Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Clin Psychopharmacol ; 43(2): 89-96, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36821406

RESUMEN

BACKGROUND: Preclinical evidence indicates that the κ-opioid receptor (KOR)/dynorphin pathway is implicated in depressive-like behaviors. Ketamine is believed to partly exert its antidepressant effects by modulating the opioid system. This post hoc study examined the following research questions: (1) at baseline, were there differences in KOR or dynorphin plasma levels between individuals with major depressive disorder (MDD) and healthy volunteers (HVs) or between men and women? (2) in individuals with MDD, did KOR or dynorphin baseline plasma levels moderate ketamine's therapeutic effects or adverse effects? and (3) in individuals with MDD, were KOR or dynorphin plasma levels affected after treatment with ketamine compared with placebo? METHODS: Thirty-nine unmedicated individuals with MDD (23 women) and 25 HVs (16 women) received intravenous ketamine (0.5 mg/kg) and placebo in a randomized, crossover, double-blind trial. Blood was obtained from all participants at baseline and at 3 postinfusion time points (230 minutes, day 1, day 3). Linear mixed model regressions were used. RESULTS: At baseline, participants with MDD had lower KOR plasma levels than HVs ( F1,60 = 13.16, P < 0.001), and women (MDD and HVs) had higher KOR plasma levels than men ( F1,60 = 4.98, P = 0.03). Diagnosis and sex had no significant effects on baseline dynorphin levels. Baseline KOR and dynorphin levels did not moderate ketamine's therapeutic or adverse effects. Compared with placebo, ketamine was not associated with postinfusion changes in KOR or dynorphin levels. CONCLUSIONS: In humans, diagnosis of MDD and biological sex are involved with changes in components of the KOR/dynorphin pathway. Neither KOR nor dynorphin levels consistently moderated ketamine's therapeutic effects or adverse effects, nor were levels altered after ketamine infusion. TRIAL REGISTRATION: NCT00088699 ( ClinicalTrials.gov ).


Asunto(s)
Trastorno Depresivo Mayor , Ketamina , Masculino , Humanos , Femenino , Trastorno Depresivo Mayor/tratamiento farmacológico , Ketamina/uso terapéutico , Receptores Opioides kappa/uso terapéutico , Dinorfinas/uso terapéutico , Antidepresivos/uso terapéutico
2.
Neurosci Res ; 188: 75-87, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36368461

RESUMEN

Panax notoginseng (Chinese ginseng, Sanqi), one of the major ginseng species, has been traditionally used to alleviate different types of chronic pain. The raw P. notoginseng powder is commonly available in China as a non-prescription drug to treat various aliments including arthritic pain. However, strong scientific evidence is needed to illustrate its pain antihypersensitive effects, effective ingredients and mechanism of action. The oral P. notoginseng powder dose-dependently alleviated formalin-induced tonic hyperalgesia, and its total ginsenosides remarkably inhibited neuropathic pain hypersensitivity. Ginsenoside Rb1, the most abundant ginsenoside of P. notoginseng, dose-dependently produced neuropathic pain antihypersensitivity. Conversely, ginsenosides Rg1, Re and notoginseng R1, the other major saponins from P. notoginseng, failed to inhibit formalin-induced tonic pain or mechanical allodynia in neuropathic pain. Ginsenoside Rb1 metabolites ginsenosides Rg3, Compound-K and protopanaxadiol also had similar antineuropathic pain efficacy to ginsenoside Rb1. Additionally, intrathecal ginsenoside Rb1 specifically stimulated dynorphin A expression which was colocalized with microglia but not neurons or astrocytes in the spinal dorsal horn and primary cultured cells. Pretreatment with microglial metabolic inhibitor minocycline, dynorphin A antiserum and specific κ-opioid receptor antagonist GNTI completely blocked Rb1-induced mechanical antiallodynia in neuropathic pain. Furthermore, the specific glucocorticoid receptor (GR) antagonist Dex-21-mesylate (but not GPR30 estrogen receptor antagonist G15) also entirely attenuated ginsenoside Rb1-related antineuropathic pain effects. All these results, for the first time, show that P. notoginseng alleviates neuropathic pain and ginsenoside Rb1 is its principal effective ingredient. Furthermore, ginsenoside Rb1 inhibits neuropathic pain by stimulation of spinal microglial dynorphin A expression following GR activation.


Asunto(s)
Ginsenósidos , Neuralgia , Panax notoginseng , Ginsenósidos/metabolismo , Ginsenósidos/farmacología , Ginsenósidos/uso terapéutico , Dinorfinas/metabolismo , Dinorfinas/farmacología , Dinorfinas/uso terapéutico , Panax notoginseng/metabolismo , Microglía/metabolismo , Polvos/metabolismo , Polvos/farmacología , Polvos/uso terapéutico , Hiperalgesia/metabolismo , Neuralgia/tratamiento farmacológico
3.
BMC Neurosci ; 23(1): 58, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36217122

RESUMEN

BACKGROUND: Opioids are among the most effective and commonly prescribed analgesics for the treatment of acute pain after spinal cord injury (SCI). However, morphine administration in the early phase of SCI undermines locomotor recovery, increases cell death, and decreases overall health in a rodent contusion model. Based on our previous studies we hypothesize that morphine acts on classic opioid receptors to alter the immune response. Indeed, we found that a single dose of intrathecal morphine increases the expression of activated microglia and macrophages at the injury site. Whether similar effects of morphine would be seen with repeated intravenous administration, more closely simulating clinical treatment, is not known. METHODS: To address this, we used flow cytometry to examine changes in the temporal expression of microglia and macrophages after SCI and intravenous morphine. Next, we explored whether morphine changed the function of these cells through the engagement of cell-signaling pathways linked to neurotoxicity using Western blot analysis. RESULTS: Our flow cytometry studies showed that 3 consecutive days of morphine administration after an SCI significantly increased the number of microglia and macrophages around the lesion. Using Western blot analysis, we also found that repeated administration of morphine increases ß-arrestin, ERK-1 and dynorphin (an endogenous kappa opioid receptor agonist) production by microglia and macrophages. CONCLUSIONS: These results suggest that morphine administered immediately after an SCI changes the innate immune response by increasing the number of immune cells and altering neuropeptide synthesis by these cells.


Asunto(s)
Morfina , Traumatismos de la Médula Espinal , Analgésicos/farmacología , Analgésicos Opioides/farmacología , Animales , Dinorfinas/metabolismo , Dinorfinas/farmacología , Dinorfinas/uso terapéutico , Macrófagos , Microglía/patología , Morfina/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Opioides kappa/metabolismo , Receptores Opioides kappa/uso terapéutico , Recuperación de la Función , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , beta-Arrestinas/metabolismo , beta-Arrestinas/farmacología , beta-Arrestinas/uso terapéutico
4.
Brain Res ; 1735: 146742, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32114059

RESUMEN

The dynorphin / kappa opioid receptor (KOR) system has been implicated in many aspects that influence neuropsychiatric disorders. Namely, this system modulates neural circuits that primarily regulate reward seeking, motivation processing, stress responsivity, and pain sensitivity, thus affecting the development of substance and alcohol use disorder (AUD). The effects of this system are often bidirectional and depend on projection targets. To date, a majority of the studies focusing on this system have examined the KOR function using agonists and antagonists. Indeed, there are studies that have examined prodynorphin and dynorphin levels by measuring mRNA and tissue content levels; however, static levels of the neuropeptide and its precursor do not explain complete and online function of the peptide as would be explained by measuring dynorphin transmission in real time. New and exciting methods using optogenetics, chemogenetics, genetic sensors, fast scan cyclic voltammetry are now being developed to detect various neuropeptides with a focus on opioid peptides, including dynorphin. In this review we discuss studies that examine dynorphin projections in areas involved in AUD, its functional involvement in AUD and vulnerability to develop AUD at various ages. Moreover, we discuss dynorphin's role in promoting AUD by dysregulation motivation circuits and how advancements in opioid peptide detection will further our understanding.


Asunto(s)
Alcoholismo/tratamiento farmacológico , Dinorfinas/farmacología , Dinorfinas/uso terapéutico , Alcoholismo/metabolismo , Animales , Dinorfinas/metabolismo , Humanos , Motivación/efectos de los fármacos , Neuropéptidos/análisis , Neuropéptidos/química , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides kappa/fisiología , Recompensa
5.
Biol Pharm Bull ; 41(6): 957-960, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29863085

RESUMEN

Olfactory bulbectomized (OBX) mice are characterized by impaired performance in the passive avoidance test and decreased number of cholinergic neurons in the hippocampus. Several studies have reported that κ-opioid receptor agonists improve cognitive function in mice. However, their influence on OBX-induced cognitive dysfunction remains unclear. To address this question, we evaluated the effects of the endogenous κ-opioid receptor agonist dynorphin A (Dyn A) and the selective agonist trans-(-)-U-50488 on the behavior of OBX mice in the passive avoidance test. The cognitive dysfunction of OBX mice was significantly recovered by the intracerebroventricular administration of Dyn A or trans-(-)-U-50488. The effects of these two agonists were counteracted by the selective κ-opioid receptor antagonist nor-binaltorphimine or the inhibitor of acetylcholine release ß-bungarotoxin. These findings suggest that κ-opioid receptor agonists produce anti-dementia effects through activation of cholinergic neurons in OBX mice.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , Receptores Opioides kappa/agonistas , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/uso terapéutico , Animales , Neuronas Colinérgicas/efectos de los fármacos , Neuronas Colinérgicas/fisiología , Disfunción Cognitiva/fisiopatología , Dinorfinas/farmacología , Dinorfinas/uso terapéutico , Hipocampo/fisiología , Masculino , Trastornos de la Memoria/fisiopatología , Ratones , Bulbo Olfatorio/cirugía , Receptores Opioides kappa/fisiología
6.
J Ethnopharmacol ; 196: 151-159, 2017 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-27989510

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Bullatine A, a C20-diterpenoid alkaloid and one of the major effective ingredients in Aconiti brachypodi Radix (Xue-shang-yi-zhi-hao), can block pain hypersensitivity in a variety of rodent models through expression of spinal microglial dynorphin A. AIM OF THE STUDY: To assess the interaction between bullatine A and morphine on antinociception in acute nociception and pain hypersensitivity states, with the exogenous synthetic dynorphin A as a comparison MATERIALS AND METHODS: Spinal nerve ligation-induced neuropathic rats and naïve mice were used for assessing the acute and chronic interactions of bullatine A/dynorphin A with morphine. RESULTS: Single subcutaneous injection of bullatine A or dynorphin A(1-17) did not either alter formalin- and thermally (hot-plate and water immersion tests)-induced acute nociception or potentiate morphine antinociception in naïve mice. In contrast, bullatine A dose-dependently inhibited formalin-induced tonic pain with the efficacy of 54% inhibition and the half-effective dose of 0.9mg/kg. Concurrent bullatine A additively enhanced morphine antinociception. In neuropathic rats, the antinociceptive effects of multiple bidaily intrathecal injections of bullatine A and dynorphin A remained consistent over 13 days, whereas morphine produced progressive and complete tolerance to antinociception, which was completely inhibited by concurrent bullatine A and dynorphin A. A single intrathecal injection of bullatine A and dynorphin A immediately reversed established morphine tolerance in neuropathic rats, although the blockade was a less degree in the thermally induced mouse acute nociceptive tests. The inhibitory effects of bullatine A and dynorphin A on morphine tolerance were immediately and completely attenuated by intrathecal dynorphin A antibody and/or selective κ-opioid receptor antagonist GNTI. CONCLUSION: These results suggest that bullatine A produces antinociception without induction of tolerance and inhibits morphine antinociceptive tolerance, and provide pharmacological basis for concurrent bullatine A and morphine treatment for chronic pain and morphine analgesic tolerance.


Asunto(s)
Alcaloides , Analgésicos , Diterpenos , Hiperalgesia/tratamiento farmacológico , Morfina , Dolor/tratamiento farmacológico , Receptores Opioides kappa/metabolismo , Alcaloides/farmacología , Alcaloides/uso terapéutico , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Conducta Animal/efectos de los fármacos , Diterpenos/farmacología , Diterpenos/uso terapéutico , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Tolerancia a Medicamentos , Dinorfinas/farmacología , Dinorfinas/uso terapéutico , Formaldehído , Calor , Hiperalgesia/metabolismo , Masculino , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Morfina/farmacología , Morfina/uso terapéutico , Dolor/metabolismo , Ratas Wistar , Columna Vertebral/efectos de los fármacos , Columna Vertebral/metabolismo
7.
Depress Anxiety ; 33(10): 895-906, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27699938

RESUMEN

Kappa-opioid receptor (KOR) antagonists are currently being considered for the treatment of a variety of neuropsychiatric conditions, including depressive, anxiety, and substance abuse disorders. A general ability to mitigate the effects of stress, which can trigger or exacerbate these conditions, may explain their putative efficacy across such a broad array of conditions. The discovery of their potentially therapeutic effects evolved from preclinical research designed to characterize the molecular mechanisms by which experience causes neuroadaptations in the nucleus accumbens (NAc), a key element of brain reward circuitry. This research established that exposure to drugs of abuse or stress increases the activity of the transcription factor CREB (cAMP response element binding protein) in the NAc, which leads to elevated expression of the opioid peptide dynorphin that in turn causes core signs of depressive- and anxiety-related disorders. Disruption of KORs-the endogenous receptors for dynorphin-produces antidepressant- and anxiolytic-like actions in screening procedures that identify standard drugs of these classes, and reduces stress effects in tests used to study addiction and stress-related disorders. Although interest in this target is high, prototypical KOR antagonists have extraordinarily persistent pharmacodynamic effects that complicate clinical trials. The development of shorter acting KOR antagonists together with more rapid designs for clinical trials may soon provide insight on whether these drugs are efficacious as would be predicted by preclinical work. If successful, KOR antagonists would represent a unique example in psychiatry where the therapeutic mechanism of a drug class is understood before it is shown to be efficacious in humans.


Asunto(s)
Ansiolíticos/uso terapéutico , Antidepresivos/uso terapéutico , Trastornos de Ansiedad/tratamiento farmacológico , Dinorfinas/fisiología , Dinorfinas/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Núcleo Accumbens/efectos de los fármacos , Receptores Opioides kappa/antagonistas & inhibidores , Animales , Trastornos de Ansiedad/fisiopatología , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Proteína de Unión a CREB/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Núcleo Accumbens/fisiopatología , Receptores Opioides kappa/fisiología , Recompensa , Estrés Psicológico/complicaciones , Estrés Psicológico/fisiopatología , Trastornos Relacionados con Sustancias/complicaciones , Trastornos Relacionados con Sustancias/fisiopatología , Investigación Biomédica Traslacional
8.
Future Med Chem ; 8(2): 165-77, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26824470

RESUMEN

Chronic pain is one of the most ubiquitous diseases in the world, but treatment is difficult with conventional methods, due to undesirable side effects of treatments and unknown mechanisms of pathological pain states. The endogenous peptide, dynorphin A has long been established as a target for the treatment of pain. Interestingly, this unique peptide has both inhibitory (opioid in nature) and excitatory activities (nonopioid) in the CNS. Both of these effects have been found to play a role in pain and much work has been done to develop therapeutics to enhance the inhibitory effects. Here we will review the dynorphin A compounds that have been designed for the modulation of pain and will discuss where the field stands today.


Asunto(s)
Dinorfinas/uso terapéutico , Neuralgia/tratamiento farmacológico , Analgésicos no Narcóticos/uso terapéutico , Enfermedad Crónica , Dinorfinas/química , Humanos , Neuralgia/patología , Neurotransmisores/química , Neurotransmisores/uso terapéutico , Receptores de Bradiquinina/química , Receptores de Bradiquinina/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides kappa/metabolismo , Relación Estructura-Actividad
9.
Neurosci Lett ; 615: 15-20, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26780565

RESUMEN

Prototypical long-acting kappa opioid receptor (KOPR) antagonists [e.g., norbinaltorphimine (norBNI)] have been reported to exert anxiolytic-like effects in several commonly used anxiety tests in rodents including the novelty-induced hypophagia (NIH) and elevated plus maze (EPM) tests. It remains unknown if the short-acting KOPR antagonists (e.g., zyklophin and LY2444296) have similar effects. In this study effects of zyklophin and LY2444296 (s.c.) were investigated in the NIH and EPM tests in mice 1h post-injection and compared with norBNI (i.p.) 48h post-administration. In the NIH test, zyklophin at 3 and 1mg/kg, but not 0.3mg/kg, or LY2444296 at 30mg/kg decreased the latency of palatable food consumption in novel cages, but had no effect in training cages, similar to norBNI (10mg/kg). Zyklophin at 3 or 1mg/kg increased or had a trend of increasing the amount of palatable food consumption in novel cages, with no effects in training cages, further indicating its anxiolytic-like effect, but norBNI (10mg/kg) and LY2444296 (30mg/kg) did not. In the EPM test, norBNI (10mg/kg) increased open arm time and % open arm entries or time, but zyklophin at all three doses and LY2444296 (30mg/kg) had no effects. In addition, zyklophin at 3mg/kg increased numbers of close and total arm entries on EPM, suggesting increased activity; however, norBNI and LY2444296 had no effects on close and total arm entries. Thus, all three KOPR antagonists had anxiolytic-like effects in the NIH test. However, only the long-acting one (norBNI), but not the short-acting ones (zyklophin and LY2444296), demonstrated anti-anxiety like effects in the EPM test. It remains to be investigated if the differences are due to the differences in their durations of action and/or pharmacodynamic properties.


Asunto(s)
Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Dinorfinas/farmacología , Naltrexona/análogos & derivados , Fragmentos de Péptidos/farmacología , Proteínas/farmacología , Receptores Opioides kappa/antagonistas & inhibidores , Animales , Ansiolíticos/uso terapéutico , Ansiedad/psicología , Dinorfinas/uso terapéutico , Conducta Alimentaria/efectos de los fármacos , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Naltrexona/farmacología , Naltrexona/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Proteínas/uso terapéutico
10.
Eur J Pharm Biopharm ; 87(1): 73-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24462793

RESUMEN

A niosomal formulation, functionalized with N-palmitoylglucosamine, was developed as potential brain targeted delivery system of dynorphin-B. In fact, this endogenous neuropeptide, selective agonist of k opioid receptors, is endowed with relevant pharmacological activities on the central nervous system, including a marked antinociceptive effect, but is unable to cross the blood brain barrier (BBB), thus requiring intracerebroventricular administration. Statistical design of experiments was utilized for a systematic evaluation of the influence of variations of the relative amounts of the components of the vesicle membrane (Span 60, cholesterol and SolulanC24) on vesicle mean diameter, polydispersity index and drug entrapment efficiency, chosen as the responses to optimize. A Scheffé simplex-centroid design was used to obtain the coefficients of the postulated mathematical model. The study of the response surface plots revealed that variations of the considered factors had different effects on the selected responses. The desirability function enabled for finding the optimal mixture composition, which represented the best compromise to simultaneously optimize all the three responses. The experimental values obtained with the optimized formulation were very similar to the predicted ones, proving the validity of the proposed regression model. The optimized niosomal formulation of dynorphin-B administered intravenously to mice (100mg/kg) showed a pronounced antinociceptive effect, significantly higher (P<0.05) than that given by i.v. administration of the simple solution of the peptide at the same concentration, proving its effectiveness in enabling the peptide brain delivery. These positive results suggest that the proposed approach could be successfully extended to other neuro-active peptides exerting a strong central action, even at low doses, but unable to cross the BBB.


Asunto(s)
Analgésicos/administración & dosificación , Encéfalo/efectos de los fármacos , Portadores de Fármacos/química , Dinorfinas/administración & dosificación , Endorfinas/administración & dosificación , Glucolípidos/química , Analgésicos/farmacocinética , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Composición de Medicamentos , Sistemas de Liberación de Medicamentos , Estabilidad de Medicamentos , Dinorfinas/farmacocinética , Dinorfinas/farmacología , Dinorfinas/uso terapéutico , Endorfinas/farmacocinética , Endorfinas/farmacología , Endorfinas/uso terapéutico , Glucolípidos/síntesis química , Inyecciones Intravenosas , Inyecciones Intraventriculares , Liposomas , Masculino , Ratones , Dolor/tratamiento farmacológico , Dolor/metabolismo , Receptores Opioides kappa/agonistas
11.
Alcohol ; 46(4): 359-70, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22459870

RESUMEN

This review represents the focus of a symposium that was presented at the "Alcoholism and Stress: A Framework for Future Treatment Strategies" conference in Volterra, Italy on May 3-6, 2011 and organized/chaired by Dr. Brendan M. Walker. The primary goal of the symposium was to evaluate and disseminate contemporary findings regarding the emerging role of kappa-opioid receptors (KORs) and their endogenous ligands dynorphins (DYNs) in the regulation of escalated alcohol consumption, negative affect and cognitive dysfunction associated with alcohol dependence, as well as DYN/KOR mediation of the effects of chronic stress on alcohol reward and seeking behaviors. Dr. Glenn Valdez described a role for KORs in the anxiogenic effects of alcohol withdrawal. Dr. Jay McLaughlin focused on the role of KORs in repeated stress-induced potentiation of alcohol reward and increased alcohol consumption. Dr. Brendan Walker presented data characterizing the effects of KOR antagonism within the extended amygdala on withdrawal-induced escalation of alcohol self-administration in dependent animals. Dr. Georgy Bakalkin concluded with data indicative of altered DYNs and KORs in the prefrontal cortex of alcohol dependent humans that could underlie diminished cognitive performance. Collectively, the data presented within this symposium identified the multifaceted contribution of KORs to the characteristics of acute and chronic alcohol-induced behavioral dysregulation and provided a foundation for the development of pharmacotherapeutic strategies to treat certain aspects of alcohol use disorders.


Asunto(s)
Alcoholismo/fisiopatología , Amígdala del Cerebelo/efectos de los fármacos , Etanol/farmacología , Receptores Opioides kappa/fisiología , Estrés Psicológico/complicaciones , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Alcoholismo/tratamiento farmacológico , Alcoholismo/psicología , Amígdala del Cerebelo/fisiopatología , Animales , Modelos Animales de Enfermedad , Dinorfinas/farmacología , Dinorfinas/fisiología , Dinorfinas/uso terapéutico , Humanos , Neurotransmisores/farmacología , Neurotransmisores/fisiología , Neurotransmisores/uso terapéutico , Receptores Opioides kappa/agonistas , Síndrome de Abstinencia a Sustancias/fisiopatología
12.
Alcohol Res ; 34(4): 516-21, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23584117

RESUMEN

Alcohol dependence is a chronic relapsing disorder characterized by neuroadaptations that may result in the emergence of negative affective states and stress responses upon discontinuation of alcohol use. Clinical studies have demonstrated that alcohol-dependent people are more sensitive to relapse provoking cues such as alcohol, negative affect, and stress. Moreover, stress relief during protracted abstinence is thought to be a major motivation for excessive alcohol consumption. The relationship between chronic alcohol use, stress, and relapse has implications for the treatment of alcohol dependence. Recent research suggests that neural systems mediating stress responses may offer useful targets for pharmacotherapy of alcoholism.


Asunto(s)
Alcoholismo/tratamiento farmacológico , Estrés Psicológico/tratamiento farmacológico , Antagonistas de Receptores Adrenérgicos alfa 1/uso terapéutico , Alcoholismo/prevención & control , Dinorfinas/uso terapéutico , Humanos , Antagonistas del Receptor de Neuroquinina-1 , Neuropéptido Y/uso terapéutico , Neurotransmisores/uso terapéutico , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores Opioides kappa/agonistas , Prevención Secundaria , Estrés Psicológico/prevención & control
13.
Nat Prod Rep ; 28(4): 741-62, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21340067

RESUMEN

Anticonvulsant neuropeptides are best known for their ability to suppress seizures and modulate pain pathways. Galanin, neuropeptide Y, somatostatin, neurotensin, dynorphin, among others, have been validated as potential first-in-class anti-epileptic or/and analgesic compounds in animal models of epilepsy and pain, but their therapeutic potential extends to other neurological indications, including neurodegenerative and psychatric disorders. Disease-modifying properties of neuropeptides make them even more attractive templates for developing new-generation neurotherapeutics. Arguably, efforts to transform this class of neuropeptides into drugs have been limited compared to those for other bioactive peptides. Key challenges in developing neuropeptide-based anticonvulsants are: to engineer optimal receptor-subtype selectivity, to improve metabolic stability and to enhance their bioavailability, including penetration across the blood­brain barrier (BBB). Here, we summarize advances toward developing systemically active and CNS-penetrant neuropeptide analogs. Two main objectives of this review are: (1) to provide an overview of structural and pharmacological properties for selected anticonvulsant neuropeptides and their analogs and (2) to encourage broader efforts to convert these endogenous natural products into drug leads for pain, epilepsy and other neurological diseases.


Asunto(s)
Anticonvulsivantes , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Neuropéptidos , Analgésicos Opioides/metabolismo , Analgésicos Opioides/uso terapéutico , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Barrera Hematoencefálica/metabolismo , Dinorfinas/metabolismo , Dinorfinas/uso terapéutico , Epilepsia/tratamiento farmacológico , Galanina/metabolismo , Galanina/uso terapéutico , Estructura Molecular , Neuropéptido Y/metabolismo , Neuropéptido Y/uso terapéutico , Neuropéptidos/metabolismo , Neuropéptidos/uso terapéutico , Neurotensina/metabolismo , Neurotensina/uso terapéutico , Convulsiones/tratamiento farmacológico , Homología de Secuencia de Aminoácido , Somatostatina/metabolismo , Somatostatina/uso terapéutico
14.
J Cereb Blood Flow Metab ; 29(4): 698-706, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19142196

RESUMEN

Using a focal cerebral ischemia model in rats, brain ischemia-induced changes in expression levels of mRNA and protein, and activities of proprotein convertase 2 (PC2) in the cortex were examined. In situ hybridization analyses revealed a transient upregulation of the mRNA level for PC2 at an early reperfusion hour, at which the level of PC2 protein was also high as determined by immunocytochemistry and western blotting. When enzymatic activities of PC2 were analyzed using a synthetic substrate, a significant decrease was observed at early reperfusion hours at which levels of PC2 protein were still high. Also decreased at these reperfusion hours were tissue levels of dynorphin-A(1-8) (DYN-A(1-8)), a PC2 substrate, as determined by radioimmunoassay. Further examination of PC2 protein biosynthesis by metabolic labeling in cultured neuronal cells showed that in ischemic cells, the proteolytic processing of PC2 was greatly attenuated. Finally, in mice, an intracerebroventricular administration of synthetic DYN-A(1-8) significantly reduced the extent of ischemic brain injury. In mice those lack an active PC2, exacerbated brain injury was observed after an otherwise non-lethal focal ischemia. We conclude that brain ischemia attenuates PC2 and PC2-mediated neuropeptide processing. This attenuation may play a role in the pathology of ischemic brain injury.


Asunto(s)
Isquemia Encefálica/metabolismo , Dinorfinas/análisis , Fragmentos de Péptidos/análisis , Proproteína Convertasa 2/análisis , Animales , Dinorfinas/administración & dosificación , Dinorfinas/uso terapéutico , Hibridación in Situ , Neuropéptidos , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/uso terapéutico , Proproteína Convertasa 2/genética , Proproteína Convertasa 2/metabolismo , ARN Mensajero/análisis , Ratas , Reperfusión , Accidente Cerebrovascular , Factores de Tiempo , Regulación hacia Arriba/genética
15.
Neurochem Int ; 52(6): 1256-65, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18294735

RESUMEN

Alterations in the opioidergic system have been found in cerebral ischemia. Neuroprotection studies have demonstrated the involvement of the opioidergic system in cerebral ischemia/reperfusion (I/R). However, the neuroprotective mechanisms remain largely unclear. This study was conducted to investigate whether intracerebroventricular administration of opioidergic agonists has a neuroprotective effect against cerebral ischemia in rats and, if this proved to be the case, to determine the potential neuroprotective mechanisms. Using a focal cerebral I/R rat model, we demonstrated that the opioidergic agents, BW373U86 (delta agonist) and Dynorphin A 1-13 (kappa agonist), but not TAPP (mu agonist), attenuated cerebral ischemic injury as manifested in the reduction of cerebral infarction and preservation of neurons. The antagonism assay showed that the neuroprotective effect of Dynorphin A was attenuated by nor-Binaltorphimine (kappa antagonist). Surprisingly, BW373U86-induced neuroprotection was not changed by Naltrindole (delta antagonist). These findings indicate that BW373U86 and Dynorphin A exerted distinct neuroprotection against ischemia via opioid-independent and -dependent mechanisms, respectively. The post-ischemic protection in beneficial treatments was accompanied by alleviations in brain edema, inflammatory cell infiltration, and pro-inflammatory cytokine interleukin 6 (IL-6) expression. In vitro cell study further demonstrated that the opioidergic agonists, delta and kappa, but not mu, attenuated IL-6 production from stimulated glial cells. Our findings indicate that opioidergic agents have a role in post-ischemic progression through both opioid-dependent and -independent mechanisms. In spite of the distinct-involved action mechanism, the potential neuroprotective effect of opioidergic compounds was associated with immune suppression. Taken together, these findings suggest a potential role for opioidergic agents in the therapeutic consideration of neuroinflammatory diseases. However, a better understanding of the mechanisms involved is necessary before this therapeutic potential can be realized.


Asunto(s)
Analgésicos Opioides/farmacología , Isquemia Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Citoprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Amidinas/farmacología , Amidinas/uso terapéutico , Analgésicos Opioides/uso terapéutico , Animales , Benzamidas/farmacología , Benzamidas/uso terapéutico , Encéfalo/irrigación sanguínea , Encéfalo/fisiopatología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Citoprotección/inmunología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Dinorfinas/farmacología , Dinorfinas/uso terapéutico , Encefalitis/tratamiento farmacológico , Encefalitis/inmunología , Encefalitis/fisiopatología , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/inmunología , Interleucina-6/inmunología , Interleucina-6/metabolismo , Masculino , Antagonistas de Narcóticos/farmacología , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Piperazinas/farmacología , Piperazinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores Opioides/agonistas , Receptores Opioides/metabolismo , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología
16.
Pharm Res ; 21(8): 1450-6, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15359581

RESUMEN

PURPOSE: Modulatory actions on morphine-induced effects, such as tolerance and withdrawal, have been noted for dynorphin A(1-13) [Dyn A(1-13)] and similar peptides. These are currently of limited therapeutic potential due to extensive metabolism by human metabolic enzymes resulting in a half-life of less than 1 min in human plasma. The purpose of this study was to identify stabilized dynorphin A (Dyn A) derivatives, to determine their metabolic routes in human plasma, and to assess whether the pharmacodynamic activity is retained. METHODS: The stability of peptides in human plasma was tested using in vitro metabolism studies with and without enzyme inhibitors. Identification of the generated metabolites was performed by mass spectrometry after high performance liquid chromatography (HPLC) separation. The in vivo activity of a stabilized dynorphin was tested by tail-flick assay in morphine-tolerant rats. RESULTS: Though amidation of the Dyn A(1-13) was able to stop the majority of C-terminal degradation, metabolism of Dyn A(1-10) amide continued by captopril sensitive enzymes, suggesting that Dyn A(1-13) amide is a better candidate for additional stabilization. Two Dyn A(1-13) amide derivatives further stabilized at the N-terminal end, [D-Tyr1]-Dyn A(1-13) amide and [N-Met-Tyr1]-Dyn A(1-13) amide, showed half-lives in plasma of 70 and 130 min, respectively. The most stable derivative [N-Met-Tyr1]-Dyn A(1-13) amide was tested successfully for retention of the pharmacological activity in modulating antinociceptive activity. CONCLUSIONS: [N-Met-Tyr1]-Dyn A(1-13) amide showed significant stability and antinociceptive activity in the tail-flick test, thus pointing to the clinical potential of this derivative in the management of pain as well as its potential activity in suppressing opiate tolerance and withdrawal.


Asunto(s)
Analgésicos/uso terapéutico , Dinorfinas/uso terapéutico , Leucina/análogos & derivados , Dependencia de Morfina/tratamiento farmacológico , Aminopeptidasas/antagonistas & inhibidores , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Captopril/farmacología , Carboxipeptidasas/antagonistas & inhibidores , Estabilidad de Medicamentos , Tolerancia a Medicamentos , Dinorfinas/química , Dinorfinas/metabolismo , Inhibidores Enzimáticos/farmacología , Semivida , Leucina/farmacología , Masculino , Dimensión del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Succinatos/farmacología , Compuestos de Sulfhidrilo/farmacología
17.
J Pharmacol Exp Ther ; 306(3): 1106-14, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12829725

RESUMEN

Topically administered capsaicin produces thermal allodynia, and this effect has been used to investigate pain transduction and its pharmacological modulation. This study investigated the parameters of topical capsaicin-induced thermal allodynia in unanesthetized rhesus monkeys and its pharmacological modulation by centrally acting compounds [a kappa-opioid agonist: (5alpha,7alpha,8beta)-(+)-N-methyl-N-(7-[1-pyrrolidinyl]-1-oxaspiro [4.5]dec-8-yl)-benzeneacetamide (U69,593); and noncompetitive N-methyl-d-aspartate (NMDA) antagonists: ketamine and MK-801 (dizocilpine)]. Rhesus monkeys (n = 4) were studied within the warm water tail withdrawal assay (20-s maximum latency), using thermal stimuli that are normally not noxious (38 and 42 degrees C). Capsaicin was applied topically on the tail (0.0013 and 0.004 M capsaicin solution on a 1-cm2 patch; 15-min contact). Topical capsaicin produced concentration-dependent thermal allodynia in both temperatures, robustly detected 15 to 90 min after topical capsaicin removal. A similar allodynic profile was observed with topical administration of the "endovanilloid" N-arachidonoyl-dopamine. The kappa-agonist U69,593 (0.01-0.1 mg/kg, s.c.) dose dependently prevented capsaicin (0.004 M)-induced allodynia in 38 and 42 degrees C, and the largest U69,593 dose also reversed ongoing allodynia within this model. Two NMDA antagonists, ketamine and MK-801 (0.32-1.8 and 0.032-0.056 mg/kg, respectively), also prevented capsaicin-induced allodynia in 38 degrees C, but only variably in 42 degrees C, at doses that did not cause robust thermal antinociceptive effects. At the largest doses studied, ketamine but not MK-801 also briefly reversed ongoing capsaicin-induced allodynia. The present model of topical capsaicin administration may be used to study antiallodynic effects of opioid and nonopioid compounds, as well as their ability to prevent and reverse allodynia, in unanesthetized nonhuman primates in the absence of tissue disruption.


Asunto(s)
Analgésicos/uso terapéutico , Bencenoacetamidas , Capsaicina , Dolor/inducido químicamente , Aminobutiratos/uso terapéutico , Animales , Modelos Animales de Enfermedad , Maleato de Dizocilpina/uso terapéutico , Dinorfinas/uso terapéutico , Femenino , Ketamina/uso terapéutico , Macaca mulatta , Dolor/tratamiento farmacológico , Dolor/prevención & control , Dimensión del Dolor , Fragmentos de Péptidos/uso terapéutico , Pirrolidinas/uso terapéutico
18.
Neurosci Lett ; 331(2): 123-7, 2002 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-12361856

RESUMEN

We compared the anticonvulsant actions of dynorphin A (1-13), galanin, neuropeptide Y and somatostatin in a model of self-sustaining status epilepticus (SSSE). SSSE was induced in adult Wistar rats by 30 min intermittent perforant path stimulation. Peptides or saline were injected into the hilus of the dentate gyrus 10 min after the end of perforant path stimulation. EEG was analyzed using Harmonie software (Stellate systems). While all neuropeptides showed significant seizure protecting effects, their anticonvulsant profiles followed different patterns: somatostatin and NPY induced strong, but transient suppression of spikes and seizures, while seizure suppression by dynorphin and galanin was more profound and irreversible.


Asunto(s)
Neuropéptidos/uso terapéutico , Estado Epiléptico/tratamiento farmacológico , Animales , Giro Dentado/efectos de los fármacos , Dinorfinas/administración & dosificación , Dinorfinas/uso terapéutico , Electroencefalografía , Galanina/administración & dosificación , Galanina/uso terapéutico , Inyecciones Intraventriculares , Masculino , Neuropéptido Y/administración & dosificación , Neuropéptido Y/uso terapéutico , Neuropéptidos/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/uso terapéutico , Vía Perforante , Ratas , Ratas Wistar , Somatostatina/administración & dosificación , Somatostatina/uso terapéutico , Estado Epiléptico/fisiopatología , Factores de Tiempo
19.
Bull Exp Biol Med ; 129(5): 423-4, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10977939

RESUMEN

In acute experiments on Nembutal-anesthetized cats, the effect of opiate receptor agonists DAGO, DSLET, and dinorphin A(1-13) on the incidence of idioventricular rhythm disturbances, including ventricular tachycardia and fibrillation, was studied under conditions of occlusion of circumflex branch of the left coronary artery and stimulation of the sensorimotor cortex. The most pronounced effects were observed with DSLET and dinorphin A(1-13). These preparations completely prevented ventricular fibrillation. DAGO produced a less pronounced protective effect probably because of parallel increase in plasma catecholamine concentration.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Encefalina Leucina/análogos & derivados , Corteza Motora/fisiopatología , Isquemia Miocárdica/fisiopatología , Receptores Opioides/fisiología , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Animales , Antiarrítmicos/farmacología , Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/tratamiento farmacológico , Gatos , Dinorfinas/farmacología , Dinorfinas/uso terapéutico , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Encefalina Ala(2)-MeFe(4)-Gli(5)/uso terapéutico , Encefalina Leucina/farmacología , Encefalina Leucina/uso terapéutico , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Receptores Opioides/agonistas
20.
Naunyn Schmiedebergs Arch Pharmacol ; 361(3): 273-8, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10731039

RESUMEN

Previous in vitro studies showed that the degradation of dynorphin-(1-8) [dyn-(1-8)] by cerebral membrane preparations is almost completely prevented by a mixture of three peptidase inhibitors (PIs), amastatin, captopril and phosphoramidon. In the present investigations, effects of the three PIs on the anti-nociception induced by the intra-third-ventricular (i.t.v.) administration of dyn-(1-8) were examined. The inhibitory effect of dyn-(1-8) on the tail-flick response was increased more than 100-fold by the i.t.v. pretreatment of rats with the three PIs. The inhibition produced by dyn-(1-8) in rats pretreated with any combination of two PIs was significantly smaller than that in rats pretreated with three PIs, indicating that any residual single peptidase could inactivate significant amounts of dyn-(1-8). The antagonistic effectiveness of naloxone, a relatively selective mu-opioid antagonist, indicates that dyn-(1-8)-induced inhibition of tail-flick response in rats pretreated with three PIs is mediated by mu-opioid receptors. Furthermore, mu-receptor-mediated inhibition induced by dyn-(1-8) was significantly greater than that produced by [Met5]-enkephalin in rats pretreated with three PIs. The data obtained in the present investigations together with those obtained in previous studies strongly indicate that dyn-(1-8) not only has well-known kappa-agonist activity but also has high mu-agonist activity.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Dinorfinas/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Fragmentos de Péptidos/antagonistas & inhibidores , Péptidos , Inhibidores de Proteasas/farmacología , Receptores Opioides mu/efectos de los fármacos , Analgésicos Opioides/farmacología , Análisis de Varianza , Animales , Antibacterianos/farmacología , Captopril/farmacología , Interacciones Farmacológicas , Dinorfinas/farmacología , Dinorfinas/uso terapéutico , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Glicopéptidos/farmacología , Inyecciones Intraventriculares , Masculino , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Dimensión del Dolor , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA