Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
Br J Pharmacol ; 180(17): 2298-2314, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37021779

RESUMO

BACKGROUND AND PURPOSE: In animal models of sepsis, increased activation of the Nociceptin/Orphanin FQ (N/OFQ) receptor NOP is associated with mortality and NOP antagonists improved survival. We have explored the role of the N/OFQ-NOP system in freshly isolated volunteer human B- and T-cells incubated with lipopolysaccharide (LPS) and peptidoglycan G (PepG) as a model of in vitro sepsis. EXPERIMENTAL APPROACH: B- and T-cell NOP expression was measured using the NOP fluorescent probe N/OFQATTO594 , N/OFQ content was measured using immunofluorescence, N/OFQ release was tracked using a CHOhNOPGαiq5 biosensor assay and NOP function was measured using transwell migration and cytokine/chemokine release using a 25-plex assay format. Cells were challenged with LPS/PepG. KEY RESULTS: CD19-positive B-cells bound N/OFQATTO594 ; they also contain N/OFQ. Stimulation with CXCL13/IL-4 increased N/OFQ release. N/OFQ trended to reduced migration to CXCL13/IL-4. Surface NOP expression was unaffected by LPS/PepG, but this treatment increased GM-CSF release in an N/OFQ sensitive manner. CD3-positive T-cells did not bind N/OFQATTO594 ; they did contain N/OFQ. Stimulation with CXCL12/IL-6 increased N/OFQ release. When incubated with LPS/PepG, NOP surface expression was induced leading to N/OFQATTO594 binding. In LPS/PepG-treated cells, N/OFQ reduced migration to CXCL12/IL-6. LPS/PepG increased GM-CSF release in an N/OFQ sensitive manner. CONCLUSIONS AND IMPLICATIONS: We suggest both a constitutive and sepsis-inducible N/OFQ-NOP receptor autocrine regulation of B- and T-cell function, respectively. These NOP receptors variably inhibit migration and reduce GM-CSF release. These data provide mechanistic insights to the detrimental role for increased N/OFQ signalling in sepsis and suggest a potential role for NOP antagonists as treatments.


Assuntos
Receptores Opioides , Sepse , Animais , Humanos , Receptores Opioides/metabolismo , Receptor de Nociceptina , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Lipopolissacarídeos/farmacologia , Interleucina-4 , Interleucina-6 , Peptídeos Opioides/fisiologia , Sepse/tratamento farmacológico , Nociceptina
2.
J Neurosci Res ; 100(1): 99-128, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34559903

RESUMO

Over the past several years, studies have highlighted the δ-opioid receptor (DOPr) as a promising therapeutic target for chronic pain management. While exhibiting milder undesired effects than most currently prescribed opioids, its specific agonists elicit effective analgesic responses in numerous animal models of chronic pain, including inflammatory, neuropathic, diabetic, and cancer-related pain. However, as compared with the extensively studied µ-opioid receptor, the molecular mechanisms governing its trafficking remain elusive. Recent advances have denoted several significant particularities in the regulation of DOPr intracellular routing, setting it apart from the other members of the opioid receptor family. Although they share high homology, each opioid receptor subtype displays specific amino acid patterns potentially involved in the regulation of its trafficking. These precise motifs or "barcodes" are selectively recognized by regulatory proteins and therefore dictate several aspects of the itinerary of a receptor, including its anterograde transport, internalization, recycling, and degradation. With a specific focus on the regulation of DOPr trafficking, this review will discuss previously reported, as well as potential novel trafficking barcodes within the opioid and nociceptin/orphanin FQ opioid peptide receptors, and their impact in determining distinct interactomes and physiological responses.


Assuntos
Dor Crônica , Receptores Opioides , Analgésicos/uso terapêutico , Analgésicos Opioides , Animais , Dor Crônica/tratamento farmacológico , Peptídeos Opioides/fisiologia , Receptores Opioides/fisiologia , Receptores Opioides mu
3.
Peptides ; 141: 170547, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33831447

RESUMO

This paper is the forty-second consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2019 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).


Assuntos
Peptídeos Opioides/farmacologia , Peptídeos Opioides/fisiologia , Receptores Opioides/fisiologia , Estresse Psicológico/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/etiologia , Analgésicos Opioides/farmacologia , Animais , Dor do Câncer/tratamento farmacológico , Dor do Câncer/genética , Dor Crônica/tratamento farmacológico , Etanol/farmacologia , Feminino , Humanos , Memória/efeitos dos fármacos , Memória/fisiologia , Dor Pós-Operatória/tratamento farmacológico , Gravidez , Receptores Opioides/agonistas , Comportamento Sexual/fisiologia , Status Social , Transtornos Relacionados ao Uso de Substâncias/genética
4.
Transl Res ; 234: 31-42, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33567346

RESUMO

Nociception and opioid antinociception in females are pliable processes, varying qualitatively and quantitatively over the reproductive cycle. Spinal estrogenic signaling via membrane estrogen receptors (mERs), in combination with multiple other signaling molecules [spinal dynorphin, kappa-opioid receptors (KOR), glutamate and metabotropic glutamate receptor 1 (mGluR1)], appears to function as a master coordinator, parsing functionality between pronociception and antinociception. This provides a window into pharmacologically accessing intrinsic opioid analgesic/anti-allodynic systems. In diestrus, membrane estrogen receptor alpha (mERα) signals via mGluR1 to suppress spinal endomorphin 2 (EM2) analgesia. Strikingly, in the absence of exogenous opioids, interfering with this suppression in a chronic pain model elicits opioid anti-allodynia, revealing contributions of endogenous opioid(s). In proestrus, robust spinal EM2 analgesia is manifest but this requires spinal dynorphin/KOR and glutamate-activated mGluR1. Furthermore, spinal mGluR1 blockade in a proestrus chronic pain animal (eliminating spinal EM2 analgesia) exacerbates mechanical allodynia, revealing tempering by endogenous opioid(s). A complex containing mu-opioid receptor, KOR, aromatase, mGluRs, and mERα are foundational to eliciting endogenous opioid anti-allodynia. Aromatase-mERα oligomers are also plentiful, in a central nervous system region-specific fashion. These can be independently regulated and allow estrogens to act intracellularly within the same signaling complex in which they are synthesized, explaining asynchronous relationships between circulating estrogens and central nervous system estrogen functionalities. Observations with EM2 highlight the translational relevance of extensively characterizing exogenous responsiveness to endogenous opioids and the neuronal circuits that mediate them along with the multiplicity of estrogenic systems that concomitantly function in phase and out-of-phase with the reproductive cycle.


Assuntos
Analgesia , Estrogênios/fisiologia , Glutamatos/fisiologia , Nociceptividade/fisiologia , Peptídeos Opioides/fisiologia , Analgesia/métodos , Analgésicos/farmacologia , Animais , Sistema Nervoso Central/fisiologia , Dor Crônica/tratamento farmacológico , Dor Crônica/fisiopatologia , Feminino , Humanos , Masculino , Modelos Neurológicos , Receptores de Estrogênio/fisiologia , Receptores de Glutamato/fisiologia , Receptores Opioides/fisiologia , Pesquisa Translacional Biomédica
5.
Biomed Pharmacother ; 131: 110722, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32916536

RESUMO

Pain sensation is characterized as a complex experience, dependent on sensory processes as well as the activation of limbic brain areas involved in emotion, among them anterior insula. This cortical area is involved in the perception and response to painful stimuli. We investigated if this area contributes to antinociception produced by NSAIDs, and underlying mechanisms. We found that administration of NSAIDs into the anterior insular cortex in rats reduced mechanical and heat hyperalgesia produced by intraplantar injection of formalin, and this was attenuated by pre- or post-treatment with the opioid receptor antagonists, naloxone and CTOP, and the cannabinoid receptor (CB1) antagonist AM-251. These data support the concept that NSAID-evoked antinociception is mediated via descending endogenous opioid and cannabinoid systems inhibiting spinal paw withdrawal reflexes in rodents.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Córtex Cerebral/efeitos dos fármacos , Endocanabinoides/fisiologia , Nociceptividade/efeitos dos fármacos , Peptídeos Opioides/fisiologia , Analgesia , Animais , Córtex Cerebral/fisiologia , Masculino , Naloxona/farmacologia , Nociceptividade/fisiologia , Piperidinas/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Wistar , Somatostatina/análogos & derivados , Somatostatina/farmacologia
6.
J Orthop Surg Res ; 15(1): 266, 2020 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-32677997

RESUMO

BACKGROUND: Osteoarthritis is a mechanical abnormality characterized by chronic joint pain associated with degeneration of the articular cartilage, synovitis, and local inflammation, leading to loss of function and pain. A connection exists between the peripheral nervous system and inflammatory joint degeneration. The process by which inflammation is influenced by the nervous system is known as neuroinflammation. One of the neuropeptides involved in peripheral neuroinflammation is nociceptin, a peptide related to the opioid class of substances. Nociceptin has both pro- and anti-inflammatory effects. Some studies show that nociceptin can be measured in synovial fluid, while other studies have not been able to detect it. The presence of nociceptin in synovial fluid could imply a molecular role for the neuropeptide in the joint, both physiologically as well as pathophysiologically. The goal of this pilot study was to determine whether nociceptin was present in the synovial fluid of osteoarthritic knees. METHODS: Patients undergoing primary total knee arthroplasty were enrolled after Institutional Review Board approval was obtained. Synovial fluid was aspirated from patients' operative knee joints and blood samples were obtained. A commercially available enzyme Immunoassay kit was used to test for nociceptin. A linear mixed-effects model was developed to account for the repeated measurements and baseline covariates. Least squares (adjusted) means were derived from the model to compare the sample types and to compare subgroups. RESULTS: Twenty patients were included in this study. Nociceptin was detected in the synovial fluid and plasma of all patients. The mean concentration (± standard deviation) of nociceptin in synovial fluid was 28.7 ± 18.2 pg/ml. The mean concentration of nociceptin in plasma was 45.2 ± 24.3 pg/ml pre-procedure, and 40.1 ± 20.6 pg/ml post-tourniquet deflation. The nociceptin concentration in synovial fluid was significantly lower than the nociceptin concentration in plasma, both pre-procedure and post-tourniquet deflation (p = 0.002 and p = 0.016 respectively). The nociceptin concentration in both plasma and synovial fluid was significantly lower in females versus males (p = 0.012). CONCLUSION: We demonstrated that nociceptin is present in synovial fluid and plasma of patients undergoing total knee arthroplasty. This implies a potential role for nociceptin in modulating inflammation in osteoarthritis. TRIAL REGISTRATION: ClinicalTrials.gov , NCT02528916 . Retrospectively registered on August 19, 2015.


Assuntos
Artroplastia do Joelho , Peptídeos Opioides/análise , Peptídeos Opioides/fisiologia , Osteoartrite do Joelho/diagnóstico , Osteoartrite do Joelho/cirurgia , Líquido Sinovial/química , Idoso , Biomarcadores/análise , Biomarcadores/sangue , Feminino , Humanos , Inflamação , Masculino , Pessoa de Meia-Idade , Peptídeos Opioides/sangue , Osteoartrite do Joelho/metabolismo , Projetos Piloto , Caracteres Sexuais , Nociceptina
7.
Am J Chin Med ; 48(4): 793-811, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32420752

RESUMO

Acupuncture reduces pain by activating specific areas called acupoints on the patient's body. When these acupoints are fully activated, sensations of soreness, numbness, fullness, or heaviness called De qi or Te qi are felt by clinicians and patients. There are two kinds of acupuncture, manual acupuncture and electroacupuncture (EA). Compared with non-acupoints, acupoints are easily activated on the basis of their special composition of blood vessels, mast cells, and nerve fibers that mediate the acupuncture signals. In the spinal cord, EA can inhibit glial cell activation by down-regulating the chemokine CX3CL1 and increasing the anti-inflammatory cytokine interleukin-10. This inhibits P38 mitogen-activated protein kinase and extracellular signal-regulated kinase pathways, which are associated with microglial activation of the C-Jun N-terminal kinase signaling pathway and subsequent astrocyte activation. The inactivation of spinal microglia and astrocytes mediates the immediate and long-term analgesic effects of EA, respectively. A variety of pain-related substances released by glial cells such as the proinflammatory cytokines tumor necrosis factor [Formula: see text], interleukin-1[Formula: see text], interleukin-6, and prostaglandins such as prostaglandins E2 can also be reduced. The descending pain modulation system in the brain, including the anterior cingulated cortex, the periaqueductal gray, and the rostral ventromedial medulla, plays an important role in EA analgesia. Multiple transmitters and modulators, including endogenous opioids, cholecystokinin octapeptide, 5-hydroxytryptamine, glutamate, noradrenalin, dopamine, [Formula: see text]-aminobutyric acid, acetylcholine, and orexin A, are involved in acupuncture analgesia. Finally, the "Acupuncture [Formula: see text]" strategy is introduced to help clinicians achieve better analgesic effects, and a newly reported acupuncture method called acupoint catgut embedding, which injects sutures made of absorbable materials at acupoints to achieve long-term effects, is discussed.


Assuntos
Analgesia por Acupuntura , Eletroacupuntura , Neurotransmissores/fisiologia , Analgesia por Acupuntura/métodos , Pontos de Acupuntura , Hormônio Adrenocorticotrópico/fisiologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/diagnóstico por imagem , Encéfalo/fisiologia , Quimiocina CX3CL1/metabolismo , Citocinas/metabolismo , Dopamina/fisiologia , Ácido Glutâmico/fisiologia , Hemodinâmica , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Neuroglia/fisiologia , Norepinefrina/fisiologia , Peptídeos Opioides/fisiologia , Serotonina/fisiologia , Sincalida/fisiologia , Medula Espinal/citologia , Ácido gama-Aminobutírico/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Pharmacol Res ; 158: 104855, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32438036

RESUMO

Hemorphins are endogenous peptides, 4-10 amino acids long, belonging to the family of atypical opioid peptides released during the sequential cleavage of hemoglobin protein. Hemorphins have been shown to exhibit diverse therapeutic effects in both human and animal models. However, the precise cellular and molecular mechanisms involved in such effects remain elusive. In this review, we summarize and propose potential mechanisms based on studies that investigated the biological activity of hemorphins of different lengths on multiple therapeutic targets. Special emphasis is given to molecular events related to renin-angiotensin system (RAS), opioid receptors and insulin-regulated aminopeptidase receptor (IRAP). This review provides a comprehensive coverage of the molecular mechanisms that underpin the therapeutic potential of hemorphins. Furthermore, it highlights the role of various hemorphin residues in pathological conditions, which could be explored further for therapeutic purposes.


Assuntos
Peptídeos Opioides/fisiologia , Peptídeos Opioides/uso terapêutico , Animais , Humanos , Proteína Antagonista do Receptor de Interleucina 1/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Peptídeos Opioides/química , Receptores Opioides/efeitos dos fármacos , Sistema Renina-Angiotensina/efeitos dos fármacos
9.
JCI Insight ; 5(4)2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-32102987

RESUMO

IL-4 is a pleiotropic antiinflammatory cytokine, which can be neuroprotective after nervous system injury. The beneficial actions of IL-4 are thought to result from the blunting of action of inflammatory mediators, such as proinflammatory cytokines. Here, we demonstrate that IL-4 induces M2 macrophages to continuously produce opioid peptides and ameliorate pain. IL-4 application at injured nerves in mice shifted F4/80+ macrophages from the proinflammatory M1 to the antiinflammatory M2 phenotype, which synthesized opioid peptides (Met-enkephalin, ß-endorphin, and dynorphin A 1-17). These effects were accompanied by a long-lasting attenuation of neuropathy-induced mechanical hypersensitivity, beyond the IL-4 treatment. This IL-4-induced analgesia was decreased by opioid peptide antibodies and opioid receptor (δ, µ, κ) antagonists applied at injured nerves, which confirms the involvement of the local opioid system. The participation of M2 macrophages was supported by analgesia in recipient mice injected at injured nerves with F4/80+ macrophages from IL-4-treated donors. Together, IL-4-induced M2 macrophages at injured nerves produced opioid peptides, which activated peripheral opioid receptors to diminish pain. Fostering the opioid-mediated actions of intrinsic M2 macrophages may be a strategy to tackle pathological pain.


Assuntos
Analgesia , Interleucina-4/farmacologia , Macrófagos/efeitos dos fármacos , Peptídeos Opioides/biossíntese , Animais , Temperatura Alta , Interleucina-4/uso terapêutico , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/tratamento farmacológico , Peptídeos Opioides/fisiologia , Tempo de Reação/efeitos dos fármacos , Receptores de Interleucina-4/antagonistas & inibidores , Receptores de Interleucina-4/fisiologia
10.
Peptides ; 124: 170223, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31805297

RESUMO

This paper is the fortieth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2017 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).


Assuntos
Analgésicos Opioides/farmacologia , Peptídeos Opioides/farmacologia , Peptídeos Opioides/fisiologia , Receptores Opioides/metabolismo , Transtornos Relacionados ao Uso de Substâncias/etiologia , Animais , Dor do Câncer/tratamento farmacológico , Dor Crônica/tratamento farmacológico , Dor Crônica/genética , Ingestão de Alimentos/efeitos dos fármacos , Emoções , Feminino , Humanos , Aprendizagem/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Peptídeos Opioides/agonistas , Peptídeos Opioides/antagonistas & inibidores , Transtornos Relacionados ao Uso de Opioides/etiologia , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Gravidez , Estresse Fisiológico , Receptor de Nociceptina
11.
Vitam Horm ; 111: 247-279, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31421703

RESUMO

More than 40years ago, the endogenous opioids were first described. Their role as important neuromodulators of pain and their influence on a variety of neuroendocrine control systems within the central nervous system has been recognized. More recently, endogenous opioids and their receptor have been identified in a variety of reproductive and non-reproductive tissues outside the central nervous system. What role the opioid system plays in these peripheral tissues and organs is not completely understood and thus the subjects of current research. In the central nervous system, endogenous opioids inhibit pulsatile Gonadotropin Releasing Hormone (GnRH) release, affecting the release of gonadotropins from the pituitary, and thus mediating stress response within the central nervous-pituitary-gonadal axes in both women and men-Peripherally, endogenous opioids have been demonstrated to be present-among other organs-in the pancreas and in the ovary, where they are produced by granulosa cells and may influence oocyte maturation. In men, endogenous opioids play a role in sperm production within the testis. Opioid antagonists such as naltrexone have been used to restore cyclicity in women through improvement in insulin resistance, GnRH-pulsatility and hyperandrogenemia stemming from specific pathophysiological conditions such as hypothalamic amenorrhea, polycystic ovarian syndrome, hyperinsulinemia, ovarian hyperstimulation syndrome. Opioid antagonists have also been used to treat male sexual disorders and male infertility. In summary, endogenous opioids exert a variety of actions within the reproductive system which are reviewed in this chapter.


Assuntos
Analgésicos Opioides/farmacologia , Peptídeos Opioides/fisiologia , Reprodução/efeitos dos fármacos , Reprodução/fisiologia , Amenorreia/etiologia , Amenorreia/fisiopatologia , Animais , Endorfinas/fisiologia , Feminino , Humanos , Doenças Hipotalâmicas/complicações , Doenças Hipotalâmicas/fisiopatologia , Masculino , Peptídeos Opioides/antagonistas & inibidores , Ocitocina/fisiologia , Síndrome do Ovário Policístico/fisiopatologia , Gravidez , Prolactina/fisiologia , Receptores Opioides/fisiologia
12.
Biol Sex Differ ; 10(1): 9, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30755252

RESUMO

BACKGROUND: Orphanin FQ (aka nociceptin; N/OFQ) binds to its nociceptin opioid peptide (NOP) receptor expressed in proopiomelanocortin (POMC) neurons within the arcuate nucleus (ARC), a critical anorexigenic component of the hypothalamic energy balance circuitry. It inhibits POMC neurons by modifying neuronal excitability both pre- and postsynaptically. We tested the hypothesis that N/OFQ inhibits neurotransmission at synapses involving steroidogenic factor (SF)-1 neurons in the ventromedial nucleus (VMN) and ARC POMC neurons in a sex- and diet-dependent fashion. METHODS: Electrophysiological recordings were done in intact male and in cycling and ovariectomized female NR5A1-Cre and eGFP-POMC mice. Energy homeostasis was assessed in wildtype animals following intra-ARC injections of N/OFQ or its saline vehicle. RESULTS: N/OFQ (1 µM) decreased light-evoked excitatory postsynaptic current (leEPSC) amplitude more so in males than in diestrus or proestrus females, which was further accentuated in high-fat diet (HFD)-fed males. N/OFQ elicited a more robust outward current and increase in conductance in males than in diestrus, proestrus, and estrus females. These pleiotropic actions of N/OFQ were abrogated by the NOP receptor antagonist BAN ORL-24 (10 µM). In ovariectomized female eGFP-POMC mice, 17ß-estradiol (E2; 100 nM) attenuated the N/OFQ-induced postsynaptic response. SF-1 neurons from NR5A1-Cre mice also displayed a robust N/OFQ-induced outward current and increase in conductance that was sexually differentiated and suppressed by E2. Finally, intra-ARC injections of N/OFQ increased energy intake and decreased energy expenditure, which was further potentiated by exposure to HFD and diminished by estradiol benzoate (20 µg/kg; s.c.). CONCLUSION: These findings show that males are more responsive to the pleiotropic actions of N/OFQ at anorexigenic VMN SF-1/ARC POMC synapses, and this responsiveness can be further enhanced under conditions of diet-induced obesity/insulin resistance.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Metabolismo Energético/fisiologia , Peptídeos Opioides/fisiologia , Pró-Opiomelanocortina/fisiologia , Fator Esteroidogênico 1/fisiologia , Transmissão Sináptica/fisiologia , Núcleo Hipotalâmico Ventromedial/fisiologia , Animais , Dieta , Feminino , Cobaias , Homeostase , Masculino , Neurônios/fisiologia , Obesidade/fisiopatologia , Caracteres Sexuais , Sinapses/fisiologia , Nociceptina
13.
Nutrients ; 11(1)2019 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-30621149

RESUMO

Opioid peptides released during digestion of dietary proteins such as casein, were suggested to contribute to autism development, leading to the announcement of opioid excess hypothesis of autism. This paper examines role of enzyme proline dipeptidyl peptidase-4 (DPPIV; EC 3.4.14.5) and it is exogenous substrate, ß-casomorphin-7 (BCM7) in autism etiology. Our study included measurements of DPPIV and BCM7 concentrations in serum and urine, which were analyzed with ELISA assays and activity of DPPIV was measured by colorimetric test. The effect of opioid peptides from hydrolysed bovine milk on DPPIV gene expression in peripheral blood mononuclear cells (PBMC) in autistic and healthy children was determined using the Real-Time PCR (Polymerase Chain Reaction) method. Our research included 51 healthy children and 86 children diagnosed with autism spectrum disorder (ASD, ICDF84). We determined that the concentration of BCM7 in serum was significantly, 1.6-fold, higher in the ASD group than in controls (p < 0.0001). Concentration of DPPIV was found to also be significantly higher in serum from ASD children compared to the control group (p < 0.01), while we did not notice significant difference in enzymatic activity of serum DPPIV between the two study groups. We confirmed correlation according to the gender between analyzed parameters. The inspiration for this study emanated from clinical experience of the daily diet role in relieving the symptoms of autism. Despite this, we have concluded that milk-derived opioid peptides and DPPIV are potentially factors in determining the pathogenesis of autism; conducted studies are still limited and require further research.


Assuntos
Transtorno do Espectro Autista/enzimologia , Dipeptidil Peptidase 4/fisiologia , Leite/química , Peptídeos Opioides/fisiologia , Animais , Transtorno do Espectro Autista/sangue , Transtorno do Espectro Autista/etiologia , Criança , Pré-Escolar , Dipeptidil Peptidase 4/sangue , Dipeptidil Peptidase 4/genética , Endorfinas/sangue , Endorfinas/farmacologia , Endorfinas/fisiologia , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Leucócitos Mononucleares/enzimologia , Masculino , Peptídeos Opioides/sangue , Peptídeos Opioides/urina , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/fisiologia , Prolina , Fatores Sexuais
14.
Lasers Surg Med ; 49(9): 844-851, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28671718

RESUMO

Low-level laser therapy (LLLT) is the direct application of light to stimulate cell responses (photobiomodulation) to promote tissue healing, reduce inflammation, and induce analgesia; the molecular basis for these effects of LLLT remains unclear. The objective of this study was to evaluate the analgesic effect of LLLT in the rat plantar incision model of postoperative pain as well as to investigate some of the possible mechanisms involved in this effect. Wistar rats were submitted to plantar incision and treated with LLLT (830 nm, continuous-mode, 30 mW/cm2 , 1-12 J/cm2 ). Postoperative thermal and mechanical hypersensitivity were monitored for 24 hours post-incision. In addition, the animals were pretreated with saline, naloxone (a nonselective opioid receptor antagonist; 20 µg/5 µl) or methysergide (5-HT2C , 5-HT2A , 5-HT7 , 5-HT5a , 5-HT6, and 5-HT1F receptors antagonist; 30 µg/5 µl). Moreover, 24 hours after incision and treatment, the TNF-α and IL-1ß levels in serum were evaluated. Our results demonstrate, for the first time, that LLLT at 3 or 8 J/cm2 , but not at 1-2, 4-7, or 9-12 J/cm2 , induced an analgesic effect on postoperative pain. Naloxone, but not methysergide, blocked the LLLT-induced anti-nociceptive effect. Additionally, IL-1-ß and TNF-α production significantly decreased after LLLT at 3 or 8 J/cm2 . Our results suggest that LLLT at 3 or 8 J/cm2 primarily modulates the endogenous opioids system and is not directly mediated by serotonergic receptors. Reduction of IL-1ß and TNF-α may play a role in the antinociceptive action of LLLT. Lasers Surg. Med. 49:844-851, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Terapia com Luz de Baixa Intensidade , Peptídeos Opioides/fisiologia , Limiar da Dor/efeitos da radiação , Dor Pós-Operatória/prevenção & controle , Animais , Citocinas/sangue , Modelos Animais de Doenças , Masculino , Metisergida , Naloxona , Antagonistas de Entorpecentes , Dor Pós-Operatória/etiologia , Dor Pós-Operatória/metabolismo , Ratos , Ratos Wistar , Antagonistas da Serotonina
15.
Neuropeptides ; 50: 51-6, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25812480

RESUMO

Nociceptin/Orphanin FQ (N/OFQ) is a 17-amino acid peptide that binds to the nociceptin receptor (NOP). N/OFQ and NOP receptors are expressed in numerous brain areas. The generation of specific agonists, antagonists and receptor-deficient mice or rats has enabled progress in elucidating the biological functions of N/OFQ. These tools have been employed to identify the biological significance of the N/OFQ system and how it interacts with other endogenous systems to regulate several body functions. The present review focuses on the role of N/OFQ in the regulation of body temperature and its relationship with energy balance. Critical evaluation of the literature data suggests that N/OFQ, acting through the NOP receptor, may cause hypothermia by influencing the complex thermoregulatory system that operates as a federation of independent thermoeffector loops to control body temperature at the hypothalamic level. Furthermore, N/OFQ counteracts hyperthermia elicited by cannabinoids or µ-opioid agonists. N/OFQ-induced hypothermia is prevented by ω-conotoxin GVIA, an N-type calcium channel blocker. Hypothermia induced by N/OFQ is considered within the framework of the complex action that this neuropeptide exerts on energy balance. Energy stores are regulated through the complex neural controls exerted on both food intake and energy expenditure. In laboratory rodents, N/OFQ stimulates consummatory behavior and decreases energy expenditure. Taken together, these studies support the idea that N/OFQ contributes to the regulation of energy balance by acting as an "anabolic" neuropeptide as it elicits effects similar to those produced in the hypothalamus by other neuropeptides such as orexins and neuropeptide Y.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Hipotálamo/fisiologia , Peptídeos Opioides/fisiologia , Animais , Química Encefálica , Citocinas/fisiologia , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Febre/fisiopatologia , Hiperfagia/fisiopatologia , Camundongos , Neuropeptídeos/fisiologia , Obesidade/fisiopatologia , Peptídeos Opioides/genética , Peptídeos Opioides/farmacologia , RNA Mensageiro/análise , Ratos , Receptores Opioides/genética , Receptores Opioides/fisiologia , Aumento de Peso/fisiologia , Receptor de Nociceptina , Nociceptina
16.
J Pharmacol Sci ; 125(2): 117-24, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24882143

RESUMO

Nicotine (NIC) is an exogenous ligand of the nicotinic acetylcholine receptor (nAChR), and it influences various functions in the central nervous system. Systemic administration of NIC elicits the release of endogenous opioids (endorphins, enkephalins, and dynorphins) in the supraspinal cord. Additionally, systemic NIC administration induces the release of methionine-enkephalin in the spinal dorsal horn. NIC has acute neurophysiological actions, including antinociceptive effects, and the ability to activate the hypothalamic-pituitary-adrenal (HPA) axis. The endogenous opioid system participates in NIC-induced antinociception, but not HPA axis activation. Moreover, NIC-induced antinociception is mediated by α4ß2 and α7 nAChRs, while NIC-induced HPA axis activation is mediated by α4ß2, not α7, suggesting that the effects of NIC on the endogenous opioid system are mediated by α7, not α4ß2. NIC has substantial physical dependence liability. The opioid-receptor antagonist naloxone (NLX) elicits NIC withdrawal after repeated NIC administration, and NLX-induced NIC withdrawal is inhibited by concomitant administration of an opioid-receptor antagonist. NLX-induced NIC withdrawal is also inhibited by concomitant administration of an α7 antagonist, but not an α4ß2 antagonist. Taken together, these findings suggest that NIC-induced antinociception and the development of physical dependence are mediated by the endogenous opioid system, via the α7 nAChR.


Assuntos
Analgésicos , Nicotina/farmacologia , Peptídeos Opioides/fisiologia , Tabagismo/etiologia , Receptor Nicotínico de Acetilcolina alfa7/fisiologia , Animais , Encefalina Metionina/metabolismo , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Naloxona/administração & dosagem , Naloxona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Nicotina/efeitos adversos , Peptídeos Opioides/metabolismo , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Medula Espinal/metabolismo , Síndrome de Abstinência a Substâncias , Tabagismo/prevenção & controle
17.
Nihon Arukoru Yakubutsu Igakkai Zasshi ; 49(5): 227-37, 2014 Oct.
Artigo em Japonês | MEDLINE | ID: mdl-25651617

RESUMO

Nicotine (NIC) regulates various cellular functions acting on the nicotinic acetylcholine receptor (nAChR). And nAChR consists of ligand-gated cation channels with pentameric structure and composed of α and ß subunits. In the central nervous system, α 4 ß 2 and α 7 nAChRs are the most abundantly expressed as nAChR subtypes. There are several lines of evidence indicating that systemic administration of NIC elicits the release of endogenous opioids, such as, endorphins, enkephalins and dynorphins, in the brain. NIC exerts numerous acute effects, for example, antinociceptive effects and the activating effects of the hypothalamic-pituitary-adrenal (HPA) axis. In these effects, NIC-induced antinociception, but not HPA axis activation, was inhibited by opioid receptor antagonist, naloxone (NLX), and was also suppressed in morphine tolerated mice, indicating the participation of the endogenous opioid system in NIC-induced antinociception, but not HPA axis activation. Moreover, NIC-induced antinociception was antagonized by both α 4 ß 2 and α 7 nAChR antagonists, while NIC-induced HPA axis activation was antagonized by α 4 ß 2 nAChR antagonist, but not by α 7 nAChR antagonist. These results suggest that the endogenous opioid system may not be located on the downstream of α 4 ß 2 nAChR. On the other hand, NIC has substantial physical dependence liability. NLX elicits NIC withdrawal after repeated NIC administration evaluated by corticosterone increase as a withdrawal sign, and NLX-precipitated NIC withdrawal is inhibited by concomitant administration of other opioid receptor antagonist, naltrexone, indicating the participation of endogenous opioid system in the development of physical dependence on NIC. NLX-precipitated NIC withdrawal was also inhibited by concomitant administration of an α 7 nAChR antagonist, but not an α 4 ß 2 nAChR antagonist. Taken together, these findings suggest that the endogenous opioid system may be located on the downstream of α 7 nAChR and participates in the development of physical dependence on NIC.


Assuntos
Nicotina/farmacologia , Peptídeos Opioides/fisiologia , Tabagismo/genética , Receptor Nicotínico de Acetilcolina alfa7/efeitos dos fármacos , Receptor Nicotínico de Acetilcolina alfa7/fisiologia , Analgésicos , Animais , Encéfalo/metabolismo , Dinorfinas/metabolismo , Dinorfinas/fisiologia , Endorfinas/metabolismo , Endorfinas/fisiologia , Encefalinas/metabolismo , Encefalinas/fisiologia , Humanos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Camundongos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Nicotina/antagonistas & inibidores , Nociceptividade/efeitos dos fármacos , Peptídeos Opioides/metabolismo , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Receptor Nicotínico de Acetilcolina alfa7/antagonistas & inibidores
18.
J Pharmacol Sci ; 124(1): 47-53, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24366190

RESUMO

We previously reported that nicotine (NIC)-induced analgesia was elicited in part by activation of the endogenous opioid system. Moreover, it is well known that NIC has physical-dependence liability, but its mechanism is unclear. Therefore, we examined whether physical dependence on NIC was mediated by activation of the endogenous opioid system in ICR mice. We evaluated increased serum corticosterone (SCS) as an indicator of NIC withdrawal, as it is a quantitative indicator of naloxone (opioid receptor antagonist, NLX)-precipitated morphine withdrawal in mice. In this study, NLX precipitated an SCS increase in mice receiving repeated NIC, by a dose-dependent mechanism, and correlated with the dose and number of days of repeated NIC administration. When an opioid receptor antagonist (naltrexone) was concomitantly administered with repeated NIC, the NLX-precipitated SCS increase was not elicited. Concomitant administration of the α7 nicotinic acetylcholine receptor (nAChR) antagonist (methyllycaconitine) with repeated NIC, but not the α4ß2 nAChR antagonist (dihydro-ß-erythroidine), did not elicit an SCS increase by NLX. Thus, a physical dependence on NIC was in part mediated by the activation of the endogenous opioid system, located downstream of α7 nAChR.


Assuntos
Nicotina/administração & dosagem , Peptídeos Opioides/genética , Peptídeos Opioides/fisiologia , Tabagismo/genética , Receptor Nicotínico de Acetilcolina alfa7/fisiologia , Aconitina/administração & dosagem , Aconitina/análogos & derivados , Aconitina/farmacologia , Animais , Biomarcadores/sangue , Corticosterona/sangue , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos ICR , Naloxona/administração & dosagem , Naloxona/farmacologia , Antagonistas de Entorpecentes , Síndrome de Abstinência a Substâncias/sangue , Síndrome de Abstinência a Substâncias/diagnóstico , Receptor Nicotínico de Acetilcolina alfa7/antagonistas & inibidores
19.
Neuropeptides ; 47(6): 439-50, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24210137

RESUMO

Dementia conditions and memory deficits of different origins (vascular, metabolic and primary neurodegenerative such as Alzheimer's and Parkinson's diseases) are getting more common and greater clinical problems recently in the aging population. Since the presently available cognitive enhancers have very limited therapeutical applications, there is an emerging need to elucidate the complex pathophysiological mechanisms, identify key mediators and novel targets for future drug development. Neuropeptides are widely distributed in brain regions responsible for learning and memory processes with special emphasis on the hippocampus, amygdala and the basal forebrain. They form networks with each other, and also have complex interactions with the cholinergic, glutamatergic, dopaminergic and GABA-ergic pathways. This review summarizes the extensive experimental data in the well-established rat and mouse models, as well as the few clinical results regarding the expression and the roles of the tachykinin system, somatostatin and the closely related cortistatin, vasoactive intestinal polypeptide (VIP) and pituitary adenylate-cyclase activating polypeptide (PACAP), calcitonin gene-related peptide (CGRP), neuropeptide Y (NPY), opioid peptides and galanin. Furthermore, the main receptorial targets, mechanisms and interactions are described in order to highlight the possible therapeutical potentials. Agents not only symptomatically improving the functional impairments, but also inhibiting the progression of the neurodegenerative processes would be breakthroughs in this area. The most promising mechanisms determined at the level of exploratory investigations in animal models of cognitive disfunctions are somatostatin sst4, NPY Y2, PACAP-VIP VPAC1, tachykinin NK3 and galanin GALR2 receptor agonisms, as well as delta opioid receptor antagonism. Potent and selective non-peptide ligands with good CNS penetration are needed for further characterization of these molecular pathways to complete the preclinical studies and decide if any of the above described targets could be appropriate for clinical investigations.


Assuntos
Encéfalo/fisiologia , Aprendizagem/fisiologia , Memória/fisiologia , Neuropeptídeos/fisiologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/fisiologia , Galanina/fisiologia , Humanos , Camundongos , Neuropeptídeo Y/fisiologia , Peptídeos Opioides/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Ratos , Somatostatina/fisiologia , Taquicininas/fisiologia , Peptídeo Intestinal Vasoativo/fisiologia
20.
Pharmacol Biochem Behav ; 110: 19-26, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23747933

RESUMO

This study aimed to assess the possible systemic antinociceptive activity of mangiferin and to clarify the underlying mechanism, using the acute models of chemical (acetic acid, formalin, and capsaicin) and thermal (hot-plate and tail-flick) nociception in mice. Mangiferin at oral doses of 10 to 100 mg/kg evidenced significant antinociception against chemogenic pain in the test models of acetic acid-induced visceral pain and in formalin- and capsaicin-induced neuro-inflammatory pain, in a naloxone-sensitive manner, suggesting the participation of endogenous opiates in its mechanism. In capsaicin test, the antinociceptive effect of mangiferin (30 mg/kg) was not modified by respective competitive and non-competitive transient receptor potential vanilloid 1 (TRPV1) antagonists, capsazepine and ruthenium red, or by pretreatment with L-NAME, a non-selective nitric oxide synthase inhibitor, or by ODQ, an inhibitor of soluble guanylyl cyclase. However, mangiferin effect was significantly reversed by glibenclamide, a blocker of K(ATP) channels and in animals pretreated with 8-phenyltheophylline, an adenosine receptor antagonist. Mangiferin failed to modify the thermal nociception in hot-plate and tail-flick test models, suggesting that its analgesic effect is only peripheral but not central. The orally administered mangiferin (10-100 mg/kg) was well tolerated and did not impair the ambulation or the motor coordination of mice in respective open-field and rota-rod tests, indicating that the observed antinociception was unrelated to sedation or motor abnormality. The findings of this study suggest that mangiferin has a peripheral antinociceptive action through mechanisms that involve endogenous opioids, K(ATP)-channels and adenosine receptors.


Assuntos
Trifosfato de Adenosina/fisiologia , Adenosina/fisiologia , Analgésicos/farmacologia , Modelos Animais de Doenças , Peptídeos Opioides/fisiologia , Dor/tratamento farmacológico , Canais de Potássio/fisiologia , Xantonas/farmacologia , Trifosfato de Adenosina/metabolismo , Analgésicos/uso terapêutico , Animais , Camundongos , Canais de Potássio/metabolismo , Xantonas/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA