Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 142(22): 1918-1927, 2023 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-37774369

RESUMO

Vaso-occlusive pain episodes (VOE) cause severe pain in patients with sickle cell disease (SCD). Vaso-occlusive events promote ischemia/reperfusion pathobiology that activates complement. We hypothesized that complement activation is linked to VOE. We used cold to induce VOE in the Townes sickle homozygous for hemoglobin S (HbSS) mouse model and complement inhibitors to determine whether anaphylatoxin C5a mediates VOE. We used a dorsal skinfold chamber to measure microvascular stasis (vaso-occlusion) and von Frey filaments applied to the plantar surface of the hind paw to assess mechanical hyperalgesia in HbSS and control Townes mice homozygous for hemoglobin A (HbAA) mice after cold exposure at 10°C/50°F for 1 hour. Cold exposure induced more vaso-occlusion in nonhyperalgesic HbSS mice (33%) than in HbAA mice (11%) or HbSS mice left at room temperature (1%). Cold exposure also produced mechanical hyperalgesia as measured by paw withdrawal threshold in HbSS mice compared with that in HbAA mice or HbSS mice left at room temperature. Vaso-occlusion and hyperalgesia were associated with an increase in complement activation fragments Bb and C5a in plasma of HbSS mice after cold exposure. This was accompanied by an increase in proinflammatory NF-κB activation and VCAM-1 and ICAM-1 expression in the liver. Pretreatment of nonhyperalgesic HbSS mice before cold exposure with anti-C5 or anti-C5aR monoclonal antibodies (mAbs) decreased vaso-occlusion, mechanical hyperalgesia, complement activation, and liver inflammatory markers compared with pretreatment with control mAb. Anti-C5 or -C5aR mAb infusion also abrogated mechanical hyperalgesia in HbSS mice with ongoing hyperalgesia at baseline. These findings suggest that C5a promotes vaso-occlusion, pain, and inflammation during VOE and may play a role in chronic pain.


Assuntos
Anemia Falciforme , Traço Falciforme , Camundongos , Humanos , Animais , Hiperalgesia/etiologia , Hiperalgesia/metabolismo , Camundongos Transgênicos , Dor , Anemia Falciforme/complicações , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Traço Falciforme/complicações , Ativação do Complemento
2.
Neuroscience ; 526: 1-12, 2023 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-37330194

RESUMO

Sickle cell disease (SCD) is an inherited blood disorder that is associated with acute episodic and chronic pain. Mice with SCD have robust hyperalgesia mediated, in part, by sensitization of spinal dorsal horn neurons. However, underlying mechanisms are not fully understood. Since the rostral ventromedial medulla (RVM) is a major component of descending circuitry that modulates nociceptive transmission in the spinal cord, we examined if the RVM contributes to hyperalgesia in mice with SCD. Injection of lidocaine, but not vehicle, into the RVM eliminated mechanical and heat hyperalgesia in sickle (HbSS-BERK) mice without altering mechanical and heat sensitivity in naïve C57B mice. These data indicate that the RVM contributes to the maintenance of hyperalgesia in mice with SCD. In electrophysiological studies, we determined the changes in response properties of RVM neurons that might contribute to hyperalgesia in sickle mice. Recordings were made from single ON, OFF, and Neutral cells in the RVM of sickle and control (HbAA-BERK) mice. Spontaneous activity and responses of ON, OFF and Neutral cells evoked by heat (50 °C) and mechanical (26 g) stimuli applied to the hind paw were compared between sickle and control mice. Although there were no differences in the proportions of functionally-identified neurons or spontaneous activity between sickle and control mice, evoked responses of ON cells to heat and mechanical stimuli were increased approximately 3-fold in sickle mice as compared to control mice. Thus, the RVM contributes to hyperalgesia in sickle mice via a specific ON cell-dependent descending facilitation of nociceptive transmission.


Assuntos
Anemia Falciforme , Hiperalgesia , Ratos , Camundongos , Animais , Nociceptividade/fisiologia , Ratos Sprague-Dawley , Neurônios/fisiologia , Anemia Falciforme/complicações , Bulbo
3.
Pain ; 164(12): 2684-2695, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37278638

RESUMO

ABSTRACT: Pain associated with bone cancer remains poorly managed, and chemotherapeutic drugs used to treat cancer usually increase pain. The discovery of dual-acting drugs that reduce cancer and produce analgesia is an optimal approach. The mechanisms underlying bone cancer pain involve interactions between cancer cells and nociceptive neurons. We demonstrated that fibrosarcoma cells express high levels of autotaxin (ATX), the enzyme synthetizing lysophosphatidic acid (LPA). Lysophosphatidic acid increased proliferation of fibrosarcoma cells in vitro. Lysophosphatidic acid is also a pain-signaling molecule, which activates LPA receptors (LPARs) located on nociceptive neurons and satellite cells in dorsal root ganglia. We therefore investigated the contribution of the ATX-LPA-LPAR signaling to pain in a mouse model of bone cancer pain in which fibrosarcoma cells are implanted into and around the calcaneus bone, resulting in tumor growth and hypersensitivity. LPA was elevated in serum of tumor-bearing mice, and blockade of ATX or LPAR reduced tumor-evoked hypersensitivity. Because cancer cell-secreted exosomes contribute to hypersensitivity and ATX is bound to exosomes, we determined the role of exosome-associated ATX-LPA-LPAR signaling in hypersensitivity produced by cancer exosomes. Intraplantar injection of cancer exosomes into naive mice produced hypersensitivity by sensitizing C-fiber nociceptors. Inhibition of ATX or blockade of LPAR attenuated cancer exosome-evoked hypersensitivity in an ATX-LPA-LPAR-dependent manner. Parallel in vitro studies revealed the involvement of ATX-LPA-LPAR signaling in direct sensitization of dorsal root ganglion neurons by cancer exosomes. Thus, our study identified a cancer exosome-mediated pathway, which may represent a therapeutic target for treating tumor growth and pain in patients with bone cancer.


Assuntos
Neoplasias Ósseas , Dor do Câncer , Exossomos , Fibrossarcoma , Humanos , Animais , Camundongos , Dor do Câncer/etiologia , Lisofosfolipídeos/metabolismo , Neoplasias Ósseas/complicações , Dor/tratamento farmacológico , Dor/etiologia
4.
Haematologica ; 108(3): 859-869, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35615929

RESUMO

Sickle cell disease (SCD) is the most common inherited disease. Pain is a key morbidity of SCD and opioids are the main treatment but their side effects emphasize the need for new analgesic approaches. Humanized transgenic mouse models have been instructive in understanding the pathobiology of SCD and mechanisms of pain. Homozygous (HbSS) Berkley mice express >99% human sickle hemoglobin and several features of clinical SCD including hyperalgesia. Previously, we reported that the endocannabinoid 2-arachidonoylglycerol (2-AG) is a precursor of the pro-nociceptive mediator prostaglandin E2-glyceryl ester (PGE2-G) which contributes to hyperalgesia in SCD. We now demonstrate the causal role of 2-AG in hyperalgesia in sickle mice. Hyperalgesia in HbSS mice correlated with elevated levels of 2-AG in plasma, its synthesizing enzyme diacylglycerol lipase ß (DAGLß) in blood cells, and with elevated levels of PGE2 and PGE2-G, pronociceptive derivatives of 2-AG. A single intravenous injection of 2-AG produced hyperalgesia in non-hyperalgesic HbSS mice, but not in control (HbAA) mice expressing normal human HbA. JZL184, an inhibitor of 2-AG hydrolysis, also produced hyperalgesia in non-hyperalgesic HbSS or hemizygous (HbAS) mice, but did not influence hyperalgesia in hyperalgesic HbSS mice. Systemic and intraplantar administration of KT109, an inhibitor of DAGLß, decreased mechanical and heat hyperalgesia in HbSS mice. The decrease in hyperalgesia was accompanied by reductions in 2-AG, PGE2 and PGE2-G in the blood. These results indicate that maintaining the physiological level of 2-AG in the blood by targeting DAGLß may be a novel and effective approach to treat pain in SCD.


Assuntos
Anemia Falciforme , Hiperalgesia , Camundongos , Humanos , Animais , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Dinoprostona , Dor/tratamento farmacológico , Dor/etiologia , Camundongos Transgênicos , Anemia Falciforme/complicações , Anemia Falciforme/tratamento farmacológico , Hemoglobina Falciforme
5.
Mol Pain ; 18: 17448069221142523, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-36408567

RESUMO

Methylene blue (MB) is an effective treatment for methemoglobinemia, ifosfamide-induced encephalopathy, cyanide poisoning, and refractory vasoplegia. However, clinical case reports and preclinical studies indicate potentially neurotoxic activity of MB at certain concentrations. The exact mechanisms of MB neurotoxicity are not known, and while the effects of MB on neuronal tissue from different brain regions and myenteric ganglia have been examined, its effects on primary afferent neurons from dorsal root ganglia (DRG) have not been studied. Mouse DRG were exposed to MB (0.3-10 µM) in vitro to assess neurite outgrowth. Increasing concentrations of MB (0.3-10 µM) were associated with neurotoxicity as shown by a substantial loss of cells with neurite formation, particularly at 10 µM. In parallel experiments, cultured rat DRG neurons were treated with MB (100 µM) to examine how MB affects electrical membrane properties of small-diameter sensory neurons. MB decreased peak inward and outward current densities, decreased action potential amplitude, overshoot, afterhyperpolarization, increased action potential rise time, and decreased action potential firing in response to current stimulation. MB induced dose-dependent toxicity in peripheral neurons, in vitro. These findings are consistent with studies in brain and myenteric ganglion neurons showing increased neuronal loss and altered membrane electrical properties after MB application. Further research is needed to parse out the toxicity profile for MB to minimize damage to neuronal structures and reduce side effects in clinical settings.


Assuntos
Gânglios Espinais , Azul de Metileno , Ratos , Camundongos , Animais , Azul de Metileno/farmacologia , Azul de Metileno/metabolismo , Gânglios Espinais/metabolismo , Células Receptoras Sensoriais/metabolismo , Eletrofisiologia , Técnicas de Cultura de Células , Células Cultivadas
6.
Neurosci Lett ; 753: 135845, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33774149

RESUMO

Although millions of people are diagnosed with cancer each year, survival has never been greater thanks to early diagnosis and treatments. Powerful chemotherapeutic agents are highly toxic to cancer cells, but because they typically do not target cancer cells selectively, they are often toxic to other cells and produce a variety of side effects. In particular, many common chemotherapies damage the peripheral nervous system and produce neuropathy that includes a progressive degeneration of peripheral nerve fibers. Chemotherapy-induced peripheral neuropathy (CIPN) can affect all nerve fibers, but sensory neuropathies are the most common, initially affecting the distal extremities. Symptoms include impaired tactile sensitivity, tingling, numbness, paraesthesia, dysesthesia, and pain. Since neuropathic pain is difficult to manage, and because degenerated nerve fibers may not grow back and regain normal function, considerable research has focused on understanding how chemotherapy causes painful CIPN so it can be prevented. Due to the fact that both therapeutic and side effects of chemotherapy are primarily associated with the accumulation of reactive oxygen species (ROS) and oxidative stress, this review focuses on the activation of endogenous antioxidant pathways, especially PPARγ, in order to prevent the development of CIPN and associated pain. The use of synthetic and natural PPARγ agonists to prevent CIPN is discussed.


Assuntos
Antineoplásicos/efeitos adversos , Neoplasias/tratamento farmacológico , Neuralgia/prevenção & controle , PPAR gama/agonistas , Animais , Modelos Animais de Doenças , Humanos , Neuralgia/induzido quimicamente , Estresse Oxidativo/efeitos dos fármacos , PPAR gama/metabolismo , Espécies Reativas de Oxigênio/metabolismo
7.
Prostaglandins Other Lipid Mediat ; 151: 106479, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32745525

RESUMO

Pain produced by bone cancer is often severe and difficult to treat. Here we examined effects of Resolvin D1 (RvD1) or E1 (RvE1), antinociceptive products of ω-3 polyunsaturated fatty acids, on cancer-induced mechanical allodynia and heat hyperalgesia. Experiments were performed using a mouse model of bone cancer produced by implantation of osteolytic ficrosarcoma into and around the calcaneus bone. Mechanical allodynia and heat hyperalgesia in the tumor-bearing paw were assessed by measuring withdrawal responses to a von Frey monofilament and to radiant heat applied on the plantar hind paw. RvD1, RvE1, and cannabinoid receptor antagonists were injected intrathecally. Spinal content of endocannabinoids was evaluated using UPLC-MS/MS analysis. RvD1 and RvE1 had similar antinociceptive potencies. ED50s for RvD1 and RvE1 in reducing mechanical allodynia were 0.2 pg (0.53 fmol) and 0.6 pg (1.71 fmol), respectively, and were 0.3 pg (0.8 fmol) and 0.2 pg (0.57 fmol) for reducing heat hyperalgesia. Comparisons of dose-response relationships showed equal efficacy for reducing mechanical allodynia, however, efficacy for reducing heat hyperalgesia was greater for of RvD1. Using UPLC-MS/MS we determined that RvD1, but not RvE1, increased levels of the endocannabinoids Anandamide and 2-Arachidonoylglycerol in the spinal cord. Importantly, Resolvins did not alter acute nociception or motor function in naïve mice. Our data indicate, that RvD1 and RvE1 produce potent antiallodynia and antihyperalgesia in a model of bone cancer pain. RvD1 also triggers spinal upregulation of endocannabinoids that produce additional antinociception predominantly through CB2 receptors.


Assuntos
Neoplasias Ósseas/complicações , Dor do Câncer/tratamento farmacológico , Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Eicosapentaenoico/análogos & derivados , Endocanabinoides/metabolismo , Hiperalgesia/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Dor do Câncer/patologia , Ácidos Docosa-Hexaenoicos/uso terapêutico , Ácido Eicosapentaenoico/farmacologia , Ácido Eicosapentaenoico/uso terapêutico , Hiperalgesia/patologia , Masculino , Camundongos
8.
Blood ; 133(18): 1989-1998, 2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-30796025

RESUMO

Pain is a characteristic feature of sickle cell disease (SCD), 1 of the most common inherited diseases. Patients may experience acute painful crises as well as chronic pain. In the Berkley transgenic murine model of SCD, HbSS-BERK mice express only human hemoglobin S. These mice share many features of SCD patients, including persistent inflammation and hyperalgesia. Cyclooxygenase-2 (COX-2) is elevated in skin, dorsal root ganglia (DRG), and spinal cord in HbSS-BERK mice. In addition to arachidonic acid, COX-2 oxidizes the endocannabinoid 2-arachidonoylglycerol (2-AG) to produce prostaglandin E2 (PGE2)-glycerol (PGE2-G); PGE2-G is known to produce hyperalgesia. We tested the hypotheses that PGE2-G is increased in DRGs of HbSS-BERK mice and sensitizes nociceptors (sensory neurons that respond to noxious stimuli), and that blocking its synthesis would decrease hyperalgesia in HbSS-BERK mice. Systemic administration of R-flurbiprofen preferentially reduced production of PGE2-G over that of PGE2 in DRGs, decreased mechanical and thermal hyperalgesia, and decreased sensitization of nociceptors in HbSS-BERK mice. The same dose of R-flurbiprofen had no behavioral effect in HbAA-BERK mice (the transgenic control), but local injection of PGE2-G into the hind paw of HbAA-BERK mice produced sensitization of nociceptors and hyperalgesia. Coadministration of a P2Y6 receptor antagonist blocked the effect of PGE2-G, indicating that this receptor is a mediator of pain in SCD. The ability of R-flurbiprofen to block the synthesis of PGE2-G and to normalize levels of 2-AG suggests that R-flurbiprofen may be beneficial to treat pain in SCD, thereby reducing the use of opioids to relieve pain.


Assuntos
Anemia Falciforme/metabolismo , Dinoprostona/análogos & derivados , Hiperalgesia/metabolismo , Nociceptores/metabolismo , Anemia Falciforme/complicações , Animais , Dinoprostona/metabolismo , Humanos , Hiperalgesia/etiologia , Camundongos , Camundongos Transgênicos
9.
Pain ; 160(3): 688-701, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30507781

RESUMO

Painful peripheral neuropathy is a dose-limiting side effect of cisplatin treatment. Using a murine model of cisplatin-induced hyperalgesia, we determined whether a PPARγ synthetic agonist, pioglitazone, attenuated the development of neuropathic pain and identified underlying mechanisms. Cisplatin produced mechanical and cold hyperalgesia and decreased electrical thresholds of Aδ and C fibers, which were attenuated by coadministration of pioglitazone (10 mg/kg, intraperitoneally [i.p.]) with cisplatin. Antihyperalgesic effects of pioglitazone were blocked by the PPARγ antagonist T0070907 (10 mg/kg, i.p.). We hypothesized that the ability of pioglitazone to reduce the accumulation of reactive oxygen species (ROS) in dorsal root ganglion (DRG) neurons contributed to its antihyperalgesic activity. Effects of cisplatin and pioglitazone on somatosensory neurons were studied on dissociated mouse DRG neurons after 24 hours in vitro. Incubation of DRG neurons with cisplatin (13 µM) for 24 hours increased the occurrence of depolarization-evoked calcium transients, and these were normalized by coincubation with pioglitazone (10 µM). Oxidative stress in DRG neurons was considered a significant contributor to cisplatin-evoked hyperalgesia because a ROS scavenger attenuated hyperalgesia and normalized the evoked calcium responses when cotreated with cisplatin. Pioglitazone increased the expression and activity of ROS-reducing enzymes in DRG and normalized cisplatin-evoked changes in oxidative stress and labeling of mitochondria with the dye MitoTracker Deep Red, indicating that the antihyperalgesic effects of pioglitazone were attributed to its antioxidant properties in DRG neurons. These data demonstrate clear benefits of broadening the use of the antidiabetic drug pioglitazone, or other PPARγ agonists, to minimize the development of cisplatin-induced painful neuropathy.


Assuntos
Hipoglicemiantes/uso terapêutico , Neuralgia/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , PPAR gama/metabolismo , Pioglitazona/uso terapêutico , Animais , Antineoplásicos/toxicidade , Células Cultivadas , Cisplatino/toxicidade , Modelos Animais de Doenças , Feminino , Gânglios Espinais/citologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Masculino , Camundongos , Mitocôndrias/efeitos dos fármacos , Neuralgia/induzido quimicamente , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Limiar da Dor/efeitos dos fármacos , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo
10.
J Neurophysiol ; 113(5): 1501-10, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25505113

RESUMO

Painful neuropathy frequently develops as a consequence of commonly used chemotherapy agents for cancer treatment and is often a dose-limiting side effect. Currently available analgesic treatments are often ineffective on pain induced by neurotoxicity. Although peripheral administration of cannabinoids, endocannabinoids, and inhibitors of endocannabinoid hydrolysis has been effective in reducing hyperalgesia in models of peripheral neuropathy, including chemotherapy-induced peripheral neuropathy (CIPN), few studies have examined cannabinoid effects on responses of nociceptors in vivo. In this study we determined whether inhibition of fatty acid amide hydrolase (FAAH), which slows the breakdown of the endocannabinoid anandamide (AEA), reduced sensitization of nociceptors produced by chemotherapy. Over the course of a week of daily treatments, mice treated with the platinum-based chemotherapy agent cisplatin developed robust mechanical allodynia that coincided with sensitization of cutaneous C-fiber nociceptors as indicated by the development of spontaneous activity and increased responses to mechanical stimulation. Administration of the FAAH inhibitor URB597 into the receptive field of sensitized C-fiber nociceptors decreased spontaneous activity, increased mechanical response thresholds, and decreased evoked responses to mechanical stimuli. Cotreatment with CB1 (AM281) or CB2 (AM630) receptor antagonists showed that the effect of URB597 was mediated primarily by CB1 receptors. These changes following URB597 were associated with an increase in the endocannabinoid anandamide in the skin. Our results suggest that enhanced signaling in the peripheral endocannabinoid system could be utilized to reduce nociceptor sensitization and pain associated with CIPN.


Assuntos
Amidoidrolases/antagonistas & inibidores , Ácidos Araquidônicos/metabolismo , Endocanabinoides/metabolismo , Nociceptividade , Nociceptores/metabolismo , Doenças do Sistema Nervoso Periférico/metabolismo , Alcamidas Poli-Insaturadas/metabolismo , Amidoidrolases/metabolismo , Animais , Benzamidas/farmacologia , Antagonistas de Receptores de Canabinoides/farmacologia , Carbamatos/farmacologia , Cisplatino/efeitos adversos , Hidrólise , Indóis/farmacologia , Masculino , Camundongos , Morfolinas/farmacologia , Fibras Nervosas Amielínicas/metabolismo , Fibras Nervosas Amielínicas/fisiologia , Nociceptores/efeitos dos fármacos , Nociceptores/fisiologia , Doenças do Sistema Nervoso Periférico/etiologia , Doenças do Sistema Nervoso Periférico/fisiopatologia , Pirazóis/farmacologia , Pele/lesões
11.
Pharmacol Res ; 90: 67-75, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25304184

RESUMO

Cisplatin has been used effectively to treat a variety of cancers but its use is limited by the development of painful peripheral neuropathy. Because the endocannabinoid 2-arachidonoyl-sn-glycerol (2-AG) is anti-hyperalgesic in several preclinical models of chronic pain, the anti-hyperalgesic effect of JZL184, an inhibitor of 2-AG hydrolysis, was tested in a murine model of cisplatin-induced hyperalgesia. Systemic injection of cisplatin (1mg/kg) produced mechanical hyperalgesia when administered daily for 7 days. Daily peripheral administration of a low dose of JZL184 in conjunction with cisplatin blocked the expression of mechanical hyperalgesia. Acute injection of a cannabinoid (CB)-1 but not a CB2 receptor antagonist reversed the anti-hyperalgesic effect of JZL184 indicating that downstream activation of CB1 receptors suppressed the expression of mechanical hyperalgesia. Components of endocannabinoid signaling in plantar hind paw skin and lumbar dorsal root ganglia (DRGs) were altered by treatments with cisplatin and JZL184. Treatment with cisplatin alone reduced levels of 2-AG and AEA in skin and DRGs as well as CB2 receptor protein in skin. Combining treatment of JZL184 with cisplatin increased 2-AG in DRGs compared to cisplatin alone but had no effect on the amount of 2-AG in skin. Evidence that JZL184 decreased the uptake of [(3)H]AEA into primary cultures of DRGs at a concentration that also inhibited the enzyme fatty acid amide hydrolase, in conjunction with data that 2-AG mimicked the effect of JZL184 on [(3)H]AEA uptake support the conclusion that AEA most likely mediates the anti-hyperalgesic effect of JZL184 in this model.


Assuntos
Analgésicos/uso terapêutico , Benzodioxóis/uso terapêutico , Hiperalgesia/tratamento farmacológico , Neuralgia/tratamento farmacológico , Piperidinas/uso terapêutico , Amidas , Analgésicos/farmacologia , Animais , Antineoplásicos , Ácidos Araquidônicos/metabolismo , Ácidos Araquidônicos/farmacologia , Benzodioxóis/farmacologia , Células Cultivadas , Cisplatino , Modelos Animais de Doenças , Endocanabinoides/metabolismo , Endocanabinoides/farmacologia , Etanolaminas/metabolismo , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Glicerídeos/metabolismo , Glicerídeos/farmacologia , Hiperalgesia/metabolismo , Indóis/farmacologia , Masculino , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Monoacilglicerol Lipases/antagonistas & inibidores , Morfolinas/farmacologia , Neuralgia/induzido quimicamente , Neuralgia/metabolismo , Ácidos Palmíticos/metabolismo , Piperidinas/farmacologia , Alcamidas Poli-Insaturadas/metabolismo , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/antagonistas & inibidores , Receptor CB2 de Canabinoide/metabolismo , Pele/efeitos dos fármacos , Pele/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
12.
Neurobiol Dis ; 58: 19-28, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23644187

RESUMO

Opioids do not effectively manage pain in many patients with advanced cancer. Because anandamide (AEA) activation of cannabinoid type-1 receptors (CB1R) on nociceptors reduces nociception, manipulation of AEA metabolism in the periphery may be an effective alternative or adjuvant therapy in the management of cancer pain. AEA is hydrolyzed by the intracellular enzyme fatty acid amide hydrolase (FAAH), and this enzyme activity contributes to uptake of AEA into neurons and to reduction of AEA available to activate CB1R. We used an in vitro preparation of adult murine dorsal root ganglion (DRG) neurons co-cultured with fibrosarcoma cells to investigate how tumors alter the uptake of AEA into neurons. Evidence that the uptake of [(3)H]AEA into dissociated DRG cells in the co-culture model mimicked the increase in uptake that occurred in DRG cells from tumor-bearing mice supported the utility of the in vitro model to study AEA uptake. Results with the fluorescent AEA analog CAY10455 confirmed that an increase in uptake in the co-culture model occurred in neurons. One factor that contributed to the increase in [(3)H]AEA uptake was an increase in total cellular cholesterol in the cancer condition. Treatment with the FAAH inhibitor URB597 reduced CAY10455 uptake in the co-culture model to the level observed in DRG neurons maintained in the control condition (i.e., in the absence of fibrosarcoma cells), and this effect was paralleled by OMDM-1, an inhibitor of AEA uptake, at a concentration that had no effect on FAAH activity. Maximally effective concentrations of the two drugs together produced a greater reduction than was observed with each drug alone. Treatment with BMS309403, which competes for AEA binding to fatty acid binding protein-5, mimicked the effect of OMDM-1 in vitro. Local injection of OMDM-1 reduced hyperalgesia in vivo in mice with unilateral tumors in and around the calcaneous bone. Intraplantar injection of OMDM-1 (5µg) into the tumor-bearing paw reduced mechanical hyperalgesia through a CB1R-dependent mechanism and also reduced a spontaneous nocifensive behavior. The same dose reduced withdrawal responses evoked by suprathreshold mechanical stimuli in naive mice. These data support the conclusion that OMDM-1 inhibits AEA uptake by a mechanism that is independent of inhibition of FAAH and provide a rationale for the development of peripherally restricted drugs that decrease AEA uptake for the management of cancer pain.


Assuntos
Ácidos Araquidônicos/metabolismo , Endocanabinoides/metabolismo , Hiperalgesia/etiologia , Limiar da Dor/fisiologia , Dor/complicações , Dor/patologia , Alcamidas Poli-Insaturadas/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Benzamidas/farmacologia , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/patologia , Antagonistas de Receptores de Canabinoides/farmacologia , Carbamatos/farmacologia , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Fibrossarcoma/complicações , Fibrossarcoma/patologia , Corantes Fluorescentes , Gânglios Espinais/citologia , Indóis/farmacologia , Lactonas , Masculino , Camundongos , Camundongos Endogâmicos C3H , Dor/etiologia , Limiar da Dor/efeitos dos fármacos , Células Receptoras Sensoriais/efeitos dos fármacos , Estatísticas não Paramétricas , Trítio/metabolismo
13.
J Neurosci ; 32(37): 12735-43, 2012 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-22972997

RESUMO

The amplitude of the depolarization-evoked Ca2+ transient is larger in dorsal root ganglion (DRG) neurons from tumor-bearing mice compared with that of neurons from naive mice, and the change is mimicked by coculturing DRG neurons with the fibrosarcoma cells used to generate the tumors (Khasabova et al., 2007). The effect of palmitoylethanolamide (PEA), a ligand for the peroxisome proliferator-activated receptor α (PPARα), was determined on the evoked-Ca2+ transient in the coculture condition. The level of PEA was reduced in DRG cells from tumor-bearing mice as well as those cocultured with fibrosarcoma cells. Pretreatment with PEA, a synthetic PPARα agonist (GW7647), or ARN077, an inhibitor of the enzyme that hydrolyzes PEA, acutely decreased the amplitude of the evoked Ca2+ transient in small DRG neurons cocultured with fibrosarcoma cells. The PPARα antagonist GW6471 blocked the effect of each. In contrast, the PPARα agonist was without effect in the control condition, but the antagonist increased the amplitude of the Ca2+ transient, suggesting that PPARα receptors are saturated by endogenous ligand under basal conditions. Effects of drugs on mechanical sensitivity in vivo paralleled their effects on DRG neurons in vitro. Local injection of ARN077 decreased mechanical hyperalgesia in tumor-bearing mice, and the effect was blocked by GW6471. These data support the conclusion that the activity of DRG neurons is rapidly modulated by PEA through a PPARα-dependent mechanism. Moreover, agents that increase the activity of PPARα may provide a therapeutic strategy to reduce tumor-evoked pain.


Assuntos
Sinalização do Cálcio/fisiologia , Endocanabinoides/farmacologia , Etanolaminas/farmacologia , Gânglios Espinais/fisiologia , PPAR alfa/metabolismo , Ácidos Palmíticos/farmacologia , Células Receptoras Sensoriais/fisiologia , Amidas , Animais , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Células Receptoras Sensoriais/efeitos dos fármacos
14.
J Neurosci ; 32(20): 7091-101, 2012 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22593077

RESUMO

Painful peripheral neuropathy is a dose-limiting complication of chemotherapy. Cisplatin produces a cumulative toxic effect on peripheral nerves, and 30-40% of cancer patients receiving this agent experience pain. By modeling cisplatin-induced hyperalgesia in mice with daily injections of cisplatin (1 mg/kg, i.p.) for 7 d, we investigated the anti-hyperalgesic effects of anandamide (AEA) and cyclohexylcarbamic acid 3'-carbamoyl-biphenyl-3-yl ester (URB597), an inhibitor of AEA hydrolysis. Cisplatin-induced mechanical and heat hyperalgesia were accompanied by a decrease in the level of AEA in plantar paw skin. No changes in motor activity were observed after seven injections of cisplatin. Intraplantar injection of AEA (10 µg/10 µl) or URB597 (9 µg/10 µl) transiently attenuated hyperalgesia through activation of peripheral CB1 receptors. Co-injections of URB597 (0.3 mg/kg daily, i.p.) with cisplatin decreased and delayed the development of mechanical and heat hyperalgesia. The effect of URB597 was mediated by CB1 receptors since AM281 (0.33 mg/kg daily, i.p.) blocked the effect of URB597. Co-injection of URB597 also normalized the cisplatin-induced decrease in conduction velocity of Aα/Aß-fibers and reduced the increase of ATF-3 and TRPV1 immunoreactivity in dorsal root ganglion (DRG) neurons. Since DRGs are a primary site of toxicity by cisplatin, effects of cisplatin were studied on cultured DRG neurons. Incubation of DRG neurons with cisplatin (4 µg/ml) for 24 h decreased the total length of neurites. URB597 (100 nM) attenuated these changes through activation of CB1 receptors. Collectively, these results suggest that pharmacological facilitation of AEA signaling is a promising strategy for attenuating cisplatin-associated sensory neuropathy.


Assuntos
Ácidos Araquidônicos/uso terapêutico , Benzamidas/uso terapêutico , Carbamatos/uso terapêutico , Cisplatino/antagonistas & inibidores , Hiperalgesia/tratamento farmacológico , Síndromes Neurotóxicas/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Alcamidas Poli-Insaturadas/uso terapêutico , Receptor CB1 de Canabinoide/agonistas , Fator 3 Ativador da Transcrição/metabolismo , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/antagonistas & inibidores , Ácidos Araquidônicos/farmacocinética , Ácidos Araquidônicos/farmacologia , Benzamidas/antagonistas & inibidores , Benzamidas/farmacologia , Moduladores de Receptores de Canabinoides/farmacocinética , Moduladores de Receptores de Canabinoides/farmacologia , Moduladores de Receptores de Canabinoides/uso terapêutico , Carbamatos/antagonistas & inibidores , Carbamatos/farmacologia , Células Cultivadas , Cisplatino/efeitos adversos , Modelos Animais de Doenças , Interações Medicamentosas , Endocanabinoides , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Hiperalgesia/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C3H , Morfolinas/farmacologia , Atividade Motora/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Alcamidas Poli-Insaturadas/farmacocinética , Alcamidas Poli-Insaturadas/farmacologia , Pirazóis/farmacologia , Canais de Cátion TRPV/metabolismo
16.
Behav Pharmacol ; 22(5-6): 607-16, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21610490

RESUMO

In light of the adverse side-effects of opioids, cannabinoid receptor agonists may provide an effective alternative for the treatment of cancer pain. This study examined the potency and efficacy of synthetic CB1 and CB2 receptor agonists in a murine model of tumor pain. Intraplantar injection of the CB1 receptor agonist arachidonylcyclopropylamide (ED(50) of 18.4 µg) reduced tumor-related mechanical hyperalgesia by activation of peripheral CB1 but not CB2 receptors. Similar injection of the CB2 receptor agonist AM1241 (ED50 of 19.5 µg) reduced mechanical hyperalgesia by activation of peripheral CB2 but not CB1 receptors. Both agonists had an efficacy comparable with that of morphine (intraplantar), but their analgesic effects were independent of opioid receptors. Isobolographic analysis of the coinjection of arachidonylcyclopropylamide and AM1241 determined that the CB1 and CB2 receptor agonists interacted synergistically to reduce mechanical hyperalgesia in the tumor-bearing paw. These data extend our previous findings that the peripheral cannabinoid receptors are a promising target for the management of cancer pain and mixed cannabinoid receptor agonists may have a therapeutic advantage over selective agonists.


Assuntos
Neoplasias Experimentais/complicações , Dor/tratamento farmacológico , Receptor CB1 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/agonistas , Analgésicos/farmacologia , Animais , Ácidos Araquidônicos/farmacologia , Canabinoides/farmacologia , Modelos Animais de Doenças , Sinergismo Farmacológico , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Morfina/farmacologia , Dor/etiologia
17.
Pharmacol Res ; 64(1): 60-7, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21440630

RESUMO

Metastatic and primary bone cancers are usually accompanied by severe pain that is difficult to manage. In light of the adverse side effects of opioids, manipulation of the endocannabinoid system may provide an effective alternative for the treatment of cancer pain. The present study determined that a local, peripheral increase in the endocannabinoid 2-arachidonoyl glycerol (2-AG) reduced mechanical hyperalgesia evoked by the growth of a fibrosarcoma tumor in and around the calcaneous bone. Intraplantar (ipl) injection of 2-AG attenuated hyperalgesia (ED(50) of 8.2 µg) by activation of peripheral CB2 but not CB1 receptors and had an efficacy comparable to that of morphine. JZL184 (10 µg, ipl), an inhibitor of 2-AG degradation, increased the local level of 2-AG and mimicked the anti-hyperalgesic effect of 2-AG, also through a CB2 receptor-dependent mechanism. These effects were accompanied by an increase in CB2 receptor protein in plantar skin of the tumor-bearing paw as well as an increase in the level of 2-AG. In naïve mice, intraplantar administration of the CB2 receptor antagonist AM630 did not alter responses to mechanical stimuli demonstrating that peripheral CB2 receptor tone does not modulate mechanical sensitivity. These data extend our previous findings with anandamide in the same model and suggest that the peripheral endocannabinoid system is a promising target for the management of cancer pain.


Assuntos
Ácidos Araquidônicos/uso terapêutico , Neoplasias Ósseas/complicações , Glicerídeos/uso terapêutico , Hiperalgesia/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Ácidos Araquidônicos/administração & dosagem , Ácidos Araquidônicos/metabolismo , Benzodioxóis/administração & dosagem , Benzodioxóis/farmacologia , Neoplasias Ósseas/metabolismo , Calcâneo/patologia , Antagonistas de Receptores de Canabinoides , Relação Dose-Resposta a Droga , Endocanabinoides , Fibrossarcoma/complicações , Fibrossarcoma/metabolismo , Gânglios Espinais/metabolismo , Glicerídeos/administração & dosagem , Glicerídeos/metabolismo , Hiperalgesia/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Monoacilglicerol Lipases/antagonistas & inibidores , Piperidinas/administração & dosagem , Piperidinas/farmacologia , Alcamidas Poli-Insaturadas/metabolismo , Receptor CB2 de Canabinoide/antagonistas & inibidores , Receptor CB2 de Canabinoide/metabolismo , Pele/efeitos dos fármacos , Pele/metabolismo , Nervo Tibial/metabolismo
18.
J Neurosci ; 28(44): 11141-52, 2008 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-18971457

RESUMO

Tumors in bone are associated with pain in humans. Data generated in a murine model of bone cancer pain suggest that a disturbance of local endocannabinoid signaling contributes to the pain. When tumors formed after injection of osteolytic fibrosarcoma cells into the calcaneus bone of mice, cutaneous mechanical hyperalgesia was associated with a decrease in the level of anandamide (AEA) in plantar paw skin ipsilateral to tumors. The decrease in AEA occurred in conjunction with increased degradation of AEA by fatty acid amide hydrolase (FAAH). Intraplantar injection of AEA reduced the hyperalgesia, and intraplantar injection of URB597, an inhibitor of FAAH, increased the local level of AEA and also reduced hyperalgesia. An increase in FAAH mRNA and enzyme activity in dorsal root ganglia (DRG) L3-L5 ipsilateral to the affected paw suggests DRG neurons contribute to the increased FAAH activity in skin in tumor-bearing mice. Importantly, the anti-hyperalgesic effects of AEA and URB597 were blocked by a CB1 receptor antagonist. Increased expression of CB1 receptors by DRG neurons ipsilateral to tumor-bearing limbs may contribute to the anti-hyperalgesic effect of elevated AEA levels. Furthermore, CB1 receptor protein-immunoreactivity as well as inhibitory effects of AEA and URB597 on the depolarization-evoked Ca(2+) transient were increased in small DRG neurons cocultured with fibrosarcoma cells indicating that fibrosarcoma cells are sufficient to evoke phenotypic changes in AEA signaling in DRG neurons. Together, the data provide evidence that manipulation of peripheral endocannabinoid signaling is a promising strategy for the management of bone cancer pain.


Assuntos
Ácidos Araquidônicos/fisiologia , Neoplasias Ósseas/metabolismo , Modelos Animais de Doenças , Hiperalgesia/metabolismo , Dor/metabolismo , Pele/metabolismo , Animais , Ácidos Araquidônicos/genética , Neoplasias Ósseas/genética , Canabinoides/genética , Canabinoides/metabolismo , Células Cultivadas , Endocanabinoides , Hiperalgesia/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Dor/genética , Estimulação Física/métodos , Alcamidas Poli-Insaturadas , Receptor CB1 de Canabinoide/genética , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais/fisiologia , Pele/patologia , Tato/fisiologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
19.
J Neurosci ; 27(38): 10289-98, 2007 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-17881535

RESUMO

In an experimental model of cancer pain, the hyperalgesia that occurs with osteolytic tumor growth is associated with the sensitization of nociceptors. We examined functional and molecular changes in small-diameter dorsal root ganglion (DRG) neurons to determine cellular mechanisms underlying this sensitization. The occurrence of a Ca2+ transient in response to either KCl (25 mM) or capsaicin (500 nM) increased in small neurons isolated from murine L3-L6 DRGs ipsilateral to fibrosarcoma cell tumors. The increased responses were associated with increased mRNA levels for the Ca2+ channel subunit alpha2delta1 and TRPV1 receptor. Pretreatment with gabapentin, an inhibitor of the alpha2delta1 subunit, blocked the increased response to KCl in vitro and the mechanical hyperalgesia in tumor-bearing mice in vivo. Similar increases in neuronal responsiveness occurred when DRG neurons from naive mice and fibrosarcoma cells were cocultured for 48 h. The CC chemokine ligand 2 (CCL2) may contribute to the tumor cell-induced sensitization because CCL2 immunoreactivity was present in tumors, high levels of CCL2 peptide were present in microperfusates from tumors, and treatment of DRG neurons in vitro with CCL2 increased the amount of mRNA for the alpha2delta1 subunit. Together, our data provide strong evidence that the chemical mediator CCL2 is released from tumor cells and evokes phenotypic changes in sensory neurons, including increases in voltage-gated Ca2+ channels that likely underlie the mechanical hyperalgesia in the fibrosarcoma cancer model. More broadly, this study provides a novel in vitro model to resolve the cellular and molecular mechanisms by which tumor cells drive functional changes in nociceptors.


Assuntos
Fibrossarcoma/metabolismo , Neurônios Aferentes/metabolismo , Dor/metabolismo , Animais , Técnicas de Cocultura , Fibrossarcoma/química , Fibrossarcoma/patologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Neurônios Aferentes/química , Neurônios Aferentes/patologia , Dor/patologia , Medição da Dor/métodos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...