Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropharmacology ; 227: 109439, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36709036

RESUMO

Currently available µ-opioid receptor agonist pharmacotherapies for opioid use disorder possess adverse effects limiting their use and, despite treatment, rates of relapse remain high. We previously showed that endomorphin analog ZH853 had no effect in rodent models that predict abuse liability in humans. Here we extended these findings by examining dependence liability and reinforcing properties in female rats and male rats with previous opioid exposure. The potential use of ZH853 in managing opioid use disorder was evaluated by examining its effect on opioid-seeking behavior and withdrawal. We found that ZH853 did not induce locomotor activation in male and female mice and was not self-administered by female rats. Relative to morphine, ZH853 led to similar somatic signs of withdrawal, but low affective-motivational signs of withdrawal, and absent changes in ventral tegmental area K(+)-Cl(-) co-transporter expression associated with reward dysregulation. The low abuse liability of ZH853 was further supported in oxycodone self-administering male rats, where ZH853 substitution extinguished opioid-seeking behavior. ZH853 priming also did not reinstate morphine conditioned place preference. Lastly, ZH853 inhibited oxycodone-seeking behavior during relapse after forced abstinence and decreased the expression of morphine withdrawal. These findings suggest the potential use of ZH853 as a safer opioid medication for long-term treatment of pain and opioid use disorder.


Assuntos
Transtornos Relacionados ao Uso de Opioides , Síndrome de Abstinência a Substâncias , Humanos , Ratos , Camundongos , Masculino , Feminino , Animais , Analgésicos Opioides/farmacologia , Oxicodona/uso terapêutico , Entorpecentes , Morfina/farmacologia , Recompensa , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Síndrome de Abstinência a Substâncias/tratamento farmacológico
2.
J Pharmacol Exp Ther ; 370(3): 369-379, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31213481

RESUMO

Opioid dependence can be difficult to manage using existing pharmacotherapies. A long-acting opioid with low abuse liability that substitutes for a shorter-acting opioid may improve treatment of opioid use disorders (OUDs). We recently characterized an endomorphin (EM) analog (ZH853) that produced a longer duration of antinociception compared with morphine, but did not produce self-administration or several other adverse effects preclinically. Here, we further characterized ZH853 in tests of antinociception, abuse liability, and drug discrimination. A conditioned place preference (CPP) procedure, that included a locomotor activity assessment, was used to test abuse liability in rats. Subsequently, dopamine (DA) cell-somas located in the ventral tegmental area (VTA) from these rats were assessed by size using immunohistochemistry and Stereo Investigator software. A hot-plate antinociception test in male and female mice confirmed central penetration. Morphine-substitution effects of several EM analogs (ZH850, ZH831, and ZH853) were tested in a drug discrimination (DD) procedure in rats. Morphine produced dose-dependent CPP and locomotor sensitization and reduced the size of DA cell somas in VTA, whereas ZH853 did not produce any of these effects relative to control. The antinociceptive effects of ZH853 were µ-receptor selective since ß-funaltrexamine antagonized these effects. Rats responded on a morphine-trained lever when injected with ZH831 and ZH853 during DD experiments. The favorable morphine-substitution effects of these EM analogs relative to their low abuse liability indicate promising novel compounds that may improve treatment of OUD. SIGNIFICANCE STATEMENT: In this experiment, we investigated the preclinical effects of novel endomorphin analogs for use as substitution therapies for opioid use disorder, a problem that has contributed to an opioid overdose epidemic. Several endomorphin analogs substituted for morphine without producing adverse effects, including reward behaviors associated with abuse liability. These compounds have the potential to become important additional tools to treat opioid use disorders.


Assuntos
Oligopeptídeos/química , Oligopeptídeos/farmacologia , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Animais , Comportamento Animal/efeitos dos fármacos , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Locomoção/efeitos dos fármacos , Masculino , Oligopeptídeos/efeitos adversos , Oligopeptídeos/uso terapêutico , Transtornos Relacionados ao Uso de Opioides/metabolismo , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Ratos , Ratos Sprague-Dawley , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
3.
J Neuroinflammation ; 16(1): 100, 2019 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-31109346

RESUMO

BACKGROUND: Numerous studies have identified the proinflammatory, pronociceptive effects of morphine which ultimately exacerbate pain. Our novel endomorphin analog ZH853 does not produce proinflammatory effects on its own and gives potent, long-lasting analgesia. This study investigates whether ZH853's lack of interaction with the neuroimmune system reduces the risk of prolonged pain. METHODS: Adult male Sprague-Dawley rats were subjected to one of two treatment paradigms. Either (1) chronic pain followed by chronic treatment with morphine, ZH853 or vehicle, or (2) chronic drug administered prior to pain induction. Complete Freund's adjuvant (CFA) was injected or paw incision surgery was performed on the left hind plantar foot pad. Drugs were administered through Alzet osmotic minipumps at a rate of 1 µl/h for 5 days at appropriate doses based on prior experiments. Animals were tested for mechanical allodynia and thermal hyperalgesia using von Frey filaments and the Hargreaves apparatus, respectively. Additionally, several gait parameters were measured using the CatWalk XT. When all animals had recovered from pain, 1 mg/kg of naltrexone was administered to test for development of latent sensitization (LS). A second set of animals was used to investigate dorsal horn inflammation following CFA and drug treatment. ANOVAs were used to assess differences between drug treatment groups. RESULTS: As expected, morphine increased and prolonged pain in all experiments compared to vehicle treatment. However, ZH853 treatment reduced the overall time spent in pain and the severity of pain scores compared to morphine. ZH853 not only reduced inflammation versus morphine treatment but also, in some instances, acted as an anti-inflammatory drug compared to vehicle treatment. Finally, ZH853 prevented the development of LS while vehicle- and morphine-treated animals showed robust relapse to pain. CONCLUSIONS: ZH853 has a favorable side effect profile versus morphine and provides superior analgesia in a number of pain states. We now know that chronic use of this compound reduces time spent in a chronic pain state, the opposite of common opioids like morphine, and reduces the risk of LS, making ZH853 an excellent candidate for clinical development in humans for inflammatory and postoperative pain.


Assuntos
Analgésicos Opioides/uso terapêutico , Analgésicos/uso terapêutico , Imunomodulação/efeitos dos fármacos , Morfina/uso terapêutico , Dor Pós-Operatória/tratamento farmacológico , Peptídeos Cíclicos/uso terapêutico , Analgésicos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Imunomodulação/fisiologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Masculino , Morfina/farmacologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Dor Pós-Operatória/imunologia , Peptídeos Cíclicos/farmacologia , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia
4.
J Pain ; 18(12): 1526-1541, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28939014

RESUMO

Activation of the mu-opioid receptor provides the gold standard for pain relief, but most opioids used clinically have adverse effects that have contributed to an epidemic of overdose deaths. We recently characterized mu-opioid receptor selective endomorphin (EM) analogs that provide potent antinociception with reduction or absence of a number of side effects of traditionally prescribed opioids including abuse liability, respiratory depression, motor impairment, tolerance, and inflammation. The current study explores the effectiveness of these EM analogs relative to morphine in four major pain models by intrathecal as well as intravenous administration in male Sprague Dawley rats and subcutaneous administration in male CD-1 mice. In the spared nerve injury model of neuropathic pain, mechanical allodynia and mechanical hyperalgesia were assessed with von Frey and Randall-Selitto tests, respectively. In the paw incision model of postoperative pain, von Frey testing was used to assess mechanical allodynia and thermal hyperalgesia was evaluated with Hargreaves testing. In the Complete Freund's Adjuvant model of inflammatory pain, thermal hyperalgesia was assessed using Hargreaves testing. In CD-1 mice, visceral pain was assessed with the acetic acid writhing test. In all cases, EM analogs had equal or greater potency and longer duration of action relative to morphine. The data suggest that EM analogs, particularly analog 4 (ZH853), could provide effective therapy for a diverse spectrum of pain conditions with low risk of adverse side effects compared with currently used opioids such as morphine. PERSPECTIVE: Novel EM analogs show equal or greater potency and effectiveness relative to morphine in multiple pain models. Together with substantially reduced side effects, including abuse liability, the compounds show promise for addressing the critical need for effective pain relief as well as reducing the opioid overdose epidemic.


Assuntos
Analgésicos Opioides/farmacologia , Hiperalgesia/tratamento farmacológico , Morfina/farmacologia , Neuralgia/tratamento farmacológico , Dor Nociceptiva/tratamento farmacológico , Oligopeptídeos/análise , Dor Pós-Operatória/tratamento farmacológico , Peptídeos Cíclicos/farmacologia , Dor Visceral/tratamento farmacológico , Analgésicos Opioides/administração & dosagem , Animais , Modelos Animais de Doenças , Inflamação/complicações , Injeções Intravenosas , Injeções Espinhais , Masculino , Camundongos , Dor Nociceptiva/etiologia , Peptídeos Cíclicos/administração & dosagem , Ratos , Ratos Sprague-Dawley
5.
Neuropharmacology ; 105: 215-227, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26748051

RESUMO

Opioids acting at the mu opioid receptor (MOR) are the most effective analgesics, however adverse side effects severely limit their use. Of particular importance, abuse liability results in major medical, societal, and economic problems, respiratory depression is the cause of fatal overdoses, and tolerance complicates treatment and increases the risk of side effects. Motor and cognitive impairment are especially problematic for older adults. Despite the host of negative side effects, opioids such as morphine are commonly used for acute and chronic pain conditions. Separation of analgesia from unwanted effects has long been an unmet goal of opioid research. Novel MOR agonist structures may prove critical for greater success. Here we tested metabolically stable analogs of the endomorphins, endogenous opioids highly selective for the MOR. Compared to morphine, the analogs showed dramatically improved analgesia-to-side-effect ratios. At doses providing equal or greater antinociception than morphine in the rat, the analogs showed reduced a) respiratory depression, b) impairment of motor coordination, c) tolerance and hyperalgesia, d) glial p38/CGRP/P2X7 receptor signaling, and e) reward/abuse potential in both conditioned place preference and self-administration tests. Differential effects on glial activation indicate a mechanism for the relative lack of side effects by the analogs compared to morphine. The results suggest that endomorphin analogs described here could provide gold standard pain relief mediated by selective MOR activation, but with remarkably safer side effect profiles compared to opioids like morphine.


Assuntos
Analgésicos Opioides/toxicidade , Discinesia Induzida por Medicamentos/fisiopatologia , Ativação de Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Morfina/toxicidade , Insuficiência Respiratória/induzido quimicamente , Transtornos Relacionados ao Uso de Substâncias/psicologia , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/farmacologia , Animais , Barreira Hematoencefálica/metabolismo , Condicionamento Operante/efeitos dos fármacos , Tolerância a Medicamentos , Hiperalgesia/induzido quimicamente , Hiperalgesia/psicologia , Masculino , Camundongos , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/efeitos dos fármacos , Insuficiência Respiratória/fisiopatologia
6.
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
7.
J Pharmacol Exp Ther ; 340(1): 56-63, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21972236

RESUMO

We studied adaptations to acute precipitated opioid withdrawal of spinal µ-opioid receptor (MOR)-coupled regulation of the release of endomorphin 2 (EM2). The release of this highly MOR-selective endogenous opioid from opioid-naive spinal tissue of male rats is subjected to MOR-coupled positive as well as negative modulation via cholera toxin-sensitive G(s) and pertussis toxin-sensitive G(i)/G(o), respectively. The net effect of this concomitant bidirectional modulation is inhibitory. MOR-coupled pleiotropic regulation of EM2 release is retained in opioid-withdrawn spinal tissue of male rats, but the balance of MOR-coupled inhibitory and facilitatory regulation shifted such that facilitatory regulation predominates. Augmented coupling of MOR to G(s) is causally associated with this change. Strikingly, pleiotropic characteristics of MOR-coupled regulation of spinal EM2 release and adaptations thereof to opioid withdrawal are male-specific. In females, MOR-coupled regulation of EM2 release from opioid-naive and -withdrawn spinal tissue does not have a significant G(s)-coupled facilitatory component, and MOR-coupled inhibition of EM2 release persists unabated in withdrawn preparations. The male-specific adaptations to chronic morphine that shift the relative predominance of opposing dual G protein-coupled MOR pathways provides a mechanism for mitigating inhibitory MOR signaling without losing MOR-coupled feedback regulation. These adaptations enable using endogenous EM2 as a substitute for morphine that had been precipitously removed. The sexually dimorphic functionality and regulation of spinal EM2/MOR-coupled signaling suggest the clinical utility of using sex-specific treatments for addiction that harness the activity of endogenous opioids.


Assuntos
Adaptação Fisiológica/fisiologia , Endorfinas/fisiologia , Oligopeptídeos/metabolismo , Coluna Vertebral/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Analgésicos Opioides/farmacologia , Animais , Western Blotting , Toxina da Cólera/administração & dosagem , Toxina da Cólera/farmacologia , Relação Dose-Resposta a Droga , Feminino , Imunoprecipitação , Masculino , Morfina/farmacologia , Antagonistas de Entorpecentes/farmacologia , Perfusão , Toxina Pertussis/administração & dosagem , Toxina Pertussis/farmacologia , Proteína Fosfatase 2/metabolismo , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides kappa/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Caracteres Sexuais , Sufentanil/farmacologia
8.
Prog Neuropsychopharmacol Biol Psychiatry ; 35(6): 1421-31, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21122812

RESUMO

Zebrafish (Danio rerio) are becoming increasingly popular in neurobehavioral research. Here, we summarize recent data on behavioral responses of adult zebrafish to a wide spectrum of putative anxiolytic and anxiogenic agents. Using the novel tank test as a sensitive and efficient behavioral assay, zebrafish anxiety-like behavior can be bi-directionally modulated by drugs affecting the gamma-aminobutyric acid, monoaminergic, cholinergic, glutamatergic and opioidergic systems. Complementing human and rodent data, zebrafish drug-evoked phenotypes obtained in this test support this species as a useful model for neurobehavioral and psychopharmacological research.


Assuntos
Ansiolíticos/farmacologia , Ansiolíticos/uso terapêutico , Ansiedade/tratamento farmacológico , Modelos Animais de Doenças , Transmissão Sináptica/efeitos dos fármacos , Animais , Ansiedade/induzido quimicamente , Comportamento Exploratório/efeitos dos fármacos , Humanos , Fenótipo , Peixe-Zebra
9.
Neuropeptides ; 44(6): 457-66, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21047684

RESUMO

Neuropeptides are essential for cell-cell communication in the nervous and neuroendocrine systems. Production of active neuropeptides requires proteolytic processing of proneuropeptide precursors in secretory vesicles that produce, store, and release neuropeptides that regulate physiological functions. This review describes recent findings indicating the prominent role of cathepsin L in secretory vesicles for production of neuropeptides from their protein precursors. The role of cathepsin L in neuropeptide production was discovered using the strategy of activity-based probes for proenkephalin-cleaving activity for identification of the enzyme protein by mass spectrometry. The novel role of cathepsin L in secretory vesicles for neuropeptide production has been demonstrated in vivo by cathepsin L gene knockout studies, cathepsin L gene expression in neuroendocrine cells, and notably, cathepsin L localization in neuropeptide-containing secretory vesicles. Cathepsin L is involved in producing opioid neuropeptides consisting of enkephalin, ß-endorphin, and dynorphin, as well as in generating the POMC-derived peptide hormones ACTH and α-MSH. In addition, NPY, CCK, and catestatin neuropeptides utilize cathepsin L for their biosynthesis. The neuropeptide-synthesizing functions of cathepsin L represent its unique activity in secretory vesicles, which contrasts with its role in lysosomes. Interesting evaluations of protease gene knockout studies in mice that lack cathepsin L compared to those lacking PC1/3 and PC2 (PC, prohormone convertase) indicate the key role of cathepsin L in neuropeptide production. Therefore, dual cathepsin L and prohormone convertase protease pathways participate in neuropeptide production. Significantly, the recent new findings indicate cathepsin L as a novel 'proprotein convertase' for production of neuropeptides that mediate cell-cell communication in health and disease.


Assuntos
Catepsina L/metabolismo , Neuropeptídeos/biossíntese , Vesículas Secretórias/enzimologia , Sequência de Aminoácidos , Animais , Catepsina L/genética , Encefalinas/genética , Encefalinas/metabolismo , Técnicas de Silenciamento de Genes , Dados de Sequência Molecular , Estrutura Molecular , Neuropeptídeos/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo
10.
J Pain ; 11(4): 388-98, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20350706

RESUMO

UNLABELLED: Nerve injury dramatically increases or decreases protein expression in the spinal cord dorsal horn. Whether the spatial distribution of these changes is restricted to the central innervation territories of injured nerves or could spread to adjacent territories in the dorsal horn is not understood. To address this question, we developed a simple computer software-assisted method to precisely distinguish and efficiently quantify immunohistochemical staining patterns across the mediolateral axis of the dorsal horn 2 weeks after transection of either the tibial and common peroneal nerves (thus sparing the sural branch, spared nerve injury, [SNI]), the tibial nerve, or the common peroneal and sural nerves. Using thiamine monophosphatase (TMP) histochemistry, we determined that central terminals of the tibial, common peroneal, sural, and posterior cutaneous nerves occupy the medial 35%, medial-central 20%, central-lateral 20%, and lateral 25% of the substantia gelatinosa, respectively. We then used these calculations to show that SNI reduced the expression of SP and TRPV1 immunoreactivity within the tibial and peroneal innervation territories in the L4 dorsal horn, without changing expression in the uninjured, sural sector. We conclude that SNI-induced loss of SP and TRPV1 in central terminals of dorsal horn is restricted to injured fibers. Our new method enables direct comparison of injured and uninjured terminals in the dorsal horn so as to better understand their relative contributions to mechanisms of chronic pain. PERSPECTIVE: A simple computer software-assisted algorithm was developed to precisely distinguish and efficiently quantify immunohistochemical staining patterns across the mediolateral axis of the dorsal horn after distal sciatic-branch transection. This method will facilitate a better understanding of the relative contribution of injured and uninjured terminals to mechanisms of chronic pain.


Assuntos
Doenças do Sistema Nervoso Periférico/metabolismo , Terminações Pré-Sinápticas/metabolismo , Raízes Nervosas Espinhais/metabolismo , Substância P/metabolismo , Substância Gelatinosa/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Modelos Animais de Doenças , Histocitoquímica/métodos , Processamento de Imagem Assistida por Computador/métodos , Masculino , Doenças do Sistema Nervoso Periférico/fisiopatologia , Terminações Pré-Sinápticas/ultraestrutura , Ratos , Ratos Sprague-Dawley , Neuropatia Ciática/metabolismo , Neuropatia Ciática/fisiopatologia , Raízes Nervosas Espinhais/citologia , Coloração e Rotulagem/métodos , Substância Gelatinosa/citologia , Transmissão Sináptica/fisiologia , Tiamina Monofosfato
11.
Mol Cell Neurosci ; 43(1): 98-107, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19837164

RESUMO

Dynorphin opioid neuropeptides mediate neurotransmission for analgesia and behavioral functions. Dynorphin A, dynorphin B, and alpha-neoendorphin are generated from prodynorphin by proteolytic processing. This study demonstrates the significant role of the cysteine protease cathepsin L for producing dynorphins. Cathepsin L knockout mouse brains showed extensive decreases in dynorphin A, dynorphin B, and alpha-neoendorphin that were reduced by 75%, 83%, and 90%, respectively, compared to controls. Moreover, cathepsin L in brain cortical neurons was colocalized with dynorphins in secretory vesicles, the primary site of neuropeptide production. Cellular coexpression of cathepsin L with prodynorphin in PC12 cells resulted in increased production of dynorphins A and B. Comparative studies of PC1/3 and PC2 convertases showed that PC1/3 knockout mouse brains had a modest decrease in dynorphin A, and PC2 knockout mice showed a minor decrease in alpha-neoendorphin. Overall, these results demonstrate a prominent role for cathepsin L, jointly with PC1/3 and PC2, for production of dynorphins in brain.


Assuntos
Catepsina L/metabolismo , Córtex Cerebral/metabolismo , Dinorfinas/metabolismo , Técnicas de Inativação de Genes , Pró-Proteína Convertase 1/genética , Pró-Proteína Convertase 2/genética , Animais , Catepsina L/genética , Córtex Cerebral/citologia , Dinorfinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , Células PC12 , Pró-Proteína Convertase 1/metabolismo , Pró-Proteína Convertase 2/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Ratos
12.
Brain Res ; 1245: 96-107, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18840417

RESUMO

Although opioids are known to influence sleep-wake regulation, the neuroanatomic substrate(s) mediating these effects remain unresolved. We hypothesized that the influence of opiates on sleep may be mediated, at least in part, by the ventrolateral preoptic nucleus (VLPO), a key cell group for producing behavioral sleep. By combining in situ hybridization for kappa and mu receptor mRNA with immunostaining of Fos expressed by VLPO cells during sleep we show that >85% of sleep-active VLPO neurons contain mRNA for either or both opioid receptors. Microinfusions of a kappa receptor agonist into the VLPO region increased NREM sleep by 51% during the subjective night, whereas a mu receptor agonist increased wakefulness by 60% during the subjective day. The sleep- and wake-promoting effects of the kappa and mu agonists were blocked by prior administration of their respective antagonist. Combining retrograde tracing from the VLPO with immunohistochemistry for dynorphin (Dyn, the endogenous kappa receptor agonist) or endomorphin 1 (EM1, the endogenous mu receptor agonist) we show that the central lateral parabrachial subnucleus (PBcl) provides Dyn inputs to the VLPO, whereas hypothalamic histaminergic neurons provide EM1 inputs to the VLPO. In summary, results from the present study suggest that central opioid inputs to the VLPO may play a role in sleep-wake regulation and that the VLPO likely mediates the hypnotic response to high levels of opioid analgesics.


Assuntos
Vias Neurais/fisiologia , Neurônios/metabolismo , Receptores Opioides/genética , Sono/fisiologia , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Animais , Dinorfinas/administração & dosagem , Dinorfinas/metabolismo , Dinorfinas/farmacologia , Eletroencefalografia , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Masculino , Antagonistas de Entorpecentes , Vias Neurais/efeitos dos fármacos , Vias Neurais/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurotransmissores/administração & dosagem , Neurotransmissores/metabolismo , Neurotransmissores/farmacologia , Oligopeptídeos/administração & dosagem , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Área Pré-Óptica/citologia , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/genética , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/genética , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Vigília/fisiologia
13.
J Comp Neurol ; 503(2): 319-33, 2007 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-17492626

RESUMO

The endomorphins are endogenous opioids with high affinity and selectivity for the mu opioid receptor (MOR, MOR-1, MOP). Endomorphin-1 (Tyr-Pro-Trp-Phe-NH(2); EM1) and endomorphin-2 (Tyr-Pro-Phe-Phe-NH(2); EM2) have been localized to many regions of the central nervous system (CNS), including those that regulate antinociception, autonomic function, and reward. Colocalization or shared distribution (overlap) of two neurotransmitters, or a transmitter and its cognate receptor, may imply an interaction of these elements in the regulation of functions mediated in that region. For example, previous evidence of colocalization of EM2 with substance P (SP), calcitonin gene-related peptide (CGRP), and MOR in primary afferent neurons suggested an interaction of these peptides in pain modulation. We therefore investigated the colocalization of EM1 and EM2 with SP, CGRP, and MOR in other areas of the CNS. EM2 was colocalized with SP and CGRP in the nucleus of the solitary tract (NTS) and with SP, CGRP and MOR in the parabrachial nucleus. Several areas in which EM1 and EM2 showed extensive shared distributions, but no detectable colocalization with other signaling molecules, are also described.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Oligopeptídeos/metabolismo , Receptores Opioides mu/metabolismo , Substância P/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Imuno-Histoquímica , Masculino , Neurônios/metabolismo , Peptídeos Opioides/metabolismo , Dor/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/agonistas , Medula Espinal/citologia , Medula Espinal/metabolismo , Distribuição Tecidual
14.
Brain Res ; 1059(2): 159-66, 2005 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-16223471

RESUMO

Opioids are among the most effective analgesics, but a major limitation for their therapeutic usefulness is their induction of respiratory depression. Endomorphin-1 (EM1), in contrast to several other mu opioids, exhibits a threshold for respiratory depression that is well above its threshold for analgesia. Its effect on sensitivity to CO(2), however, remains unknown. Minute ventilation (V(E)) in 2, 4, and 6% CO(2) was measured before and after systemic administration of EM1, endomorphin-2 (EM2), DAMGO, and morphine in the conscious rat. EM1 and EM2 attenuated the hypercapnic ventilatory response (HCVR) only in high doses, while DAMGO and morphine diminished the HCVR in much lower doses. The ventilatory effects of high doses of all 4 agonists were blocked by the mu-opioid antagonist naloxone (0.4 mg/kg i.v.), but not by the peripherally restricted mu-opioid antagonist, methyl-naloxone (0.4 mg/kg i.v.). It was concluded that the endomorphins attenuated the HCVR only in large doses, well beyond the analgesic threshold, and did so through a centrally mediated mu-opioid mechanism.


Assuntos
Analgésicos Opioides/farmacologia , Oligopeptídeos/farmacologia , Ventilação Pulmonar/efeitos dos fármacos , Receptores Opioides mu/agonistas , Respiração/efeitos dos fármacos , Adaptação Fisiológica , Animais , Dióxido de Carbono , Relação Dose-Resposta a Droga , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Hipercapnia/induzido quimicamente , Masculino , Morfina/farmacologia , Naloxona/farmacologia , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/prevenção & controle
15.
Brain Res ; 1028(2): 121-32, 2004 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-15527737

RESUMO

The human neuroblastoma cell line, SH-SY5Y, was used to examine the effects of morphine and the endogenous opioid peptides, endomorphin-1 (EM-1) and endomorphin-2 (EM-2), on mu opioid receptor (MOR) internalization and down-regulation. Treatment for 24 h with EM-1, EM-2 or morphine at 100 nM, 1 microM and 10 microM resulted in a dose-dependent down-regulation of mu receptors. Exposure of cells to 10 microM EM-1 for 2.5, 5 and 24 h resulted in a time-dependent down-regulation of mu receptors. Down-regulation of mu receptors by morphine and EM-1 was blocked by treatment with hypertonic sucrose, consistent with an endocytosis-dependent mechanism. Sensitive cell-surface binding studies with a radiolabeled mu antagonist revealed that morphine was able to induce internalization of mu receptors naturally expressed in SH-SY5Y cells. EM-1 produced a more rapid internalization of mu receptors than morphine, but hypertonic sucrose blocked the internalization induced by each of these agonists. This study demonstrates that, like morphine, the endomorphins down-regulate mu opioid receptors in a dose- and time-dependent manner. This study also demonstrates that morphine, as well as EM-1, can induce rapid, endocytosis-dependent internalization of mu opioid receptors in SH-SY5Y cells. These results may help elucidate the ability of mu agonists to regulate the number and responsiveness of their receptors.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Oligopeptídeos/farmacologia , Receptores Opioides mu/metabolismo , Analgésicos Opioides/farmacocinética , Sítios de Ligação/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Interações Medicamentosas , Endocitose/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacocinética , Humanos , Soluções Hipotônicas/farmacologia , Antagonistas de Entorpecentes/farmacocinética , Neuroblastoma , Peptídeos/farmacocinética , Ensaio Radioligante/métodos , Receptores Opioides mu/efeitos dos fármacos , Sacarose/farmacologia , Fatores de Tempo , Trítio/farmacocinética
16.
Exp Brain Res ; 156(2): 224-30, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15344852

RESUMO

Endomorphin-1 (EM-1) and endomorphin-2 (EM-2) are two highly selective mu-opiate receptor agonists. We recently demonstrated that EM-1 and EM-2 have a saturable transport system from brain-to-blood in vivo. Since the endothelial cells are the main component of the non-fenestrated microvessels of the blood-brain barrier (BBB), we examined whether these endogenous tetrapeptides have a saturable transport system in cultured cerebral endothelial cells. EM-1 and EM-2 binding and transport were studied in a transwell system in which primary mouse endothelial cells were co-cultured with rat glioma cells. We found that binding of both endomorphins was greater on the basolateral than the apical cell surface. Flux of EM-1 and EM-2 occurred predominantly in the basolateral to apical direction, each showing self-inhibition with an excess of the respective endomorphin. Transport was not influenced by the addition of the P-glycoprotein inhibitor, cyclosporin A. Neither the mu-opiate receptor agonist DAMGO nor the delta-opiate receptor agonist DPDPE had any effect on the transport. Thus, the results show that a saturable transport system for EM-1 and EM-2 occurs at the level of endothelial cells of the BBB, and unlike beta-endorphin and morphine, P-glycoprotein is not needed for the brain-to-blood transport. Cross-inhibition of the transport of each endomorphin by the other suggests a shared transport system that is different from mu- or delta-opiate receptors. As endormorphins are mainly produced in the CNS, the presence of the efflux system at the BBB could play an important role in pain modulation and neuroendocrine control.


Assuntos
Células Endoteliais/metabolismo , Neocórtex/metabolismo , Oligopeptídeos/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Neocórtex/citologia , Neocórtex/efeitos dos fármacos , Fatores de Tempo
17.
Neurosci Lett ; 366(2): 211-4, 2004 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-15276249

RESUMO

The present study examined the development of analgesic tolerance to endomorphin-1 (EM1), endomorphin-2 (EM2), and morphine, and cross-tolerance among these drugs. Male Swiss Webster mice were injected i.c.v. with EM1, EM2, morphine, or vehicle once daily for 5 days, and tested for analgesia in the tail flick test. To determine the extent of cross-tolerance, on the sixth day mice from each of the above groups received i.c.v. injections of EM1, EM2, morphine, or vehicle before analgesic testing. The development of tolerance to EM1 and EM2 closely resembled that of morphine. Complete, symmetrical cross-tolerance was observed between all drugs in the study. These results demonstrate a time-course and extent of tolerance similar to morphine, and support a common mechanism of action through the mu-opioid receptor.


Assuntos
Analgésicos Opioides/farmacologia , Tolerância a Medicamentos , Morfina/farmacologia , Oligopeptídeos/farmacologia , Analgésicos Opioides/administração & dosagem , Animais , Injeções Intraventriculares , Masculino , Camundongos , Morfina/administração & dosagem , Oligopeptídeos/administração & dosagem , Oligopeptídeos/fisiologia , Dor/tratamento farmacológico , Receptores Opioides mu/agonistas
18.
Brain Res ; 977(2): 190-8, 2003 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-12834879

RESUMO

The endomorphins represent a novel group of endogenous opioid peptides that have high affinity for the mu-opioid receptor (MOR1). Endomorphin-2 is present in high density in the spinal and trigeminal dorsal horns and is localized to primary afferents. If endomorphin-2 were an endogenous ligand for the MOR1, we would expect to find the receptor at cellular sites in close association with the peptide. We used dual-labeling immunocytochemical methods combined with electron microscopy to determine if a cellular substrate exists for functional interactions between endomorphin-2 and MOR1. We confirmed the localization of endomorphin-2 to unmyelinated axons and axon terminals in the trigeminal dorsal horn. A small proportion of these endomorphin-2 axons contained MOR1, but many of the dendritic targets of endomorphin-2 terminals contained MOR1. Consistent with previous studies, endomorphin-2 was contained primarily in dense core vesicles and MOR1 was located primarily at non-synaptic sites. These morphological characteristics are consistent with the hypothesis that peptides are released extra-synaptically and their receptors may be located at sites distal to the synaptic junction. These anatomical data support the hypothesis that endomorphin-2 is a ligand for MORs in the trigeminal dorsal horn, particularly at postsynaptic sites.


Assuntos
Dendritos/metabolismo , Oligopeptídeos/metabolismo , Terminações Pré-Sinápticas/metabolismo , Receptores Opioides mu/metabolismo , Núcleo Espinal do Trigêmeo/citologia , Animais , Dendritos/ultraestrutura , Imuno-Histoquímica/métodos , Masculino , Microscopia Eletrônica , Células do Corno Posterior/citologia , Terminações Pré-Sinápticas/ultraestrutura , Ratos , Ratos Sprague-Dawley
19.
Neurosci Lett ; 341(2): 107-10, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12686377

RESUMO

Endomorphin 2 is a newly discovered peptide that has high affinity and specificity for the mu-opioid receptor. One criterion for establishing that endomorphin serves as an endogenous agonist for the mu receptor is that it be anatomically distributed in close proximity to that receptor. We tested this idea with a preembedding double immunostaining technique to study synaptic relationships between them. The distributions of both endomorphin 2 and the mu-opioid receptor were similar in the dorsal horn of the cervical spinal cord at the light microscopic level. At the electron microscopic level, axon terminals with dense-cored vesicles containing endomorphin 2-like immunoreactivity were observed making mostly asymmetrical synapses on profiles immunostained for the mu-opioid receptor. The immunostaining for the mu-opioid receptor was found mostly in postsynaptic membranes in profiles having dendrite-like appearance. The results support the idea that endomorphin 2 is an endogenous ligand for the mu-opioid receptor. Furthermore, the results indicate that such a role is mediated at least in part through synaptic relationships.


Assuntos
Oligopeptídeos/farmacologia , Receptores Opioides mu/agonistas , Medula Espinal/efeitos dos fármacos , Animais , Masculino , Microscopia Imunoeletrônica/métodos , Oligopeptídeos/metabolismo , Células do Corno Posterior/metabolismo , Células do Corno Posterior/ultraestrutura , Ratos , Ratos Wistar , Receptores Opioides mu/metabolismo , Receptores Opioides mu/ultraestrutura , Vesículas Secretórias/metabolismo , Vesículas Secretórias/ultraestrutura , Medula Espinal/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/ultraestrutura
20.
J Comp Neurol ; 454(3): 320-8, 2002 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-12442322

RESUMO

Endomorphin-1 and -2 (EM1, EM2) are endogenous opioids with high affinity and selectivity for the mu-opioid receptor. Cells expressing EM-like immunoreactivity (EM-LI) are present in the hypothalamus, and fibers containing EM-LI project to many brain regions, including the ventral tegmental area (VTA). The VTA is one of the most sensitive brain regions for the rewarding and locomotor effects of opioids. It contains mu-opioid receptors, which are thought to mediate gamma-aminobutyric acid-dependent disinhibition of dopamine transmission to the nucleus accumbens. We investigated whether hypothalamic EM-LI cells project to the VTA, where they could play a natural role in this circuitry. The retrograde tracer Fluoro-Gold (FG) was microinjected into the anterior or posterior VTA in rats. Nine days later, colchicine was injected, and 24 hours later, the animals were perfused and processed for fluorescence immunocytochemistry. Numerous FG-labeled cells were detected in the hypothalamus. Both EM1-LI and EM2-LI cells were present in the periventricular nucleus, between the dorsomedial and ventromedial hypothalamus and between the ventromedial and arcuate nuclei. Subpopulations of EM1-LI and EM2-LI cells were labeled by FG. Injections of FG to the anterior and posterior VTA were both effective in producing double-labeled cells, and an anterior-posterior topographical organization between the VTA and hypothalamus was observed. The results support the idea that some endomorphin-containing neurons in the hypothalamus project to the VTA, where they may modulate reward and locomotor circuitry.


Assuntos
Corantes Fluorescentes/análise , Hipotálamo/química , Oligopeptídeos/análise , Estilbamidinas , Área Tegmentar Ventral/química , Animais , Imuno-Histoquímica , Masculino , Vias Neurais/química , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...