Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Mol Pain ; 19: 17448069231183902, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37285551

RESUMO

Background: Opioids are efficacious and safe analgesic drugs in short-term use for acute pain but chronic use can lead to tolerance and dependence. Opioid-induced microglial activation may contribute to the development of tolerance and this process may differ between males and females. A link is suggested between this microglial activation and inflammation, disturbances of circadian rhythms, and neurotoxic effects. We set out to further delineate the effects of chronic morphine on pain behaviour, microglial and neuronal staining, and the transcriptome of spinal microglia, to better understand the role of microglia in the consequences of long-term high-dose opioid administration. Experimental Approach: In two experiments, we administered increasing subcutaneous doses of morphine hydrochloride or saline to male and female rats. Thermal nociception was assessed with the tail flick and hot plate tests. In Experiment I, spinal cord (SC) samples were prepared for immunohistochemical staining for microglial and neuronal markers. In Experiment II, the transcriptome of microglia from the lumbar SC was analysed. Key Results: Female and male rats had similar antinociceptive responses to morphine and developed similar antinociceptive tolerance to thermal stimuli following chronic increasing high doses of s.c. morphine. The area of microglial IBA1-staining in SC decreased after 2 weeks of morphine administration in both sexes. Following morphine treatment, the differentially expressed genes identified in the microglial transcriptome included ones related to the circadian rhythm, apoptosis, and immune system processes. Conclusions: Female and male rats showed similar pain behaviour following chronic high doses of morphine. This was associated with decreased staining of spinal microglia, suggesting either decreased activation or apoptosis. High-dose morphine administration also associated with several changes in gene expression in SC microglia, e.g., those related to the circadian rhythm (Per2, Per3, Dbp). These changes should be considered in the clinical consequences of long-term high-dose administration of opioids.


Assuntos
Analgésicos Opioides , Morfina , Ratos , Masculino , Feminino , Animais , Morfina/uso terapêutico , Analgésicos Opioides/farmacologia , Analgésicos Opioides/uso terapêutico , Microglia , Transcriptoma/genética , Analgésicos/farmacologia , Dor/metabolismo , Medula Espinal/metabolismo
2.
J Neurosci Res ; 100(1): 329-338, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-32459013

RESUMO

Opioids are effective analgesics in the management of severe pain. However, tolerance, leading to dose escalation and adverse effects are significant limiting factors in their use. The role of peripheral opioid receptors in analgesia has been discussed especially under inflammatory conditions. The results from pharmacological and conditional knockout studies together do not provide a clear picture of the contribution of peripheral opioid receptors on antinociceptive tolerance and this needs to be evaluated. Therefore, we studied whether the peripherally restricted opioid receptor antagonist, methylnaltrexone (MNTX), could prevent morphine tolerance without attenuating the antinociceptive effect of morphine. Male Sprague-Dawley rats were treated for 7 days with increasing subcutaneous doses of morphine (5-30 mg/kg) and were coadministered saline, MNTX (0.5 or 2 mg/kg), or naltrexone (NTX; 2 mg/kg). Nociception was assessed with tail-flick, hotplate, and von Frey tests. Morphine, MNTX, and NTX concentrations in the plasma, brain, and spinal cord were measured by liquid chromatography-tandem mass spectrometry. In acute coadministration, NTX, but not MNTX, abolished the acute antinociceptive effects of morphine in all nociceptive tests. The antinociceptive tolerance after repeated morphine administration was also prevented by NTX but not by MNTX. MNTX penetrated to the spinal cord and the brain to some extent after repeated administration. The results do not support the use of MNTX for preventing opioid tolerance and also suggest that morphine tolerance is mediated by central rather than peripheral opioid receptors in the rat.


Assuntos
Morfina , Naltrexona , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Masculino , Morfina/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Compostos de Amônio Quaternário , Ratos , Ratos Sprague-Dawley , Receptores Opioides , Receptores Opioides mu
3.
J Neurophysiol ; 124(3): 790-801, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32755331

RESUMO

Descending facilitatory circuitry that involves the rostroventromedial medulla (RVM) exerts a significant role in the development of antinociceptive tolerance and hyperalgesia following chronic morphine treatment. The role of the RVM in the development of antinociceptive tolerance to oxycodone, another clinically used strong opioid, is not yet known. Ketamine, an N-methyl-d-aspartate (NMDA) receptor antagonist, attenuates opioid antinociceptive tolerance, but its effect on RVM cell discharge in opioid-tolerant animals is not known. Here, we compared chronic effects of morphine and oxycodone on the discharge properties of RVM cells and attempted to attenuate chronic treatment-induced changes with ketamine. Parallel recordings of RVM cell discharge and limb withdrawal response were performed under light pentobarbital anesthesia in male rats following sustained systemic treatment with morphine or oxycodone at equianalgesic doses. Ongoing activity and the response to noxious heat and pinch were determined in pronociceptive RVM ON-cells and antinociceptive OFF-cells on the sixth treatment day. Proportions of RVM cell types were not changed. Chronic oxycodone induced antinociceptive tolerance both in limb withdrawal and RVM cell activity. Chronic morphine induced antinociceptive tolerance in limb withdrawal that was accompanied by pronociceptive heat response changes in RVM ON- and OFF-cells. A behaviorally subantinociceptive dose of acute ketamine reversed antinociceptive tolerance both to morphine and oxycodone in limb withdrawal and reversed the chronic morphine-induced pronociceptive discharge changes in RVM cells. The results indicate that an NMDA receptor-dependent descending pronociceptive circuitry involving the RVM has an important role in behavioral antinociceptive tolerance to morphine but not oxycodone.NEW & NOTEWORTHY Morphine and oxycodone are two clinically used strong opioids. Chronic treatment with oxycodone as well as morphine can lead to analgesic tolerance and paradoxical hyperalgesia. Here we show that an N-methyl-d-aspartate receptor-dependent pronociceptive change in discharge properties of rostroventromedial medullary neurons controlling spinal nociception has an important role in antinociceptive tolerance to morphine but not oxycodone. Interestingly, chronic oxycodone did not induce pronociceptive changes in the rostroventromedial medulla.


Assuntos
Analgésicos Opioides/farmacologia , Tolerância a Medicamentos , Hiperalgesia/induzido quimicamente , Ketamina/farmacologia , Bulbo/efeitos dos fármacos , Morfina/farmacologia , Nociceptividade/efeitos dos fármacos , Oxicodona/farmacologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Analgésicos Opioides/administração & dosagem , Animais , Comportamento Animal/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios , Ketamina/administração & dosagem , Masculino , Morfina/administração & dosagem , Oxicodona/administração & dosagem , Ratos , Ratos Sprague-Dawley
4.
Mol Pain ; 16: 1744806920950866, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32811276

RESUMO

The glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) alleviate symptoms of experimental neuropathy, protect and stimulate regeneration of sensory neurons in animal models of neuropathic pain, and restore their functional activity. However, clinical development of GFL proteins is complicated by their poor pharmacokinetic properties and multiple effects mediated by several receptors. Previously, we have identified a small molecule that selectively activates the major signal transduction unit of the GFL receptor complex, receptor tyrosine kinase RET, as an alternative to GFLs, for the treatment of neuropathic pain. We then introduced a series of chemical changes to improve the biological activity of these compounds and tested an optimized compound named BT44 in a panel of biological assays. BT44 efficiently and selectively stimulated the GFL receptor RET and activated the intracellular mitogene-activated protein kinase/extracellular signal-regulated kinase pathway in immortalized cells. In cultured sensory neurons, BT44 stimulated neurite outgrowth with an efficacy comparable to that of GFLs. BT44 alleviated mechanical hypersensitivity in surgery- and diabetes-induced rat models of neuropathic pain. In addition, BT44 normalized, to a certain degree, the expression of nociception-related neuronal markers which were altered by spinal nerve ligation, the neuropathy model used in this study. Our results suggest that the GFL mimetic BT44 is a promising new lead for the development of novel disease-modifying agents for the treatment of neuropathy and neuropathic pain.


Assuntos
Biomimética/métodos , Neuralgia/tratamento farmacológico , Proteínas Proto-Oncogênicas c-ret/agonistas , Proteínas Proto-Oncogênicas c-ret/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Nervos Espinhais/efeitos dos fármacos , Animais , Escala de Avaliação Comportamental , Linhagem Celular , Neuropatias Diabéticas/tratamento farmacológico , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fatores Neurotróficos Derivados de Linhagem de Célula Glial , Imuno-Histoquímica , Masculino , Proteínas do Tecido Nervoso/metabolismo , Neuralgia/metabolismo , Nociceptividade/efeitos dos fármacos , Fosforilação , Ratos , Ratos Wistar , Células Receptoras Sensoriais/metabolismo , Nervos Espinhais/lesões
5.
IBRO Neurosci Rep ; 17: 38-51, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38933596

RESUMO

First-line pharmacotherapy for peripheral neuropathic pain (NP) of diverse pathophysiology consists of antidepressants and gabapentinoids, but only a minority achieve sufficient analgesia with these drugs. Opioids are considered third-line analgesics in NP due to potential severe and unpredictable adverse effects in long-term use. Also, opioid tolerance and NP may have shared mechanisms, raising further concerns about opioid use in NP. We set out to further elucidate possible shared and separate mechanisms after chronic morphine treatment and oxaliplatin-induced and diabetic polyneuropathies, and to identify potential diagnostic markers and therapeutic targets. We analysed thermal nociceptive behaviour, the transcriptome of dorsal root ganglia (DRG) and the metabolome of cerebrospinal fluid (CSF) in these three conditions, in rats. Several genes were differentially expressed, most following oxaliplatin and least after chronic morphine treatment, compared with saline-treated rats. A few genes were differentially expressed in the DRGs in all three models (e.g. Csf3r and Fkbp5). Some, e.g. Alox15 and Slc12a5, were differentially expressed in both diabetic and oxaliplatin models. Other differentially expressed genes were associated with nociception, inflammation, and glial cells. The CSF metabolome was most significantly affected in the diabetic rats. Interestingly, we saw changes in nicotinamide metabolism, which has been associated with opioid addiction and withdrawal, in the CSF of morphine-tolerant rats. Our results offer new hypotheses for the pathophysiology and treatment of NP and opioid tolerance. In particular, the role of nicotinamide metabolism in opioid addiction deserves further study.

6.
Anesth Analg ; 114(6): 1353-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22556211

RESUMO

BACKGROUND: Opioid analgesics are effective in the treatment of chronic pain, but they have serious adverse effects such as development of tolerance and dependence. Adrenergic α(2) agonists and µ-opioid receptor agonists show synergistic potentiation and cross-tolerance in spinal analgesia, whereas α(2)-adrenergic antagonists have shown pronociceptive effects. However, at ultralow doses, spinal α(2)-adrenergic antagonists have been reported to paradoxically enhance opioid antinociception. New data have suggested a functional µ-opioid-α(2)-adrenoceptor complex, which may help in interpreting the paradoxical effect of the α(2)-adrenergic antagonists. In the present study we assessed the effects of low doses of atipamezole, a nonselective α(2)-adrenergic antagonist, on both systemic and spinal morphine antinociception and tolerance. METHODS: Antinociception was assessed in male Sprague-Dawley rats using hotplate, tail-flick, and paw pressure tests. Spinal or systemic opioid tolerance was induced for 4 days. The effects of both intrathecal and subcutaneous atipamezole on acute morphine-induced antinociception and established morphine tolerance were studied. RESULTS: Systemic or spinal atipamezole itself did not produce antinociception at the doses studied (subcutaneous 0.03, 0.3, 3 µg/kg or intrathecal 0.1, 1, 10 ng). The combined administration of spinal morphine and 1 ng of atipamezole increased the antinociceptive effect of acute spinal morphine 30 minutes after the administration of test drugs in the tail-flick test. Furthermore, 10 ng of intrathecal atipamezole attenuated established morphine tolerance 30 minutes after the administration of test drugs in the tail-flick test. However, subcutaneous atipamezole had no significant effect on systemic morphine antinociception, and it did not attenuate morphine tolerance. CONCLUSIONS: Spinal coadministration of low doses of atipamezole augmented the antinociceptive effect of morphine in naïve and tolerant rats. Heterodimerization of µ-opioid- and α(2A)-adrenoceptors with consequent changes in function and interaction could explain these results. This also suggests an interesting explanation for the variability in opioid response and tolerance in patients experiencing stress or having an increased noradrenergic tone due to other causes, e.g., drugs.


Assuntos
Antagonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Analgésicos Opioides/administração & dosagem , Imidazóis/administração & dosagem , Morfina/administração & dosagem , Dor/prevenção & controle , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Tolerância a Medicamentos , Temperatura Alta , Injeções Espinhais , Injeções Subcutâneas , Masculino , Dor/diagnóstico , Dor/etiologia , Dor/fisiopatologia , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Pressão , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
7.
J Control Release ; 344: 214-224, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35301056

RESUMO

The blood-brain barrier significantly limits effective drug delivery to central nervous system (CNS) targets. The recently characterized glymphatic system offers a perivascular highway for intrathecally (i.t.) administered drugs to reach deep brain structures. Although periarterial cerebrospinal fluid (CSF) influx and concomitant brain drug delivery can be enhanced by pharmacological or hyperosmotic interventions, their effects on drug delivery to the spinal cord, an important target for many drugs, have not been addressed. Hence, we studied in rats whether enhancement of periarterial flow by systemic hypertonic solution might be utilized to enhance spinal delivery and efficacy of i.t. morphine. We also studied whether the hyperosmolar intervention affects brain or cerebrospinal fluid drug concentrations after systemic administration. Periarterial CSF influx was enhanced by intraperitoneal injection of hypertonic saline (HTS, 5.8%, 20 ml/kg, 40 mOsm/kg). The antinociceptive effects of morphine were characterized, using tail flick, hot plate and paw pressure tests. Drug concentrations in serum, tissue and microdialysis samples were determined by liquid chromatography-tandem mass spectrometry. Compared with isotonic solution, HTS increased concentrations of spinal i.t. administered morphine by 240% at the administration level (T13-L1) at 60 min and increased the antinociceptive effect of morphine in tail flick, hot plate, and paw pressure tests. HTS also independently increased hot plate and paw pressure latencies but had no effect in the tail flick test. HTS transiently increased the penetration of intravenous morphine into the lateral ventricle, but not into the hippocampus. In conclusion, acute systemic hyperosmolality is a promising intervention for enhanced spinal delivery of i.t. administered morphine. The relevance of this intervention should be expanded to other i.t. drugs and brought to clinical trials.


Assuntos
Morfina , Medula Espinal , Animais , Injeções Espinhais , Medição da Dor , Ratos , Ratos Sprague-Dawley
8.
Biomed Chromatogr ; 24(3): 294-300, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19629963

RESUMO

Novel HPLC method utilizing UV-detection was developed to analyse catechol-O-methyltransferase (COMT) products, vanillic acid and isovanillic acid, S-adenosylhomocysteine (SAH) and adenosine formed from dihydroxybenzoic acid and S-adenosyl-L-methionine (SAM) by incubation of the rat tissues. Entacapone, a COMT inhibitor, prevented the formation of SAH only partially in the striatal homogenate whereas in the kidney homogenate the increase of SAH was prevented by entacapone. In conclusion, this method was reliable, rapid and simple. COMT seemed to be partially responsible on the SAM utilizing methylations in the striatal homogenates while in the high COMT activity tissue, COMT was the main SAH producing methyltransferase.


Assuntos
Adenosina/metabolismo , Catecol O-Metiltransferase/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , S-Adenosil-Homocisteína/metabolismo , Adenosina/análise , Animais , Ratos , Ratos Wistar , S-Adenosil-Homocisteína/análise , S-Adenosilmetionina/metabolismo , Ácido Vanílico/análise , Ácido Vanílico/metabolismo
9.
Eur J Pharmacol ; 875: 173021, 2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-32112778

RESUMO

Morphine-3-glucuronide (M3G), the main metabolite of morphine, has been implicated in the development of tolerance and of opioid-induced hyperalgesia, both limiting the analgesic use of morphine. We evaluated the acute and chronic effects of M3G and morphine as well as development of antinociceptive cross-tolerance between morphine and M3G after intrathecal administration and assessed the expression of pain-associated neurotransmitter substance P in the spinal cord. Sprague-Dawley rats received intrathecal M3G or morphine twice daily for 6 days. Nociception and tactile allodynia were measured with von Frey filaments after acute and chronic treatments. Substance P levels in the dorsal horn of the spinal cord were determined by immunohistochemistry after 4-day treatments. Acute morphine caused antinociception as expected, whereas acute M3G caused tactile allodynia, as did both chronic M3G and morphine. Chronic M3G also induced antinociceptive cross-tolerance to morphine. M3G and morphine increased substance P levels similarly in the nociceptive laminae of the spinal cord. This study shows that chronic intrathecal M3G sensitises animals to mechanical stimulation and elevates substance P levels in the nociceptive laminae of the spinal cord. Chronic M3G also induces antinociceptive cross-tolerance to morphine. Thus, chronic M3G exposure might contribute to morphine-induced tolerance and opioid-induced hyperalgesia.


Assuntos
Estimulantes do Sistema Nervoso Central/farmacologia , Hiperalgesia/induzido quimicamente , Derivados da Morfina/farmacologia , Morfina/farmacologia , Nociceptividade/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Esquema de Medicação , Tolerância a Medicamentos , Humanos , Hiperalgesia/diagnóstico , Injeções Espinhais , Masculino , Morfina/metabolismo , Derivados da Morfina/metabolismo , Medição da Dor , Ratos , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Substância P/metabolismo
10.
Anesthesiology ; 111(6): 1356-64, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19934882

RESUMO

BACKGROUND: Opioid analgesics are effective in relieving chronic pain, but they have serious adverse effects, including development of tolerance and dependence. Ibudilast, an inhibitor of glial activation and cyclic nucleotide phosphodiesterases, has shown potential in the treatment of neuropathic pain and opioid withdrawal. Because glial cell activation could also be involved in the development of opioid tolerance in rats, the authors studied the antinociceptive effects of ibudilast and morphine in different models of coadministration. METHODS: Antinociception was assessed using male Sprague- Dawley rats in hot plate and tail-flick tests. The effects of ibudilast on acute morphine-induced antinociception, induction of morphine tolerance, and established morphine tolerance were studied. RESULTS: Systemic ibudilast produced modest dose-related antinociception and decreased locomotor activity at the studied doses of 2.5-22.5 mg/kg. The highest tested dose of 22.5 mg/kg produced 52% of the maximum possible effect in the tail-flick test. It had an additive antinociceptive effect when combined with systemic morphine. Coadministration of ibudilast with morphine did not attenuate the development of morphine tolerance. However, in morphine-tolerant rats, ibudilast partly restored morphine-induced antinociception. CONCLUSIONS: Ibudilast produces modest antinociception, and it is effective in restoring but not in preventing morphine tolerance. The mechanisms of the effects of ibudilast should be better understood before it is considered for clinical use.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Piridinas/farmacologia , Animais , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Tolerância a Medicamentos , Masculino , Atividade Motora/efeitos dos fármacos , Medição da Dor/efeitos dos fármacos , Equilíbrio Postural/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos
11.
J Control Release ; 304: 29-38, 2019 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-31067483

RESUMO

Drug delivery to the central nervous system remains a major problem due to biological barriers. The blood-brain-barrier can be bypassed by administering drugs intrathecally directly to the cerebrospinal fluid (CSF). The glymphatic system, a network of perivascular spaces promoting fluid exchange between CSF and interstitial space, could be utilized to enhance convective drug delivery from the CSF to the parenchyma. Glymphatic flow is highest during sleep and anesthesia regimens that induce a slow-wave sleep-like state. Here, using mass spectrometry and fluorescent imaging techniques, we show that the clinically used α2-adrenergic agonist dexmedetomidine that enhances EEG slow-wave activity, increases brain and spinal cord drug exposure of intrathecally administered drugs in mice and rats. Using oxycodone, naloxone, and an IgG-sized antibody as relevant model drugs we demonstrate that modulation of glymphatic flow has a distinct impact on the distribution of intrathecally administered therapeutics. These findings can be exploited in the clinic to improve the efficacy and safety of intrathecally administered therapeutics.


Assuntos
Encéfalo/metabolismo , Dexmedetomidina/administração & dosagem , Sistemas de Liberação de Medicamentos , Sistema Glinfático/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Anticorpos/administração & dosagem , Anticorpos/metabolismo , Dexmedetomidina/farmacologia , Sistema Glinfático/metabolismo , Injeções Espinhais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Naloxona/administração & dosagem , Naloxona/farmacocinética , Oxicodona/administração & dosagem , Oxicodona/farmacocinética , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
12.
Free Radic Biol Med ; 45(6): 749-55, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18572022

RESUMO

Nitrosation of enzyme regulatory cysteines is one of the key posttranslational modification mechanisms of enzyme function. Frequently such modifications are readily reversible; however, cysteine proteases, such as cathepsin B, have been shown to be covalently and permanently inactivated by nitroxyl (HNO), the one-electron reduction product of NO. Owing to the high reactivity of HNO with NO, endogenous NO production could provide direct protection for the less reactive protein cysteines by scavenging HNO. Additionally, endogenous cellular production of NO could rescue enzyme function by protective nitrosation of cysteines prior to exposure to HNO. Thus, we studied the effect of endogenous NO production, induced by LPS or IFN-gamma, on inhibition of cysteine protease cathepsin B in RAW macrophages. Both LPS and IFN-gamma induce iNOS with generation of nitrate up to 9 muM in the media after a 24-h stimulation, while native RAW 264.7 macrophages neither express iNOS nor generate nitrate. After the 24-h stimulation, the HNO-releasing Angeli's salt (0-316 microM) caused dose-dependent and DTT-irreversible loss of cathepsin B activity, and induction of iNOS activity did not protect the enzyme. The lack of protection was also verified in an in vitro setup, where papain, a close structural analogue of cathepsin B, was inhibited by Angeli's salt (2.7 microM) in the presence of the NO donor DEA/NO (0-316 microM). This clearly showed that a high molar excess of DEA/NO (EC(50) 406 microM) is needed to protect papain from the DTT-irreversible covalent modification by HNO. Our results provide first evidence on a cellular level for the remarkably high sensitivity of active-site cysteines in cysteine proteases for modification by HNO.


Assuntos
Catepsina B/antagonistas & inibidores , Óxido Nítrico/fisiologia , Óxidos de Nitrogênio/metabolismo , Animais , Western Blotting , Catepsina B/metabolismo , Linhagem Celular , Glutationa/farmacologia , Imuno-Histoquímica , Interferon gama/farmacologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/metabolismo , Camundongos
13.
Basic Clin Pharmacol Toxicol ; 122(5): 481-488, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29171155

RESUMO

Ketamine and its primary metabolite norketamine attenuate morphine tolerance by antagonising N-methyl-d-aspartate (NMDA) receptors. Ketamine is extensively metabolized to several other metabolites. The major secondary metabolite (2S,6S;2R,6R)-hydroxynorketamine (6-hydroxynorketamine) is not an NMDA antagonist. However, it may modulate nociception through negative allosteric modulation of α7 nicotinic acetylcholine receptors. We studied whether 6-hydroxynorketamine could affect nociception or the effects of morphine in acute or chronic administration settings. Male Sprague Dawley rats received subcutaneous 6-hydroxynorketamine or ketamine alone or in combination with morphine, as a cotreatment during induction of morphine tolerance, and after the development of tolerance induced by subcutaneous minipumps administering 9.6 mg morphine daily. Tail flick, hot plate, paw pressure and rotarod tests were used. Brain and serum drug concentrations were quantified with high-performance liquid chromatography-tandem mass spectrometry. Ketamine (10 mg/kg), but not 6-hydroxynorketamine (10 and 30 mg/kg), enhanced antinociception and decreased rotarod performance following acute administration either alone or combined with morphine. Ketamine efficiently attenuated morphine tolerance. Acutely administered 6-hydroxynorketamine increased the brain concentration of morphine (by 60%), and brain and serum concentrations of 6-hydroxynorketamine were doubled by morphine pre-treatment. This pharmacokinetic interaction did not, however, lead to altered morphine tolerance. Co-administration of 6-hydroxynorketamine 20 mg/kg twice daily did not influence development of morphine tolerance. Even though morphine and 6-hydroxynorketamine brain concentrations were increased after co-administration, the pharmacokinetic interaction had no effect on acute morphine nociception or tolerance. These results indicate that 6-hydroxynorketamine does not have antinociceptive properties or attenuate opioid tolerance in a similar way as ketamine.


Assuntos
Analgésicos Opioides/farmacologia , Anestésicos Dissociativos/farmacologia , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Tolerância a Medicamentos , Ketamina/análogos & derivados , Morfina/farmacologia , Dor Nociceptiva/prevenção & controle , Analgésicos Opioides/sangue , Analgésicos Opioides/farmacocinética , Anestésicos Dissociativos/sangue , Anestésicos Dissociativos/farmacocinética , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Interações Medicamentosas , Ketamina/sangue , Ketamina/farmacocinética , Ketamina/farmacologia , Masculino , Morfina/sangue , Morfina/farmacocinética , Atividade Motora/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Dor Nociceptiva/sangue , Dor Nociceptiva/fisiopatologia , Dor Nociceptiva/psicologia , Limiar da Dor/efeitos dos fármacos , Ratos Sprague-Dawley
14.
Neuroscience ; 375: 10-24, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29421434

RESUMO

Development of tolerance is a well known pharmacological characteristic of opioids and a major clinical problem. In addition to the known neuronal mechanisms of opioid tolerance, activation of glia has emerged as a potentially significant new mechanism. We studied activation of microglia and astrocytes in morphine tolerance and opioid-induced hyperalgesia in rats using immunohistochemistry, flow cytometry and RNA sequencing in spinal- and supraspinal regions. Chronic morphine treatment that induced tolerance and hyperalgesia also increased immunoreactivity of spinal microglia in the dorsal and ventral horns. Flow cytometry demonstrated that morphine treatment increased the proportion of M2-polarized spinal microglia, but failed to impact the number or the proportion of M1-polarized microglia. In the transcriptome of microglial cells isolated from the spinal cord (SC), morphine treatment increased transcripts related to cell activation and defense response. In the studied brain regions, no activation of microglia or astrocytes was detected by immunohistochemistry, except for a decrease in the number of microglial cells in the substantia nigra. In flow cytometry, morphine caused a decrease in the number of microglial cells in the medulla, but otherwise no change was detected for the count or the proportion of M1- and M2-polarized microglia in the medulla or sensory cortex. No evidence for the activation of glia in the brain was seen. Our results suggest that glial activation associated with opioid tolerance and opioid-induced hyperalgesia occurs mainly at the spinal level. The transcriptome data suggest that the microglial activation pattern after chronic morphine treatment has similarities with that of neuropathic pain.


Assuntos
Analgésicos Opioides/farmacologia , Encéfalo/efeitos dos fármacos , Morfina/farmacologia , Neuroglia/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Tolerância a Medicamentos , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Masculino , Modelos Animais , Neuroglia/metabolismo , Neuroglia/patologia , Dor Nociceptiva/tratamento farmacológico , Dor Nociceptiva/metabolismo , Dor Nociceptiva/patologia , Ratos Sprague-Dawley , Medula Espinal/metabolismo , Medula Espinal/patologia , Transcriptoma/efeitos dos fármacos
15.
Neurochem Int ; 51(6-7): 412-23, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17573159

RESUMO

Activated microglial cells are found in the substantia nigra and the striatum of Parkinson's disease patients. These cells have been shown to express catechol-O-methyltransferase activity which may increase during pathological conditions. Lipopolysaccharides are potent activators of microglial cells. After paranigral lipopolysaccharide infusion to rats we observed intense microglial activation around the lesion area followed by a delayed injury in nigrostriatal pathway in 2 weeks. Simultaneously, catechol-O-methyltransferase activity in the substantia nigra was gradually increased up to 213%. In the Western blot the amount of soluble COMT and membrane bound COMT proteins were increased by 255% and 86%, respectively. Increased catechol-O-methyltransferase immunoreactivity was located primarily into the activated microglial cells in the lesion area. Interestingly, catechol-O-methyltransferase and OX-42 stained also intensively microglia/macrophage-like cells which surrounded the adjacent blood vessels. Inhibition of catechol-O-methyltransferase activity by tolcapone or entacapone did not increase lipopolysaccharide-induced neurotoxicity. We conclude that catechol-O-methyltransferase activity and protein expression were increased in the substantia nigra after inflammation induced by lipopolysaccharides. These changes in glial and perivascular catechol-O-methyltransferase activity may have clinical relevance for Parkinson's disease drug treatment due to increased metabolism of levodopa in the brain.


Assuntos
Catecol O-Metiltransferase/metabolismo , Dopamina/metabolismo , Encefalite/enzimologia , Gliose/enzimologia , Microglia/enzimologia , Substância Negra/enzimologia , Animais , Biomarcadores/metabolismo , Antígeno CD11b/metabolismo , Encefalite/induzido quimicamente , Encefalite/fisiopatologia , Ativação Enzimática/fisiologia , Gliose/induzido quimicamente , Gliose/fisiopatologia , Imuno-Histoquímica , Mediadores da Inflamação/farmacologia , Levodopa/metabolismo , Levodopa/farmacologia , Levodopa/uso terapêutico , Lipopolissacarídeos/farmacologia , Masculino , Microglia/efeitos dos fármacos , Doença de Parkinson/enzimologia , Doença de Parkinson/fisiopatologia , Ratos , Ratos Wistar , Substância Negra/fisiopatologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
16.
Basic Clin Pharmacol Toxicol ; 120(1): 38-45, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27312359

RESUMO

Spironolactone, eplerenone, chlorothiazide and furosemide are diuretics that have been suggested to have antinociceptive properties, for example via mineralocorticoid receptor antagonism. In co-administration, diuretics might enhance the antinociceptive effect of opioids via pharmacodynamic and pharmacokinetic mechanisms. Effects of spironolactone (100 mg/kg, i.p.), eplerenone (100 mg/kg, i.p.), chlorothiazide (50 mg/kg, i.p.) and furosemide (100 mg/kg, i.p.) were studied on acute oxycodone (0.75 mg/kg, s.c.)- and morphine (3 mg/kg, s.c.)-induced antinociception using tail-flick and hot plate tests in male Sprague Dawley rats. The diuretics were administered 30 min. before the opioids, and behavioural tests were performed 30 and 90 min. after the opioids. Concentrations of oxycodone, morphine and their major metabolites in plasma and brain were quantified by mass spectrometry. In the hot plate test at 30 and 90 min., spironolactone significantly enhanced the antinociceptive effect (% of maximum possible effect) of oxycodone from 10% to 78% and from 0% to 50%, respectively, and that of morphine from 12% to 73% and from 4% to 83%, respectively. The brain oxycodone and morphine concentrations were significantly increased at 30 min. (oxycodone, 46%) and at 90 min. (morphine, 190%). We did not detect any independent antinociceptive effects with the diuretics. Eplerenone and chlorothiazide did not enhance the antinociceptive effect of either opioid. The results suggest that spironolactone enhances the antinociceptive effect of both oxycodone and morphine by increasing their concentrations in the central nervous system.


Assuntos
Analgésicos/uso terapêutico , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Diuréticos/uso terapêutico , Neurônios/efeitos dos fármacos , Dor/prevenção & controle , Espironolactona/uso terapêutico , Analgésicos Opioides/sangue , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Clorotiazida/uso terapêutico , Interações Medicamentosas , Quimioterapia Combinada , Eplerenona , Furosemida/uso terapêutico , Masculino , Morfina/sangue , Morfina/metabolismo , Morfina/farmacocinética , Morfina/uso terapêutico , Neurônios/metabolismo , Oxicodona/sangue , Oxicodona/metabolismo , Oxicodona/farmacocinética , Oxicodona/uso terapêutico , Dor/sangue , Dor/metabolismo , Ratos Sprague-Dawley , Espironolactona/análogos & derivados , Distribuição Tecidual
17.
Front Pharmacol ; 8: 365, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28680400

RESUMO

Neuropathic pain caused by nerve damage is a common and severe class of chronic pain. Disease-modifying clinical therapies are needed as current treatments typically provide only symptomatic relief; show varying clinical efficacy; and most have significant adverse effects. One approach is targeting either neurotrophic factors or their receptors that normalize sensory neuron function and stimulate regeneration after nerve damage. Two candidate targets are glial cell line-derived neurotrophic factor (GDNF) and artemin (ARTN), as these GDNF family ligands (GFLs) show efficacy in animal models of neuropathic pain (Boucher et al., 2000; Gardell et al., 2003; Wang et al., 2008, 2014). As these protein ligands have poor drug-like properties and are expensive to produce for clinical use, we screened 18,400 drug-like compounds to develop small molecules that act similarly to GFLs (GDNF mimetics). This screening identified BT13 as a compound that selectively targeted GFL receptor RET to activate downstream signaling cascades. BT13 was similar to NGF and ARTN in selectively promoting neurite outgrowth from the peptidergic class of adult sensory neurons in culture, but was opposite to ARTN in causing neurite elongation without affecting initiation. When administered after spinal nerve ligation in a rat model of neuropathic pain, 20 and 25 mg/kg of BT13 decreased mechanical hypersensitivity and normalized expression of sensory neuron markers in dorsal root ganglia. In control rats, BT13 had no effect on baseline mechanical or thermal sensitivity, motor coordination, or weight gain. Thus, small molecule BT13 selectively activates RET and offers opportunities for developing novel disease-modifying medications to treat neuropathic pain.

18.
Free Radic Biol Med ; 41(1): 120-31, 2006 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16781460

RESUMO

We previously showed that the one-electron reduction product of nitric oxide (NO), nitroxyl (HNO), irreversibly inhibits the proteolytic activity of the model cysteine protease papain. This result led us to investigate the differential effects of the nitrogen oxides, such as nitroxyl (HNO), NO, and in situ-generated peroxynitrite on cysteine modification-sensitive cellular proteolytic enzymes. We used Angeli's salt, diethylaminenonoate (DEA/NO), and 3-morpholinosydnoniminehydrochloride (SIN-1), as donors of HNO, NO, and peroxynitrite, respectively. In this study we evaluated their inhibitory activities on the lysosomal mammalian papain homologue cathepsin B and on the cytosolic 26S proteasome in THP-1 monocyte/macrophages after LPS activation or TPA differentiation. HNO-generating Angeli's salt caused a concentration-dependent (62 +/- 4% at 316 muM) inhibition of the 26S proteasome activity, resulting in accumulation of protein-bound polyubiquitinylated proteins in LPS-activated cells, whereas neither DEA/NO nor SIN-1 showed any effect. Angeli's salt, but not DEA/NO or SIN-1, also caused (94 +/- 2% at 316 muM) inhibition of lysosomal cathepsin B activity in LPS-activated cells. Induction of macrophage differentiation did not significantly alter the inhibitory effect of HNO on lysosomal cathepsin B activity, but protected the proteasome from HNO-induced inhibition. The protection awarded by macrophage differentiation was associated with induction of the GSH synthesis rate-limiting enzyme gamma-glutamylcysteine synthetase, as well as with increased intracellular GSH. In conclusion, HNO abrogates both lysosomal and cytosolic proteolysis in THP-1 cells. Macrophage differentiation, associated with upregulation of antioxidant defenses such as increased cellular GSH, does not protect the lysosomal cysteine protease cathepsin B from inhibition.


Assuntos
Catepsina B/metabolismo , Macrófagos/enzimologia , Doadores de Óxido Nítrico/farmacologia , Óxidos de Nitrogênio/farmacologia , Diferenciação Celular/efeitos dos fármacos , Citotoxicidade Imunológica , Humanos , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , NADP/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico/metabolismo , Óxido Nítrico/farmacologia , Nitritos/farmacologia , Óxidos de Nitrogênio/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Ácido Peroxinitroso/metabolismo , Poliubiquitina/química , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Acetato de Tetradecanoilforbol/farmacologia
19.
Free Radic Biol Med ; 38(8): 1102-11, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15780768

RESUMO

Protein oxidation, irreversible modification, and inactivation may play key roles in various neurodegenerative disorders. Therefore, we studied the effects of the potentially in vivo occurring nitric oxide-related species on two different markers of protein oxidation: protein carbonyl generation on bovine serum albumine (BSA) and loss of activity of a cysteine-dependent protease, papain, in vitro by using Angeli's salt, papanonoate, SIN-1, and S-nitrosoglutathione (GSNO) as donors of nitroxyl, nitric oxide, peroxynitrite, and nitrosonium ions, respectively. Angeli's salt, SIN-1, and papanonoate (0-1000 microM) all generated a concentration-dependent increase in carbonyl formation on BSA (107, 60, and 45%, respectively). GSNO did not affect carbonyl formation. Papain was inhibited by Angeli's salt, SIN-1, papanonoate, and GSNO with IC50 values of 0.62, 2.3, 54, and 80 microM, respectively. Angeli's salt (3.16 microM)-induced papain inactivation was only partially reversible, while the effects of GSNO (316 microM) and papanonoate (316 microM) were reversible upon addition of excess DTT. The Angeli's salt-mediated DTT-irreversible inhibition of papain was prevented by GSNO or papanonoate pretreatment, hypothetically through mixed disulfide formation or S-nitrosylation of the catalytically critical thiol group of papain. These results, for the first time, compare the generation of carbonyls in proteins by Angeli's salt, papanonoate, and SIN-1. Furthermore, these results suggest that S-nitrosothiols may have a novel function in protecting critical thiols from irreversible oxidative damage.


Assuntos
Óxido Nítrico/fisiologia , Papaína/metabolismo , Antioxidantes/farmacologia , Oxirredução , Papaína/antagonistas & inibidores , Compostos de Sulfidrila/metabolismo
20.
Toxicol Appl Pharmacol ; 207(2 Suppl): 91-5, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15987648

RESUMO

Oxidative stress and apoptosis may play an important role in the neurodegeneration. The present paper outlines antioxidative and antiapototic mechanisms of nitric oxide and S-nitrosothiols, which could mediate neuroprotection. Nitric oxide generated by nitric oxide synthase or released from an endogenous S-nitrosothiol, S-nitrosoglutathione may up-regulate antioxidative thioredoxin system and antiapototic Bcl-2 protein through a cGMP-dependent mechanism. Moreover, nitric oxide radicals have been shown to have direct antioxidant effect through their reaction with free radicals and iron-oxygen complexes. In addition to serving as a stabilizer and carrier of nitric oxide, S-nitrosoglutathione may have protective effect through transnitrosylation reactions. Based on these new findings, a hypothesis arises that the homeostasis of nitric oxide, S-nitrosothiols, glutathione, and thioredoxin systems is important for protection against oxidative stress, apoptosis, and related neurodegenerative disorders.


Assuntos
Fármacos Neuroprotetores/farmacologia , Óxido Nítrico/farmacologia , S-Nitrosoglutationa/farmacologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , GMP Cíclico/farmacologia , Humanos , Estresse Oxidativo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA