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











Intervalo de ano de publicação
1.
CNS Drugs ; 36(6): 617-632, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35616826

RESUMO

Opioids are widely used in chronic pain management, despite major concerns about their risk of adverse events, particularly abuse, misuse, and respiratory depression from overdose. Multi-mechanistic opioids, such as tapentadol and buprenorphine, have been widely studied as a valid alternative to traditional opioids for their safer profile. Special interest was focused on the role of the nociceptin opioid peptide (NOP) receptor in terms of analgesia and improved tolerability. Nociceptin opioid peptide receptor agonists were shown to reinforce the antinociceptive effect of mu opioid receptor (MOR) agonists and modulate some of their adverse effects. Therefore, multi-mechanistic opioids involving both MOR and NOP receptor activation became a major field of pharmaceutical and clinical investigations. Buprenorphine was re-discovered in a new perspective, as an atypical analgesic and as a substitution therapy for opioid use disorders; and buprenorphine derivatives have been tested in animal models of nociceptive and neuropathic pain. Similarly, cebranopadol, a full MOR/NOP receptor agonist, has been clinically evaluated for its potent analgesic efficacy and better tolerability profile, compared with traditional opioids. This review overviews pharmacological mechanisms of the NOP receptor system, including its role in pain management and in the development of opioid tolerance. Clinical data on buprenorphine suggest its role as a safer alternative to traditional opioids, particularly in patients with non-cancer pain; while data on cebranopadol still require phase III study results to approve its introduction on the market. Other bifunctional MOR/NOP receptor ligands, such as BU08028, BU10038, and AT-121, are currently under pharmacological investigations and could represent promising analgesic agents for the future.


Assuntos
Analgésicos Opioides , Buprenorfina , Analgésicos Opioides/efeitos adversos , Animais , Buprenorfina/farmacologia , Buprenorfina/uso terapêutico , Tolerância a Medicamentos , Humanos , Isoquinolinas , Naltrexona/análogos & derivados , Peptídeos Opioides/uso terapêutico , Dor/tratamento farmacológico , Fenilpropionatos , Receptores Opioides mu/agonistas , Receptores Opioides mu/uso terapêutico , Nociceptina
2.
Int J Mol Sci ; 22(24)2021 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-34948415

RESUMO

Opioids are the most potent widely used analgesics, primarily, but not exclusively, in palliative care. However, they are associated with numerous side effects, such as tolerance, addiction, respiratory depression, and cardiovascular events. This, in turn, can result in their overuse in cases of addiction, the need for dose escalation in cases of developing tolerance, and the emergence of dose-related opioid toxicity, resulting in respiratory depression or cardiovascular problems that can even lead to unintentional death. Therefore, a very important challenge for researchers is to look for ways to counteract the side effects of opioids. The use of peptides and their related compounds, which have been shown to modulate the effects of opioids, may provide such an opportunity. This short review is a compendium of knowledge about the most important and recent findings regarding selected peptides and their modulatory effects on various opioid actions, including cardiovascular and respiratory responses. In addition to the peptides more commonly reported in the literature in the context of their pro- and/or anti-opioid activity-such as neuropeptide FF (NPFF), cholecystokinin (CCK), and melanocyte inhibiting factor (MIF)-we also included in the review nociceptin/orphanin (N/OFQ), ghrelin, oxytocin, endothelin, and venom peptides.


Assuntos
Analgésicos Opioides/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Peptídeos/uso terapêutico , Analgésicos Opioides/farmacologia , Animais , Colecistocinina/farmacologia , Colecistocinina/uso terapêutico , Tolerância a Medicamentos , Grelina/farmacologia , Grelina/uso terapêutico , Humanos , Antagonistas de Entorpecentes/farmacologia , Oligopeptídeos/farmacologia , Oligopeptídeos/uso terapêutico , Peptídeos Opioides/farmacologia , Peptídeos Opioides/uso terapêutico , Peptídeos/farmacologia , Receptores Opioides/metabolismo , Nociceptina
3.
Pharmacol Res ; 158: 104855, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32438036

RESUMO

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


Assuntos
Peptídeos Opioides/fisiologia , Peptídeos Opioides/uso terapêutico , Animais , Humanos , Proteína Antagonista do Receptor de Interleucina 1/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Peptídeos Opioides/química , Receptores Opioides/efeitos dos fármacos , Sistema Renina-Angiotensina/efeitos dos fármacos
4.
Pharmacol Ther ; 210: 107519, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32165137

RESUMO

The scarcity and limited risk/benefit ratio of painkillers available on the market, in addition to the opioid crisis, warrant reflection on new innovation strategies. The pharmacopoeia of analgesics is based on products that are often old and derived from clinical empiricism, with limited efficacy or spectrum of action, or resulting in an unsatisfactory tolerability profile. Although they are reference analgesics for nociceptive pain, opioids are subject to the same criticism. The use of opium as an analgesic is historical. Morphine was synthesized at the beginning of the 19th century. The efficacy of opioids is limited in certain painful contexts and these drugs can induce potentially serious and fatal adverse effects. The current North American opioid crisis, with an ever-rising number of deaths by opioid overdose, is a tragic illustration of this. It is therefore legitimate to develop research into molecules likely to maintain or increase opioid efficacy while improving their tolerability. Several avenues are being explored including targeting of the mu opioid receptor (MOR) splice variants, developing biased agonists or targeting of other receptors such as heteromers with MOR. Ion channels acting as MOR effectors, are also targeted in order to offer compounds without MOR-dependent adverse effects. Another route is to develop opioid analgesics with peripheral action or limited central nervous system (CNS) access. Finally, endogenous opioids used as drugs or compounds that modify the metabolism of endogenous opioids (Dual ENKephalinase Inhibitors) are being developed. The aim of the present review is to present these various targets/strategies with reference to current indications for opioids, concerns about their widespread use, particularly in chronic non-cancer pains, and ways of limiting the risk of opioid abuse and misuse.


Assuntos
Analgésicos Opioides/efeitos adversos , Analgésicos/uso terapêutico , Sistema Nervoso Central/efeitos dos fármacos , Descoberta de Drogas , Peptídeos Opioides/uso terapêutico , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Dor/tratamento farmacológico , Receptores Opioides mu/agonistas , Analgésicos/efeitos adversos , Animais , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiopatologia , Humanos , Ligantes , Terapia de Alvo Molecular , Epidemia de Opioides , Peptídeos Opioides/efeitos adversos , Peptídeos Opioides/metabolismo , Transtornos Relacionados ao Uso de Opioides/diagnóstico , Transtornos Relacionados ao Uso de Opioides/epidemiologia , Dor/metabolismo , Dor/fisiopatologia , Limiar da Dor/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Transdução de Sinais
5.
Behav Brain Res ; 374: 112123, 2019 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-31376441

RESUMO

Previous studies have uncovered a potential role of the opioid system in iron hemostasis and dopamine metabolism. Abnormalities in both of these systems have been noted in human RLS. Autopsy studies of human RLS have shown an endogenous opioid deficiency in the thalamus. Opioids, particularly prolonged-release oxycodone/naloxone, have been approved in Europe to be a second-line therapy for severe restless legs syndrome (RLS). To study the role of opioid receptors in the pathogenesis of RLS, we used a triple knockout (KO) mouse strain that lack mu, delta, and kappa opioid receptors and explored the behavioral and biochemical parameters relevant to RLS. The triple KO mice showed hyperactivity and a trend of increased probability of waking during the rest period (day) akin to that in human RLS (night). Surprisingly, triple KO mice also exhibit decreased serum iron concentration, evidence of anemia, a significant dysfunction in dopamine metabolism akin to that noted in human RLS, as well as an increased latency in response to thermal stimuli. To our knowledge, this is the first study to demonstrate that the endogenous opioid system may play a role in iron metabolism and subsequently in the pathogenesis of anemia. It is also the first study showing that opioid receptors are involved in the production of motor restlessness with a circadian predominance. Our findings support the role of endogenous opioids in the pathogenesis of RLS, and the triple KO mice can be used to understand the relationship between iron deficiency, anemia, dopaminergic dysfunction, and RLS.


Assuntos
Deficiências de Ferro , Ferro/metabolismo , Receptores Opioides mu/fisiologia , Analgésicos Opioides/uso terapêutico , Anemia/metabolismo , Anemia Ferropriva/metabolismo , Animais , Dopamina/metabolismo , Dopamina/fisiologia , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/fisiologia , Masculino , Camundongos , Camundongos Knockout , Naloxona/uso terapêutico , Peptídeos Opioides/uso terapêutico , Agitação Psicomotora/tratamento farmacológico , Receptores Opioides mu/genética , Síndrome das Pernas Inquietas/metabolismo , Síndrome das Pernas Inquietas/fisiopatologia
6.
Peptides ; 116: 63-67, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31014958

RESUMO

The current opioid crisis has created a tragic problem in medicine and society. Pain is the most ubiquitous and costly disease in society and yet all of our "treatments" have toxicities, especially for prolonged use. However, there are several alternatives that have been discovered in the past fifteen years that have been demonstrated in animals to have none of the toxicities of current drugs. Many of the compounds are multivalent and have novel biological activity profiles. Unfortunately, none of these have been in clinical trials in humans, perhaps because they were discovered in academic laboratories. A review of these novel chemicals are given in this paper.


Assuntos
Analgésicos Opioides/uso terapêutico , Dor/tratamento farmacológico , Peptídeos/uso terapêutico , Peptidomiméticos/uso terapêutico , Animais , Humanos , Ligantes , Peptídeos Opioides/química , Peptídeos Opioides/uso terapêutico , Dor/patologia , Manejo da Dor , Peptídeos/efeitos adversos , Peptidomiméticos/efeitos adversos , Receptores Opioides/química , Receptores Opioides/uso terapêutico , Receptores Opioides delta/química , Receptores Opioides delta/genética , Receptores Opioides mu/química , Receptores Opioides mu/genética
8.
Peptides ; 110: 40-46, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30391421

RESUMO

It has been shown that the hippocampus plays an essential role in the regulation of reward and memory as indicated by the conditioned place preference (CPP) paradigm. Morphine-induced CPP is a common method to consider motivational properties of morphine in animals. Recently, this model has been used in many laboratories to investigate neuronal mechanisms underlying reinstatement of morphine seeking induced by drug re-exposure. Our previous studies indicate that the hippocampus especially CA1 region is involved in reinstatement of drug-seeking behaviors. Also, several studies have shown that orexin attenuates key functional and behavioral effects of its co-transmitter dynorphin. The present study evaluates the role of orexinergic receptors within the CA1 region of the hippocampus in the reinstatement of morphine-induced CPP. Therefore, after the extinction period, the different doses (SB 334867; 0.3, 3, and 30 nM/0.5 µl DMSO) of either orexin-1 or -2 receptor antagonists were bilaterally microinjected into the CA1, 15 min before receiving an effective priming dose of morphine (1 mg/kg). The results revealed that administration of both SB 334867 and TCS OX2 29 prior to injection of the priming dose of morphine significantly reduced the reinstatement of morphine-induced CPP without altering the animal's locomotor activity. Also, the 50% effective dose value of SB 334867 on the reinstatement of morphine seeking behavior was close three times more than that in TCS OX2 29 treatment group. Therefore, the consequences suggested that both orexin receptors in the CA1 play a considerable role in the reinstatement of morphine-induced CPP.


Assuntos
Hipocampo/metabolismo , Morfina/farmacologia , Antagonistas dos Receptores de Orexina/farmacologia , Receptores de Orexina/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Western Blotting , Comportamento de Procura de Droga , Feminino , Hipocampo/efeitos dos fármacos , Masculino , Peptídeos Opioides/uso terapêutico , Ratos
9.
Eur J Pharmacol ; 766: 155-62, 2015 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-26450087

RESUMO

Oxaliplatin and paclitaxel are considered central components in the treatment of colorectal and breast cancer, respectively. The development of neuropathy during chronic treatment represents the major dose-limiting side effect that leads to discontinuation or interruption of therapies. The management of neuropathy is a challenge to individuate innovative therapeutic strategies based on new targets and correct routes of administration. We evaluated the hypersensitivity reliever effect of different opioid receptor agonists in rat models of oxaliplatin and paclitaxel-induced neuropathy. Compounds were spinally infused by intrathecal catheter. In oxaliplatin-treated rats, 0.3 nmol morphine induced the reversion of the mechanical hypersensitivity (Paw-pressure test), nociceptin/orphanin FQ (N/OFQ; 0.3-3 nmol) significantly increased the pain threshold without reaching the values of the control animals. The N/OFQ peptide (NOP) receptor full agonist UFP-112 reverted pain threshold alterations at lower dosage (0.1 nmol) vs morphine and N/OFQ, the partial agonist UFP-113 (0.1-1 nmol) was similar to N/OFQ. The higher efficacy of morphine vs N/OFQ was highlighted also in paclitaxel-treated rats. The mechanical hypersensitivity was fully reverted by 0.1 nmol UFP-112 and UFP-113. In conclusion, intrathecal µ opioid peptide (MOP) and NOP receptor agonists relieved chemotherapy-induced neuropathic pain. The synthetic peptides showed valuable potency and efficacy suggesting the NOP system as an exploitable target.


Assuntos
Analgésicos Opioides/uso terapêutico , Hiperalgesia/tratamento farmacológico , Neuralgia/tratamento farmacológico , Peptídeos Opioides/uso terapêutico , Receptores Opioides/agonistas , Animais , Antineoplásicos , Hiperalgesia/induzido quimicamente , Injeções Espinhais , Masculino , Neuralgia/induzido quimicamente , Compostos Organoplatínicos , Oxaliplatina , Paclitaxel , Ratos , Ratos Sprague-Dawley , Receptor de Nociceptina
10.
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
11.
Mol Pain ; 10: 10, 2014 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-24499354

RESUMO

BACKGROUND: Leukocytes containing opioid peptides locally control inflammatory pain. In the early phase of complete Freund's adjuvant (CFA)-induced hind paw inflammation, formyl peptides (derived e.g. from Mycobacterium butyricum) trigger the release of opioid peptides from neutrophils contributing to tonic basal antinociception. In the later phase we hypothesized that toll-like-receptor-(TLR)-4 activation of monocytes/macrophages triggers opioid peptide release and thereby stimulates peripheral opioid-dependent antinociception. RESULTS: In Wistar rats with CFA hind paw inflammation in the later inflammatory phase (48-96 h) systemic leukocyte depletion by cyclophosphamide (CTX) or locally injected naloxone (NLX) further decreased mechanical and thermal nociceptive thresholds. In vitro ß-endorphin (ß-END) content increased during human monocyte differentiation as well as in anti-inflammatory CD14+CD16- or non-classical M2 macrophages. Monocytes expressing TLR4 dose-dependently released ß-END after stimulation with lipopolysaccharide (LPS) dependent on intracellular calcium. Despite TLR4 expression proinflammatory M1 and anti-inflammatory M2 macrophages only secreted opioid peptides in response to ionomycin, a calcium ionophore. Intraplantar injection of LPS as a TLR4 agonist into the inflamed paw elicited an immediate opioid- and dose-dependent antinociception, which was blocked by TAK-242, a small-molecule inhibitor of TLR4, or by peripheral applied NLX. In the later phase LPS lowered mechanical and thermal nociceptive thresholds. Furthermore, local peripheral TLR4 blockade worsened thermal and mechanical nociceptive pain thresholds in CFA inflammation. CONCLUSION: Endogenous opioids from monocytes/macrophages mediate endogenous antinociception in the late phase of inflammation. Peripheral TLR4 stimulation acts as a transient counter-regulatory mechanism for inflammatory pain in vivo, and increases the release of opioid peptides from monocytes in vitro. TLR4 antagonists as new treatments for sepsis and neuropathic pain might unexpectedly transiently enhance pain by impairing peripheral opioid analgesia.


Assuntos
Analgesia , Inflamação/tratamento farmacológico , Peptídeos Opioides/uso terapêutico , Receptor 4 Toll-Like/metabolismo , Animais , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Adjuvante de Freund/administração & dosagem , Adjuvante de Freund/farmacologia , Humanos , Hiperalgesia/complicações , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Inflamação/complicações , Inflamação/metabolismo , Inflamação/patologia , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Nociceptividade/efeitos dos fármacos , Peptídeos Opioides/farmacologia , Ratos , Ratos Wistar , Receptores de IgG/metabolismo , Receptores Opioides/metabolismo , Receptor 2 Toll-Like/metabolismo , beta-Endorfina/metabolismo
12.
Invest New Drugs ; 31(4): 1066-70, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23275062

RESUMO

Hepatoblastoma is the most common liver malignancy in children, typically diagnosed before age 2. The survival rate for hepatoblastoma has increased dramatically in the last 30 years, but the typical chemotherapeutic agents used for treatment are associated with significant toxicity. In this report, the authors present two cases of hepatoblastoma treated with surgical resection and a novel biotherapeutic regimen that included opioid growth factor (OGF). Case #1 is an infant diagnosed with a large mass on prenatal ultrasound. After subsequent diagnosis of hepatoblastoma, she was treated with one course of neoadjuvant chemotherapy at approximately 1 week of age. Following significant complications from the chemotherapy (neutropenic fever, pneumonia and sepsis), the patient's parents declined further chemotherapy, and the infant was treated with surgical resection and opioid growth factor (OGF)/low dose naltrexone (LDN). She is currently at close to 10 years disease-free survival. Case #2 is a child diagnosed with a liver mass on ultrasound at 20 months of age, later biopsy-proven to represent hepatoblastoma. Due to existing co-morbidities including autosomal recessive polycystic kidney disease and hypertension, and indications from the biopsy that the tumor might be insensitive to chemotherapy, the parents elected not to proceed with neoadjuvant chemotherapy. The patient was treated with surgical resection and OGF/LDN, and is currently at more than 5 years disease-free survival. This case series highlights the need for less toxic treatment options than conventional chemotherapy. Modulation of the OGF-OGF receptor axis represents a promising safe and therapeutic avenue for effective treatment of hepatoblastoma.


Assuntos
Encefalina Metionina/uso terapêutico , Hepatoblastoma/tratamento farmacológico , Peptídeos Opioides/uso terapêutico , Quimioterapia Adjuvante , Criança , Pré-Escolar , Feminino , Hepatoblastoma/diagnóstico por imagem , Humanos , Lactente , Recém-Nascido , Gravidez , Ultrassonografia
13.
J Dent Res ; 91(5): 447-53, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21972258

RESUMO

Cancer pain is an ever-present public health concern. With innovations in treatment, cancer patients are surviving longer, but uncontrollable pain creates a poor quality of life for these patients. Oral cancer is unique in that it causes intense pain at the primary site and significantly impairs speech, swallowing, and masticatory functions. We propose that oral cancer pain has underlying biologic mechanisms that are generated within the cancer microenvironment. A comprehensive understanding of key mediators that control cross-talk between the cancer and peripheral nervous system, and possible interventions, underlies effective cancer pain management. The purpose of this review is to explore the current studies on oral cancer pain and their implications in clinical management for cancer pain in general. Furthermore, we will explore the endogenous opioid systems and novel cancer pain therapeutics that target these systems, which could solve the issue of opiate tolerance and improve quality of life in oral cancer patients.


Assuntos
Carcinoma de Células Escamosas/fisiopatologia , Dor Facial/etiologia , Neoplasias Bucais/fisiopatologia , Dor Intratável/etiologia , Analgésicos Opioides/farmacologia , Animais , Carcinoma de Células Escamosas/complicações , Tolerância a Medicamentos/fisiologia , Endotelina-1/fisiologia , Dor Facial/tratamento farmacológico , Dor Facial/fisiopatologia , Humanos , Neoplasias Bucais/complicações , Fator de Crescimento Neural/fisiologia , Nociceptores/fisiologia , Peptídeos Opioides/genética , Peptídeos Opioides/fisiologia , Peptídeos Opioides/uso terapêutico , Manejo da Dor/métodos , Dor Intratável/tratamento farmacológico , Dor Intratável/fisiopatologia , Cuidados Paliativos , Qualidade de Vida , Receptores Ativados por Proteinase/fisiologia
14.
Headache ; 51(8): 1245-53, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21797854

RESUMO

BACKGROUND: Alterations in the levels of nociceptin/orphanin FQ (N/OFQ) have been reported in patients with primary headaches, including migraines and cluster headaches. These clinical observations suggest that N/OFQ is involved in the pathogenesis of primary headaches. OBJECTIVES: The present study was conducted to determine the role of N/OFQ in the control of trigeminal nociception and cortical excitation. METHODS: Cortical spreading depression (CSD) was elicited in Wistar rats by cortical application of potassium chloride, and electrocorticograms were recorded. N/OFQ was administered via an intracisternal injection. The presence of CSD-evoked trigeminal nociception was determined with Fos and transient receptor potential vanilloid 1 (TRPV1) immunoreactivity. RESULTS: Nociceptin/orphanin FQ produced a biphasic effect on CSD generation, characterized by an initial attenuation followed by delayed potentiation. The amplitude of CSD waves were lower in the initial period but increased in the later period. The total number of CSD waves recorded in 1 hour was greater in the N/OFQ-treated group. Exposure to N/OFQ significantly increased the number of Fos-immunoreactive cells in the trigeminal nucleus caudalis and the number of TRPV1-immunoreactive cells in the trigeminal ganglia, indicating the enhancement of trigeminal nociception. CONCLUSION: These results indicate that N/OFQ can lead to biphasic effect characterized by an initial inhibition, and delay potentiation that eventually intensify CSD-evoked trigeminal nociception.


Assuntos
Depressão Alastrante da Atividade Elétrica Cortical/efeitos dos fármacos , Nociceptividade/fisiologia , Peptídeos Opioides/uso terapêutico , Nervo Trigêmeo/fisiopatologia , Animais , Área Sob a Curva , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Modelos Animais de Doenças , Eletroencefalografia/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Receptores Opioides/metabolismo , Canais de Cátion TRPV/metabolismo , Núcleo Inferior Caudal do Nervo Trigêmeo/metabolismo , Gânglio Trigeminal/metabolismo , Neuralgia do Trigêmeo , Nociceptina
15.
Neuroscience ; 168(2): 543-50, 2010 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-20394808

RESUMO

Microinjection of opioids into the ventrolateral periaqueductal gray (vlPAG) produces antinociception in part by binding to mu-opioid receptors (MOPrs). Although both high and low efficacy agonists produce antinociception, low efficacy agonists such as morphine produce limited MOPr internalization suggesting that MOPr internalization and signaling leading to antinociception are independent. This hypothesis was tested in awake, behaving rats using DERM-A594, a fluorescently labeled dermorphin analog, and internalization blockers. Microinjection of DERM-A594 into the vlPAG produced both antinociception and internalization of DERM-A594. Administration of the irreversible opioid receptor antagonist beta-chlornaltrexamine (beta-CNA) prior to DERM-A594 microinjection reduced both the antinociceptive effect and the number of DERM-A594 labeled cells demonstrating that both effects are opioid receptor-mediated. Pretreatment with the internalization blockers dynamin dominant-negative inhibitory peptide (dynamin-DN) and concanavalinA (ConA) attenuated both DERM-A594 internalization and antinociception. Microinjection of dynamin-DN and ConA also decreased the antinociceptive potency of the unlabeled opioid agonist dermorphin when microinjected into the vlPAG as demonstrated by rightward shifts in the dose-response curves. In contrast, administration of dynamin-DN had no effect on the antinociceptive effect of microinjecting the GABA(A) receptor antagonist bicuculline into the vlPAG. The finding that dermorphin-induced antinociception is attenuated by blocking receptor internalization indicates that key parts of opioid receptor-mediated signaling depend on internalization.


Assuntos
Analgésicos Opioides/farmacologia , Peptídeos Opioides/farmacologia , Dor/tratamento farmacológico , Receptores Opioides mu/metabolismo , Analgésicos Opioides/química , Analgésicos Opioides/uso terapêutico , Animais , Bicuculina/farmacologia , Concanavalina A/farmacologia , Dinaminas/antagonistas & inibidores , Corantes Fluorescentes/química , Antagonistas de Receptores de GABA-A , Masculino , Microinjeções , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Neurônios/metabolismo , Peptídeos Opioides/química , Peptídeos Opioides/uso terapêutico , Dor/metabolismo , Dor/fisiopatologia , Medição da Dor , Peptídeos/farmacologia , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Substância Cinzenta Periaquedutal/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/antagonistas & inibidores
16.
Int. braz. j. urol ; 32(6): 620-630, Nov.-Dec. 2006.
Artigo em Inglês | LILACS | ID: lil-456568

RESUMO

Contemporary, the management of overactive bladder (OAB), a medical condition characterized by urgency, with or without urge urinary incontinence, frequency and nocturia, in absence of genitourinary pathologies or metabolic factors that could explain these symptoms, is complex, and a wide range of conservative treatments has been offered, including bladder training, biofeedback, behavioral changes, oral or intravesical anticholinergic agents, S3 sacral neuromodulation and peripheral electrical stimulation. Clinical efficacy of these treatments remains an open issue and several experimental and clinical studies were carried out in the last years improving the results of medical treatment.Here we review the pathophysiology of micturition reflex, the current therapies for OAB and the rationale for alternative treatments. Furthermore we critically address the potential use of medications targeting the central nervous system (CNS) and the primary sensory nerves of the bladder wall, we review the use of agonists of nociceptin/orphanin protein (NOP) receptor and finally we report the results obtained by intradetrusor injection of botulinum toxin.


Assuntos
Humanos , Antagonistas Muscarínicos/uso terapêutico , Bexiga Urinária Hiperativa/tratamento farmacológico , Toxinas Botulínicas/uso terapêutico , Sistema Nervoso Central/efeitos dos fármacos , Antagonistas Muscarínicos/farmacologia , Peptídeos Opioides/farmacologia , Peptídeos Opioides/uso terapêutico , Receptores Muscarínicos/efeitos dos fármacos , Bexiga Urinária Hiperativa/fisiopatologia
17.
Gastroenterology ; 131(3): 900-10, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16952558

RESUMO

BACKGROUND & AIMS: The endogenous opioid system is involved in modulating the experience of pain, the response to stress, and the action of analgesic therapies. Recent human imaging studies have shown a significant tonic modulation of visceral pain, raising the question of whether endogenous opioids tonically modulate the pain of visceral cancer. METHODS: Transgenic mice expressing the first 127 amino acids of simian virus 40 large T antigen, under the control of the rat elastase-1 promoter, that spontaneously develop pancreatic cancer were used to investigate the role of endogenous opioids in the modulation of pancreatic cancer pain. Visceral pain behaviors were assessed as degree of hunching and vocalization. RESULTS: Although mice with late-stage pancreatic cancer displayed spontaneous, morphine-reversible, visceral pain-related behaviors such as hunching and vocalization, these behaviors were absent in mice with early-stage pancreatic cancer. After systemic administration of the central nervous system (CNS)-penetrant opioid receptor antagonists naloxone or naltrexone, mice with early-stage pancreatic cancer displayed significant visceral pain-related behaviors, whereas systemic administration of the CNS-nonpenetrant opioid antagonist naloxone-methiodide did not induce an increase in visceral pain behaviors. CONCLUSIONS: Our findings suggest that a CNS opioid-dependent mechanism tonically modulates early and late-stage pancreatic cancer pain. Understanding the mechanisms that mask this pain in early stage disease and drive this pain in late-stage disease may allow improved diagnosis, treatment, and care of patients with pancreatic cancer.


Assuntos
Dor Abdominal/tratamento farmacológico , Neurotransmissores/uso terapêutico , Peptídeos Opioides/uso terapêutico , Neoplasias Pancreáticas/complicações , Dor Abdominal/etiologia , Dor Abdominal/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Índice de Gravidade de Doença , Resultado do Tratamento
18.
ScientificWorldJournal ; 6: 1066-74, 2006 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-16951899

RESUMO

Chronic, unremitting pain is perhaps the most common reason that patients seek medical care. In general, conservative techniques, such as medical management, are implemented as first-line therapy. Local anesthesia and lytic procedures, followed by interventional techniques, such as dorsal column stimulation and intrathecal drug delivery systems, are second-line therapies. However, for refractory and severe pain, which is not adequately controlled by other modes of therapy, new emerging options, including molecular or gene therapy, may become more widely utilized as experimental results are translated into clinical options.


Assuntos
Terapia Genética , Manejo da Dor , Adenoviridae/genética , Analgesia/métodos , Animais , Ensaios Clínicos como Assunto , DNA/administração & dosagem , Endorfinas/uso terapêutico , Terapia Genética/métodos , Vetores Genéticos , Herpesvirus Humano 1/genética , Humanos , Inflamação/tratamento farmacológico , Lipossomos , Neoplasias/terapia , Doenças do Sistema Nervoso/terapia , Peptídeos Opioides/uso terapêutico , Cuidados Paliativos , Transgenes/fisiologia
19.
Curr Pain Headache Rep ; 8(5): 393-8, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15361324

RESUMO

There now is one realized and several attractive targets for the treatment of acute attacks of migraine that will follow and augment the use of serotonin 5-HT1B/1D receptor agonists, the triptans. Calcitonin gene-related peptide (CGRP) receptor blockade recently has been shown to be an effective acute antimigraine strategy; therefore, blockade of CGRP release by inhibition of trigeminal nerves would seem a logical approach. A number of targets are reviewed in this article including serotonin 5-HT1F and 5-HT1D receptors, adenosine A1 receptors, nociceptin, vanilloid TRPV1 receptors, and anandamide CB1 receptors. Development of one or more such compound offers the exciting prospect of new non-vasoconstrictor treatments for migraine and cluster headache.


Assuntos
Antidepressivos de Segunda Geração/uso terapêutico , Capsaicina/farmacologia , Capsaicina/uso terapêutico , Transtornos de Enxaqueca/tratamento farmacológico , Peptídeos Opioides/farmacologia , Peptídeos Opioides/uso terapêutico , Receptor A1 de Adenosina/uso terapêutico , Agonistas do Receptor de Serotonina/farmacologia , Agonistas do Receptor de Serotonina/uso terapêutico , Nervo Trigêmeo/efeitos dos fármacos , Triptofano/uso terapêutico , Vasodilatadores/farmacologia , Vasodilatadores/uso terapêutico , Doença Aguda , Cefaleia Histamínica/tratamento farmacológico , Humanos , Nociceptina
20.
Neuroreport ; 9(10): 2243-7, 1998 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-9694207

RESUMO

The recently isolated peptide nociceptin has a primary structure similar to that of opioid peptides. Early functional studies suggested that it may act in opposition to opioid systems. To determine whether nociceptin influences the rewarding properties of heroin, nociceptin was given intracerebroventricularly (i.c.v.) to rats trained to self-administer heroin. Rats (n = 8) were given doses of 0.01 microg, 0.1 microg, 1.0 microg and 10.0 microg in a Latin square design. None of the doses significantly affected heroin self-administration rates compared to vehicle. The highest dose of nociceptin used inhibited spontaneous locomotor activity, evidence that the peptide retained its biological activity after i.c.v. infusion. These studies suggest that nociceptin does not affect the rewarding value of heroin.


Assuntos
Dependência de Heroína/tratamento farmacológico , Dependência de Heroína/psicologia , Peptídeos Opioides/uso terapêutico , Receptores Opioides/agonistas , Animais , Relação Dose-Resposta a Droga , Técnicas In Vitro , Injeções Intravenosas , Injeções Intraventriculares , Masculino , Atividade Motora/efeitos dos fármacos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Wistar , Receptores de Hormônio Liberador da Corticotropina/agonistas , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Esquema de Reforço , Autoadministração , Nociceptina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA