Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Curr Protein Pept Sci ; 21(6): 622-637, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32338216

RESUMO

All life forms typically possess homochirality, with rare exceptions. In the case of peptides and proteins, only L-amino acids are known to be encoded by genes. Nevertheless, D-amino acids have been identified in a variety of peptides, synthesized by animal cells. They include neuroexcitatory and neuroprotective peptides, cardioexcitatory peptides, hyperglycemic hormones, opioid peptides, antimicrobial peptides, natriuretic and defensin-like peptides, and fibrinopeptides. This article is a review of their occurrence, structure and bioactivity. It further explores the pharmacology and potential medical applications of some of the peptides.


Assuntos
Aminoácidos/química , Conotoxinas/química , Hormônios de Invertebrado/síntese química , Proteínas do Tecido Nervoso/química , Peptídeos Opioides/química , Proteínas Citotóxicas Formadoras de Poros/química , Sequência de Aminoácidos , Aminoácidos/metabolismo , Animais , Fármacos Cardiovasculares/química , Fármacos Cardiovasculares/metabolismo , Fármacos Cardiovasculares/farmacologia , Conotoxinas/biossíntese , Conotoxinas/farmacologia , Crustáceos/química , Crustáceos/metabolismo , Fibrinopeptídeo A/biossíntese , Fibrinopeptídeo A/química , Fibrinopeptídeo A/farmacologia , Humanos , Hormônios de Invertebrado/biossíntese , Hormônios de Invertebrado/química , Hormônios de Invertebrado/farmacologia , Moluscos/química , Moluscos/metabolismo , Peptídeos Natriuréticos/biossíntese , Peptídeos Natriuréticos/química , Peptídeos Natriuréticos/farmacologia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/farmacologia , Peptídeos Opioides/biossíntese , Peptídeos Opioides/farmacologia , Proteínas Citotóxicas Formadoras de Poros/biossíntese , Proteínas Citotóxicas Formadoras de Poros/farmacologia , Especificidade da Espécie , Aranhas/química , Aranhas/metabolismo , Estereoisomerismo
2.
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
3.
Psychopharmacology (Berl) ; 234(9-10): 1603-1614, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28280884

RESUMO

RATIONALE: Mood disorders can be triggered by stress and are characterized by deficits in reward processing, including disrupted reward learning (the ability to modulate behavior according to past rewards). Reward learning is regulated by the anterior cingulate cortex (ACC) and striatal circuits, both of which are implicated in the pathophysiology of mood disorders. OBJECTIVES: Here, we assessed in rats the effects of a potent stressor (social defeat) on reward learning and gene expression in the ACC, ventral tegmental area (VTA), and striatum. METHODS: Adult male Wistar rats were trained on an operant probabilistic reward task (PRT) and then exposed to 3 days of social defeat before assessment of reward learning. After testing, the ACC, VTA, and striatum were dissected, and expression of genes previously implicated in stress was assessed. RESULT: Social defeat blunted reward learning (manifested as reduced response bias toward a more frequently rewarded stimulus) and was associated with increased nociceptin/orphanin FQ (N/OFQ) peptide mRNA levels in the striatum and decreased Fos mRNA levels in the VTA. Moreover, N/OFQ peptide and nociceptin receptor mRNA levels in the ACC, VTA and striatum were inversely related to reward learning. CONCLUSIONS: The behavioral findings parallel previous data in humans, suggesting that stress similarly disrupts reward learning in both species. Increased striatal N/OFQ mRNA in stressed rats characterized by impaired reward learning is consistent with accumulating evidence that antagonism of nociceptin receptors, which bind N/OFQ, has antidepressant-like effects. These results raise the possibility that nociceptin systems represent a molecular substrate through which stress produces reward learning deficits in mood disorders.


Assuntos
Corpo Estriado/metabolismo , Aprendizagem/fisiologia , Peptídeos Opioides/biossíntese , RNA Mensageiro/biossíntese , Recompensa , Estresse Psicológico/metabolismo , Animais , Feminino , Humanos , Relações Interpessoais , Masculino , Transtornos do Humor/genética , Transtornos do Humor/metabolismo , Transtornos do Humor/psicologia , Peptídeos Opioides/genética , RNA Mensageiro/genética , Ratos , Ratos Long-Evans , Ratos Wistar , Estresse Psicológico/psicologia , Área Tegmentar Ventral/metabolismo , Nociceptina
4.
Br J Anaesth ; 117(2): 250-7, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27307289

RESUMO

BACKGROUND: Nociceptin in the peripheral circulation has been proposed to have an immunoregulatory role with regards to inflammation and pain. However, the mechanisms involved in its regulation are still not clear. The aim of this study was to investigate signalling pathways contributing to the regulation of the expression of nociceptin under inflammatory conditions. METHODS: Mono Mac 6 cells (MM6) were cultured with or without phorbol-12-myristate-13-acetate (PMA). Prepronociceptin (ppNOC) mRNA was detected by RT-qPCR and extracellular nociceptin by fluorescent-enzyme immunoassay. Intracellular nociceptin and phosphorylated kinases were measured using flow cytometry. To evaluate the contribution of various signalling pathways to the regulation of ppNOC mRNA and nociceptin protein, cells were pre-treated with specific kinase inhibitors before co-culturing with PMA. RESULTS: ppNOC mRNA was expressed in untreated MM6 at low concentrations. Exposure of cells to PMA upregulated ppNOC after nine h compared with controls without PMA (median normalized ratio with IQR: 0.18 (0.15-0.26) vs. 0 (0-0.02), P<0.01). Inhibition of mitogen-activated protein kinases specific for signal transduction reversed the PMA effects (all P<0.001). Induction of nociceptin protein concentrations in PMA stimulated MM6 was prevented predominantly by identity of ERK inhibitor (P<0.05). CONCLUSIONS: Upregulation of nociceptin expression by PMA in MM6 cells involves several pathways. Underlying mechanisms involved in nociceptin expression may lead to new insights in the treatment of pain and inflammatory diseases.


Assuntos
Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Peptídeos Opioides/biossíntese , Inibidores de Proteínas Quinases/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Relação Dose-Resposta a Droga , Humanos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Nociceptina
5.
Mol Pain ; 11: 2, 2015 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-25563474

RESUMO

BACKGROUND: The treatment of spinal cord injury (SCI)-induced neuropathic pain presents a challenging healthcare problem. The lack of available robust pharmacological treatments underscores the need for novel therapeutic methods and approaches. Due to the complex character of neuropathic pain following SCI, therapies targeting multiple mechanisms may be a better choice for obtaining sufficient long-term pain relief. Previous studies in our lab showed analgesic effects using combinations of an NMDA antagonist peptide [Ser1]histogranin (SHG), and the mu-opioid peptides endomorphins (EMs), in several pain models. As an alternative to drug therapy, this study evaluated the analgesic potential of these peptides when delivered via gene therapy. RESULTS: Lentiviruses encoding SHG and EM-1 and EM-2 were intraspinally injected, either singly or in combination, into rats with clip compression SCI 2 weeks following injury. Treated animals showed significant reduction in mechanical and thermal hypersensitivity, compared to control groups injected with GFP vector only. The antinociceptive effects of individually injected components were modest, but the combination of EMs and SHG produced robust and sustained antinociception. The onset of the analgesic effects was observed between 1-5 weeks post-injection and sustained without decrement for at least 7 weeks. No adverse effects on locomotor function were observed. The involvement of SHG and EMs in the observed antinociception was confirmed by pharmacologic inhibition using intrathecal injection of either the opioid antagonist naloxone or an anti-SHG antibody. Immunohistochemical analysis showed the presence of SHG and EMs in the spinal cord of treated animals, and immunodot-blot analysis of CSF confirmed the presence of these peptides in injected animals. In a separate group of rats, delayed injection of viral vectors was performed in order to mimic a more likely clinical scenario. Comparable and sustained antinociceptive effects were observed in these animals using the SHG-EMs combination vectors compared to the group with early intervention. CONCLUSIONS: Findings from this study support the potential for direct gene therapy to provide a robust and sustained alleviation of chronic neuropathic pain following SCI. The combination strategy utilizing potent mu-opioid peptides with a naturally-derived NMDA antagonist may produce additive or synergistic analgesic effects without the tolerance development for long-term management of persistent pain.


Assuntos
Neuralgia/tratamento farmacológico , Neuralgia/etiologia , Peptídeos Opioides/uso terapêutico , Proteínas/uso terapêutico , Traumatismos da Medula Espinal/complicações , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Vetores Genéticos/fisiologia , Humanos , Hiperalgesia/tratamento farmacológico , Lentivirus/genética , Masculino , Neuroblastoma/patologia , Neuropeptídeos/biossíntese , Neuropeptídeos/uso terapêutico , Peptídeos Opioides/biossíntese , Peptídeos Opioides/genética , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Proteínas/genética , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos
6.
Artigo em Inglês | MEDLINE | ID: mdl-24184686

RESUMO

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


Assuntos
Cocaína/farmacologia , Corpo Estriado/efeitos dos fármacos , Dinorfinas/genética , Epigênese Genética/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Receptores Opioides kappa/genética , Receptores Opioides/genética , Animais , Corpo Estriado/metabolismo , Dinorfinas/biossíntese , Encefalinas/biossíntese , Encefalinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Masculino , Núcleo Accumbens/metabolismo , Peptídeos Opioides/biossíntese , Peptídeos Opioides/genética , Precursores de Proteínas/biossíntese , Precursores de Proteínas/genética , Ratos , Receptores Opioides/biossíntese , Receptores Opioides kappa/biossíntese , Receptor de Nociceptina , Nociceptina
7.
J Neurosci Res ; 91(12): 1533-40, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24038231

RESUMO

The peptides dynorphin and enkephalin modulate many physiological processes, such as motor activity and the control of mood and motivation. Their expression in the caudate putamen (CPu) is regulated by dopamine and opioid receptors. The current work was designed to explore the early effects of the acute activation of D4 and/or µ opioid receptors by the agonists PD168,077 and morphine, respectively, on the regulation of the expression of these opioid peptides in the rat CPu, on transcription factors linked to them, and on the expression of µ opioid receptors. In situ hybridization experiments showed that acute treatment with morphine (10 mg/kg) decreased both enkephalin and dynorphin mRNA levels in the CPu after 30 min, but PD168,077 (1 mg/kg) did not modify their expression. Coadministration of the two agonists demonstrated that PD168,077 counteracted the morphine-induced changes and even increased enkephalin mRNA levels. The immunohistochemistry studies showed that morphine administration also increased striatal µ opioid receptor immunoreactivity but reduced P-CREB expression, effects that were blocked by the PD168,077-induced activation of D4 receptors. The current results present evidence of functional D4 -µ opioid receptor interactions, with consequences for the opioid peptide mRNA levels in the rat CPu, contributing to the integration of DA and opioid peptide signaling.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Peptídeos Opioides/biossíntese , Putamen/metabolismo , Receptores de Dopamina D4/metabolismo , Animais , Dinorfinas/biossíntese , Encefalinas/biossíntese , Imuno-Histoquímica , Hibridização In Situ , Masculino , Putamen/efeitos dos fármacos , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/metabolismo
8.
Dev Cell ; 26(2): 195-203, 2013 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-23906067

RESUMO

Few families of signaling factors have been implicated in the control of development. Here, we identify the neuropeptides nociceptin and somatostatin, a neurotransmitter and neuroendocrine hormone, as a class of developmental signals in both chick and zebrafish. We show that signals from the anterior mesendoderm are required for the formation of anterior placode progenitors, with one of the signals being somatostatin. Somatostatin controls ectodermal expression of nociceptin, and both peptides regulate Pax6 in lens and olfactory progenitors. Consequently, loss of somatostatin and nociceptin signaling leads to severe reduction of lens formation. Our findings not only uncover these neuropeptides as developmental signals but also identify a long-sought-after mechanism that initiates Pax6 in placode progenitors and may explain the ancient evolutionary origin of neuropeptides, predating a complex nervous system.


Assuntos
Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Cristalino/embriologia , Bulbo Olfatório/embriologia , Mucosa Olfatória/embriologia , Peptídeos Opioides/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , Somatostatina/metabolismo , Células-Tronco/citologia , Animais , Embrião de Galinha , Proteínas do Olho/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Cristalino/citologia , Cristalino/metabolismo , Bulbo Olfatório/citologia , Bulbo Olfatório/metabolismo , Mucosa Olfatória/citologia , Mucosa Olfatória/metabolismo , Peptídeos Opioides/biossíntese , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/biossíntese , Proteínas Repressoras/biossíntese , Transdução de Sinais , Células-Tronco/fisiologia , Peixe-Zebra/embriologia , Proteínas de Peixe-Zebra/metabolismo , Nociceptina
9.
Biochem Biophys Res Commun ; 421(4): 678-83, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22542942

RESUMO

In postnatal testes, follicle-stimulating hormone (FSH) acts on somatic Sertoli cells to activate gene expression directly via an intracellular signaling pathway composed of cAMP, cAMP-dependent protein kinase (PKA), and cAMP-response element-binding protein (CREB), and promotes germ cell development indirectly. Yet, the paracrine factors mediating the FSH effects to germ cells remained elusive. Here we show that nociceptin, known as a neuropeptide, is upregulated by FSH through cAMP/PKA/CREB pathway in Sertoli cells in murine testes. Chromatin immunoprecipitation from Sertoli cells shows that CREB phosphorylated at Ser133 associates with prepronociceptin gene encoding nociceptin. Analyses with Sertoli cells and testes demonstrates that both prepronociceptin mRNA and the nociceptin peptide are induced after FSH signaling is activated. In addition, the nociceptin peptide is induced in testes after 9days post partum following FSH surge. Thus, our findings may identify nociceptin as a novel paracrine mediator of the FSH effects in the regulation of spermatogenesis.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Peptídeos Opioides/biossíntese , Células de Sertoli/metabolismo , Espermatogênese , Animais , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Hormônio Foliculoestimulante/farmacologia , Masculino , Camundongos , Peptídeos Opioides/genética , Fosforilação , Células de Sertoli/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/metabolismo , Regulação para Cima , Nociceptina
10.
Stress ; 15(4): 378-84, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22074385

RESUMO

Nociceptin/orphanin FQ (N/OFQ) peptide and its receptor are not only ubiquitously expressed in mammalian brain and spinal cord but are also abundant in limbic structures, particularly in the hippocampus. The widespread distribution of N/OFQ reflects the broad spectrum of its biological actions such as nociception, food intake, spontaneous locomotor activity, and learning and memory processes. Since the hippocampus is involved in the control of adrenocortical activity, its role in stress-related phenomena is well characterized. In male Wistar rats, we first examined the effects of acute restraint stress (120 min) on the brain immunohistochemical localization of N/OFQ. The analysis carried out on sections obtained at the onset of stress revealed enhanced expression of N/OFQ in CA1, CA3, and the dentate gyrus as well as increased plasma corticosterone concentrations. Next, we examined whether endogenous glucocorticoid hormone plays a role in the modulation of hippocampal N/OFQ expression in response to stress. To this end, rats were injected with corticosterone (1 mg/kg) or subjected to restraint stress 1 week after adrenalectomy. Two hours after corticosterone administration, plasma glucocorticoid concentrations were comparable to those observed after restraint stress, while N/OFQ expression had significantly increased in all the hippocampal subfields examined. By contrast, in adrenalectomized rats, stress did not modify protein expression. These results confirm that stress can affect N/OFQ expression and that glucocorticoids may constitute hormonal mediators of this complex interplay.


Assuntos
Peptídeos Opioides/biossíntese , Estresse Psicológico/fisiopatologia , Adrenalectomia , Animais , Corticosterona/fisiologia , Hipocampo , Imuno-Histoquímica , Masculino , Ratos , Ratos Wistar , Restrição Física , Nociceptina
11.
Exp Dermatol ; 20(1): 24-8, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20955200

RESUMO

Opioids exert major effects not only in the central nervous system but also in immune responses. We investigated the effects of µ-opioid peptides, secreted by tumor cells, on anti-tumor immune responses. For this purpose, tumor growth was studied in wild-type and µ-opioid receptor-deficient (MOR-/-) mice injected with B16 melanoma cells. The ability of these cells to produce opioids was studied by Western blots in vitro. Finally, biopsy material from human melanomas was investigated by immunohistochemistry for ß endorphin expression. Injection of B16 melanoma cells, producing endogenous ß endorphin, in the flank of MOR-/- mice revealed a profound reduction in tumor growth, paralleled by a significantly higher infiltration of immune cells into the tumors, when compared to tumor growth after injection of B16 melanoma cells into wild-type mice. Opioids present in B16 cell supernatant significantly reduced the proliferation of normal but not MOR-/- leucocytes. Immunohistochemical analyses of biopsies from human melanoma tissues showed a positive correlation between expression of ß endorphin and tumor progression. Our data provide evidence that µ-opioid peptides may play a major role in cancer progression by modulating immune response. This finding may have implications for the future optimization of immunointerventions for cancer.


Assuntos
Melanoma/imunologia , Peptídeos Opioides/imunologia , Neoplasias Cutâneas/imunologia , Animais , Progressão da Doença , Humanos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Melanoma/metabolismo , Melanoma/patologia , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Knockout , Peptídeos Opioides/biossíntese , Receptores Opioides mu/agonistas , Receptores Opioides mu/deficiência , Receptores Opioides mu/genética , Receptores Opioides mu/imunologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , beta-Endorfina/imunologia , beta-Endorfina/metabolismo
12.
Brain Res ; 1379: 71-85, 2011 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-20828542

RESUMO

Circulating estrogen levels and hippocampal-dependent cognitive functions decline with aging. Moreover, the responses of hippocampal synaptic structure to estrogens differ between aged and young rats. We recently reported that estrogens increase levels of post-synaptic proteins, including PSD-95, and opioid peptides leu-enkephalin and dynorphin in the hippocampus of young animals. However, the influence of ovarian hormones on synaptic protein and opioid peptide levels in the aging hippocampus is understudied. Here, young (3- to 5-month-old), middle-aged (9- to 12-month-old), and aged (about 22-month-old) female rats were ovariectomized and then, 4 weeks later, subcutaneously implanted with a silastic capsule containing vehicle or 17ß-estradiol. After 48 h, rats were subcutaneously injected with progesterone or vehicle and sacrificed 1 day later. Coronal sections through the dorsal hippocampus were processed for quantitative peroxidase immunohistochemistry of leu-enkephalin, dynorphin, synaptophysin, and PSD-95. With age, females showed opposing changes in leu-enkephalin and dynorphin levels in the mossy fiber pathway, particularly within the hilus, and regionally specific changes in synaptic protein levels. 17ß-estradiol, with or without progesterone, altered leu-enkephalin levels in the dentate gyrus and synaptophysin levels in the CA1 of young but not middle-aged or aged females. Additionally, 17ß-estradiol decreased synaptophysin levels in the CA3 of middle-aged females. Our results support and extend previous findings indicating 17ß-estradiol modulation of hippocampal opioid peptides and synaptic proteins while demonstrating regional and age-specific effects. Moreover, they lend credence to the "window of opportunity" hypothesis during which hormone replacement can modulate hippocampal structure and circuitry to improve cognitive outcomes.


Assuntos
Envelhecimento/metabolismo , Estradiol/fisiologia , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Peptídeos Opioides/biossíntese , Progesterona/fisiologia , Sinapses/química , Fatores Etários , Animais , Feminino , Proteínas do Tecido Nervoso/metabolismo , Ovariectomia , Ratos , Ratos Sprague-Dawley , Sinapses/metabolismo
13.
J Neuroimmunol ; 232(1-2): 83-93, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21129785

RESUMO

Previous studies have demonstrated that inflammatory cells produce several mediators that can effectively counteract pain. This study was designed to test the hypothesis that exogenous administration of recombinant mouse granulocyte-colony-stimulating factor (rmG-CSF) to enhance the recruitment of inflammatory cells to painful inflamed sites could attenuate pain in a chronic neuropathic pain model in mice. Our results indicate that treatment with rmG-CSF increased several cytokines and opioid peptides content; however, it did not attenuate but exacerbate neuropathic pain. Our study highlights the potent pro-inflammatory potential of G-CSF and suggests they may be targets for therapeutic intervention in chronic neuropathic pain.


Assuntos
Fator Estimulador de Colônias de Granulócitos/farmacologia , Neuralgia/imunologia , Limiar da Dor/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Separação Celular , Citocinas/biossíntese , Citometria de Fluxo , Imuno-Histoquímica , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos Opioides/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nervo Isquiático/lesões , Nervo Isquiático/cirurgia
14.
Brain Behav Evol ; 76(2): 154-62, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21079395

RESUMO

The various means by which the body perceives, transmits, and resolves the experiences of pain and nociception are mediated by a host of molecules, including neuropeptides within the opioid gene signaling pathway. The peptide ligands and receptors encoded by this group of genes have been linked to behavioral disorders as well as a number of psychiatric affective disorders. Our aim was to explore the recent evolutionary history of these two gene families by taking a comparative genomics approach, specifically through a comparison between humans and chimpanzees. Our analyses indicate differential expression of these genes between the two species, more than expected based on genome-wide comparisons, indicating that differential expression is pervasive among the opioid genes. Of the 8 family members, three genes showed significant expression differences (PENK, PNOC, and OPRL1), with two others marginally significant (OPRM1 and OPRD1). Accelerated substitution rates along human and chimpanzee lineages within the putative regulatory regions of OPRM1, POMC, and PDYN between the human and chimpanzee branches are consistent with positive selection. Collectively, these results suggest that there may have been a selective advantage to modulating the expression of the opioid genes in humans compared with our closest living relatives. Information about the cognitive roles mediated by these genes in humans may help to elucidate the trait consequences of these putatively adaptive expression changes.


Assuntos
Química Encefálica/genética , Regulação da Expressão Gênica/genética , Peptídeos Opioides/biossíntese , Peptídeos Opioides/genética , Pan troglodytes , Animais , Evolução Molecular , Estudo de Associação Genômica Ampla/métodos , Humanos , Especificidade da Espécie
15.
Neuroscience ; 169(1): 269-78, 2010 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-20417255

RESUMO

Antagonists selectively inhibiting activation of the nociceptin/orphanin FQ (N/OFQ) receptor reduce motor symptoms in experimental models of Parkinson's disease, and genetic deletion of the ppN/OFQ gene offers partial protection of mid-brain dopamine neurons against the neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). MPTP increased ppN/OFQ mRNA expression in the substantia nigra (SN). We have evaluated the temporal relationship of dopamine cell loss to increased ppN/OFQ mRNA expression in the substantia nigra after MPTP treatment, and characterized the cellular locations in which increased ppN/OFQ mRNA expression was observed after MPTP treatment. MPTP increased by about 5-fold the number of neurons expressing ppN/OFQ mRNA in the pars reticulata of SN (SNr) by 24 h after treatment and the elevation remained significant for at least 7 days. This period coincided with the timing of the loss of dopamine neurons from the pars compacta of substantia nigra (SNc) after MPTP. The increased expression of ppN/OFQ mRNA co-localized with a neuronal marker in the SNr. MPTP treatment resulted in a small increase in the numbers of neurons expressing ppN/OFQ in the SNc in mice from one mouse colony but the increase did not reach statistical significance in mice from another colony. No changes in ppN/OFQ-mRNA expression were observed in the ventral tegmental area (VTA), the caudate-putamen, the subthalamic nucleus, or in two other brains areas. These results demonstrate that increased N/OFQ expression in the SNr is closely associated with the MPTP-induced loss of dopamine neurons in the SNc in a widely used animal model of Parkinson's disease.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Intoxicação por MPTP/genética , Neurônios/efeitos dos fármacos , Transtornos Parkinsonianos/genética , Precursores de Proteínas/biossíntese , RNA Mensageiro/biossíntese , Receptores Opioides/biossíntese , Substância Negra/efeitos dos fármacos , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/classificação , Neurônios/metabolismo , Peptídeos Opioides/biossíntese , Peptídeos Opioides/genética , Precursores de Proteínas/deficiência , Precursores de Proteínas/genética , RNA Mensageiro/genética , Receptores Opioides/deficiência , Receptores Opioides/genética , Substância Negra/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo , Nociceptina
16.
Eur J Pharmacol ; 615(1-3): 246-51, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19445924

RESUMO

Cirrhosis is associated with impairment of the male reproductive system, hypogonadism and feminization. It is important to rule out whether the impairment in the reproductive system exists earlier in the course of cholestatic liver disease to target effective therapies at the best time point. In this study we investigated the role of endogenous opioid and nitric oxide system in alterations of the reproductive system in male rats. We performed sham or bile duct ligation surgery on male Sprague-Dawley rats and treated the animals for seven days with saline, naltrexone, an opioid receptor blocker (20 mg/kg) and N (G)-nitro-L-arginine methyl ester (L-NAME), a nitric oxide synthase inhibitor (10 mg/kg). We then evaluated the plasma level of testosterone, luteinizing hormone (LH) and follicle stimulating hormone (FSH), sperm count and motility as well as biomarkers of cholestasis and nitric oxide productions. The results showed that following cholestasis, total testosterone level decrease and LH level increase in plasma of cholestatic rats and treatment with L-NAME and naltrexone could improve the plasma level of testosterone. Naltrexone could decrease the elevated level of LH in cholestatic animals. In addition, the weight of seminal vesicles and prostate significantly decreased in cholestasis as compared to the control group and treatment with L-NAME and naltrexone could improve the weights of the two organs in cholestasis. Our results demonstrate for the first time that the male reproductive system is impaired early in cholestasis and that endogenous opioid and nitric oxide system contribute to these impairments in the early course of the disease.


Assuntos
Colestase/fisiopatologia , Genitália Masculina/fisiopatologia , Óxido Nítrico/biossíntese , Peptídeos Opioides/biossíntese , Animais , Ductos Biliares , Colestase/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hormônio Foliculoestimulante/sangue , Ligadura , Hormônio Luteinizante/sangue , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes , Óxido Nítrico Sintase/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Contagem de Espermatozoides , Motilidade dos Espermatozoides , Testosterona/sangue
17.
J Neurochem ; 106(6): 2337-44, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18643791

RESUMO

Morphine, the most used compound among narcotic analgesics, has been shown to be endogenously present in different mammalian/invertebrate normal tissues. In this study, we used mice that cannot make dopamine due to a genetic deletion of tyrosine hydroxylase specifically in dopaminergic neurons, to test the hypothesis that endogenous dopamine is necessary to endogenous morphine formation in vivo in mammalian brain. When dopamine was lacking in brain neurons, endogenous morphine was missing in brain mouse whereas it could be detected in brain from wild type rodent at a picogram range. Our data prove for the first time that endogenous dopamine is necessary to endogenous morphine formation in normal mammalian brain. Morphine synthesis appears to be originated from dopamine through L-tyrosine in normal brain tissue. Morphine synthesis is not considered to occur inside the same neuron in normal tissue; released dopamine might be transported into morphinergic neuron and further transformed into morphine. A physiological role for endogenous morphine is suggested considering that dopamine could modulate thermal threshold through endogenous morphine formation in vivo. Thus, dopamine and endogenous opiates/opioid peptides may be interconnected in the physiological processes; yet, endogenous morphine may represent a basic link of this chain.


Assuntos
Encéfalo/metabolismo , Dopamina/deficiência , Morfina/metabolismo , Peptídeos Opioides/biossíntese , Animais , Regulação da Temperatura Corporal/fisiologia , Encéfalo/anatomia & histologia , Dopamina/biossíntese , Camundongos , Camundongos Knockout , Limiar Sensorial/fisiologia , Sensação Térmica/fisiologia , Tirosina/metabolismo , Tirosina 3-Mono-Oxigenase/genética
18.
Psychopharmacology (Berl) ; 196(4): 523-31, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17989958

RESUMO

RATIONALE: Nociceptin/orphanin FQ (N/OFQ) has been proposed to be a functional antagonist of corticotropin-releasing factor (CRF) in relation to its anti-stress action and its ability to antagonize the anorectic effect of CRF in rats without exhibiting affinity for CRF receptors. The bed nucleus of the stria terminalis (BST) is highly sensitive to the inhibitory effect of N/OFQ on CRF-induced anorexia. OBJECTIVE: The present study was aimed at further evaluating the role of the BST in the functional antagonism between N/OFQ and CRF by examining it at molecular level and in the context of CRF-induced anxiety in the rat. MATERIALS AND METHODS: First, in situ hybridization experiments investigated the expression of the pro-N/OFQ precursor and of NOP receptors in several brain areas 6 h after injection of CRF (0.2 and 1 microg/rat) into the lateral cerebroventricle (LV). Second, the elevated plus maze test was used to evaluate whether N/OFQ, injected into the BST (0.05 and 0.5 microg/rat) or into the LV (0.5, 1.8, and 2.4 microg/rat), inhibits the anxiogenic-like effect evoked by LV injection of CRF (1 microg/rat) in rats. RESULTS: The in situ hybridization study showed that LV injection of CRF 1 microg/rat increases NOP receptor expression in the BST, while no change of the N/OFQ precursor was observed. On the other hand, N/OFQ injection into the BST blocks the anxiogenic effect of CRF at doses lower than those required by LV injection (0.5 vs 1.8 microg/rat, respectively). CONCLUSION: These data provide further support for the hypothesis that N/OFQ may behave as functional antagonist of CRF and suggest that this antagonism may occur within the BST.


Assuntos
Hormônio Liberador da Corticotropina/fisiologia , Peptídeos Opioides/fisiologia , Núcleos Septais/fisiologia , Animais , Ansiedade/induzido quimicamente , Ansiedade/tratamento farmacológico , Hormônio Liberador da Corticotropina/farmacologia , Hibridização In Situ , Injeções Intraventriculares , Masculino , Aprendizagem em Labirinto , Peptídeos Opioides/biossíntese , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Receptores Opioides/biossíntese , Núcleos Septais/metabolismo , Receptor de Nociceptina , Nociceptina
19.
Neuropeptides ; 41(6): 355-63, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17640727

RESUMO

It is accepted that inflammatory mediators released from leukocytes contribute to the generation of pain. However, it is less well known that immune cells also produce mediators that can effectively counteract pain. These include anti-inflammatory cytokines and opioid peptides. This article concentrates on recent evidence that interactions between leukocyte-derived opioid peptides and their receptors on peripheral sensory neurons can result in potent, clinically relevant inhibition of pathological pain. Inflammation of peripheral tissues leads to increased synthesis and axonal transport of opioid receptors in dorsal root ganglion neurons. This results in opioid receptor upregulation and enhanced G-protein coupling at peripheral sensory nerve terminals. These events are dependent on neuronal electrical activity, production of proinflammatory cytokines and nerve growth factor within the inflamed tissue. Together with the disruption of the perineurial barrier, all these changes lead to an enhanced peripheral analgesic efficacy of opioids. The major source of local endogenous opioid ligands (beta-endorphin, enkephalins, endomorphins and dynorphin) are leukocytes. These cells contain and upregulate signal-sequence encoding mRNA of the beta-endorphin precursor proopiomelanocortin and the entire enzymatic machinery necessary for its processing into the functionally active peptide. Opioid-containing immune cells extravasate using adhesion molecules and chemokines to accumulate in inflamed tissues. Upon stressful stimuli or in response to releasing agents such as corticotropin-releasing factor, cytokines, chemokines and catecholamines, leukocytes secrete opioids. Depending on the cell type, this release is contingent on extracellular Ca(2+) or on inositol triphosphate receptor-triggered release of Ca(2+) from endoplasmic reticulum. Once secreted opioid peptides activate peripheral opioid receptors and produce analgesia by inhibiting the excitability of sensory nerves and/or the release of excitatory neuropeptides. These effects occur without central untoward side effects such as depression of breathing, clouding of consciousness or addiction. Future aims include the selective targeting of opioid-containing leukocytes to sites of painful injury and the augmentation of opioid peptide and receptor synthesis.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Leucócitos/metabolismo , Peptídeos Opioides/biossíntese , Dor/prevenção & controle , Animais , Quimiotaxia de Leucócito , Humanos , Inflamação/imunologia , Inflamação/patologia , Leucócitos/imunologia , Peptídeos Opioides/metabolismo , Manejo da Dor , Receptores Opioides , Regulação para Cima/imunologia , Regulação para Cima/fisiologia
20.
Neurologia ; 22(2): 99-105, 2007 Mar.
Artigo em Espanhol | MEDLINE | ID: mdl-17323235

RESUMO

INTRODUCTION: All pharmacological treatment is surrounded by a psychosocial context that contributes in part to the outcome of the therapy. The placebo effect is the improvement observed after a simulated treatment in which the subject, without his or her knowledge, is exposed only to the psychosocial context surrounding the treatment, without receiving the pure "pharmacodynamic" effect of the medication. The aim of this paper is to examine which brain regions and neurochemical mechanisms are responsible for the analgesia resulting from the placebo. METHOD AND RESULTS: Recent studies using functional neuroimaging methods have suggested that when expectations of analgesia arise because of a simulated treatment (placebo) there is an increase in the activity of the dorsolateral and orbitofrontal prefrontal cortices. These increments are detected early on, before the experience of pain, which suggests that these structures could be part of a system that is involved in the cognitive control of pain. Several studies have found that once the subject experiences the nociceptive stimulus, the placebo causes a significant reduction in the activity of cortical regions classically associated with nociceptive processing, mainly the anterior insula and the rostral cingulate. In these regions the attenuation of the pain processing seems to be mediated by the release of opioids. CONCLUSIONS: In placebo analgesia at least two anatomically differentiated cerebral systems work together. The guiding mechanism, which appears to be located prefrontally, is activated first. This system would influence other cortical regions (cingulate and insula), attenuating the processing of nociceptive information at these higher levels.


Assuntos
Analgésicos Opioides/uso terapêutico , Analgésicos/uso terapêutico , Encéfalo/fisiologia , Peptídeos Opioides/biossíntese , Dor/tratamento farmacológico , Dor/psicologia , Efeito Placebo , Placebos/uso terapêutico , Giro do Cíngulo/fisiologia , Humanos , Nociceptores/fisiologia , Córtex Pré-Frontal/fisiologia , Receptores Opioides/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA