Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 203
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(4): 2140-2148, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31932450

RESUMO

Approximately 25% of patients who are prescribed opioids for chronic pain misuse them, and 5 to 10% develop an opioid use disorder. Although the neurobiological target of opioids is well known, the molecular mechanisms that are responsible for the development of addiction-like behaviors in some but not all individuals are poorly known. To address this issue, we used a unique outbred rat population (heterogeneous stock) that better models the behavioral and genetic diversity that is found in humans. We characterized individual differences in addiction-like behaviors using an addiction index that incorporates the key criteria of opioid use disorder: escalated intake, highly motivated responding, and hyperalgesia. Using in vitro electrophysiological recordings in the central nucleus of the amygdala (CeA), we found that rats with high addiction-like behaviors (HA) exhibited a significant increase in γ-aminobutyric acid (GABA) transmission compared with rats with low addiction-like behaviors (LA) and naive rats. The superfusion of CeA slices with nociceptin/orphanin FQ peptide (N/OFQ; 500 nM), an endogenous opioid-like peptide, normalized GABA transmission in HA rats. Intra-CeA levels of N/OFQ were lower in HA rats than in LA rats. Intra-CeA infusions of N/OFQ (1 µg per site) reversed the escalation of oxycodone self-administration in HA rats but not in LA rats. These results demonstrate that the downregulation of N/OFQ levels in the CeA may be responsible for hyper-GABAergic tone in the CeA that is observed in individuals who develop addiction-like behaviors. Based on these results, we hypothesize that small molecules that target the N/OFQ system might be useful for the treatment of opioid use disorder.


Assuntos
Tonsila do Cerebelo/metabolismo , Peptídeos Opioides/administração & dosagem , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Oxicodona/efeitos adversos , Ácido gama-Aminobutírico/metabolismo , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Humanos , Masculino , Transtornos Relacionados ao Uso de Opioides/etiologia , Transtornos Relacionados ao Uso de Opioides/metabolismo , Ratos , Autoadministração , Nociceptina
2.
Biol Pharm Bull ; 45(8): 1203-1207, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35908903

RESUMO

The opioid system in the central nervous system regulates depressive-like behavior in animals. Opioid receptors and their endogenous ligands have been focused on as novel therapeutic targets for depression. We synthesized dermorphin (DRM)-dynorphin (DYN) analogs (DRM-DYN001-004) using the message-address concept concerning interactions with opioid receptors. It has previously been reported that DRM-DYN001, 003, and 004 have shown high affinities for µ- and κ-opioid receptors, whereas all analogs had a lower affinity for the δ-opioid receptor than for other receptors using a receptor binding assay. However, it remains unknown whether these analogs show antidepressant-like effects in mice. We examined the effects of DRM-DYN analogs on the duration of immobile behavior in a tail suspension test. Intracerebroventricular administration of DRM-DYN001 in mice shortened the duration of immobile behavior, but did not affect locomotion. The DRM-DYN001-induced antidepressant-like effect was inhibited by co-administration of naloxone (non-selective opioid receptor antagonist), naloxonazine (selective µ1-opioid receptor antagonist), or nor-BNI (κ-opioid receptor antagonist), but not naltrindole (δ-opioid receptor antagonist). These data suggest that DRM-DYN001 exerts an antidepressant-like effect via activation of the central µ1- and κ-opioid receptors in mice and may represent a new lead peptide for further investigation for the development of novel therapeutic approaches for depression.


Assuntos
Antidepressivos , Dinorfinas , Peptídeos Opioides , Receptores Opioides kappa , Animais , Antidepressivos/administração & dosagem , Antidepressivos/farmacologia , Dinorfinas/administração & dosagem , Dinorfinas/química , Camundongos , Antagonistas de Entorpecentes/farmacologia , Peptídeos Opioides/administração & dosagem , Peptídeos Opioides/química , Receptores Opioides , Receptores Opioides kappa/metabolismo
3.
Anesthesiology ; 135(3): 482-493, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34237134

RESUMO

BACKGROUND: Cebranopadol, a mixed nociceptin/opioid receptor full agonist, can effectively relieve pain in rodents and humans. However, it is unclear to what degree different opioid receptor subtypes contribute to its antinociception and whether cebranopadol lacks acute opioid-associated side effects in primates. The authors hypothesized that coactivation of nociceptin receptors and µ receptors produces analgesia with reduced side effects in nonhuman primates. METHODS: The antinociceptive, reinforcing, respiratory-depressant, and pruritic effects of cebranopadol in adult rhesus monkeys (n = 22) were compared with µ receptor agonists fentanyl and morphine using assays, including acute thermal nociception, IV drug self-administration, telemetric measurement of respiratory function, and itch-scratching responses. RESULTS: Subcutaneous cebranopadol (ED50, 2.9 [95% CI, 1.8 to 4.6] µg/kg) potently produced antinociception compared to fentanyl (15.8 [14.6 to 17.1] µg/kg). Pretreatment with antagonists selective for nociceptin and µ receptors, but not δ and κ receptor antagonists, caused rightward shifts of the antinociceptive dose-response curve of cebranopadol with dose ratios of 2 and 9, respectively. Cebranopadol produced reinforcing effects comparable to fentanyl, but with decreased reinforcing strength, i.e., cebranopadol (mean ± SD, 7 ± 3 injections) versus fentanyl (12 ± 3 injections) determined by a progressive-ratio schedule of reinforcement. Unlike fentanyl (8 ± 2 breaths/min), systemic cebranopadol at higher doses did not decrease the respiratory rate (17 ± 2 breaths/min). Intrathecal cebranopadol (1 µg) exerted full antinociception with minimal scratching responses (231 ± 137 scratches) in contrast to intrathecal morphine (30 µg; 3,009 ± 1,474 scratches). CONCLUSIONS: In nonhuman primates, the µ receptor mainly contributed to cebranopadol-induced antinociception. Similar to nociceptin/µ receptor partial agonists, cebranopadol displayed reduced side effects, such as a lack of respiratory depression and pruritus. Although cebranopadol showed reduced reinforcing strength, its detectable reinforcing effects and strength warrant caution, which is critical for the development and clinical use of cebranopadol.


Assuntos
Indóis/administração & dosagem , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Receptores Opioides/agonistas , Compostos de Espiro/administração & dosagem , Analgésicos Opioides/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Feminino , Fentanila/administração & dosagem , Injeções Espinhais , Macaca mulatta , Masculino , Peptídeos Opioides/administração & dosagem , Receptores Opioides/fisiologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiologia , Receptor de Nociceptina , Nociceptina
4.
Turk J Med Sci ; 51(4): 2185-2192, 2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-33862670

RESUMO

Background/aim: Physical exercise is a state of physiological stress that requires adaptation of the organism to physical activity. Glycogen is an important and essential energy source for muscle contraction. Skeletal muscle and liver are two important glycogen stores, and the energy required to maintain exercise in rodents are provided by destruction of this glycogen depot. In this study, the effects of endogenous opioid peptide antagonism at the central nervous system level on tissue glycogen content after exhaustive exercise were investigated. Materials and methods: Rats had intracerebroventricularly (icv) received nonspecific opioid peptide receptor antagonist, naloxone (50 µg/10 µL in saline) and δ-opioid receptor-selective antagonist naltrindole (50 µg/10 µL in saline) and then exercised till exhaustion. After exhaustion, skeletal muscle, heart, and liver were excised immediately. Results: Both opioid peptide antagonists decreased glycogen levels in skeletal muscle. Although, in soleus muscle, this decrease was not statistically significant (p > 0.05), in gastrocnemius muscle, it was significant in the icv naloxone administered group compared with control (p < 0.05). Heart glycogen levels increased significantly in both naloxone and naltrindole groups compared to control and sham-operated groups (p < 0.05). Heart glycogen levels were higher in the naloxone group than naltrindole (p < 0.05). Liver glycogen levels were elevated significantly with icv naloxone administration compared with the control group (p < 0.05). Glycogen levels in the naloxone group was also significantly higher than the naltrindole group (p < 0.05). Conclusion: Our findings indicate that icv administered opioid peptide antagonists may play a role in glycogen metabolism in peripheral tissues such as skeletal muscle, heart, and liver.


Assuntos
Glicogênio/sangue , Antagonistas de Entorpecentes/farmacologia , Peptídeos Opioides/antagonistas & inibidores , Condicionamento Físico Animal , Animais , Encefalinas/administração & dosagem , Encefalinas/farmacologia , Infusões Intraventriculares , Naloxona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Neurotransmissores/administração & dosagem , Neurotransmissores/farmacologia , Peptídeos Opioides/administração & dosagem , Ratos , Receptores Opioides delta
5.
Br Poult Sci ; 60(3): 317-322, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30892928

RESUMO

1. The aim of the current study was to determine the effects of the central dopaminergic system on N/OFQ-induced feed intake in 3-h feed-deprived neonatal broilers. 2. In experiment 1, chicken received intracerebroventricular (ICV) injections of a control solution, SCH 23 390 (D1 receptors antagonist, 5 nmol), N/OFQ (16 nmol) or their combination (SCH23 390 + N/OFQ). In experiment 2, a control solution, AMI-193 (D2 receptors antagonist, 5 nmol), N/OFQ (16 nmol) or their combination (AMI-193 + N/OFQ) were ICV injected into chickens. In experiment 3, birds received ICV injections of a control solution, NGB2904 (D3 receptors antagonist, 6.4 nmol), N/OFQ (16 nmol) and co-injection of NGB2904 + N/OFQ. In experiment 4, ICV injections of the control solution, L-741,742 (D4 receptors antagonist, 6 nmol), N/OFQ (16 nmol) or their combination (L-741,742 + N/OFQ) were applied to broilers. In experiment 5, birds were ICV injected with control solution, L-DOPA (dopamine precursor, 125 nmol), N/OFQ (16 nmol) and L-DOPA + N/OFQ. Cumulative feed intake was recorded until 120 min after injection. 3. According to the results, ICV injection of N/OFQ significantly increased feed intake (P < 0.05). Co-injection of N/OFQ and D1 receptor antagonist (SCH 23390) amplified hyperphagic effect of N/OFQ (P < 0.05). The N/OFQ-induced feed intake was increased by the D2 receptor antagonist (P < 0.05). The hyperphagic effect of N/PFQ was weakened by co-injection of L-DOPA + N/OFQ (P < 0.05). 4. These results suggested that an interaction exists between dopamine and N/OFQ via D1 and D2 receptors on central feed intake in neonatal broiler chickens.


Assuntos
Estimulantes do Apetite/farmacologia , Galinhas/fisiologia , Comportamento Alimentar/efeitos dos fármacos , Peptídeos Opioides/farmacologia , Ração Animal , Animais , Animais Recém-Nascidos/fisiologia , Estimulantes do Apetite/administração & dosagem , Benzazepinas/administração & dosagem , Injeções Intraventriculares/veterinária , Peptídeos Opioides/administração & dosagem , Nociceptina
6.
Drug Dev Ind Pharm ; 43(10): 1694-1702, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28589736

RESUMO

AIM: In this study, self-emulsifying drug delivery system (SEDDS) for oral delivery of opioid peptide dalargin were developed and characterized in vitro. METHODS: Dalargin lipophilicity was increased by O-esterification of tyrosine OH group, hydrophobic ion pairing, or a combination thereof. Distribution coefficients (log D) of lipidized dalargin derivatives were determined. Then, dalargin was incorporated in chosen SEDDS, namely SEDDS-1, composed of 50% Capmul 907, 40% Cremophor EL, and 10% propylene glycol and comparatively more lipophilic SEDDS-2 composed of 30% Captex 8000, 30% Capmul MCM, 30% Cremophor EL, and 10% propylene glycol. Additionally, SEDDS were characterized regarding droplet size, polydispersity index (PDI), cloudy point, physical stability and stability against pancreatic lipase. Furthermore, mucus permeating properties of SEDDS and their ability to protect the incorporated dalargin against proteolysis by trypsin, α-chymotrypsin, elastase, simulated gastric fluid (SGF), and simulated intestinal fluid (SIF) were evaluated. RESULTS: The highest dalargin drug payload of 4.57% in SEDDS-2 was achieved when dalargin palmitate (pDAL) was ion paired with sodium dodecyl sulfate (SDS) in molar ratio 1:1. Moreover, SEDDS-1 and SEDDS-2 had a narrow droplet size distribution with average droplet sizes of 42.1 and 33.1 nm with PDI of 0.042 and 0.034, respectively. Lipolysis study showed that within 30 min 78.5% of SEDDS-1 and 92.1% of SEDDS-2 were digested. In addition, both SEDDS exhibited mucus permeating properties as well as a protective effect against enzymatic degradation by trypsin, α-chymotrypsin, elastase, SGF and SIF. CONCLUSION: The results of this study suggest that the developed SEDDS could be considered for oral opioid peptide delivery.


Assuntos
Caprilatos/química , Quimotripsina/química , Sistemas de Liberação de Medicamentos/métodos , Emulsões/química , Glicerídeos/química , Lipídeos/química , Muco/química , Peptídeos Opioides/química , Polietilenoglicóis/química , Propilenoglicol/química , Administração Oral , Disponibilidade Biológica , Peptídeos Opioides/administração & dosagem , Peptídeos Opioides/farmacologia , Solubilidade
7.
Eur J Neurosci ; 43(11): 1499-508, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27038234

RESUMO

Pathological chronic pain, which is frequently associated with prolonged tissue damage, inflammation, tumour invasion, and neurodegenerative diseases, gives rise to hyperalgesia and allodynia. We previously reported that intrathecal administration of nociceptin/orphanin FQ (N/OFQ), an endogenous ligand for the orphan opioid receptor-like receptor, in the femtomole range induces touch-evoked allodynia. N/OFQ has been implicated in multiple signalling pathways, such as inhibition of cAMP production and Ca(2+) channels, or activation of K(+) channels and mitogen-activated protein kinase, although the signalling pathways of N/OFQ-induced allodynia remain unclear. To address these issues, we developed an ex vivo mitogen-activated protein kinase assay by using intact slices of mouse spinal cord. N/OFQ markedly increased the phosphorylation of c-Jun N-terminal kinase (JNK) in the superficial dorsal horn of the spinal cord. The N/OFQ-stimulated JNK phosphorylation was significantly inhibited by pertussis toxin, the phospholipase C inhibitor U73122, and the inositol trisphosphate receptor antagonist Xestospongin C. Intrathecal administration of the JNK inhibitor SP600125 inhibited N/OFQ-evoked allodynia. The N/OFQ-induced increase in JNK phosphorylation was observed in astrocytes that expressed glial fibrillary acidic protein. N/OFQ also induced monocyte chemoattractant protein-1 (MCP-1) release via the JNK pathway, and N/OFQ-induced JNK phosphorylation was observed in MCP-1-immunoreactive astrocytes. Intrathecal administration of the MCP-1 receptor antagonist RS504393 inhibited N/OFQ-evoked allodynia. These results suggest that, in the spinal dorsal horn, N/OFQ induces allodynia through activation of JNK via the phospholipase C-inositol trisphosphate pathway, which is coupled to pertussis toxin-sensitive G-protein, and following the release of MCP-1 from astrocytes.


Assuntos
Quimiocina CCL2/metabolismo , Hiperalgesia/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Peptídeos Opioides/administração & dosagem , Medula Espinal/metabolismo , Animais , Astrócitos/metabolismo , Hiperalgesia/induzido quimicamente , Injeções Espinhais , Camundongos , Fosforilação , Transdução de Sinais , Nociceptina
8.
Bioorg Med Chem ; 22(14): 3703-12, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24878361

RESUMO

Branched peptides have been found to be useful in several research fields however their synthesis and purification is complicated. Here we present a novel and facile synthesis of tetra branched derivatives of nociceptin/orphanin FQ (N/OFQ). Three N/OFQ tetra branched derivatives were prepared using novel cores (PWT1, PWT2 and PWT3) containing a maleimido moiety. [Cys(18)]N/OFQ-NH2 was linked to the cores via thiol-Michael reaction characterized by high yield and purity of the desired final product. In the electrically stimulated mouse vas deferens PWT-N/OFQ derivatives mimicked the inhibitory action of the natural sequence showing similar maximal effects and 3 fold higher potencies. The NOP selective antagonist SB-612111 antagonized the effects of N/OFQ and PWT derivatives with similar pKB values (8.02-8.48). In vivo after supraspinal administration PWT2-N/OFQ stimulated food intake in mice mimicking the action of N/OFQ. Compared to the natural peptide PWT2-N/OFQ was 40 fold more potent and elicited larger effects. These findings suggest that the PWT chemical strategy can be successfully applied to biologically active peptides to generate, with unprecedented high purity and yield, tetra branched derivatives displaying an in vitro pharmacological profile similar to that of the natural sequence associated, in vivo, to increased potency and effectiveness.


Assuntos
Ingestão de Alimentos/efeitos dos fármacos , Peptídeos Opioides/farmacologia , Receptores Opioides/agonistas , Animais , Relação Dose-Resposta a Droga , Estimulação Elétrica , Injeções Intraventriculares , Ligantes , Masculino , Camundongos , Conformação Molecular , Peptídeos Opioides/administração & dosagem , Peptídeos Opioides/química , Relação Estrutura-Atividade , Receptor de Nociceptina , Nociceptina
10.
Mol Med ; 19: 7-17, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23348513

RESUMO

It is well known that genotypic differences can account for the subject-specific responses to opiate administration. In this regard, the basal activity of the endogenous system (either at the receptor or ligand level) can modulate the effects of exogenous agonists as morphine and vice versa. The µ opioid receptor from zebrafish, dre-oprm1, binds endogenous peptides and morphine with similar affinities. Morphine administration during development altered the expression of the endogenous opioid propeptides proenkephalins and proopiomelanocortin. Treatment with opioid peptides (Met-enkephalin [Met-ENK], Met-enkephalin-Gly-Tyr [MEGY] and ß-endorphin [ß-END]) modulated dre-oprm1 expression during development. Knocking down the dre-oprm1 gene significantly modified the mRNA expression of the penk and pomc genes, thus indicating that oprm1 is involved in shaping penk and pomc expression. In addition, the absence of a functional oprm1 clearly disrupted the embryonic development, since proliferation was disorganized in the central nervous system of oprm1-morphant embryos: mitotic cells were found widespread through the optic tectum and were not restricted to the proliferative areas of the mid- and hindbrain. Transferase-mediated dUTP nick-end labeling (TUNEL) staining revealed that the number of apoptotic cells in the central nervous system (CNS) of morphants was clearly increased at 24-h postfertilization. These findings clarify the role of the endogenous opioid system in CNS development. Our results will also help unravel the complex feedback loops that modulate opioid activity and that may be involved in establishing a coordinated expression of both receptors and endogenous ligands. Further knowledge of the complex interactions between the opioid system and analgesic drugs will provide insights that may be relevant for analgesic therapy.


Assuntos
Analgésicos Opioides/administração & dosagem , Regulação da Expressão Gênica , Rim/metabolismo , Morfina/administração & dosagem , Peptídeos Opioides/administração & dosagem , Receptores Opioides mu/metabolismo , Peixe-Zebra/metabolismo , Analgésicos Opioides/metabolismo , Animais , Feminino , Células HEK293 , Humanos , Rim/citologia , Rim/embriologia , Morfina/metabolismo , Peptídeos Opioides/metabolismo , Gravidez , Receptores Opioides mu/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética
11.
Pharmacol Res ; 70(1): 72-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23353033

RESUMO

Nociceptin/orphanin FQ (N/OFQ) and nociceptin orphanin peptide (NOP) receptors represent an endogenous system modulating gastrointestinal functions and inflammation. We investigated the peripheral effect of N/OFQ and of UFP-101, the NOP antagonist, in a model of colitis induced by TNBS (2,4,6 trinitrobenzenesulphonic acid; 60mg/kg). Male rats received two intraperitoneal injections per day of N/OFQ, UFP-101 or saline for 3 days after colitis induction. Four days after TNBS, animals were sacrificed and colonic histological damage, myeloperoxidase (MPO) activity and cytokine (IL-1ß and IL-10) levels were evaluated. N/OFQ plasmatic levels were assessed by radioimmunoassay. TNBS increased all the inflammatory variables considered. In colitic rats, N/OFQ (0.02 and 0.2nmol/kg) improved microscopic damage, MPO activity and decreased IL-1ß levels in comparison with TNBS group, whereas at the highest dose (20nmol/kg) the peptide worsened colitis. UFP-101 at the dose of 1nmol/kg, without pharmacological activity, antagonised the protective effect of N/OFQ (0.2nmol/kg) on colitis, but at a dose level of 3 and 10nmol/kg worsened inflammation, revealing the endogenous N/OFQergic system protective role. N/OFQ plasmatic levels were not modified in TNBS-treated rats compared with controls, whereas they were reduced in rats treated with the doses of UFP-101 aggravating colitis. In conclusion, peripheral low doses of N/OFQ have a beneficial effect on colonic inflammation in rats. In contrast, N/OFQ at a dose 100-1000-fold higher than those that protect worsens colitis, probably through different mechanisms. The peripheral N/OFQergic system can represent a new field of investigation in some intestinal inflammatory conditions.


Assuntos
Colite/metabolismo , Colite/prevenção & controle , Peptídeos Opioides/farmacologia , Receptores Opioides/metabolismo , Animais , Colite/induzido quimicamente , Colite/patologia , Citocinas/imunologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Masculino , Peptídeos Opioides/administração & dosagem , Peptídeos Opioides/antagonistas & inibidores , Peptídeos Opioides/sangue , Ligação Proteica , Radioimunoensaio , Ratos , Ratos Wistar , Ácido Trinitrobenzenossulfônico/farmacologia , Receptor de Nociceptina , Nociceptina
12.
Neurol Sci ; 33(6): 1233-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22240716

RESUMO

Nocistatin and nociceptin/orphanin FQ (N/OFQ) are two neuropeptides which may have opposite effects in several biological functions but their neuro-anatomical sites of interaction are not fully clear. We investigated interaction between the effect of intracerebroventricular (i.c.v.) injection of nocistatin and N/OFQ, on c-Fos expression in the mouse thalamus, using c-Fos immunohistochemistry. We found that co-injection of nocistatin with N/OFQ significantly modulates c-Fos expression in the thalamus. The present study strongly suggests that "Nocistatin-Nociceptin" interaction system in the thalamus may be the promising neuromodulatory sites in the investigation of unlocking their possible therapeutic circuit in nociception, memory and anxiety.


Assuntos
Regulação da Expressão Gênica , Genes fos/fisiologia , Peptídeos Opioides/administração & dosagem , Tálamo/metabolismo , Animais , Genes fos/efeitos dos fármacos , Humanos , Injeções Intraventriculares , Camundongos , Peptídeos Opioides/fisiologia , Tálamo/efeitos dos fármacos , Nociceptina
13.
J Neurophysiol ; 105(3): 1179-84, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21191090

RESUMO

Targeting the melanin-concentrating hormone (MCH) system has been suggested as a potential treatment for obesity, anxiety disorders, as well as addiction. Despite the therapeutic potential of MCH agonists and antagonists, the endogenous factors regulating MCH activity, in particular those implicated in anxiety and reward, are ill-defined. The present study investigated the cellular effects of nociceptin/orphanin FQ (N/OFQ), an endogenous opioid with anxiolytic and antireward properties, on MCH neurons. We found that N/OFQ induced a concentration-dependent reversible outward current in MCH neurons (EC(50) = 50.7 nM), an effect that was blocked by the competitive antagonist of the nociceptin opioid peptide (NOP) receptor UFP-101. N/OFQ-induced outward currents persisted in TTX, reversed near the potassium equilibrium potential, and displayed inward rectification, suggesting direct postsynaptic potassium channel activation. Tertiapin-Q completely abolished the N/OFQ effect, whereas glibenclamide did not, implicating protein G-dependent inwardly rectifying potassium (GIRK) and not ATP-sensitive potassium (K(ATP)) channels as the effector ion channel. The N/OFQ-induced outward current desensitized during repeated applications and occluded the inhibitory effect of dynorphin, suggesting that dynorphin and N/OFQ activate the same pathway. N/OFQ also reversibly inhibited voltage-gated calcium currents in MCH neurons. In conclusion, our study indicates N/OFQ as a robust endogenous regulator of MCH neurons, which may play a role in anxiety and drug addiction.


Assuntos
Encéfalo/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Hormônios/metabolismo , Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Inibição Neural/fisiologia , Neurônios/fisiologia , Peptídeos Opioides/administração & dosagem , Hormônios Hipofisários/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Masculino , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Nociceptina
14.
Horm Behav ; 60(5): 540-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21872598

RESUMO

Sexual receptivity, lordosis, can be induced by sequential estradiol and progesterone or extended exposure to high levels of estradiol in the female rat. In both cases estradiol initially inhibits lordosis through activation of ß-endorphin (ß-END) neurons of the arcuate nucleus of the hypothalamus (ARH) that activate µ-opioid receptors (MOP) in the medial preoptic nucleus (MPN). Subsequent progesterone or extended estradiol exposure deactivates MPN MOP to facilitate lordosis. Opioid receptor-like receptor-1 (ORL-1) is expressed in ARH and ventromedial hypothalamus (VMH). Infusions of its endogenous ligand, orphanin FQ (OFQ/N, aka nociceptin), into VMH-ARH region facilitate lordosis. Whether OFQ/N acts in ARH and/or VMH and whether OFQ/N is necessary for steroid facilitation of lordosis are unclear. In Exp I, OFQ/N infusions in VMH and ARH that facilitated lordosis also deactivated MPN MOP indicating that OFQ/N facilitation of lordosis requires deactivation of ascending ARH-MPN projections by directly inhibiting ARH ß-END neurons and/or through inhibition of excitatory VMH-ARH pathways to proopiomelanocortin neurons. It is unclear whether OFQ/N activates the VMH output motor pathways directly or via the deactivation of MPN MOP. In Exp II we tested whether ORL-1 activation is necessary for estradiol-only or estradiol+progesterone lordosis facilitation. Blocking ORL-1 with UFP-101 inhibited estradiol-only lordosis and MPN MOP deactivation but had no effect on estradiol+progesterone facilitation of lordosis and MOP deactivation. In conclusion, steroid facilitation of lordosis inhibits ARH ß-END neurons to deactivate MPN MOP, but estradiol-only and estradiol+progesterone treatments appear to use different neurotransmitter systems to inhibit ARH-MPN signaling.


Assuntos
Hipotálamo/efeitos dos fármacos , Peptídeos Opioides/fisiologia , Postura/fisiologia , Comportamento Sexual Animal/fisiologia , Animais , Estradiol/administração & dosagem , Feminino , Hipotálamo/fisiologia , Masculino , Peptídeos Opioides/administração & dosagem , Peptídeos Opioides/antagonistas & inibidores , Peptídeos Opioides/farmacologia , Área Pré-Óptica , Progesterona/administração & dosagem , Ratos , Ratos Long-Evans , Receptores Opioides mu/fisiologia , Comportamento Sexual Animal/efeitos dos fármacos , Nociceptina
15.
J Med Chem ; 63(5): 2673-2687, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-31834798

RESUMO

Four novel fluorinated cyclic analogues of biphalin with excellent to modest binding affinity for µ-, δ-, and κ-receptors were synthesized. The cyclic peptides have a combination of piperazine or hydrazine linker with or without a xylene bridge. Among the ligands, MACE3 demonstrated a better activity than biphalin after intravenous administration, and its corresponding analogue incorporating the hydrazine linker (MACE2) was able to induce longer lasting analgesia following subcutaneous administration. An analogue of MACE2 containing 2,6-dimethyl-l-tyrosine (MACE4) showed the best potency and in vivo antinociceptive activity of this series.


Assuntos
Analgésicos Opioides/uso terapêutico , Peptídeos Opioides/uso terapêutico , Dor/tratamento farmacológico , Peptídeos Cíclicos/uso terapêutico , Administração Intravenosa , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/farmacologia , Animais , Células CHO , Cricetulus , Feminino , Humanos , Infusões Subcutâneas , Masculino , Camundongos , Modelos Moleculares , Peptídeos Opioides/administração & dosagem , Peptídeos Opioides/farmacocinética , Peptídeos Opioides/farmacologia , Dor/metabolismo , Peptídeos Cíclicos/administração & dosagem , Peptídeos Cíclicos/farmacocinética , Peptídeos Cíclicos/farmacologia , Receptores Opioides/metabolismo
16.
Regul Pept ; 154(1-3): 32-8, 2009 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-19046996

RESUMO

The participation of hypothalamus-pituitary-adrenal axis in the gastroprotective effects of nociceptin/orphanin FQ (N/OFQ) has been investigated. Gastric mucosal lesions were induced by intragastric administration of 50% ethanol, 1 ml/rat. Rats received N/OFQ either by the intracerebroventricular (icv) route, at 3 microg/rat, or by the intraperitoneal (ip) route, at 10 microg/kg, 30 min before ethanol administration. The protective effect of icv and ip administered N/OFQ was assessed in adrenalectomized rats and in rats pretreated with the glucocorticoid receptor antagonist, mifepristone, or with the CRF receptor antagonist, alpha-helical CRF(9-41). The damaging effect of ethanol was apparently not influenced by adrenalectomy. N/OFQ markedly reduced macroscopically and histologically assessed gastric mucosal damage. The extent of reduction by N/OFQ was comparable in adrenalectomized and in sham-operated rats, with either icv or ip route of administration. Pretreatment with mifepristone, both icv (80 microg/rat) and ip (10 mg/kg) injected, did not modify the response to icv and ip N/OFQ. Pretreatment with alpha-helical CRF(9-41) (25 microg/rat icv or 250 microg/kg ip), had no effect on the reduction of gastric damage produced by icv or ip N/OFQ. Present findings suggest that the gastroprotective effects of N/OFQ on ethanol-induced damage do not involve the endocrine pathway through the HPA axis.


Assuntos
Sistema Hipotálamo-Hipofisário/metabolismo , Peptídeos Opioides/farmacologia , Sistema Hipófise-Suprarrenal/metabolismo , Gastropatias/patologia , Gastropatias/prevenção & controle , Adrenalectomia , Animais , Hormônio Liberador da Corticotropina/antagonistas & inibidores , Relação Dose-Resposta a Droga , Etanol/toxicidade , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/patologia , Injeções Intraperitoneais , Injeções Intraventriculares , Masculino , Mifepristona/administração & dosagem , Peptídeos Opioides/administração & dosagem , Fragmentos de Peptídeos/antagonistas & inibidores , Ratos , Ratos Wistar , Receptores de Glucocorticoides/antagonistas & inibidores , Gastropatias/induzido quimicamente , Fatores de Tempo , Nociceptina
17.
Pharmacol Biochem Behav ; 91(3): 436-40, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18804120

RESUMO

Nociceptin, also known as orphanin FQ, is a opioid-like neuropeptide that mediates its effects at the nociceptin receptor, a member of the G protein-coupled receptor superfamily. In mammals, nociceptin produces analgesia after spinal administration, however the role of nociceptin and nociceptin receptors in the modulation of noxious stimuli in non-mammalian species has not been examined. In an amphibian pain model using the acetic acid test with Rana pipiens, nociceptin and nociceptin1-13 amide produced dose-dependent antinociception (1-100 nmol), blocked by the nociceptin antagonist, [Nphe1]-nociceptin1-13 amide (30 nmol), but not the opioid antagonist, naltrexone (100 nmol/g, s.c.). Conversely, the antinociceptive effects of micro, delta, and kappa opioid receptor agonists were not blocked by the nociceptin antagonist. Nociceptin and nociceptin1-13 amide were the least potent of the opioid agonists tested. These studies demonstrate that spinal nociceptin receptors and not opioid receptors mediate the antinociceptive effect of nociceptin. Considered with previous findings, these behavioral data supports a role for nociceptin inhibition of spinal nociception in amphibians and perhaps all vertebrates.


Assuntos
Analgésicos , Peptídeos Opioides/farmacologia , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Ácido Acético , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Interações Medicamentosas , D-Penicilina (2,5)-Encefalina/farmacologia , Fentanila/farmacologia , Injeções Espinhais , Naltrexona/farmacologia , Peptídeos Opioides/administração & dosagem , Limiar da Dor/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Rana pipiens , Nociceptina
18.
Eur J Pharmacol ; 856: 172396, 2019 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-31103632

RESUMO

Addiction to conventional opioid pain analgesics is a major societal problem that is increasing at an alarming rate. New drugs to combat the effects of opioid abuse are desperately needed. Kappa-opioid agonists are efficacious in peripheral pain models but suffer from centrally-mediated effects. In this article, we discuss our efforts in developing peripheral kappa-based opioid receptor agonists that have the potential analgesic activity of opioids but do not manifest the negative side-effects of opioid use and abuse. Further, derivatives of the tetra-peptide D-Phe-D-Phe-D-Nle-D-Arg-NH2, such as CR665, exhibit high peripheral to central selectivity in analgesic models when administered intravenously (i.v.); however, they are inactive when administered orally. Application of our laboratory's proprietary non-natural amino acid technology to CR665 produced derivatives that exhibit peripheral analgesic activity when dosed orally but do not promote CNS-based effects. Lead compound JT09 activates the kappa-opioid receptor with EC50s in the low nM range, while agonist selectivity for kappa over other peripheral opioid receptors was >33,400 fold. Results indicate that JT09 is approximately as efficacious as morphine in alleviating peripheral pain, while failing to produce undesired CNS-mediated activity. Additionally, JT09 did not promote other CNS-mediated effects associated with morphine (addiction, sedation, dysphoria, tolerance, addiction). Thus, we propose that JT09 has potential for development as a novel analgesic. PERSPECTIVE: This article presents data supporting the analgesic properties of an orally available, peripherally-restricted, kappa-opioid agonist for peripheral pain. A potential out-patient pharmaceutical that acts as efficacious as morphine in alleviating peripheral pain, while failing to produce undesired CNS-mediated effects, could help reduce the current health care burden associated with prescription opioids.


Assuntos
Analgésicos/farmacologia , Peptídeos Opioides/farmacologia , Dor/tratamento farmacológico , Receptores Opioides kappa/agonistas , Administração Oral , Analgésicos/administração & dosagem , Analgésicos/uso terapêutico , Analgésicos/toxicidade , Animais , Condicionamento Psicológico/efeitos dos fármacos , Locomoção/efeitos dos fármacos , Masculino , Peptídeos Opioides/administração & dosagem , Peptídeos Opioides/uso terapêutico , Peptídeos Opioides/toxicidade , Ratos , Ratos Sprague-Dawley
19.
Vitam Horm ; 111: 195-225, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31421701

RESUMO

The neuropeptide Oxytocin (ΟΤ) is involved as a neurohormone, a neurotransmitter, or a neuromodulator in an extensive range of central and peripheral effects, complex emotional and social human behaviors, memory and learning processes. It is implicated in homeostatic, neuroadaptive processes associated with stress responses and substance use via interactions with the hypothalamic-pituitary-adrenal (HPA) axis and the dopamine mesolimbic reward stress system. This chapter reviews the preclinical and clinical literature on the complicated relationships between endogenous and exogenous opioids and ΟΤ systems and attempts to highlight key findings to date on the effectiveness of intranasal OT administration to treat opioid use disorders. OΤ seems to attenuate, even inhibit, the development of opioid use disorders in preclinical models but is still under clinical research as a promising pharmacological agent in the treatment of opioid use related behaviors. Evidence suggests a role for OT as an adjunctive or stand-alone treatment of behavioral, cognitive and emotional deficits associated with substance use, which may be responsible for seeking behavior and relapse. The mechanisms by which oxytocin acts to reverse the neural substrates of these deficits, partially due to substance induced alterations of the endogenous OT system, and thus modify the behavioral response to substance use are discussed. Other clinically relevant issues are also discussed.


Assuntos
Analgésicos Opioides , Peptídeos Opioides , Ocitocina , Analgésicos Opioides/administração & dosagem , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Química Encefálica , Tolerância a Medicamentos/fisiologia , Humanos , Neurônios/química , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Peptídeos Opioides/administração & dosagem , Peptídeos Opioides/fisiologia , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Ocitocina/administração & dosagem , Ocitocina/efeitos dos fármacos , Ocitocina/fisiologia , Receptores de Ocitocina/antagonistas & inibidores , Síndrome de Abstinência a Substâncias/fisiopatologia
20.
J Pharmacol Exp Ther ; 326(3): 897-904, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18539652

RESUMO

Intracerebroventricular administration of the opioid-like peptide nociceptin/orphanin FQ (N/OFQ) produces bradycardia, hypotension, and diuresis in mice. We hypothesized that these responses are solely caused by selective activation of central N/OFQ peptide (NOP) receptors. To test this premise, we first examined whether i.c.v. N/OFQ produced dose-dependent diuretic and cardiovascular depressor responses in commercially available C57BL/6 mice. Next, using doses established in these studies, we examined the renal excretory and cardiovascular responses to i.c.v. N/OFQ in conscious transgenic NOP receptor knockout mice (NOP(-/-)). In metabolic studies, i.c.v. N/OFQ, but not saline vehicle, dose-dependently increased urine output (V) in NOP(+/+); this response was significant at 3 nmol (N/OFQ, V = 0.39 +/- 0.10 ml/2 h; saline, 0.08 +/- 0.05 ml/2 h). The N/OFQ-evoked diuresis was absent in littermate NOP(-/-) (N/OFQ, V = 0.06 +/- 0.06 ml/2 h; saline, 0.03 +/- 0.03 ml/2 h). There were no significant changes in urinary sodium or potassium excretion or free water clearance in either group. In telemetry studies, i.c.v. N/OFQ dose dependently lowered heart rate (HR) and mean arterial pressure (MAP). At 3 nmol N/OFQ, both HR and MAP were reduced in NOP(+/+) (peak DeltaHR = -217 +/- 31 bpm; peak DeltaMAP =-47 +/- 7 mm Hg) compared with saline (peak DeltaHR =-14 +/- 5 bpm; peak DeltaMAP = 2 +/- 3 mm Hg). These N/OFQ-evoked bradycardic and hypotensive responses were absent in NOP(-/-) (peak DeltaHR =-13 +/- 17 bpm; peak DeltaMAP =-2 +/- 4 mm Hg, respectively). Basal 24-h cardiovascular and renal excretory function were not different between NOP(-/-) and NOP(+/+) mice. These results establish that the bradycardia, hypotension and diuresis produced by centrally administered N/OFQ are mediated by selective activation of NOP receptors.


Assuntos
Bradicardia/metabolismo , Diurese/fisiologia , Hipotensão/metabolismo , Peptídeos Opioides/administração & dosagem , Peptídeos Opioides/deficiência , Receptores Opioides/metabolismo , Animais , Bradicardia/induzido quimicamente , Diurese/efeitos dos fármacos , Hipotensão/induzido quimicamente , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Peptídeos Opioides/genética , Receptores Opioides/agonistas , Nociceptina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA