Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Pain Med ; 15(1): 93-110, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24433468

RESUMO

OBJECTIVE: The pathobiology of prostate cancer (PCa)-induced bone pain (PCIBP) has both inflammatory and neuropathic components. Previously, we showed that small molecule angiotensin II type 2 receptor (AT2 R) antagonists with >1,000-fold selectivity over the angiotensin II type 1 receptor produced dose-dependent analgesia in a rat model of neuropathic pain. Here, we assessed the analgesic efficacy and mode of action of the AT2 R antagonist, EMA200, in a rat model of PCIBP. METHODS: At 14-21 days after unilateral intratibial injection of AT3B PCa cells, rats exhibiting hindpaw hypersensitivity received single intravenous bolus doses of EMA200 (0.3-10 mg/kg) or vehicle, and analgesic efficacy was assessed. The mode of action was investigated using immunohistochemical, Western blot, and/or molecular biological methods in lumbar dorsal root ganglia (DRGs) removed from drug-naïve and EMA200-treated PCIBP rats relative to sham-control rats. RESULTS: Intravenous bolus doses of EMA200 produced dose-dependent analgesia in PCIBP rats. Lumbar DRG levels of angiotensin II, nerve growth factor (NGF), tyrosine kinase A (TrkA), phospho-p38 mitogen-activated protein kinase (MAPK), and phospho-p44/p42 MAPK, but not the AT2 R, were increased significantly (P < 0.05) in PCIBP rats, c.f. the corresponding levels for sham controls. EMA200 produced analgesia in PCIBP rats by reducing elevated angiotensin II levels in the lumbar DRGs to attenuate augmented angiotensin II/AT2 R signaling. This in turn reduced augmented NGF/TrkA signaling in the lumbar DRGs. The net result was inhibition of p38 MAPK and p44/p42 MAPK activation. CONCLUSION: Small molecule AT2 R antagonists are worthy of further investigation as novel analgesics for relief of intractable PCIBP and other pain types where hyperalgesia worsens symptoms.


Assuntos
Analgésicos/uso terapêutico , Bloqueadores do Receptor Tipo 2 de Angiotensina II/uso terapêutico , Neoplasias Ósseas/secundário , Imidazóis/uso terapêutico , Dor/tratamento farmacológico , Neoplasias da Próstata/complicações , Piridinas/uso terapêutico , Receptor Tipo 2 de Angiotensina/efeitos dos fármacos , Analgésicos/farmacologia , Bloqueadores do Receptor Tipo 2 de Angiotensina II/farmacologia , Animais , Neoplasias Ósseas/fisiopatologia , Linhagem Celular Tumoral/transplante , Relação Dose-Resposta a Droga , Gânglios Espinais/química , Gânglios Espinais/patologia , Hiperalgesia/etiologia , Imidazóis/farmacologia , Injeções Intravenosas , Região Lombossacral , Masculino , Proteínas Quinases Ativadas por Mitógeno/análise , Proteínas de Neoplasias/análise , Fator de Crescimento Neural/análise , Neuralgia/tratamento farmacológico , Neuralgia/etiologia , Dor/etiologia , Piridinas/farmacologia , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Receptor Tipo 2 de Angiotensina/biossíntese , Receptor Tipo 2 de Angiotensina/genética , Receptor Tipo 2 de Angiotensina/fisiologia , Células Receptoras Sensoriais/fisiologia
2.
Pain Med ; 14(10): 1557-68, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23742186

RESUMO

OBJECTIVE: There is an unmet clinical need for novel analgesics for neuropathic pain. This study was designed to elucidate the mechanism through which EMA300, a small molecule antagonist of the angiotensin II type 2 receptor (AT2R) with >1,000-fold selectivity over the angiotensin II type 1 receptor, produces analgesia in a rodent model of neuropathic pain. DESIGN AND METHODS: Groups of AT2R knockout, hemizygotes, and wild-type mice with a chronic constriction injury (CCI) of the sciatic nerve received single intraperitoneal (i.p.) bolus doses of EMA300 (100 or 300 mg/kg), and analgesic efficacy was assessed. Groups of control, sham-operated, and CCI rats were euthanized and perfusion fixed. Lumbar dorsal root ganglia (DRGs) were removed for investigation of the mechanism through which EMA300 alleviates neuropathic pain. RESULTS: EMA300 analgesia was abolished in AT2R knockout CCI mice with intermediate responses in the hemizygotes, affirming the AT2R as the target mediating EMA300 analgesia. In CCI rats, DRG immunofluorescence (IF) levels for angiotensin II, the main endogenous ligand of the AT2R, were increased ∼1.5-2.0-fold (P < 0.05) cf. sham-controls. Mean DRG IF levels for activated p38 (pp38) and activated p44/p42 (pp44/pp42) MAPK were also increased ∼1.5-2.0-fold (P < 0.05) cf. sham-controls. At the time of peak EMA300 analgesia in CCI rats, mean DRG levels of pp38 MAPK and pp44/pp42 MAPK (but not angiotensin II) were reduced to match the respective levels in sham-controls. CONCLUSION: Augmented angiotensin II/AT2R signaling in the DRGs of CCI rats is attenuated by EMA300 to block p38 MAPK and p44/p42 MAPK activation, a mechanism with clinical validity for alleviating neuropathic pain.


Assuntos
Analgésicos/farmacologia , Bloqueadores do Receptor Tipo 2 de Angiotensina II/farmacologia , Gânglios Espinais/efeitos dos fármacos , Imidazóis/farmacologia , Neuralgia/metabolismo , Piridinas/farmacologia , Animais , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Gânglios Espinais/enzimologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Neuralgia/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Inflammopharmacology ; 21(5): 339-63, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23918298

RESUMO

Prostate cancer (PCa) has a high propensity for metastasis to bone. Despite the availability of multiple treatment options for relief of PCa-induced bone pain (PCIBP), satisfactory relief of intractable pain in patients with advanced bony metastases is challenging for the clinicians because currently available analgesic drugs are often limited by poor efficacy and/or dose-limiting side effects. Rodent models developed in the past decade show that the pathobiology of PCIBP comprises elements of inflammatory, neuropathic and ischemic pain arising from ectopic sprouting and sensitization of sensory nerve fibres within PCa-invaded bones. In addition, at the cellular level, PCIBP is underpinned by dynamic cross talk between metastatic PCa cells, cellular components of the bone matrix, factors associated with the bone microenvironment as well as peripheral components of the somatosensory system. These insights are aligned with the clinical management of PCIBP involving use of a multimodal treatment approach comprising analgesic agents (opioids, NSAIDs), radiotherapy, radioisotopes, cancer chemotherapy agents and bisphosphonates. However, a major drawback of most rodent models of PCIBP is their short-term applicability due to ethical concerns. Thus, it has been difficult to gain insight into the mal(adaptive) neuroplastic changes occurring at multiple levels of the somatosensory system that likely contribute to intractable pain at the advanced stages of metastatic disease. Specifically, the functional responsiveness of noxious circuitry as well as the neurochemical signature of a broad array of pro-hyperalgesic mediators in the dorsal root ganglia and spinal cord of rodent models of PCIBP is relatively poorly characterized. Hence, recent work from our laboratory to develop a protocol for an optimized rat model of PCIBP will enable these knowledge gaps to be addressed as well as identification of novel targets for drug discovery programs aimed at producing new analgesics for the improved relief of intractable PCIBP.


Assuntos
Neoplasias Ósseas/secundário , Osso e Ossos/patologia , Dor Nociceptiva/tratamento farmacológico , Manejo da Dor/métodos , Neoplasias da Próstata/patologia , Adjuvantes Farmacêuticos/administração & dosagem , Adjuvantes Farmacêuticos/efeitos adversos , Adjuvantes Farmacêuticos/uso terapêutico , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/uso terapêutico , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/efeitos da radiação , Terapia Combinada , Modelos Animais de Doenças , Quimioterapia Combinada , Humanos , Masculino , Dor Nociceptiva/etiologia , Dor Nociceptiva/radioterapia , Manejo da Dor/tendências
4.
J Clin Invest ; 132(8)2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35426375

RESUMO

Mice with experimental nerve damage can display long­lasting neuropathic pain behavior. We show here that 4 months and later after nerve injury, male but not female mice displayed telomere length (TL) reduction and p53­mediated cellular senescence in the spinal cord, resulting in maintenance of pain and associated with decreased lifespan. Nerve injury increased the number of p53­positive spinal cord neurons, astrocytes, and microglia, but only in microglia was the increase male­specific, matching a robust sex specificity of TL reduction in this cell type, which has been previously implicated in male­specific pain processing. Pain hypersensitivity was reversed by repeated intrathecal administration of a p53­specific senolytic peptide, only in male mice and only many months after injury. Analysis of UK Biobank data revealed sex-specific relevance of this pathway in humans, featuring male­specific genetic association of the human p53 locus (TP53) with chronic pain and a male-specific effect of chronic pain on mortality. Our findings demonstrate the existence of a biological mechanism maintaining pain behavior, at least in males, occurring much later than the time span of virtually all extant preclinical studies.


Assuntos
Dor Crônica , Neuralgia , Animais , Senescência Celular , Dor Crônica/genética , Dor Crônica/metabolismo , Feminino , Hiperalgesia/metabolismo , Masculino , Camundongos , Microglia/metabolismo , Neuralgia/genética , Neuralgia/metabolismo , Medula Espinal/metabolismo , Telômero/genética , Telômero/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
5.
Br J Pharmacol ; 178(13): 2709-2726, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33782947

RESUMO

BACKGROUND AND PURPOSE: The µ-opioid receptor (µ receptor) is the primary target for opioid analgesics. The 7-transmembrane (TM) and 6TM µ receptor isoforms mediate inhibitory and excitatory cellular effects. Here, we developed compounds selective for 6TM- or 7TM-µ receptors to further our understanding of the pharmacodynamic properties of µ receptors. EXPERIMENTAL APPROACH: We performed virtual screening of the ZINC Drug Now library of compounds using in silico 7TM- and 6TM-µ receptor structural models and identified potential compounds that are selective for 6TM- and/or 7TM-µ receptors. Subsequently, we characterized the most promising candidate compounds in functional in vitro studies using Be2C neuroblastoma transfected cells, behavioural in vivo pain assays using various knockout mice and in ex vivo electrophysiology studies. KEY RESULTS: Our virtual screen identified 30 potential candidate compounds. Subsequent functional in vitro cellular assays shortlisted four compounds (#5, 10, 11 and 25) that demonstrated 6TM- or 7TM-µ receptor-dependent NO release. In in vivo pain assays these compounds also produced dose-dependent hyperalgesic responses. Studies using mice that lack specific opioid receptors further established the µ receptor-dependent nature of identified novel ligands. Ex vivo electrophysiological studies on spontaneous excitatory postsynaptic currents in isolated spinal cord slices also validated the hyperalgesic properties of the most potent 6TM- (#10) and 7TM-µ receptor (#5) ligands. CONCLUSION AND IMPLICATIONS: Our novel compounds represent a new class of ligands for µ receptors and will serve as valuable research tools to facilitate the development of opioids with significant analgesic efficacy and fewer side-effects.


Assuntos
Analgésicos Opioides , Receptores Opioides mu , Analgésicos Opioides/farmacologia , Animais , Camundongos , Camundongos Knockout , Dor , Isoformas de Proteínas
6.
Pain Rep ; 5(3): e824, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32903926

RESUMO

INTRODUCTION: Increasing attention is being paid to the effects of organismic factors like age on pain sensitivity. However, very little data exist on this topic using modern algesiometric assays and measures in laboratory rodents. OBJECTIVES: We investigated the effect of age and duration of nerve injury on baseline mechanical thresholds, neuropathic allodynia, and the antiallodynic and analgesic efficacy of 4 systemically administered analgesics: amitriptyline, diclofenac, morphine, and pregabalin. METHODS: Mice of both sexes and 3 conditions were compared: Young-Young, in which baseline testing (von Frey thresholds), the injury producing neuropathic pain (spared nerve injury [SNI]) and subsequent drug testing occurred while mice were young (8-10 weeks); Young-Old, in which mice received the nerve injury while young but were tested for drug efficacy over 10 months later; and Old-Old, in which both the nerve injury and drug testing occurred at approximately 1 year of age. RESULTS: Old-Old mice were found to display higher baseline mechanical sensitivity than other groups. No group differences were seen in SNI-induced allodynia in males; female Young-Old mice were found to display greatly reduced allodynia. With respect to drug efficacy, no differences among conditions were observed for amitriptyline, diclofenac, or morphine. For pregabalin, however, Young-Old mice displayed significantly reduced antiallodynia, and the drug was completely ineffective in Old-Old mice. CONCLUSION: Novel findings include the apparent remission of SNI-induced allodynia in female mice 10 months after injury and reduced pregabalin antiallodynic effects produced by both the passage of time after nerve injury and aging.

7.
Philos Trans R Soc Lond B Biol Sci ; 374(1785): 20190287, 2019 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-31544607

RESUMO

Nerve injury leads to devastating and often untreatable neuropathic pain. While acute noxious sensation (nociception) is a crucial survival mechanism and is conserved across phyla, chronic neuropathic pain is considered a maladaptive response owing to its devastating impact on a patient's quality of life. We have recently shown that a neuropathic pain-like response occurs in adult Drosophila. However, the mechanisms underlying this phenomenon are largely unknown. Previous studies have shown that the α2δ peripheral calcium channel subunit straightjacket (stj) is a conserved factor required for thermal pain perception. We demonstrate here that stj is required in peripheral ppk+ sensory neurons for acute thermal responses and that it mediates nociceptive hypersensitivity in an adult Drosophila model of neuropathic pain-like disease. Given that calcium channels are the main targets of gabapentinoids (pregabalin and gabapentin), we assessed if these drugs can alleviate nociceptive hypersensitivity. Our findings suggest that gabapentinoids may prevent nociceptive hypersensitivity by preserving central inhibition after nerve injury. Together, our data further highlight the similarity of some mechanisms for pain-like conditions across phyla and validates the scientific use of Drosophila neuropathic sensitization models for analgesic drug discovery. This article is part of the Theo Murphy meeting issue 'Evolution of mechanisms and behaviour important for pain'.


Assuntos
Canais de Cálcio/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiologia , Neuralgia/genética , Animais , Canais de Cálcio/metabolismo , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Expressão Gênica/fisiologia , Larva/genética , Larva/fisiologia , Neuralgia/fisiopatologia
10.
Front Mol Neurosci ; 10: 389, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29200998

RESUMO

Recent preclinical and proof-of-concept clinical studies have shown promising analgesic efficacy of selective small molecule angiotensin II type 2 (AT2) receptor antagonists in the alleviation of peripheral neuropathic pain. However, their cellular and molecular mechanism of action requires further investigation. To address this issue, groups of adult male Sprague-Dawley rats with fully developed unilateral hindpaw hypersensitivity, following chronic constriction injury (CCI) of the sciatic nerve, received a single intraperitoneal bolus dose of the small molecule AT2 receptor antagonist, EMA300 (10 mg kg-1), or vehicle. At the time of peak EMA300-mediated analgesia (∼1 h post-dosing), groups of CCI-rats administered either EMA300 or vehicle were euthanized. A separate group of rats that underwent sham surgery were also included. The lumbar (L4-L6) dorsal root ganglia (DRGs) were obtained from all experimental cohorts and processed for immunohistochemistry and western blot studies. In vehicle treated CCI-rats, there was a significant increase in the expression levels of angiotensin II (Ang II), but not the AT2 receptor, in the ipsilateral lumbar DRGs. The elevated levels of Ang II in the ipsilateral lumbar DRGs of CCI-rats were at least in part contributed by CD3+ T-cells, satellite glial cells (SGCs) and subsets of neurons. Our findings suggest that the analgesic effect of EMA300 in CCI-rats involves multimodal actions that appear to be mediated at least in part by a significant reduction in the otherwise increased expression levels of Ang II as well as the number of Ang II-expressing CD3+ T-cells in the ipsilateral lumbar DRGs of CCI-rats. Additionally, the acute anti-allodynic effects of EMA300 in CCI-rats were accompanied by rescue of the otherwise decreased expression of mature nerve growth factor (NGF) in the ipsilateral lumbar DRGs of CCI-rats. In contrast, the increased expression levels of TrkA and glial fibrillary acidic protein in the ipsilateral lumbar DRGs of vehicle-treated CCI-rats were not attenuated by a single bolus dose of EMA300. Consistent with our previous findings, there was also a significant decrease in the augmented levels of the downstream mediators of Ang II/AT2 receptor signaling, i.e., phosphorylated-p38 mitogen-activated protein kinase (MAPK) and phosphorylated-p44/p42 MAPK, in the ipsilateral lumbar DRGs.

11.
Front Pharmacol ; 8: 493, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28798688

RESUMO

Chronic low back pain (LBP), the leading cause of disability globally, is notoriously difficult to treat. Most rodent models of LBP mimic lumbar radicular pain rather than mechanical LBP. Here, we describe establishment of a new rat model of mechanical LBP that is devoid of a neuropathic component. Groups of adult male Sprague Dawley rats were anesthetized and their lumbar L4/L5 and L5/L6 intervertebral disks (IVDs) were punctured (0.5 mm outer diameter, 2mm-deep) 5 (LPB-5X), or 10 (LBP-10X) times per disk. Sham-rats underwent similar surgery, but without disk puncture. Baseline noxious pressure hyperalgesia of lumbar axial deep tissues, mechanical allodynia in the hindpaws and gait were assessed prior to surgery and once-weekly until study completion on day 49. The model was also characterized using pharmacologic and histologic methods. Good animal health was maintained for ≥ 49 days post-surgery. For LBP- but not sham-rats, there was temporal development of noxious pressure hyperalgesia in lumbar axial deep tissues at days 14-49 post-surgery. Importantly, there were no between-group differences in von Frey paw withdrawal thresholds or gait parameters until study completion. On day 49, significant histologic changes were observed in the L4/L5 and L5/L6 IVDs for LBP-10X rats, but not sham-rats. In LBP-10X rats, single bolus doses of morphine produced dose-dependent relief of primary and secondary mechanical hyperalgesia in lumbar axial deep tissues at L4/L5 and L1, respectively. In conclusion, our new rat model has considerable potential for providing novel insight on the pathobiology of mechanical LBP and for analgesic efficacy assessment of novel compounds.

12.
Front Behav Neurosci ; 10: 88, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27242458

RESUMO

Establishment and validation of ethologically-relevant, non-evoked behavioral end-points as surrogate measures of spontaneous pain in rodent pain models has been proposed as a means to improve preclinical to clinical research translation in the pain field. Here, we compared the utility of burrowing behavior with hypersensitivity to applied mechanical stimuli for pain assessment in rat models of chronic inflammatory and peripheral neuropathic pain. Briefly, groups of male Sprague-Dawley rats were habituated to the burrowing environment and trained over a 5-day period. Rats that burrowed ≤ 450 g of gravel on any 2 days of the individual training phase were excluded from the study. The remaining rats received either a unilateral intraplantar injection of Freund's complete adjuvant (FCA) or saline, or underwent unilateral chronic constriction injury (CCI) of the sciatic nerve- or sham-surgery. Baseline burrowing behavior and evoked pain behaviors were assessed prior to model induction, and twice-weekly until study completion on day 14. For FCA- and CCI-rats, but not the corresponding groups of sham-rats, evoked mechanical hypersensitivity developed in a temporal manner in the ipsilateral hindpaws. Although burrowing behavior also decreased in a temporal manner for both FCA-and CCI- rats, there was considerable inter-animal variability. By contrast, mechanical hyperalgesia and mechanical allodynia in the ipsilateral hindpaws of FCA- and CCI-rats respectively, exhibited minimal inter-animal variability. Our data collectively show that burrowing behavior is altered in rodent models of chronic inflammatory pain and peripheral neuropathic pain. However, large group sizes are needed to ensure studies are adequately powered due to considerable inter-animal variability.

13.
Expert Opin Ther Targets ; 19(1): 25-35, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25315162

RESUMO

INTRODUCTION: Neuropathic pain and chronic inflammatory pain are large unmet medical needs. Over the past two decades, numerous 'pain targets' have been identified for analgesic drug discovery. Despite promising results in rodent pain models, many compounds modulating such targets lacked efficacy in clinical trials. An exception is oral EMA401, a small-molecule angiotensin II type 2 receptor (AT2R) antagonist. AREAS COVERED: Herein, angiotensin II/AT2R signaling-induced hyperexcitability and abnormal sprouting of cultured dorsal root ganglion neurons, together with radioligand binding, pharmacokinetics, analgesic efficacy and mode of action of small-molecule AT2R antagonists in rodent models of peripheral neuropathic and chronic inflammatory pain, are reviewed. The findings of a successful Phase IIa clinical trial of EMA401 in patients with neuropathic pain are presented in brief. EXPERT OPINION: The functional importance of angiotensin II/AT2R signaling has remained enigmatic for decades, and there are no clinically available medications that target the AT2R. However, on the basis of preclinical findings and recent clinical trial data showing that the peripherally restricted, small-molecule AT2R antagonist, EMA401, successfully alleviated neuropathic pain in a Phase II clinical trial, the AT2R is receiving considerable attention as a new therapeutic target with human validation for the relief of peripheral neuropathic and chronic inflammatory pain conditions.


Assuntos
Analgésicos/farmacologia , Bloqueadores do Receptor Tipo 2 de Angiotensina II/farmacologia , Inflamação/metabolismo , Dor/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Analgésicos/uso terapêutico , Bloqueadores do Receptor Tipo 2 de Angiotensina II/uso terapêutico , Animais , Humanos , Inflamação/tratamento farmacológico , Dor/tratamento farmacológico , Sistema Renina-Angiotensina
14.
Pharmacol Biochem Behav ; 106: 33-46, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23500189

RESUMO

The major limitation of currently utilized rodent models of prostate cancer (PCa)-induced bone pain (PCIBP) involving intra-osseous injection of PCa cells, is their relatively short-term applicability due to progressive deterioration of animal health necessitating euthanasia. Here, we describe establishment of an optimized rat model of PCIBP where good animal health was maintained for at least 90-days following unilateral intra-tibial injection (ITI) of PCa cells. We have characterized this model using behavioral, pharmacological, radiological, histological and immunohistochemical methods. Our findings show that following unilateral ITI of 4×10(4) AT3B PCa cells (APCCs), there was temporal development of bilateral hindpaw hypersensitivity that was fully developed between days 14 and 21 post-ITI. Although there was apparent spontaneous reversal of bilateral hindpaw sensitivity that was maintained until at least day 90 post-ITI, administration of bolus doses of the opioid receptor antagonist, naloxone, rescued the pain phenotype in these animals. Hence, upregulation of endogenous opioid signaling mechanisms appears to underpin apparent spontaneous resolution of hindpaw hypersensitivity. Importantly, the histological and radiological assessments confirmed that tumor formation and development of osteosclerotic metastases was confined to the APCC-injected tibial bones. In our rat model of PCIBP, single bolus doses of morphine, gabapentin, meloxicam and amitriptyline produced dose-dependent relief of mechanical allodynia and thermal hyperalgesia in the bilateral hindpaws. The optimized rat model of PCIBP characterized herein has potential to provide new insights into the pathophysiological mechanisms associated with long-term (mal)adaptive pain due to advanced PCa-induced bony metastases and for screening novel compounds with potential for improved alleviation of this condition.


Assuntos
Comportamento Animal , Neoplasias Ósseas/secundário , Modelos Animais de Doenças , Dor/etiologia , Neoplasias da Próstata/complicações , Animais , Neoplasias Ósseas/complicações , Neoplasias Ósseas/diagnóstico por imagem , Linhagem Celular Tumoral , Imuno-Histoquímica , Masculino , Dor/fisiopatologia , Neoplasias da Próstata/patologia , Ratos , Ratos Wistar , Tomografia Computadorizada por Raios X
15.
J Pharm Pharmacol ; 63(11): 1387-400, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21988420

RESUMO

OBJECTIVES: In the clinical setting, there is marked intersubject variability in the intensity of pain reported by patients with apparently similar pain states, as well as widely differing analgesic dosing requirements between individuals to produce satisfactory pain relief with tolerable side-effects. Genetic and environmental factors as well as their interaction are implicated, and these are discussed in this review. KEY FINDINGS: Pioneering work undertaken in mice more than a decade ago, showed a strong genetic contribution to levels of nociception/hypersensitivity as well as levels of antinociception produced by commonly available analgesic agents. To date more than 300 candidate 'pain' genes have been identified as potentially contributing to heritable differences in pain sensitivity and analgesic responsiveness in animals and humans, with this information available in a publicly accessible database http://www.jbldesign.com/jmogil/enter.html. Since then, many genetic association studies have been conducted in humans to investigate the possibility that single nucleotide polymorphisms (SNPs) in an individual gene may explain drug inefficacy or excessive toxicity experienced by a small subset of the whole population who have the rare allele for a particular SNP. SUMMARY: Despite the fact that SNPs in more than 20 genes that affect pain sensitivity or contribute to interindividual variability in responses to analgesic medications have been identified in the human genome, much of the data is conflicting. Apart from deficiencies in the design and conduct of human genetic association studies, recent research from other fields has implicated epigenetic mechanisms that facilitate dynamic gene-environment communication, as a possible explanation.


Assuntos
Analgésicos/uso terapêutico , Relação Dose-Resposta a Droga , Dor/genética , Analgesia/métodos , Animais , Genoma Humano , Humanos , Dor/tratamento farmacológico , Manejo da Dor , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Farmacogenética , Polimorfismo de Nucleotídeo Único/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa