
12.
J Neuroendocrinol ; 26(5): 329-40, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24617903

RESUMEN

Orphanin FQ/nociceptin (OFQ/N) inhibits the activity of pro-opiomelanocortin (POMC) neurones located in the hypothalamic arcuate nucleus (ARH) that regulate female sexual behaviour and energy balance. We tested the hypothesis that gonadal steroids differentially modulate the ability of OFQ/N to inhibit these cells via presynaptic inhibition of transmitter release and postsynaptic activation of G protein-gated, inwardly-rectifying K(+) (GIRK)-1 channels. Whole-cell patch clamp recordings were performed in hypothalamic slices prepared from ovariectomised rats. OFQ/N (1 µm) decreased the frequency of miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs), and also caused a robust outward current in the presence of tetrodotoxin, in ARH neurones from vehicle-treated animals. A priming dose of oestradiol benzoate (EB; 2 µg) increased basal mEPSC frequency, markedly diminished both the OFQ/N-induced decrease in mEPSC frequency and the activation of GIRK-1 currents, and potentiated the OFQ/N-induced decrease in mIPSC frequency. Steroid treatment regimens that facilitate sexual receptivity reinstate the basal mEPSC frequency, the OFQ/N-induced decrease in mEPSC frequency and the activation of GIRK-1 currents to levels observed in vehicle-treated controls, and largely abolish the ability of OFQ/N to decrease mIPSC frequency. These effects were observed in an appreciable population of identified POMC neurones, almost one-half of which projected to the medial preoptic nucleus. Taken together, these data reveal that gonadal steroids influence the pleiotropic actions of OFQ/N on ARH neurones, including POMC neurones, in a disparate manner. These temporal changes in OFQ/N responsiveness further implicate this neuropeptide system as a critical mediator of the gonadal steroid regulation of reproductive behaviour.


Asunto(s)
Hormonas Esteroides Gonadales/farmacología , Péptidos Opioides/efectos de los fármacos , Conducta Sexual Animal/efectos de los fármacos , Animales , Estradiol/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Ovariectomía , Técnicas de Placa-Clamp , Canales de Potasio/efectos de los fármacos , Proopiomelanocortina/fisiología , Progesterona/farmacología , Ratas , Ratas Long-Evans , Receptores Opioides/biosíntesis , Receptores Opioides/genética , Receptor de Nociceptina , Nociceptina
13.
Artículo en Inglés | MEDLINE | ID: mdl-24184686

RESUMEN

Cocaine induces neurochemical changes of endogenous prodynorphin-kappa opioid receptor (pDYN-KOP) and pronociceptin/orphaninFQ-nociceptin receptor (pN/OFQ-NOP) systems. Both systems play an important role in rewarding mechanisms and addictive stimulus processing by modulating drug-induced dopaminergic activation in the mesocortico-limbic brain areas. They are also involved in regulating stress mechanisms related to addiction. The aim of this study was to investigate possible changes of gene expression of the dynorphinergic and nociceptinergic system components in the nucleus accumbens (NA) and in medial and lateral caudate putamen (mCPu and lCPu, respectively) of rats, following chronic subcutaneous infusion of cocaine. In addition, the epigenetic histone modifications H3K4me3 and H3K27me3 (an activating and a repressive marker, respectively) at the promoter level of the pDYN, KOP, pN/OFQ and NOP genes were investigated. Results showed that cocaine induced pDYN gene expression up-regulation in the NA and lCPu, and its down-regulation in the mCPu, whereas KOP mRNA levels were unchanged. Moreover, cocaine exposure decreased pN/OFQ gene expression in the NA and lCPu, while NOP mRNA levels appeared significantly increased in the NA and decreased in the lCPu. Specific changes of the H3K4me3 and H3K27me3 levels were found at pDYN, pN/OFQ, and NOP gene promoter, consistent with the observed gene expression alterations. The present findings contribute to better define the role of endogenous pDYN-KOP and pN/OFQ-NOP systems in neuroplasticity mechanisms following chronic cocaine treatment. The epigenetic histone modifications underlying the gene expression changes likely mediate the effects of cocaine on transcriptional regulation of specific gene promoters that result in long-lasting drug-induced plasticity.


Asunto(s)
Cocaína/farmacología , Cuerpo Estriado/efectos de los fármacos , Dinorfinas/genética , Epigénesis Genética/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Receptores Opioides kappa/genética , Receptores Opioides/genética , Animales , Cuerpo Estriado/metabolismo , Dinorfinas/biosíntesis , Encefalinas/biosíntesis , Encefalinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Masculino , Núcleo Accumbens/metabolismo , Péptidos Opioides/biosíntesis , Péptidos Opioides/genética , Precursores de Proteínas/biosíntesis , Precursores de Proteínas/genética , Ratas , Receptores Opioides/biosíntesis , Receptores Opioides kappa/biosíntesis , Receptor de Nociceptina , Nociceptina
14.
Oncotarget ; 4(5): 677-90, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23633472

RESUMEN

Cyclic AMP (cAMP) regulates a number of cellular processes and modulates cell death induction. cAMP levels are altered upon stimulation of specific G-protein-coupled receptors inhibiting or activating adenylyl cyclases. Opioid receptor stimulation can activate inhibitory Gi-proteins which in turn block adenylyl cyclase activity reducing cAMP. Opioids such as D,L-methadone induce cell death in leukemia cells. However, the mechanism how opioids trigger apoptosis and activate caspases in leukemia cells is not understood. In this study, we demonstrate that downregulation of cAMP induced by opioid receptor activation using the opioid D,L-methadone kills and sensitizes leukemia cells for doxorubicin treatment. Enhancing cAMP levels by blocking opioid-receptor signaling strongly reduced D,L-methadone-induced apoptosis, caspase activation and doxorubicin-sensitivity. Induction of cell death in leukemia cells by activation of opioid receptors using the opioid D,L-methadone depends on critical levels of opioid receptor expression on the cell surface. Doxorubicin increased opioid receptor expression in leukemia cells. In addition, the opioid D,L-methadone increased doxorubicin uptake and decreased doxorubicin efflux in leukemia cells, suggesting that the opioid D,L-methadone as well as doxorubicin mutually increase their cytotoxic potential. Furthermore, we found that opioid receptor activation using D,L-methadone alone or in addition to doxorubicin inhibits tumor growth significantly in vivo. These results demonstrate that opioid receptor activation via triggering the downregulation of cAMP induces apoptosis, activates caspases and sensitizes leukemia cells for doxorubicin treatment. Hence, opioid receptor activation seems to be a promising strategy to improve anticancer therapies.


Asunto(s)
Adenilil Ciclasas/metabolismo , Apoptosis , AMP Cíclico/metabolismo , Leucemia de Células B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Receptores Opioides/metabolismo , Inhibidores de Adenilato Ciclasa , Analgésicos Opioides/farmacología , Animales , Antibióticos Antineoplásicos/farmacología , Caspasas/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Doxorrubicina/sangre , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Humanos , Masculino , Metadona/sangre , Metadona/farmacología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Receptores Opioides/biosíntesis , Transducción de Señal/efectos de los fármacos
15.
Epilepsy Res ; 105(1-2): 13-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23337899

RESUMEN

The opioid/nociceptin receptors are involved in many neurological disorders such as Alzheimer's disease, Parkinson's disease and epilepsy. Kainic acid (KA) is an analog of the excitatory amino acid transmitter glutamate and the systemic administration of KA induces status epilepticus (SE) in rodents. In this study, we examined the alterations in the G-protein activity and the gene expression levels of mu, kappa, delta opioid and nociceptin receptors (MOPr, KOPr, DOPr and NOPr) as well as PNOC, the precursor polypeptide of nociceptin-OFQ (N/OFQ) in KA-induced seizures in the rat brain cortex. KA was used to create seizures with the dose of 10 mg/kg body weight i.p. Following the KA administration, the rats were observed for 3 h to assess seizure activity. Seizures occurred approximately 45 min after the KA injection. Only rats exhibiting full limbic seizures, forelimb clonus with rearing, were used in this study. All animals were decapitated 4 h after the administration of KA. Our [(35)S]GTPγS binding results showed that there was a significant difference in both the affinity and efficacy particularly one of NOPr stimulation following KA treatment. Slight, but significant increase was observed for MOPr. Moreover PNOC, NOPr and MOPr mRNA levels were increased by KA treatment but there were no significant changes in the levels of DOPr and KOPr mRNAs. These results show that the activities of opioid/nociceptin receptors can be modified by KA-treatment, and MOPr, PNOC and NOPr are the most responsive to KA-induced seizures in the rat brain cortex.


Asunto(s)
Corteza Cerebral/metabolismo , Proteínas de Unión al GTP/biosíntesis , Ácido Kaínico/toxicidad , ARN Mensajero/biosíntesis , Receptores Opioides/biosíntesis , Convulsiones/metabolismo , Animales , Corteza Cerebral/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Encefalina Ala(2)-MeFe(4)-Gli(5)/metabolismo , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Regulación de la Expresión Génica , Masculino , Péptidos Opioides/metabolismo , Péptidos Opioides/farmacología , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Receptor de Nociceptina
16.
Parasite Immunol ; 34(11): 536-46, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22889318

RESUMEN

Primary exposure of mice to the nematode Heligmosomoides polygyrus infection reduces inflammation in an experimental model of colitis. The aim of the present investigation was to evaluate whether the reduced inflammation provoked by H. polygyrus L4 larvae in BALB/c mice treated with dextran sulphate sodium is associated with changed expression of opioids in the small intestine and colon. Colitis was induced by 5% Dextran sulphate sodium (DSS) oral administration for 3 days before oral infection with 200 infective larvae (L3) H. polygyrus until the end of the experiment, 6 days post-infection. Clinical disease symptoms were monitored daily. The expressions of proopiomelanocortin POMC1, MOR1 (Oprm1) - opioid receptor and ß-endorphin were determined by RT-PCR, Western blot and immunoassay, respectively, in the colon and small intestine of mice. RT-PCR analysis of colon tissues showed up-regulation of the expression of POMC and MOR1 opioid-dependent genes in mice with DSS-induced colitis. H. polygyrus L4 larvae inhibited DSS-induced colitis symptoms that were correlated with increased IL-1ß, TNF-α, IL-6, myeloperoxidase (MPO) concentration, macrophages infiltration and MOR1, POMC and ß-endorphin increased expression in the small intestine and inhibition of those in the colon.


Asunto(s)
Colitis/prevención & control , Intestinos/fisiología , Nematospiroides dubius/inmunología , Proopiomelanocortina/biosíntesis , Receptores Opioides/biosíntesis , betaendorfina/biosíntesis , Animales , Western Blotting , Colitis/inducido químicamente , Colitis/patología , Citocinas/metabolismo , Sulfato de Dextran , Perfilación de la Expresión Génica , Larva/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Neutrófilos/inmunología , Peroxidasa/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Índice de Severidad de la Enfermedad
17.
Psychopharmacology (Berl) ; 224(2): 303-12, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22669129

RESUMEN

INTRODUCTION: Agonists at the mu opioid receptor (MOR) are widely recognized for their effects on reward and pain. Although prior studies have attributed some of these effects to MORs on GABA neurons in the ventral tegmental area (VTA), recent studies have identified a region of particularly strong MOR immunostaining residing caudal to the VTA, in a region denoted the rostromedial tegmental nucleus (RMTg). METHODS: Hence, we examined whether rats would self-administer small doses (50-250 pmol) of the selective MOR agonist endomorphin-1 (EM1) into the RMTg and adjacent sites. EM1 was chosen due to its short half-life, thus limiting drug spread, and due to its presence endogenously in brain neurons, including some afferents to the RMTg. RESULTS: The highest rates of EM1 self-administration occurred within 0.5 mm of the RMTg center, in a region roughly 0.8-1.6 mm caudal to the majority of VTA DA neurons. In contrast, self-administration rates were much lower in the adjacent VTA, interpeduncular nucleus, central linear nucleus, or median raphe nucleus. Furthermore, EM1 infusions into the RMTg, but not surrounding regions, produced conditioned place preference, while EM1 infusions into the RMTg but not anterior VTA markedly reduced formalin-induced pain behaviors. EM1 effects were mimicked by infusions of the GABA agonist muscimol into the same region, consistent with EM1 having inhibitory actions on its target neurons. CONCLUSION: These results implicate a novel brain region in modulating MOR influences on both appetitive and aversive behavior.


Asunto(s)
Analgésicos Opioides/farmacología , Oligopéptidos/farmacología , Receptores Opioides mu/agonistas , Tegmento Mesencefálico/metabolismo , Analgésicos Opioides/administración & dosificación , Animales , Dopamina/metabolismo , Relación Dosis-Respuesta a Droga , Agonistas de Receptores de GABA-A/farmacología , Masculino , Muscimol/farmacología , Oligopéptidos/administración & dosificación , Dolor/tratamiento farmacológico , Precursores de Proteínas/biosíntesis , Ratas , Ratas Wistar , Receptores Opioides/biosíntesis , Refuerzo en Psicología , Recompensa , Autoadministración
18.
Vis Neurosci ; 29(3): 203-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22643230

RESUMEN

The presence of opioid receptors has been confirmed by a variety of techniques in vertebrate retinas including those of mammals; however, in most reports, the location of these receptors has been limited to retinal regions rather than specific cell types. Concurrently, our knowledge of the physiological functions of opioid signaling in the retina is based on only a handful of studies. To date, the best-documented opioid effect is the modulation of retinal dopamine release, which has been shown in a variety of vertebrate species. Nonetheless, it is not known if opioids can affect dopaminergic amacrine cells (DACs) directly, via opioid receptors expressed by DACs. This study, using immunohistochemical methods, sought to determine whether (1) µ- and δ-opioid receptors (MORs and DORs, respectively) are present in the mouse retina, and if present, (2) are they expressed by DACs. We found that MOR and DOR immunolabeling were associated with multiple cell types in the inner retina, suggesting that opioids might influence visual information processing at multiple sites within the mammalian retinal circuitry. Specifically, colabeling studies with the DAC molecular marker anti-tyrosine hydroxylase antibody showed that both MOR and DOR immunolabeling localize to DACs. These findings predict that opioids can affect DACs in the mouse retina directly, via MOR and DOR signaling, and might modulate dopamine release as reported in other mammalian and nonmammalian retinas.


Asunto(s)
Células Amacrinas/metabolismo , Neuronas Dopaminérgicas/metabolismo , Receptores Opioides/biosíntesis , Retina/metabolismo , Animales , Anticuerpos Monoclonales/biosíntesis , Interpretación Estadística de Datos , Femenino , Cabras/inmunología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Receptores Opioides delta/inmunología , Receptores Opioides delta/fisiología , Receptores Opioides mu/inmunología , Receptores Opioides mu/fisiología , Tirosina 3-Monooxigenasa/metabolismo
19.
J Mol Neurosci ; 46(2): 285-92, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21603976

RESUMEN

Several studies demonstrated a cross-talk between the opioid and cannabinoid system. The NOP receptor and its endogenous ligand nociceptin/orphanin FQ represent an opioid-related functional entity that mediates some non-classical opioid effects. The relationship between cannabinoid and nociceptin/NOP system is yet poorly explored. In this study, we used the neuroblastoma SH-SY5Y cell line to investigate the effect of delta-9-tetrahydrocannabinol (∆(9)-THC) on nociceptin/NOP system. Results revealed that the exposure to ∆(9)-THC (100, 150, and 200 nM) for 24 h produces a dose-dependent NOP receptor B (max) down-regulation. Moreover, ∆(9)-THC caused a dose-dependent decrease in NOP mRNA levels. The selective cannabinoid receptor CB1 antagonist AM251 (1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-1-piperidinyl-1H-pyrazole-3-carboxamide) reduces both effects, suggesting that ∆(9)-THC activation of CB1 receptor is involved in the observed effects. These data show evidence of a cross-talk between NOP and CB1 receptors, thus suggesting a possible interplay between cannabinoid and nociceptin/NOP system.


Asunto(s)
Dronabinol/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptores Opioides/biosíntesis , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Dronabinol/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Neuroblastoma/patología , Piperidinas/farmacología , Unión Proteica , Pirazoles/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Receptores Opioides/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor de Nociceptina
20.
Gynecol Oncol ; 124(2): 319-24, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22037317

RESUMEN

OBJECTIVE: The opioid growth factor (OGF) and its receptor (OGFr), serve as inhibitory axis regulating cell proliferation in normal cells and cancer. We investigated the presence and relative expression of OGF and OGFr in normal human ovarian surface epithelial (HOSE) cells, benign ovarian cysts, and ovarian cancers. METHODS: Surgical samples of 16 patients with ovarian cancer and 27 patients with ovarian benign cysts were obtained intraoperatively. HOSE were collected by scraping the surface of normal ovaries of 10 post menopausal women undergoing hysterectomy and oophorectomy. Semiquantitative immunohistochemistry was used to assess the presence, distribution, and levels of OGF and OGFr. Receptor binding assays measured binding capacity and affinity of OGFr for radiolabeled OGF. RESULTS: OGF and OGFr were present in HOSE cells, ovarian cysts, and ovarian cancers. Compared to HOSE cells, OGF and OGFr protein levels were reduced 29% and 34% (p<0.001), respectively, in ovarian cysts, and decreased 58% and 48% (p<0.001), respectively, in ovarian cancers. Binding assays revealed 5.4 fold fewer OGFr binding sites in cancers than cysts (p<0.05). Levels of OGF and OGFr were comparable in primary, metastatic, or recurrent ovarian cancers. CONCLUSION: We have shown that a native opioid pathway, the OGF-OGFr axis, is present in human ovarian cancer. Importantly, the expression of OGF and OGFr is diminished in human ovarian cancer. As OGF and OGFr normally function in maintaining cell proliferation, therapy to harness OGF/OGFr function could provide a useful biologic-based treatment for human ovarian cancer.


Asunto(s)
Neoplasias Ováricas/metabolismo , Receptores Opioides/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Células Epiteliales/metabolismo , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Quistes Ováricos/metabolismo , Ovario/citología , Ovario/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA