Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Neuropharmacology ; 160: 107690, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31271770

ABSTRACT

Pain is among the most common symptoms in cancer and approximately 90% of patients experience end-stage cancer pain. The management of cancer pain is challenging due to the significant side effects associated with opioids, and novel therapeutic approaches are needed. MMG22 is a bivalent ligand containing MOR agonist and mGluR5 antagonist pharmacophores joined by a 22-atom spacer. MMG22 exhibited extraordinary analgesia following intrathecal administration in a mouse model of bone cancer pain. Here, we assessed the effectiveness of systemic administration of MMG22 in reducing cancer pain and evaluated whether MMG22 displays side effects associated with opioids. Fibrosarcoma cells were injected into and around the calcaneus bone in C3H mice. Mechanical hyperalgesia was defined as an increase in the paw withdrawal frequencies (PWFs) evoked by application of a von Frey monofilament (3.9 mN bending force) applied to the plantar surface of the hind paw Subcutaneous (s.c.), intramuscular (i.m.), and oral (p.o.) administration of MMG22 produced robust dose-dependent antihyperalgesia, whose ED50 was orders of magnitude lower than morphine. Moreover, the ED50 for MMG22 decreased with disease progression. Importantly, s.c. administration of MMG22 did not produce acute (24 h) or long-term (9 days) tolerance, was not rewarding (conditioned place preference test), and did not produce naloxone-induced precipitated withdrawal or alter motor function. A possible mechanism of action of MMG22 is discussed in terms of inhibition of spinal NMDAR via antagonism of its co-receptor, mGluR5, and concomitant activation of neuronal MOR. We suggest that MMG22 may be a powerful alternative to traditional opioids for managing cancer pain. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.


Subject(s)
Cancer Pain/drug therapy , Cancer Pain/metabolism , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Disease Models, Animal , Drug Administration Routes , Fibrosarcoma/drug therapy , Fibrosarcoma/metabolism , Hyperalgesia/drug therapy , Ligands , Male , Mice , Mice, Inbred C3H , Morphine/therapeutic use , Receptors, Kainic Acid/administration & dosage , Receptors, Opioid, mu/administration & dosage
2.
J Innov Health Inform ; 24(2): 900, 2017 Jun 23.
Article in English | MEDLINE | ID: mdl-28749314

ABSTRACT

BACKGROUND: Changes in health insurance policies have increased coverage opportunities, but enrollees are required to annually reapply for benefits which, if not managed appropriately, can lead to insurance gaps. Electronic health records (EHRs) can automate processes for assisting patients with health insurance enrollment and re-enrollment. OBJECTIVE: We describe community health centers' (CHC) workflow, documentation, and tracking needs for assisting families with insurance application processes, and the health information technology (IT) tool components that were developed to meet those needs. METHOD: We conducted a qualitative study using semi-structured interviews and observation of clinic operations and insurance application assistance processes. Data were analyzed using a grounded theory approach. We diagramed workflows and shared information with a team of developers who built the EHR-based tools. RESULTS: Four steps to the insurance assistance workflow were common among CHCs: 1) Identifying patients for public health insurance application assistance; 2) Completing and submitting the public health insurance application when clinic staff met with patients to collect requisite information and helped them apply for benefits; 3) Tracking public health insurance approval to monitor for decisions; and 4) assisting with annual health insurance reapplication. We developed EHR-based tools to support clinical staff with each of these steps. CONCLUSION: CHCs are uniquely positioned to help patients and families with public health insurance applications. CHCs have invested in staff to assist patients with insurance applications and help prevent coverage gaps. To best assist patients and to foster efficiency, EHR based insurance tools need comprehensive, timely, and accurate health insurance information.


Subject(s)
Electronic Health Records , Insurance Coverage/organization & administration , Insurance, Health/organization & administration , Medical Informatics/organization & administration , Community Health Centers/organization & administration , Grounded Theory , Health Policy , Humans , Interviews as Topic , Medicaid , Qualitative Research , United States
3.
Acad Med ; 91(9): 1293-304, 2016 09.
Article in English | MEDLINE | ID: mdl-27028034

ABSTRACT

PURPOSE: To report findings from a national effort initiated by three primary care certifying boards to catalyze change in primary care training. METHOD: In this mixed-method pilot study (2012-2014), 36 faculty in 12 primary care residencies (family medicine, internal medicine, pediatrics) from four institutions participated in a professional development program designed to prepare faculty to accelerate change in primary care residency training by uniting them in a common mission to create effective ambulatory clinical learning environments. Surveys administered at baseline and 12 months after initial training measured changes in faculty members' confidence and skills, continuity clinics, and residency training programs. Feasibility evaluation involved assessing participation. The authors compared quantitative data using Wilcoxon signed-rank and Bhapkar tests. Observational field notes underwent narrative analysis. RESULTS: Most participants attended two in-person training sessions (92% and 72%, respectively). Between baseline and 12 months, faculty members' confidence in leadership improved significantly for 15/19 (79%) variables assessed; their self-assessed skills improved significantly for 21/22 (95%) competencies. Two medical home domains ("Continuity of Care," "Support/Care Coordination") improved significantly (P < .05) between the two time periods. Analyses of qualitative data revealed that interdisciplinary learning communities formed during the program and served to catalyze transformational change. CONCLUSIONS: Results suggest that improvements in faculty perceptions of confidence and skills occurred and that the creation of interdisciplinary learning communities catalyzed transformation. Lengthening the intervention period, engaging other professions involved in training the primary care workforce, and a more discriminating evaluation design are needed to scale this model nationally.


Subject(s)
Curriculum , Education, Medical, Graduate/trends , Family Practice/education , Internal Medicine/education , Interprofessional Relations , Pediatrics/education , Primary Health Care/trends , Adult , Female , Forecasting , Humans , Male , Middle Aged , Pilot Projects , Program Evaluation , United States
4.
Implement Sci ; 10: 123, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26652866

ABSTRACT

BACKGROUND: Patients with gaps in health insurance coverage often defer or forgo cancer prevention services. These delays in cancer detection and diagnoses lead to higher rates of morbidity and mortality and increased costs. Recent advances in health information technology (HIT) create new opportunities to enhance insurance support services that reduce coverage gaps through automated processes applied in healthcare settings. This study will assess the implementation of insurance support HIT tools and their effectiveness at improving patients' insurance coverage continuity and cancer screening rates. METHODS/DESIGN: This study uses a hybrid cluster-randomized design-a combined effectiveness and implementation trial-in community health centers (CHCs) in the USA. Eligible CHC clinic sites will be randomly assigned to one of two groups in the trial's implementation component: tools + basic training (Arm I) and tools + enhanced training + facilitation (Arm II). A propensity score-matched control group of clinics will be selected to assess the tools' effectiveness. Quantitative analyses of the tools' impact will use electronic health record and Medicaid data to assess effectiveness. Qualitative data will be collected to evaluate the implementation process, understand how the HIT tools are being used, and identify facilitators and barriers to their implementation and use. DISCUSSION: This study will test the effectiveness of HIT tools to enhance insurance support in CHCs and will compare strategies for facilitating their implementation in "real-world" practice settings. Findings will inform further development and, if indicated, more widespread implementation of insurance support HIT tools. TRIAL REGISTRATION: Clinical trial NTC02355262.


Subject(s)
Community Health Centers/organization & administration , Insurance Coverage/organization & administration , Insurance, Health/organization & administration , Medical Informatics/organization & administration , Primary Health Care/organization & administration , Early Detection of Cancer/economics , Electronic Health Records/statistics & numerical data , Humans , Medicaid/statistics & numerical data , Primary Health Care/economics , Propensity Score , Research Design , United States
5.
J Comp Eff Res ; 4(4): 351-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26274796

ABSTRACT

The Patient-Centered Outcomes Research Institute has accelerated conversations about the importance of actively engaging stakeholders in all aspects of comparative effectiveness research (CER). Other scientific disciplines have a history of stakeholder engagement, yet few empirical examples exist of how these stakeholders can inform and enrich CER. Here we present a case study which includes the methods used to engage stakeholders, what we learned from them, and how we incorporated their ideas in a CER project. We selected stakeholders from key groups, built relationships with them and collected their feedback through interviews, observation and ongoing meetings during the four research process phases: proposal development, adapting study methods, understanding the context and information technology tool design and refinement.


Subject(s)
Community Health Centers , Community Participation/methods , Comparative Effectiveness Research/methods , Patient Outcome Assessment , Patient-Centered Care , Program Evaluation , Academies and Institutes , Community Participation/trends , Comparative Effectiveness Research/trends , Humans , Interviews as Topic , Oregon , Research Design
6.
Neuroscience ; 247: 84-94, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23673278

ABSTRACT

Pain from cancer can be severe, difficult to treat, and greatly diminishes patients' quality of life. It is therefore important to gain new information on the mechanisms of cancer pain and develop new treatment strategies. We have used a murine model of bone cancer pain to investigate underlying peripheral neural mechanisms and novel treatment approaches. In this model, implantation of fibrosarcoma cells into and around the calcaneous bone produces mechanical and thermal hyperalgesia in mice. C-fiber nociceptors in tumor-bearing mice develop spontaneous ongoing activity and sensitization to thermal stimuli. However, it is unclear whether sensitization of nociceptors to mechanical stimuli underlies the mechanical hyperalgesia seen in tumor-bearing mice. We therefore examined responses of C-fiber nociceptors to suprathreshold mechanical stimuli in tumor-bearing mice and found they did not differ from those of C-nociceptors in control mice. Thus, sensitization of C-fiber nociceptors to mechanical stimulation does not appear to underlie tumor-evoked mechanical hyperalgesia in this murine model of bone cancer pain. We also examined the effect of the non-selective cannabinoid receptor agonist, WIN 55,212-2, on spontaneous activity and responses evoked by mechanical stimuli of C-fiber nociceptors innervating the tumor-bearing paw. Selective CB1 and CB2 antagonists were administered to determine the contribution of each receptor subtype to the effects of WIN 55,212-2. Intraplantar administration of WIN 55,212-2 attenuated spontaneous discharge and responses evoked by mechanical stimulation of C-fiber nociceptors. These effects were inhibited by prior intraplantar administration of selective CB1 (AM281) or CB2 (AM630) receptor antagonists but not by vehicle. These results indicate that activation of either CB1 or CB2 receptors reduced the spontaneous activity of C-fiber nociceptors associated with tumor growth as well as their evoked responses. Our results provide further evidence that activation of peripheral cannabinoid receptors may be a useful target for the treatment of cancer pain.


Subject(s)
Benzoxazines/therapeutic use , Cannabinoid Receptor Agonists/therapeutic use , Disease Models, Animal , Morpholines/therapeutic use , Naphthalenes/therapeutic use , Neoplasms/drug therapy , Nerve Fibers, Unmyelinated/drug effects , Pain/drug therapy , Animals , Benzoxazines/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Male , Mice , Mice, Inbred C3H , Morpholines/pharmacology , Naphthalenes/pharmacology , Neoplasms/pathology , Nerve Fibers, Unmyelinated/physiology , Nociceptors/drug effects , Nociceptors/physiology , Pain/pathology
7.
Neurobiol Dis ; 58: 19-28, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23644187

ABSTRACT

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.


Subject(s)
Arachidonic Acids/metabolism , Endocannabinoids/metabolism , Hyperalgesia/etiology , Pain Threshold/physiology , Pain/complications , Pain/pathology , Polyunsaturated Alkamides/metabolism , Sensory Receptor Cells/metabolism , Animals , Benzamides/pharmacology , Brain Neoplasms/complications , Brain Neoplasms/pathology , Cannabinoid Receptor Antagonists/pharmacology , Carbamates/pharmacology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Fibrosarcoma/complications , Fibrosarcoma/pathology , Fluorescent Dyes , Ganglia, Spinal/cytology , Indoles/pharmacology , Lactones , Male , Mice , Mice, Inbred C3H , Pain/etiology , Pain Threshold/drug effects , Sensory Receptor Cells/drug effects , Statistics, Nonparametric , Tritium/metabolism
8.
J Neurosci ; 32(20): 7091-101, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22593077

ABSTRACT

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.


Subject(s)
Arachidonic Acids/therapeutic use , Benzamides/therapeutic use , Carbamates/therapeutic use , Cisplatin/antagonists & inhibitors , Hyperalgesia/drug therapy , Neurotoxicity Syndromes/drug therapy , Peripheral Nervous System Diseases/drug therapy , Polyunsaturated Alkamides/therapeutic use , Receptor, Cannabinoid, CB1/agonists , Activating Transcription Factor 3/metabolism , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/antagonists & inhibitors , Arachidonic Acids/pharmacokinetics , Arachidonic Acids/pharmacology , Benzamides/antagonists & inhibitors , Benzamides/pharmacology , Cannabinoid Receptor Modulators/pharmacokinetics , Cannabinoid Receptor Modulators/pharmacology , Cannabinoid Receptor Modulators/therapeutic use , Carbamates/antagonists & inhibitors , Carbamates/pharmacology , Cells, Cultured , Cisplatin/adverse effects , Disease Models, Animal , Drug Interactions , Endocannabinoids , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Hyperalgesia/chemically induced , Male , Mice , Mice, Inbred C3H , Morpholines/pharmacology , Motor Activity/drug effects , Neurites/drug effects , Peripheral Nervous System Diseases/chemically induced , Polyunsaturated Alkamides/pharmacokinetics , Polyunsaturated Alkamides/pharmacology , Pyrazoles/pharmacology , TRPV Cation Channels/metabolism
9.
J Endod ; 37(8): 1098-101, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21763901

ABSTRACT

INTRODUCTION: Studies to examine sex differences in response to pain have suggested that females exhibit lower threshold responses to painful stimuli and that threshold response varies greatly at different stages of the menstrual cycle. Additional studies suggest that sex differences may be caused by societal sex roles or differences in anxiety responses by men and women. OBJECTIVE: The purpose of this study was to evaluate biologically evident sex differences in male and female rats chronically treated with a systemic algogen, the nerve growth factor (NGF), by measuring neuropeptides (calcitonin gene-related peptide) content and release from isolated dental pulp. METHODS: Rats were injected subcutaneously every other day with either murine NGF (1 mg/kg) or vehicle for 7 or 13 days. Isolated incisor pulp tissue was evaluated from these male and female rats (n = 96). Capsaicin-evoked neurosecretion of CGRP and tissue content were measured using a previously validated radioimmunoassay. RESULTS: Dental pulp from female rats at 7 days showed significantly increased capsaicin-evoked immunoreactive CGRP release (>50% increase) compared with tissue from male rats. After 13 days, this release was significantly increased only in NGF-treated female rats (3-fold increase) when compared with control females or both male groups. The CGRP content in tissue from both female groups was also significantly increased after 7 days of treatment (>3 fold), but after 13 days this content was only significantly increased in tissue from NGF-treated female rats (P = .0001). CONCLUSIONS: These data suggest that sex differences affect the role of NGF in the modulation of inflammation through the regulation of peripheral neuropeptide release and content.


Subject(s)
Calcitonin Gene-Related Peptide/biosynthesis , Dental Pulp/metabolism , Hyperalgesia/metabolism , Neurogenic Inflammation/metabolism , Sex Characteristics , Animals , Capsaicin/adverse effects , Dental Pulp/drug effects , Female , Male , Mice , Nerve Growth Factor/adverse effects , Proestrus/metabolism , Rats
10.
Behav Pharmacol ; 22(5-6): 607-16, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21610490

ABSTRACT

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.


Subject(s)
Neoplasms, Experimental/complications , Pain/drug therapy , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Analgesics/pharmacology , Animals , Arachidonic Acids/pharmacology , Cannabinoids/pharmacology , Disease Models, Animal , Drug Synergism , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Male , Mice , Mice, Inbred C3H , Morphine/pharmacology , Pain/etiology
11.
Pharmacol Res ; 64(1): 60-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21440630

ABSTRACT

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.


Subject(s)
Arachidonic Acids/therapeutic use , Bone Neoplasms/complications , Glycerides/therapeutic use , Hyperalgesia/drug therapy , Signal Transduction/drug effects , Animals , Arachidonic Acids/administration & dosage , Arachidonic Acids/metabolism , Benzodioxoles/administration & dosage , Benzodioxoles/pharmacology , Bone Neoplasms/metabolism , Calcaneus/pathology , Cannabinoid Receptor Antagonists , Dose-Response Relationship, Drug , Endocannabinoids , Fibrosarcoma/complications , Fibrosarcoma/metabolism , Ganglia, Spinal/metabolism , Glycerides/administration & dosage , Glycerides/metabolism , Hyperalgesia/etiology , Male , Mice , Mice, Inbred C3H , Monoacylglycerol Lipases/antagonists & inhibitors , Piperidines/administration & dosage , Piperidines/pharmacology , Polyunsaturated Alkamides/metabolism , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/metabolism , Skin/drug effects , Skin/metabolism , Tibial Nerve/metabolism
12.
J Neurosci ; 28(44): 11141-52, 2008 Oct 29.
Article in English | MEDLINE | ID: mdl-18971457

ABSTRACT

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.


Subject(s)
Arachidonic Acids/physiology , Bone Neoplasms/metabolism , Disease Models, Animal , Hyperalgesia/metabolism , Pain/metabolism , Skin/metabolism , Animals , Arachidonic Acids/genetics , Bone Neoplasms/genetics , Cannabinoids/genetics , Cannabinoids/metabolism , Cells, Cultured , Endocannabinoids , Hyperalgesia/genetics , Male , Mice , Mice, Inbred C3H , Pain/genetics , Physical Stimulation/methods , Polyunsaturated Alkamides , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/physiology , Skin/pathology , Touch/physiology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
13.
Brain Res ; 1215: 69-75, 2008 Jun 18.
Article in English | MEDLINE | ID: mdl-18486111

ABSTRACT

Several lines of evidence suggest that cannabinoids can attenuate various types of pain and hyperalgesia through peripheral mechanisms. The development of rodent cancer pain models has provided the opportunity to investigate novel approaches to treat this common form of pain. In the present study, we examined the ability of peripherally administered cannabinoids to attenuate tumor-evoked mechanical hyperalgesia in a murine model of cancer pain. Unilateral injection of osteolytic fibrosarcoma cells into and around the calcaneus bone resulted in tumor formation and mechanical hyperalgesia in the injected hindpaw. Mechanical hyperalgesia was defined as an increase in the frequency of paw withdrawals to a suprathreshold von Frey filament (3.4 mN) applied to the plantar surface of the hindpaw. WIN 55, 212-2 (1.5 to 10 microg) injected subcutaneously into the tumor-bearing hindpaw produced a dose-dependent decrease in paw withdrawal frequencies to suprathreshold von Frey filament stimulation. Injection of WIN 55,212-2 (10 microg) into the contralateral hindpaw did not decrease paw withdrawal frequencies in the tumor-bearing hindpaw. Injection of the highest antihyperalgesic dose of WIN 55,212-2 (10 microg) did not produce catalepsy as determined by the bar test. Co-administration of WIN 55,212-2 with either cannabinoid 1 (AM251) or cannabinoid 2 (AM630) receptor antagonists attenuated the antihyperalgesic effects of WIN 55, 212-2. In conclusion, peripherally administered WIN 55,212-2 attenuated tumor-evoked mechanical hyperalgesia by activation of both peripheral cannabinoid 1 and cannabinoid 2 receptors. These results suggest that peripherally-administered cannabinoids may be effective in attenuating cancer pain.


Subject(s)
Analgesics/pharmacology , Benzoxazines/pharmacology , Fibrosarcoma/complications , Hyperalgesia/drug therapy , Morpholines/pharmacology , Naphthalenes/pharmacology , Receptors, Cannabinoid/metabolism , Anesthetics, Local/pharmacology , Animals , Cannabinoid Receptor Agonists , Disease Models, Animal , Dose-Response Relationship, Drug , Hyperalgesia/etiology , Male , Mice , Mice, Inbred C3H , Neoplasms, Experimental/complications , Random Allocation , Receptors, Cannabinoid/classification , Touch
14.
Brain Res ; 1180: 7-19, 2007 Nov 14.
Article in English | MEDLINE | ID: mdl-17935703

ABSTRACT

Pain associated with cancer, particularly when tumors metastasize to bone, is often severe and debilitating. Better understanding of the neurobiological mechanisms underlying cancer pain will likely lead to the development of more effective treatments. The aim of this study was to characterize changes in response properties of nociceptive dorsal horn neurons following implantation of fibrosarcoma cells into and around the calcaneus bone, an established model of cancer pain. Extracellular electrophysiological recordings were made from wide dynamic range (WDR) and high threshold (HT) dorsal horn neurons in mice with tumor-evoked hyperalgesia and control mice. WDR and HT neurons were examined for ongoing activity and responses to mechanical, heat, and cold stimuli applied to the plantar surface of the hind paw. Behavioral experiments showed that mice exhibited hyperalgesia to mechanical and heat stimuli applied to their tumor-bearing hind paw. WDR, but not HT, nociceptive dorsal horn neurons in tumor-bearing mice exhibited sensitization to mechanical, heat, and cold stimuli and may contribute to tumor-evoked hyperalgesia. Specifically, the proportion of WDR neurons that exhibited ongoing activity and their evoked discharge rates were greater in tumor-bearing than in control mice. In addition, WDR neurons exhibited lower response thresholds for mechanical and heat stimuli, and increased responses to suprathreshold mechanical, heat, and cold stimuli. Our findings show that sensitization of WDR neurons contributes to cancer pain and supports the notion that the mechanisms underlying cancer pain differ from those that contribute to inflammatory and neuropathic pain.


Subject(s)
Bone Neoplasms/complications , Fibrosarcoma/complications , Hyperalgesia/physiopathology , Nociceptors/physiopathology , Pain/physiopathology , Analysis of Variance , Animals , Electrophysiology , Hot Temperature , Hyperalgesia/etiology , Male , Mice , Mice, Inbred C3H , Neoplasms, Experimental , Pain/etiology , Pain Threshold/physiology , Posterior Horn Cells/physiopathology , Statistics, Nonparametric , Touch/physiology
15.
J Neurosci ; 27(38): 10289-98, 2007 Sep 19.
Article in English | MEDLINE | ID: mdl-17881535

ABSTRACT

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.


Subject(s)
Fibrosarcoma/metabolism , Neurons, Afferent/metabolism , Pain/metabolism , Animals , Coculture Techniques , Fibrosarcoma/chemistry , Fibrosarcoma/pathology , Male , Mice , Mice, Inbred C3H , Neurons, Afferent/chemistry , Neurons, Afferent/pathology , Pain/pathology , Pain Measurement/methods , Tumor Cells, Cultured
16.
Neurosci Lett ; 403(3): 305-8, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16777323

ABSTRACT

Nerve growth factor (NGF) plays an important role in inflammation and pain and has been suggested to regulate the responsiveness and sensitivity of nociceptive fibers. However, no study has evaluated whether chronic NGF alters the exocytotic capacity of peripheral terminals of peptidergic fibers. To test this hypothesis, rats were injected subcutaneously every other day with either murine recombinant NGF (mNGF; 1.0 mg/kg) or vehicle for 7 days; or mNGF (0.1 mg/kg), mNGF (1 mg/kg) or vehicle every other day for 13 days. Treatment of rats with NGF over a 13-day period produced a significant increase in capsaicin-evoked iCGRP release from isolated biopsies of hindpaw skin, as assessed by in vitro superfusion and RIA. This effect was dose-dependent and exhibited a temporal requirement, because the enhancement was only observed after 13 days of treatment and was not evident after 7 days of treatment. This NGF enhancement of capsaicin-evoked iCGRP release was not due solely to increases in peripheral iCGRP content since only the 1mg/kg dose of NGF elevated cutaneous pools of iCGRP, whereas both doses significantly increased capsaicin-evoked peptide release. Moreover, NGF also enhanced capsaicin-evoked thermal hyperalgesia under similar dose- and time-related conditions. Collectively, the chronic administration of NGF not only increases capsaicin-evoked hyperalgesia, but also significantly primes peripheral fibers to enhanced peptidergic exocytosis following activation of the capsaicin receptor. Collectively, these data are consistent with the hypothesis that persistently elevated NGF levels may contribute to enhanced neurogenic regulation of inflammatory and wound healing processes in injured tissue.


Subject(s)
Capsaicin/pharmacology , Exocytosis , Nerve Growth Factor/physiology , Neurons/metabolism , Skin/innervation , Animals , Calcitonin Gene-Related Peptide/metabolism , Dose-Response Relationship, Drug , Hot Temperature , Hyperalgesia/physiopathology , In Vitro Techniques , Injections, Subcutaneous , Male , Nerve Endings/metabolism , Nerve Growth Factor/administration & dosage , Nerve Growth Factor/pharmacology , Neurogenic Inflammation/metabolism , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Skin/cytology
17.
Brain Res ; 1044(2): 197-205, 2005 May 24.
Article in English | MEDLINE | ID: mdl-15885218

ABSTRACT

To determine whether ATP and P2X3 receptors contribute to bone-cancer pain in a mouse model, immunohistochemical techniques were used to identify whether changes in the labeling of P2X3 receptors on epidermal nerve fibers (ENFs) occurred during tumor development. C3H mice were injected with osteolytic fibrosarcoma cells in and around the calcaneus bone. These mice exhibited mechanical hyperalgesia by day 10 post-implantation as assessed using von Frey monofilaments. Biopsies of the plantar skin overlying the tumor were obtained at days 10, 14, and 18 post-implantation. Confocal images were analyzed for the number of PGP 9.5, P2X3, and CGRP immunoreactive (ir) ENFs. The overall ENF population (PGP-ir) decreased progressively over time, whereas the subsets of P2X3-ir fibers demonstrated a modest increase and CGRP-ir nerve fibers remained fairly constant. Importantly, the proportion of CGRP-ir fibers that labeled for P2X3 increased from approximately 6% in control animals to nearly 30% at day 14 following tumor cell implantation. These studies demonstrate increased expression of P2X3 receptors on CGRP-ir ENFs during tumor growth and suggest a role for ATP in cancer-related pain.


Subject(s)
Bone Neoplasms/metabolism , Fibrosarcoma/metabolism , Nerve Fibers , Pain/metabolism , Receptors, Purinergic P2/metabolism , Skin/innervation , Animals , Bone Neoplasms/complications , Calcaneus/pathology , Calcaneus/surgery , Calcitonin Gene-Related Peptide/metabolism , Cell Line, Tumor , Disease Models, Animal , Fibrosarcoma/complications , Gene Expression Regulation, Neoplastic/physiology , Male , Mice , Mice, Inbred C3H , Neoplasm Transplantation/methods , Nerve Fibers/metabolism , Nerve Fibers/pathology , Nerve Fibers/physiology , Pain/etiology , Pain Measurement/methods , Receptors, Purinergic P2X3 , Skin/metabolism , Skin/pathology , Time Factors , Ubiquitin Thiolesterase/metabolism
18.
Neurosci Lett ; 363(3): 239-42, 2004 Jun 17.
Article in English | MEDLINE | ID: mdl-15182951

ABSTRACT

The effect of systemic nerve growth factor (NGF) on neuropeptide content and capsaicin-evoked release of neuropeptide from in vitro spinal cord dorsal horn slices was examined. Rats were injected subcutaneously every other day with murine NGF (mNGF) 1 mg/kg or saline for 7 days, or mNGF 0.1/kg, mNGF 1 mg/kg or saline for 13 days. Lumbar dorsal horn slices of the rat spinal cord from all groups showed a significant increase in immunoreactive calcitonin gene-related peptide (CGRP) release upon exposure to capsaicin. This release was enhanced in rats pretreated with mNGF 1 mg/kg for 7 days, but not after 13 days. No enhancement was seen after 7 or 13 days in any treatment group for immunoreactive substance P release. Upon examination of neuropeptide content in dorsal horn, no significant differences were noted between treatment groups. The increased iCGRP release from dorsal horn slices suggests a preferential release of CGRP and provides further evidence that NGF indirectly plays a role in the modulation of inflammation through the regulation of neuropeptide release.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Nerve Growth Factors/pharmacology , Posterior Horn Cells/drug effects , Spinal Cord/cytology , Substance P/metabolism , Animals , Capsaicin/pharmacology , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , In Vitro Techniques , Male , Mice , Posterior Horn Cells/metabolism , Radioimmunoassay , Rats , Rats, Sprague-Dawley
19.
J Neurosci ; 24(7): 1744-53, 2004 Feb 18.
Article in English | MEDLINE | ID: mdl-14973253

ABSTRACT

Inhibition of primary afferent neurons contributes to the antihyperalgesic effects of opioid and CB1 receptor agonists. Two bioassays were used to compare the effects of the CB1 receptor agonist CP 55,940 and morphine on dissociated adult rat DRG neurons. Both agonists inhibited the increase in free intracellular Ca2+ concentration evoked by depolarization; however, effects of CP 55,940 occurred primarily in large neurons (cell area, >800 microm2), whereas morphine inhibited the response in smaller neurons. Cotreatment with selective blockers of L-, N-, and P/Q-type voltage-dependent Ca2+ channels indicated that CB1 receptors on DRG neurons couple solely with N-type channels but opioid receptors couple with multiple subtypes. Experiments with selective agonists and antagonists of opioid receptors indicated that mu and delta, but not kappa, receptors contributed to the inhibitory effect of morphine on voltage-dependent Ca2+ influx. Because Ca2+ channels underlie release of transmitters from neurons, the effects of opioid agonists and CP 55,940 on depolarization-evoked release of calcitonin gene-related peptide (CGRP) were compared. Morphine inhibited release through delta receptors but CP 55,940 had no effect. Colocalization of CGRP with delta-opioid but not mu-opioid or CB1 receptor immunoreactivity in superficial laminae of the dorsal horn of the spinal cord was consistent with the data for agonist inhibition of peptide release. Therefore, CB1 and opioid agonists couple with different voltage-dependent Ca2+ channels in different populations of DRG neurons. Furthermore, differences occur in the distribution of receptors between the cell body and terminals of DRG neurons. The complementary action of CB1 and opioid receptor agonists on populations of DRG neurons provides a rationale for their combined use in modulation of somatosensory input to the spinal cord.


Subject(s)
Ganglia, Spinal/cytology , Narcotics/pharmacology , Neurons/drug effects , Receptor, Cannabinoid, CB1/agonists , Receptors, Opioid/agonists , Animals , Biological Assay , Calcitonin Gene-Related Peptide/biosynthesis , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels/metabolism , Cells, Cultured , Cyclohexanols/pharmacology , Male , Morphine/pharmacology , Narcotic Antagonists/pharmacology , Neurons/classification , Neurons/metabolism , Pertussis Toxin/pharmacology , Potassium/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/metabolism , Receptors, Opioid/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism
20.
Eur J Neurosci ; 14(7): 1113-20, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11683903

ABSTRACT

Many of the physiological hallmarks associated with neurogenic inflammatory processes in cutaneous tissues are similarly present within orofacial structures. Such attributes include the dependence upon capsaicin-sensitive sensory neurons and the involvement of certain inflammatory mediators derived therein, including calcitonin gene-related peptide (CGRP). However, there are also important differences between the trigeminal and spinal nervous systems, and the potential contributions of neurogenic processes to inflammatory disease within the trigeminal system have yet to be fully elucidated. We present here a model system that affords the ability to study mechanisms regulating the efferent functions of peptidergic terminals that may subserve neurogenic inflammation within the oral cavity. Freshly dissected buccal mucosa tissue from adult, male, Sprague-Dawley rats was placed into chambers and superfused with oxygenated, Krebs buffer. Serial aliquots of the egressing superfusate were acquired and analysed by radioimmunoassay for immunoreactive CGRP (iCGRP). Addition of the selective excitotoxin, capsaicin (10-300 microm), to the superfusion buffer resulted in a significant, concentration-dependent increase in superfusate levels of iCGRP. Similarly, release of iCGRP from the buccal mucosa could also be evoked by a depolarizing concentration of potassium chloride (50 mm) or by the calcium ionophore A23187 (1 microm). The specific, capsaicin receptor antagonist, capsazepine (300 microm), completely abolished the capsaicin-evoked release of iCGRP while having no effect whatsoever on the potassium-evoked release. Moreover, capsaicin-evoked release was dependent upon the presence of extracellular calcium ions and was significantly, though incompletely, attenuated by neonatal capsaicin denervation. Collectively, these data indicate that the evoked neurosecretion of iCGRP in response to capsaicin occurs via a vanilloid receptor-mediated, exocytotic mechanism. The model system described here should greatly facilitate future investigations designed to identify and characterize the stimuli that regulate the release of CGRP or other neurosecretory substances in isolated tissues. This system may also be used to elucidate the role of these mediators in the aetiology of inflammatory processes within the trigeminal field of innervation.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Inflammation Mediators/metabolism , Mouth Mucosa/innervation , Mouth Mucosa/metabolism , Neurogenic Inflammation/metabolism , Trigeminal Nerve/metabolism , Animals , Bradykinin/pharmacology , Calcimycin/pharmacology , Calcium/metabolism , Dinoprostone/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Histamine/pharmacology , Ionophores/pharmacology , Male , Mouth Mucosa/drug effects , Neurogenic Inflammation/chemically induced , Neurogenic Inflammation/physiopathology , Nociceptors/drug effects , Nociceptors/metabolism , Organ Culture Techniques , Pain Measurement/drug effects , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin/pharmacology , Trigeminal Nerve/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...