Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 559
Filtrar
1.
Eur J Pharmacol ; 933: 175214, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-36007608

RESUMO

Opioids are one of the most effective anti-nociceptive agents used in patients with cancer pain or after serious surgery in most countries. The endogenous opioid system participates in pain perception, but recently its role in inflammation was determined. κ-opioid receptors (KOP receptors), a member of the opioid receptor family, are expressed in the central and peripheral nervous system as well as on the surface of different types of immune cells, e.g. T cells, B cells and monocytes. In this review, we focused on the involvement of KOP receptors in the inflammatory process and described their function in a number of conditions in which the immune system plays a key role (e.g. inflammatory bowel disease, arthritis, subarachnoid hemorrhage, vascular dysfunction) and inflammatory pain. We summed up the application of known KOP ligands in pathophysiology and we aimed to shed new light on KOP receptors as important elements during inflammation.


Assuntos
Analgésicos Opioides , Receptores Opioides kappa , Humanos , Sistema Imunitário , Inflamação , Receptores Opioides/fisiologia
2.
Adv Biol (Weinh) ; 6(9): e2200020, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35531616

RESUMO

Cancer is a significant public health problem worldwide. While there has been a steady decrease in the cancer death rate over the last two decades, the number of survivors has increased and, thus, cancer-related sequela. Pain affects the life of patients with cancer and survivors. Prescription opioids continue as the analgesic of choice to treat moderate-to-severe cancer-related pain. There has been controversy on whether opioids impact cancer progression by acting on cancer cells or the tumor microenvironment. The µ-opioid receptor is the site of action of prescription opioids. This receptor can participate in an important mechanism of cancer spread, such as perineural invasion. In this review, current evidence on the role of the µ-opioid receptor in cancer growth is summarized and preliminary evidence about its effect on the cross-talk between sensory neurons and malignant cells is provided.


Assuntos
Neoplasias , Receptores Opioides , Analgésicos/uso terapêutico , Analgésicos Opioides/uso terapêutico , Humanos , Dor/tratamento farmacológico , Receptores Opioides/fisiologia , Microambiente Tumoral
3.
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
4.
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
5.
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
6.
Fundam Clin Pharmacol ; 35(1): 174-183, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32564393

RESUMO

Fentanyl is a common sedative/analgesic used for intrathecal chemotherapy injection in children with acute leukemia. Given the contradictory findings that fentanyl has both inhibitory and stimulatory activities in cancer cells, we investigated the biological effects of fentanyl alone and its combination with standard of care in acute myeloid leukemia (AML) cells at all stages of development. We showed that fentanyl at clinically relevant concentration inhibited growth and colony formation of AML differentiated cells and committed progenitors without affecting their survival. Compared to AML cells without FLT3 mutation, cells harboring FLT3-ITD mutation are likely to be more sensitive to fentanyl. However, fentanyl did not affect the most primitive AML stem cells. Fentanyl significantly augmented the efficacy of cytarabine but not midostaurin in AML differentiated cells and committed progenitors. We further demonstrated that fentanyl inhibited AML cells via suppressing Ras/Raf/MEK/ERK and STAT5 pathway, and this was not dependent on opioid receptor system. Our findings demonstrate the anti-leukemia activity of fentanyl and synergistic effects between fentanyl and cytarabine in AML, via opioid receptor-independent suppression of Ras and STAT5 pathways. Our work is the first to suggest the beneficial effects of fentanyl in children with leukemia.


Assuntos
Fentanila/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Receptores Opioides/fisiologia , Fator de Transcrição STAT5/antagonistas & inibidores , Proteínas ras/antagonistas & inibidores , Linhagem Celular Tumoral , Humanos , Leucemia Mieloide Aguda/patologia , Transdução de Sinais/efeitos dos fármacos , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores
7.
Front Immunol ; 11: 300, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194554

RESUMO

Opioid receptors comprise µ (MOP), δ (DOP), κ (KOP), and nociceptin/orphanin FQ (NOP) receptors. Opioids are agonists of MOP, DOP, and KOP receptors, whereas nociceptin/orphanin FQ (N/OFQ) is an agonist of NOP receptors. Activation of all four opioid receptors in neurons can induce analgesia in animal models, but the most clinically relevant are MOP receptor agonists (e.g., morphine, fentanyl). Opioids can also affect the function of immune cells, and their actions in relation to immunosuppression and infections have been widely discussed. Here, we analyze the expression and the role of opioid receptors in peripheral immune cells and glia in the modulation of pain. All four opioid receptors have been identified at the mRNA and protein levels in immune cells (lymphocytes, granulocytes, monocytes, macrophages) in humans, rhesus monkeys, rats or mice. Activation of leukocyte MOP, DOP, and KOP receptors was recently reported to attenuate pain after nerve injury in mice. This involved intracellular Ca2+-regulated release of opioid peptides from immune cells, which subsequently activated MOP, DOP, and KOP receptors on peripheral neurons. There is no evidence of pain modulation by leukocyte NOP receptors. More good quality studies are needed to verify the presence of DOP, KOP, and NOP receptors in native glia. Although still questioned, MOP receptors might be expressed in brain or spinal cord microglia and astrocytes in humans, mice, and rats. Morphine acting at spinal cord microglia is often reported to induce hyperalgesia in rodents. However, most studies used animals without pathological pain and/or unconventional paradigms (e.g., high or ultra-low doses, pain assessment after abrupt discontinuation of chronic morphine treatment). Therefore, the opioid-induced hyperalgesia can be viewed in the context of dependence/withdrawal rather than pain management, in line with clinical reports. There is convincing evidence of analgesic effects mediated by immune cell-derived opioid peptides in animal models and in humans. Together, MOP, DOP, and KOP receptors, and opioid peptides in immune cells can ameliorate pathological pain. The relevance of NOP receptors and N/OFQ in leukocytes, and of all opioid receptors, opioid peptides and N/OFQ in native glia for pain control is yet to be clarified.


Assuntos
Leucócitos/fisiologia , Macrófagos/fisiologia , Neuroglia/fisiologia , Dor/fisiopatologia , Receptores Opioides/fisiologia , Animais , Humanos , Receptor de Nociceptina
8.
Front Immunol ; 11: 94, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32076421

RESUMO

The opioid family of GPCRs consists of the classical opioid receptors, designated µ-, κ-, and δ-opioid receptors, and the orphanin-FQ receptor, and these proteins are expressed on both neuronal and hematopoietic cells. A number of laboratories have reported that an important degree of cross-talk can occur between the opioid receptors and the chemokine and chemokine receptor families. As a part of this, the opioid receptors are known to regulate the expression of certain chemokines and chemokine receptors, including those that possess strong pro-inflammatory activity. At the level of receptor function, it is clear that certain members of the chemokine family can mediate cross-desensitization of the opioid receptors. Conversely, the opioid receptors are all able to induce heterologous desensitization of some of the chemokine receptors. Consequently, activation of one or more of the opioid receptors can selectively cross-desensitize chemokine receptors and regulate chemokine function. These cross-talk processes have significant implications for the inflammatory response, since the regulation of both the recruitment of inflammatory cells, as well as the sensation of pain, can be controlled in this way.


Assuntos
Receptores de Quimiocinas/fisiologia , Receptores Opioides/fisiologia , Animais , Humanos
9.
Vitam Horm ; 111: 147-193, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31421699

RESUMO

The pentapeptides methionine-enkephalin and leucine-enkephalin belong to the opioid family of peptides, and the non-opiate peptide adrenocorticotropin hormone (ACTH) to the melanocortin peptide family. Enkephalins/ACTH are derived from pro-enkephalin, pro-dynorphin or pro-opiomelanocortin precursors and, via opioid and melanocortin receptors, are responsible for many biological activities. Enkephalins exhibit the highest affinity for the δ receptor, followed by the µ and κ receptors, whereas ACTH binds to the five subtypes of melanocortin receptor, and is the only member of the melanocortin family of peptides that binds to the melanocortin-receptor 2 (ACTH receptor). Enkephalins/ACTH and their receptors exhibit a widespread anatomical distribution. Enkephalins are involved in analgesia, angiogenesis, blood pressure, embryonic development, emotional behavior, feeding, hypoxia, limbic system modulation, neuroprotection, peristalsis, and wound repair; as well as in hepatoprotective, motor, neuroendocrine and respiratory mechanisms. ACTH plays a role in acetylcholine release, aggressive behavior, blood pressure, bone maintenance, hyperalgesia, feeding, fever, grooming, learning, lipolysis, memory, nerve injury repair, neuroprotection, sexual behavior, sleep, social behavior, tissue growth and stimulates the synthesis and secretion of glucocorticoids. Enkephalins/ACTH are also involved in many pathologies. Enkephalins are implicated in alcoholism, cancer, colitis, depression, heart failure, Huntington's disease, influenza A virus infection, ischemia, multiple sclerosis, and stress. ACTH plays a role in Addison's disease, alcoholism, cancer, Cushing's disease, dermatitis, encephalitis, epilepsy, Graves' disease, Guillain-Barré syndrome, multiple sclerosis, podocytopathies, and stress. In this review, we provide an updated description of the enkephalinergic and ACTH systems.


Assuntos
Hormônio Adrenocorticotrópico/fisiologia , Encefalinas/fisiologia , Fenômenos Fisiológicos do Sistema Nervoso , Hormônio Adrenocorticotrópico/análise , Hormônio Adrenocorticotrópico/genética , Sequência de Aminoácidos , Animais , Encefalinas/análise , Encefalinas/genética , Humanos , Sistema Nervoso , Precursores de Proteínas/metabolismo , Receptores da Corticotropina/fisiologia , Receptores Opioides/fisiologia , Transdução de Sinais , Distribuição Tecidual
10.
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
11.
Vitam Horm ; 111: 299-311, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31421705

RESUMO

The development, progression, metastasis and drug resistance of the most common human cancers are driven by cyclic adenosine monophosphate (cAMP)-signaling downstream of beta-adrenergic receptors (ß-Ars) coupled to the stimulatory G-protein Gs. Receptors coupled to the inhibitory G-protein Gi inhibit this signaling cascade by blocking the activation of the enzyme adenylyl cyclase that catalyzes the formation of cAMP and function as the physiological inhibitors of this signaling cascade. Members of the Gi-coupled receptor family widely expressed in the mammalian organism are GABA B receptors (GABAB-Rs) for the inhibitory neurotransmitter γ-aminobutyric acid (GABA), opioid receptors for endogenous opioid peptides and cannabinoid receptors for endogenous cannabinoids. This review summarizes current evidence for the concept that the activation of Gi-receptor signaling by pharmacological and psychological means is a promising tool for the long-term management of cAMP-driven cancers with special emphasis on the inhibitory effects of opioids on lung adenocarcinoma and its stem cells.


Assuntos
Adenocarcinoma de Pulmão/tratamento farmacológico , AMP Cíclico/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Receptores Acoplados a Proteínas G/fisiologia , Receptores Opioides/fisiologia , Inibidores de Adenilil Ciclases , Adenilil Ciclases/farmacologia , Animais , Anticarcinógenos/farmacologia , Canabinoides/farmacologia , AMP Cíclico/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Humanos , Metadona/farmacologia , Antagonistas de Entorpecentes , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia
12.
Neurourol Urodyn ; 37(8): 2462-2469, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29862555

RESUMO

AIM: To examine the roles of opioid receptors in the inhibition of nociceptive and nonnociceptive bladder reflexes by sacral dorsal root ganglion (DRG) stimulation in cats. METHODS: Hook electrodes were placed in the right S1 and S2 DRG of cats. The bladders were infused with physiologic saline or 0.25% acetic acid (AA). Naloxone (0.1, 0.3, and 1 mg/kg), an opioid receptor antagonist, was administered intravenously. S1 or S2 DRG stimulation was applied before and after administering the drug. Multiple cystometrograms were performed to determine the effects of DRG stimulation and opioid receptors on the micturition reflex under nociceptive and non-nociceptive conditions. RESULTS: AA significantly (P < 0.01) reduced bladder capacity (BC). DRG stimulation at threshold (T) and 1.5 T significantly increased BC of the saline control under nociceptive and non-nociceptive conditions. When saline was infused, naloxone (0.1-1 mg/kg) significantly (P < 0.01) reduced BC; however, naloxone did not change BC during AA irritation. During saline infusion, naloxone (0.3 and 1 mg/kg) partly blocked S1 DRG stimulation-induced inhibition but had only a slight effect on S2 DRG stimulation. During AA infusion, naloxone (0.3 and 1 mg/kg) only partially blocked S1 DRG stimulation at T intensity but not during 1.5 T stimulation. However, no doses of naloxone significantly affected S2 DRG stimulation. CONCLUSION: Opioid receptors play a role in sacral DRG stimulation on non-nociceptive condition but are not involved in the inhibitory effect of stimulation in nociceptive conditions.


Assuntos
Gânglios Espinais/fisiopatologia , Nociceptores/fisiologia , Receptores Opioides/fisiologia , Animais , Gatos , Estimulação Elétrica , Eletrodos Implantados , Feminino , Masculino , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Dor/fisiopatologia , Reflexo/efeitos dos fármacos , Micção/efeitos dos fármacos
13.
Pancreatology ; 18(6): 615-623, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29937364

RESUMO

Insulin, a key hormone produced by pancreatic beta cells precisely regulates glucose metabolism in vertebrates. In type 1 diabetes, the beta cell mass is destroyed, a process triggered by a combination of environmental and genetic factors. This ultimately results in absolute insulin deficiency and dysregulated glucose metabolism resulting in a number of detrimental pathophysiological effects. The traditional focus of treating type 1 diabetes has been to control blood sugar levels through the administration of exogenous insulin. Newer approaches aim to replace the beta cell mass through pancreatic or islet transplantation. Type 2 diabetes results from a relative insulin deficiency for the prevailing insulin resistance. Treatments are generally aimed at reducing insulin resistance and/or augmenting insulin secretion and the use of insulin itself is often required. It is increasingly being recognized that the beta cell mass is dynamic and increases insulin secretion in response to beta cell mitogens and stress signals to maintain glycemia within a very narrow physiological range. This review critically discusses the role of adrenergic, adenosine and opioid pathways and their interrelationship in insulin secretion, beta cell proliferation and regeneration.


Assuntos
Adenosina/fisiologia , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/fisiologia , Receptores Opioides/fisiologia , Transdução de Sinais/fisiologia , Sistema Nervoso Simpático/fisiologia , Animais , Proliferação de Células , Diabetes Mellitus/fisiopatologia , Humanos , Regeneração
14.
Psychopharmacology (Berl) ; 234(20): 3009-3025, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28856406

RESUMO

RATIONALE: Gamma-aminobutyric acid (GABA)ergic neurons of the substantia nigra pars reticulata (SNpr) are connected to the deep layers of the superior colliculus (dlSC). The dlSC, in turn, connect with the SNpr through opioid projections. Nociceptin/orphanin FQ peptide (N/OFQ) is a natural ligand of a Gi protein-coupled nociceptin receptor (ORL1; NOP) that is also found in the SNpr. Our hypothesis is that tectonigral opioid pathways and intranigral orphanin-mediated mechanisms modulate GABAergic nigrotectal connections. OBJECTIVES: Therefore, the aim of this work was to study the role of opioid and NOP receptors in the SNpr during the modulation of defence reactions organised by the dlSC. METHODS: The SNpr was pretreated with either opioid or NOP receptor agonists and antagonists, followed by dlSC treatment with bicuculline. RESULTS: Blockade of GABAA receptors in the dlSC elicited fear-related defensive behaviour. Pretreatment of the SNpr with naloxone benzoylhydrazone (NalBzoH), a µ-, δ-, and κ1-opioid receptor antagonist as well as a NOP receptor antagonist, decreased the aversive effect of bicuculline treatment on the dlSC. Either µ-opioid receptor activation or blockade by SNpr microinjection of endomorphin-1 (EM-1) and CTOP promoted pro-aversive and anti-aversive actions, respectively, that modulated the defensive responses elicited by bicuculline injection into the dlSC. Pretreatment of the SNpr with the selective NOP receptor antagonist JTC801 decreased the aversive effect of bicuculline, and microinjections of the selective NOP receptor agonist NNC 63-0532 promoted the opposite effect. CONCLUSIONS: These results demonstrate that opioid pathways and orphanin-mediated mechanisms have a critical role in modulating the activity of nigrotectal GABAergic pathways during the organisation of defensive behaviours.


Assuntos
Aminoquinolinas/administração & dosagem , Benzamidas/administração & dosagem , Medo/efeitos dos fármacos , Oligopeptídeos/administração & dosagem , Parte Reticular da Substância Negra/efeitos dos fármacos , Receptores Opioides , Somatostatina/análogos & derivados , Analgésicos Opioides/administração & dosagem , Animais , Bicuculina/administração & dosagem , Relação Dose-Resposta a Droga , Medo/fisiologia , Masculino , Naloxona/administração & dosagem , Naloxona/análogos & derivados , Peptídeos Opioides/administração & dosagem , Parte Reticular da Substância Negra/fisiologia , Ratos , Ratos Wistar , Receptores Opioides/fisiologia , Somatostatina/administração & dosagem , Colículos Superiores/efeitos dos fármacos , Colículos Superiores/fisiologia , Ácido gama-Aminobutírico/administração & dosagem , Receptor de Nociceptina , Nociceptina
15.
J Basic Clin Physiol Pharmacol ; 27(6): 557-561, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27226099

RESUMO

BACKGROUND: Pain is the most common cause of patients seeking medical advice as a result of its association with different pathologies. This study evaluated the antinociceptive property of Haematostaphis barteri as well as the possible mechanism(s) associated with its antinociceptive property. METHODS: Mice were administered H. barteri (30-300 mg kg-1; p.o.), followed by intraplantar injection of 10 µL of 5% formalin into the hind paws. The pain score was determined for 1 h in the formalin test. The possible nociceptive pathways involved in the antinociceptive action of H. barteri were determined by pre-treating mice with theophylline (5 mg kg-1, a non-selective adenosine receptor antagonist), naloxone (2 mg kg-1, a non-selective opioid receptor antagonist), glibenclamide (8 mg kg-1; an ATP-sensitive K+ channel inhibitor), and atropine (3 mg kg-1; non-selective muscarinic antagonist). RESULTS: H. barteri (30-300 mg kg-1) significantly and dose dependently precluded both first and second phases of nociception. Pre-treatment with naloxone had no effect on the analgesic activities of H. barteri in the first phase. Again, pre-treatment with atropine and glibenclamide did not significantly reverse the neurogenic antinociception of the extract in phase 1. However, theophylline reversed the analgesic effect of the extract in the first phase. In phase 2, theophylline had no effect on the analgesic activities of the extract. Naloxone, atropine, and glibenclamide significantly blocked the antinociception of H. barteri in the inflammatory phase of the formalin test. CONCLUSIONS: H. barteri possesses antinociceptive property mediated via the opioidergic, adrenergic, muscarinic, ATP-sensitive K+ channels, and adenosinergic nociceptive pathways.


Assuntos
Anacardiaceae , Analgésicos/farmacologia , Canais KATP/antagonistas & inibidores , Medição da Dor/efeitos dos fármacos , Extratos Vegetais/farmacologia , Folhas de Planta , Neurônios Adrenérgicos/efeitos dos fármacos , Neurônios Adrenérgicos/fisiologia , Analgésicos/isolamento & purificação , Animais , Feminino , Canais KATP/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Medição da Dor/métodos , Extratos Vegetais/isolamento & purificação , Receptores Muscarínicos/fisiologia , Receptores Opioides/fisiologia , Receptores Purinérgicos P1/fisiologia , Neurônios Serotoninérgicos/efeitos dos fármacos , Neurônios Serotoninérgicos/fisiologia
16.
J Ethnopharmacol ; 186: 205-208, 2016 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-27060632

RESUMO

ETHNAPHARMACOLOGICAL RELEVANCE: Stephania japonica is a common plant, widely distributed in all over Bangladesh. Traditionally, this plant is considered as one of the important ingredients in treatment of a variety of ailments including inflammation, pain, rheumatism, cancer, bone fracture, fever etc. However, the scientific reports regarding the antinociceptive effect of this plant are very limited. This study evaluated the antinociceptive effect of methanolic extract of S. japonica (MESJ) leaves. MATERIALS AND METHODS: The antinociceptive effect of MESJ was investigated using both heat- and chemical-induced nociceptive models such as hot plate, tail immersion, acetic acid-induced writhing, formalin and glutamate tests at the doses of 50, 100 and 200mg/kg. Morphine (5mg/kg) and diclofenac sodium (10mg/kg) were used as reference drugs in thermal and chemical models, respectively. Moreover, naloxone (2mg/kg) was used in the thermal models to justify the possible role of the opioid receptors. RESULTS: MESJ produced a significant and dose-dependent increase in the hot plate and tail immersion latencies which were reversed by the treatment with naloxone, suggests the possible involvement of opioid receptors in this activity. Moreover, MESJ inhibited acetic acid-induced writhing, formalin and glutamate-induced lickings in a dose-dependent manner. In parallel, the reference drugs also produced desired antinociceptive effects in this study. CONCLUSION: These results strongly support the antinociceptive activity of the leaves of Stephania japonica and rationalize the traditional use of the leaves in treatment of different painful conditions.


Assuntos
Analgésicos/uso terapêutico , Dor/tratamento farmacológico , Extratos Vegetais/uso terapêutico , Stephania , Ácido Acético , Animais , Formaldeído , Ácido Glutâmico , Temperatura Alta , Masculino , Camundongos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Dor/etiologia , Fitoterapia , Folhas de Planta , Receptores Opioides/fisiologia
17.
Mol Psychiatry ; 21(8): 1057-62, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26460230

RESUMO

Positron emission tomography (PET) studies suggest opioidergic system dysfunction in morbid obesity, while evidence for the role of the dopaminergic system is less consistent. Whether opioid dysfunction represents a state or trait in obesity remains unresolved, but could be assessed in obese subjects undergoing weight loss. Here we measured brain µ-opioid receptor (MOR) and dopamine D2 receptor (D2R) availability in 16 morbidly obese women twice-before and 6 months after bariatric surgery-using PET with [(11)C]carfentanil and [(11)C]raclopride. Data were compared with those from 14 lean control subjects. Receptor-binding potentials (BPND) were compared between the groups and between the pre- and postoperative scans among the obese subjects. Brain MOR availability was initially lower among obese subjects, but weight loss (mean=26.1 kg, s.d.=7.6 kg) reversed this and resulted in ~23% higher MOR availability in the postoperative versus preoperative scan. Changes were observed in areas implicated in reward processing, including ventral striatum, insula, amygdala and thalamus (P's<0.005). Weight loss did not influence D2R availability in any brain region. Taken together, the endogenous opioid system plays an important role in the pathophysiology of human obesity. Because bariatric surgery and concomitant weight loss recover downregulated MOR availability, lowered MOR availability is associated with an obese phenotype and may mediate excessive energy uptake. Our results highlight that understanding the opioidergic contribution to overeating is critical for developing new treatments for obesity.


Assuntos
Obesidade Mórbida/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores Opioides mu/metabolismo , Adulto , Cirurgia Bariátrica , Encéfalo/metabolismo , Dopamina/metabolismo , Feminino , Fentanila/análogos & derivados , Humanos , Pessoa de Meia-Idade , Tomografia por Emissão de Pósitrons/métodos , Receptores de Dopamina D2/fisiologia , Receptores Opioides/metabolismo , Receptores Opioides/fisiologia , Receptores Opioides mu/fisiologia , Redução de Peso
19.
Pharm Biol ; 53(11): 1583-90, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25856703

RESUMO

CONTEXT: Pain corresponds to the most frequent reason for visits to physicians, and its control by conventional drugs is accompanied by several side effects, making treatment difficult. For this reason, new chemical entities derived from natural products still hold great promise for the future of drug discovery to pain treatment. OBJECTIVE: The objective of this study was to evaluate the antinociceptive and anti-inflammatory profiles of p-cymene (PC), a monocyclic monoterpene, and its possible mechanisms of action. MATERIALS AND METHODS: Mice treated acutely with PC (25, 50, or 100 mg/kg, i.p.) were screened for carrageenan-induced hyperalgesia and the inflammatory components of its cascade (30-180 min), carrageenan-induced pleurisy (4 h), and tail-flick test (1-8 h). Also, we observed the PC effect on the generation of nitric oxide by macrophages and the activation of neurons in the periaqueductal gray (PAG) by immunofluorescence. RESULTS: PC reduced (p < 0.001) the hyperalgesia induced by carrageenan, TNF-α, dopamine, and PGE2. PC decrease total leukocyte migration (100 mg/kg: p < 0.01), neutrophils (50 and 100 mg/kg: p < 0.05 and 0.001), and TNF-α (25, 50, and 100 mg/kg: p < 0.01, 0.05, and 0.001, respectively), besides reducing NO production (p < 0.05) in vitro. PC produced antinociceptive effect in tail-flick test (p < 0.05), which was antagonized by naloxone, naltrindole, nor-BNI, and CTOP, and increased (p < 0.001) the number of c-Fos-immunoreactive neurons in PAG. DISCUSSION AND CONCLUSION: These results provide information about the anti-hyperalgesic and anti-inflammatory properties of PC suggesting a possible involvement of the opioid system and modulating some pro-inflammatory cytokines.


Assuntos
Analgésicos/farmacologia , Anti-Inflamatórios/farmacologia , Citocinas , Hiperalgesia/tratamento farmacológico , Monoterpenos/farmacologia , Receptores Opioides , Analgésicos/uso terapêutico , Animais , Anti-Inflamatórios/uso terapêutico , Cimenos , Citocinas/fisiologia , Relação Dose-Resposta a Droga , Hiperalgesia/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Masculino , Camundongos , Monoterpenos/uso terapêutico , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Receptores Opioides/agonistas , Receptores Opioides/fisiologia
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA