Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Anesthesiology ; 128(6): 1220-1236, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29601322

RESUMO

BACKGROUND: Ongoing neuropathic pain is difficult to treat. The authors examined whether dermorphin [D-Arg2, Lys4] (1-4) amide, a peripherally acting µ-opioid receptor agonist, attenuates ongoing pain-associated manifestations after nerve injury in rats and mice. METHODS: Using conditioned place preference assay, the authors tested whether animals show a preference to the environment associated with drug treatment. Wide-dynamic range and dorsal root ganglion neuronal activities were measured by electrophysiology recording and calcium imaging. RESULTS: Nerve-injured animals stayed longer in dermorphin [D-Arg2, Lys4] (1-4) amide-paired chamber after conditioning than during preconditioning (rats: 402.4 ± 61.3 vs. 322.1 ± 45.0 s, 10 mg/kg, n = 9, P = 0.009; mice: 437.8 ± 59.4 vs. 351.3 ± 95.9 s, 2 mg/kg, n = 8, P = 0.047). Topical ganglionic application of dermorphin [D-Arg2, Lys4] (1-4) amide (5 µM, 1 µl, n = 5) reduced the numbers of small-diameter dorsal root ganglion neurons that showed spontaneous activity (1.1 ± 0.4 vs. 1.5 ± 0.3, P = 0.044) and that were activated by test stimulation (15.5 ± 5.5 vs. 28.2 ± 8.2, P = 0.009) after injury. In neuropathic rats, dermorphin [D-Arg2, Lys4] (1-4) amide (10 mg/kg, n = 8) decreased spontaneous firing rates in wide-dynamic range neurons to 53.2 ± 46.6% of predrug level, and methylnaltrexone (5 mg/kg, n = 9) blocked dermorphin [D-Arg2, Lys4] (1-4) amide-induced place preference and inhibition of wide-dynamic range neurons. Dermorphin [D-Arg2, Lys4] (1-4) amide increased paw withdrawal threshold (17.5 ± 2.2 g) from baseline (3.5 ± 0.7 g, 10 mg/kg, n = 8, P = 0.002) in nerve-injured rats, but the effect diminished after repeated administrations. CONCLUSIONS: Peripherally acting µ-opioids may attenuate ongoing pain-related behavior and its neurophysiologic correlates. Yet, repeated administrations cause antiallodynic tolerance.


Assuntos
Analgésicos Opioides/uso terapêutico , Neuralgia/tratamento farmacológico , Neuralgia/fisiopatologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiologia , Nervos Espinhais/fisiologia , Analgésicos Opioides/farmacologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuralgia/psicologia , Ratos , Ratos Sprague-Dawley , Nervos Espinhais/efeitos dos fármacos
2.
Pain ; 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38815196

RESUMO

ABSTRACT: Many medications commonly used to treat neuropathic pain are associated with significant, dose-limiting adverse effects, including sedation, dizziness, and fatigue. These adverse effects are due to the activity of these medications within the central nervous system. The objective of this work was to investigate the interactions between peripherally restricted cannabinoid receptor and mu-opioid receptor (MOR) agonists on ongoing and evoked neuropathic pain behaviors in mouse models. RNAscope analysis of cannabinoid receptor type 1 (CB1R) and MOR mRNA demonstrated that the mRNA of both receptors is colocalized in both mouse and human dorsal root ganglion. Single-cell RNAseq of dorsal root ganglion from chronic constriction injury mice showed that the mRNA of both receptors (Cnr1 and Oprm1) is coexpressed across different neuron clusters. Myc-CB1R and FLAG-MOR were cotransfected into immortalized HEK-293T cells and were found to interact at a subcellular level. We also find that CB-13 (a peripherally restricted dual CB1R and cannabinoid receptor type 2 agonist) and DALDA (a peripherally restricted MOR agonist) both attenuate mechanical hypersensitivity in a murine model of neuropathic pain. Using isobolographic analysis, we demonstrate that when coadministered, these agents synergistically attenuate mechanical hypersensitivity. Importantly, combination dosing of these agents does not cause any detectable preferential behaviors or motor impairment. However, repeated dosing of these agents is associated with the development of tolerance to these drugs. Collectively, these findings suggest that leveraging synergistic pain inhibition between cannabinoid receptor and MOR agonists in peripheral sensory neurons may be worth examining in patients with neuropathic pain.

3.
Pain ; 164(11): 2463-2476, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37326644

RESUMO

ABSTRACT: The propensity for breast cancer to metastasize to bone is coupled to the most common complaint among breast cancer patients: bone pain. Classically, this type of pain is treated using escalating doses of opioids, which lack long-term efficacy due to analgesic tolerance, opioid-induced hypersensitivity, and have recently been linked to enhanced bone loss. To date, the molecular mechanisms underlying these adverse effects have not been fully explored. Using an immunocompetent murine model of metastatic breast cancer, we demonstrated that sustained morphine infusion induced a significant increase in osteolysis and hypersensitivity within the ipsilateral femur through the activation of toll-like receptor-4 (TLR4). Pharmacological blockade with TAK242 (resatorvid) as well as the use of a TLR4 genetic knockout ameliorated the chronic morphine-induced osteolysis and hypersensitivity. Genetic MOR knockout did not mitigate chronic morphine hypersensitivity or bone loss. In vitro studies using RAW264.7 murine macrophages precursor cells demonstrated morphine-enhanced osteoclastogenesis that was inhibited by the TLR4 antagonist. Together, these data indicate that morphine induces osteolysis and hypersensitivity that are mediated, in part, through a TLR4 receptor mechanism.


Assuntos
Neoplasias da Mama , Osteólise , Camundongos , Humanos , Animais , Feminino , Morfina/farmacologia , Receptor 4 Toll-Like/genética , Osteólise/induzido quimicamente , Osteólise/tratamento farmacológico , Modelos Animais de Doenças , Analgésicos Opioides/uso terapêutico , Dor/tratamento farmacológico
4.
Sci Signal ; 11(535)2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29921657

RESUMO

The µ-opioid receptor (MOR) agonist morphine is commonly used for pain management, but it has severe adverse effects and produces analgesic tolerance. Thus, alternative ways of stimulating MOR activity are needed. We found that MrgC11, a sensory neuron-specific G protein-coupled receptor, may form heteromeric complexes with MOR. Peptide-mediated activation of MrgC11 enhanced MOR recycling by inducing coendocytosis and sorting of MOR for membrane reinsertion. MrgC11 activation also inhibited the coupling of MOR to ß-arrestin-2 and enhanced the morphine-dependent inhibition of cAMP production. Intrathecal coadministration of a low dose of an MrgC agonist potentiated acute morphine analgesia and reduced chronic morphine tolerance in wild-type mice but not in Mrg-cluster knockout (Mrg KO) mice. BAM22, a bivalent agonist of MrgC and opioid receptors, enhanced the interaction between MrgC11 and MOR and produced stronger analgesia than did the individual monovalent agonists. Morphine-induced neuronal and pain inhibition was reduced in Mrg KO mice compared to that in wild-type mice. Our results uncover MrgC11-MOR interactions that lead to positive functional modulation of MOR. MrgC shares genetic homogeneity and functional similarity with human MrgX1. Thus, harnessing this positive modulation of MOR function by Mrg signaling may enhance morphine analgesia in a sensory neuron-specific fashion to limit central side effects.


Assuntos
Analgesia/métodos , Morfina/farmacologia , Dor/tratamento farmacológico , Receptores Acoplados a Proteínas G/fisiologia , Receptores Opioides mu/fisiologia , Células Receptoras Sensoriais/metabolismo , Animais , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Manejo da Dor , Receptores Acoplados a Proteínas G/química , Receptores Opioides mu/química , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/efeitos dos fármacos
5.
Pain ; 158(9): 1733-1742, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28570482

RESUMO

Metastatic bone pain is the single most common form of cancer pain and persists as a result of peripheral and central inflammatory, as well as neuropathic mechanisms. Here, we provide the first characterization of sphingolipid metabolism alterations in the spinal cord occurring during cancer-induced bone pain (CIBP). Following femoral arthrotomy and syngenic tumor implantation in mice, ceramides decreased with corresponding increases in sphingosine and the bioactive sphingolipid metabolite, sphingosine 1-phosphate (S1P). Intriguingly, de novo sphingolipid biosynthesis was increased as shown by the elevations of dihydro-ceramides and dihydro-S1P. We next identified the S1P receptor subtype 1 (S1PR1) as a novel target for therapeutic intervention. Intrathecal or systemic administration of the competitive and functional S1PR1 antagonists, TASP0277308 and FTY720/Fingolimod, respectively, attenuated cancer-induced spontaneous flinching and guarding. Inhibiting CIBP by systemic delivery of FTY720 did not result in antinociceptive tolerance over 7 days. FTY720 administration enhanced IL-10 in the lumbar ipsilateral spinal cord of CIBP animals and intrathecal injection of an IL-10 neutralizing antibody mitigated the ability of systemic FTY720 to reverse CIBP. FTY720 treatment was not associated with alterations in bone metabolism in vivo. Studies here identify a novel mechanism to inhibit bone cancer pain by blocking the actions of the bioactive metabolites S1P and dihydro-S1P in lumbar spinal cord induced by bone cancer and support potential fast-track clinical application of the FDA-approved drug, FTY720, as a therapeutic avenue for CIBP.


Assuntos
Dor do Câncer/etiologia , Dor do Câncer/metabolismo , Inflamação Neurogênica/etiologia , Inflamação Neurogênica/metabolismo , Pró-Proteína Convertases/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Serina Endopeptidases/metabolismo , Animais , Neoplasias da Mama/complicações , Neoplasias da Mama/patologia , Dor do Câncer/tratamento farmacológico , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Inflamação Neurogênica/tratamento farmacológico , Pró-Proteína Convertases/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Receptores de Esfingosina-1-Fosfato , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Sulfonas/farmacologia , Sulfonas/uso terapêutico , Triazóis/farmacologia , Triazóis/uso terapêutico
6.
Neuropharmacology ; 116: 59-70, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28007501

RESUMO

The misuse of prescription opiates is on the rise with combination therapies (e.g. acetaminophen or NSAIDs) resulting in severe liver and kidney damage. In recent years, cannabinoid receptors have been identified as potential modulators of pain and rewarding behaviors associated with cocaine, nicotine and ethanol in preclinical models. Yet, few studies have identified whether mu opioid agonists and CB2 agonists act synergistically to inhibit chronic pain while reducing unwanted side effects including reward liability. We determined if analgesic synergy exists between the mu-opioid agonist morphine and the selective CB2 agonist, JWH015, in rodent models of acute and chronic inflammatory, post-operative, and neuropathic pain using isobolographic analysis. We also investigated if the MOR-CB2 agonist combination decreased morphine-induced conditioned place preference (CPP) and slowing of gastrointestinal transit. Co-administration of morphine with JWH015 synergistically inhibited preclinical inflammatory, post-operative and neuropathic-pain in a dose- and time-dependent manner; no synergy was observed for nociceptive pain. Opioid-induced side effects of impaired gastrointestinal transit and CPP were significantly reduced in the presence of JWH015. Here we show that MOR + CB2 agonism results in a significant synergistic inhibition of preclinical pain while significantly reducing opioid-induced unwanted side effects. The opioid sparing effect of CB2 receptor agonism strongly supports the advancement of a MOR-CB2 agonist combinatorial pain therapy for clinical trials.


Assuntos
Analgésicos não Narcóticos/farmacologia , Analgésicos Opioides/farmacologia , Agonistas de Receptores de Canabinoides/farmacologia , Dor Crônica/tratamento farmacológico , Indóis/farmacologia , Morfina/farmacologia , Analgésicos Opioides/efeitos adversos , Animais , Dor Crônica/metabolismo , Constipação Intestinal/induzido quimicamente , Constipação Intestinal/tratamento farmacológico , Constipação Intestinal/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Masculino , Camundongos Endogâmicos ICR , Morfina/efeitos adversos , Ratos Sprague-Dawley , Receptor CB2 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Recompensa
7.
Expert Opin Drug Discov ; 9(11): 1345-54, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25307021

RESUMO

INTRODUCTION: Since ancient times, the opium poppy has been used in a variety of settings, including pain management. Natural and synthetic derivatives of opium are commonly used in medicine today and include drugs, such as morphine, codeine, hydromorphone and oxycodone. Although excellent at inhibiting pain, these narcotics often produce a state of euphoria leading to misuse and abuse by the general population, particularly in young adults. The misuse of prescription opiates has continually increased over the past 10 years despite associated negative outcomes, resulting in opiate psychological dependence, withdrawal and relapse. AREAS COVERED: This paper briefly refers to the history of opiate use and the modern challenges associated with chronic exposure. The authors present the prevalence of addiction and misuse of prescription opiates and discuss some of the opiate-associated effects. This includes activation of reward circuitry and compensatory receptor mechanisms. Finally, the authors provide a review on neuroadaptive changes that manifest during opiate dependence, withdrawal and relapse in animal models. EXPERT OPINION: In spite of the various methods available to treat opiate addiction, there is still a huge unmet need for its management, including the creative design of novel, non-addictive pain medications. The authors believe that multifunctional compounds or combinations of compounds that inhibit pain pathways, whereas not activating the reward pathways, will begin to subdue the opiate addiction endemic.


Assuntos
Modelos Animais de Doenças , Descoberta de Drogas , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Animais , Tolerância a Medicamentos , Humanos , Transtornos Relacionados ao Uso de Opioides/psicologia , Recidiva , Recompensa , Síndrome de Abstinência a Substâncias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA