Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Harm Reduct J ; 21(1): 71, 2024 03 28.
Article in English | MEDLINE | ID: mdl-38549074

ABSTRACT

BACKGROUND: This study compares emergency department (ED) revisits for patients receiving hospital-based substance-use support compared to those who did not receive specialized addiction services at Health Sciences North in Sudbury, Ontario, Canada. METHODS: The study is a retrospective observational study using administrative data from all patients presenting with substance use disorder (SUD) at Health Sciences North from January 1, 2018, and August 31, 2022 with ICD-10 codes from the Discharge Abstract Database (DAD) and the National Ambulatory Care Database (NACRS). There were two interventions under study: addiction medicine consult services (AMCS group), and specialized addiction medicine unit (AMU group). The AMCS is a consult service offered for patients in the ED and those who are admitted to the hospital. The AMU is a specialized inpatient medical unit designed to offer addiction support to stabilize patients that operates under a harm-reduction philosophy. The primary outcome was all cause ED revisit within 30 days of the index ED or hospital visit. The secondary outcome was all observed ED revisits in the study period. Kaplan-Meier curves were used to measure the proportion of 30-day revisits by exposure group. Odds ratios and Hazard Ratios were calculated using logistic regression models with random effects and Cox-proportional hazard model respectively. RESULTS: A total of 5,367 patients with 10,871 ED index visits, and 2,127 revisits between 2018 and 2022 are included in the study. 45% (2,340/5,367) of patient were not admitted to hospital. 30-day revisits were less likely among the intervention group: Addiction Medicine Consult Services (AMCS) in the ED significantly reduced the odds of revisits (OR 0.53, 95% CI 0.39-0.71, p < 0.01) and first revisits (OR 0.42, 95% CI 0.33-0.53, p < 0.01). The AMU group was associated with lower revisits odds (OR 0.80, 95% CI 0.66-0.98, p = 0.03). For every additional year of age, the odds of revisits slightly decreased (OR 0.99, 95% CI 0.98-1.00, p = 0.01) and males were found to have an increased risk compared to females (OR 1.50, 95% CI 1.35-1.67, p < 0.01). INTERPRETATION: We observe statistically significant differences in ED revisits for patients receiving hospital-based substance-use support at Health Sciences North. Hospital-based substance-use supports could be applied to other hospitals to reduce 30-day revisits.


Subject(s)
Patient Readmission , Substance-Related Disorders , Male , Female , Humans , Retrospective Studies , Emergency Service, Hospital , Substance-Related Disorders/epidemiology , Substance-Related Disorders/therapy , Hospitals , Ontario/epidemiology
2.
Neurosci Lett ; 575: 13-8, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24861519

ABSTRACT

Mitogen-activated protein kinases (MAPKs) play critical roles in the central nervous system immune responses through glial function, which are regulated with relative selectivity (or preference) by MAPK phosphatases (MKP). Phosphorylated extracellular signal-regulated protein kinase (p-ERK) is preferentially dephosphorylated by MKP-3, which display little activity over p-p38 and p-c-Jun NH2-terminal kinases (p-JNK). It has been proposed that these substrate preferences may vary depending on tissue or functional cellular processes. Since astrocytes display a prominent activity of JNK>ERK under stressed or reactive phenotype, we hypothesize that MKP-3 possess a similar or differential substrate preference in astrocytes for JNK and ERK (ERK=JNK or JNK>ERK). We generated transient expression of MKP-3 by transfecting a specific cDNA in primary rat neonatal brain cortex astrocytes. Cells were stimulated with lipopolysaccharide (LPS), and MAPKs and downstream pro-inflammatory products were measured by Western blot and ELISA analyses. MKP-3 expression in primary astrocytes reduced LPS-induced p-ERK and p-p38 by ∼50%, and p-JNK by ∼75%, and moderately reduced nitrite oxide (NO), while completely blocked Interleukin (IL)-6 and tumor necrosis factor alpha (TNFα). We confirmed MKP-3 specific activity by developing a BV-2 microglia cell line stably overexpressing MKP-3 and using a specific siRNA against MKP-3. Our data demonstrate MKP-3 has differential substrate preference in astrocytes compared to other cells types, since it preferentially dephosphorylated p-JNK over p-ERK. Our results indicate also that astrocytic immune functions can be modulated by MKP-3 induction, a strategy that could be beneficial in neurological conditions in which astrocytes play a pathophysiological role, i.e. persistent pain.


Subject(s)
Astrocytes/metabolism , Dual Specificity Phosphatase 6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Dual Specificity Phosphatase 6/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Lipopolysaccharides/pharmacology , Microglia/drug effects , Microglia/metabolism , Phosphorylation , Rats, Sprague-Dawley , Substrate Specificity
3.
J Pain ; 13(9): 836-48, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22901764

ABSTRACT

UNLABELLED: Peripheral nerve injury generally results in spinal neuronal and glial plastic changes associated with chronic behavioral hypersensitivity. Spinal mitogen-activated protein kinases (MAPKs), eg, p38 or extracellular signal-regulated kinases (ERKs), are instrumental in the development of chronic allodynia in rodents, and new p38 inhibitors have shown potential in acute and neuropathic pain patients. We have previously shown that the cannabinoid type 2 receptor agonist JWH015 inhibits ERK activity by inducing MAPK phosphatase (MKP)-1 and MKP-3 (the major regulators of MAPKs) in vitro in microglial cells. Therefore, we decided to investigate the role of these phosphatases in the mechanisms of action of JWH015 in vivo using the rat L5 nerve transection model of neuropathic pain. We observed that peripheral nerve injury reduced spinal MKP-1/3 expression and activity and that intrathecal JWH015 reduced established L5 nerve injury-induced allodynia, enhanced spinal MKP-1/3 expression and activity, and reduced the phosphorylated form of p38 and ERK-1/2. Triptolide, a pharmacological blocker of MKP-1 and MKP-3 expression, inhibited JWH015's effects, suggesting that JWH015 exerts its antinociceptive effects by modulating MKP-1 and MKP-3. JWH015-induced antinociception and MKP-1 and MKP-3 expression were inhibited by the cannabinoid type 2 receptor antagonist AM630. Our data suggest that MKP-1 and MKP-3 are potential targets for novel analgesic drugs. PERSPECTIVE: MAPKs are pivotal in the development of chronic allodynia in rodent models of neuropathic pain. A cannabinoid type 2 receptor agonist, JWH015, reduced neuropathic allodynia in rats by reducing MAPK phosphorylation and inducing spinal MAPK phosphatases 1 and 3, the major regulators of MAPKs.


Subject(s)
Hyperalgesia , Indoles/therapeutic use , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Neuralgia/complications , Receptor, Cannabinoid, CB2/agonists , 4-Nitrophenylphosphatase/metabolism , Animals , Disease Models, Animal , Diterpenes/therapeutic use , Dual Specificity Phosphatase 1/metabolism , Dual Specificity Phosphatase 6/metabolism , Epoxy Compounds/therapeutic use , Gene Expression Regulation/drug effects , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/pathology , Immunosuppressive Agents/therapeutic use , Indoles/pharmacology , Male , Mitogen-Activated Protein Kinase Phosphatases/classification , Nerve Tissue Proteins/metabolism , Phenanthrenes/therapeutic use , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Signal Transduction/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism , Time Factors
4.
Mol Pain ; 8: 34, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22540262

ABSTRACT

BACKGROUND: Neuropathic pain due to nerve injury is one of the most difficult types of pain to treat. Following peripheral nerve injury, neuronal and glial plastic changes contribute to central sensitization and perpetuation of mechanical hypersensitivity in rodents. The mitogen activated protein kinase (MAPK) family is pivotal in this spinal cord plasticity. MAPK phosphatases (MKPs) limit inflammatory processes by dephosphorylating MAPKs. For example, MKP-1 preferentially dephosphorylates p-p38. Since spinal p-p38 is pivotal for the development of chronic hypersensitivity in rodent models of pain, and p-p38 inhibitors have shown clinical potential in acute and chronic pain patients, we hypothesize that induction of spinal MKP-1 will prevent the development of peripheral nerve-injury-induced hypersensitivity and p-p38 overexpression. RESULTS: We cloned rat spinal cord MKP-1 and optimize MKP-1 cDNA in vitro using transfections to BV-2 cells. We observed that in vitro overexpression of MKP-1 blocked lipopolysaccharide-induced phosphorylation of p38 (and other MAPKs) as well as release of pro-algesic effectors (i.e., cytokines, chemokines, nitric oxide). Using this cDNA MKP-1 and a non-viral, in vivo nanoparticle transfection approach, we found that spinal cord overexpression of MKP-1 prevented development of peripheral nerve-injury-induced tactile hypersensitivity and reduced pro-inflammatory cytokines and chemokines and the phosphorylated form of p38. CONCLUSIONS: Our results indicate that MKP-1, the natural regulator of p-p38, mediates resolution of the spinal cord pro-inflammatory milieu induced by peripheral nerve injury, resulting in prevention of chronic mechanical hypersensitivity. We propose that MKP-1 is a potential therapeutic target for pain treatment or prevention.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Neuralgia/enzymology , Neuralgia/physiopathology , Touch/physiology , Animals , Behavior, Animal , Cell Line , Cytokines/biosynthesis , Disease Models, Animal , Gene Knockdown Techniques , Inflammation Mediators , Lumbar Vertebrae/enzymology , Lumbar Vertebrae/physiopathology , Lumbar Vertebrae/surgery , Microglia/enzymology , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Spinal Nerves/physiopathology , Spinal Nerves/surgery , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Neuro Oncol ; 14(2): 119-31, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086978

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain cancer, with a median survival of less than 2 years after diagnosis. The tumor microenvironment plays a critical role in tumor invasion and progression. Microglia and infiltrating macrophages are the most abundant immune cells in the tumor. In the present study, we demonstrate that systemic propentofylline (PPF), an atypical methylxanthine with central nervous system (CNS) glial modulating and anti-inflammatory actions, significantly decreased tumor growth in a CNS-1 rat model of GBM by targeting microglia and not tumor cells. Rats received tumor injections of 1 × 10(5) CNS-1 cells in the right striatum with daily intraperitonial injections of PPF (50 mg/kg) or saline beginning the day of tumor injection. PPF did not cause apoptosis or decrease proliferation of CNS-1 tumor cells. Furthermore, we demonstrate, using in vitro methods, that PPF decreased microglial migration toward CNS-1 tumor cells and decreased MMP-9 expression. The effects of PPF were shown to be specific to microglia and not peripheral macrophages. These results support a differential functional role of resident microglia and infiltrating macrophages in the brain tumor environment. Our data highlight microglia as a crucial target for future therapeutic development and present PPF as a possible drug for treatment of human GBM.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Macrophages/drug effects , Microglia/drug effects , Xanthines/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Central Nervous System/drug effects , Central Nervous System/immunology , Central Nervous System/metabolism , Disease Models, Animal , Glioblastoma/immunology , Glioblastoma/metabolism , Humans , Macrophages/immunology , Male , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Microglia/pathology , Neoplasm Invasiveness/pathology , Rats , Tumor Cells, Cultured , Tumor Microenvironment , Xanthines/therapeutic use
6.
Exp Neurol ; 234(2): 340-50, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22119425

ABSTRACT

There is a growing body of preclinical evidence for the potential involvement of glial cells in neuropathic pain conditions. Several glial-targeted agents are in development for the treatment of pain conditions. Here we report the failure of a glial modulating agent, propentofylline, to decrease pain reported in association with post-herpetic neuralgia. We offer new evidence to help explain why propentofylline failed in patients by describing in vitro functional differences between rodent and human microglia and macrophages. We directly compared the proinflammatory response induced by lipopolysaccharide (LPS) with or without propentofylline using rat postnatal microglia, rat peritoneal macrophages, human fetal microglia, human peripheral macrophages and human immortalized THP-1 cells. We measured tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß) and nitrite release (as an indicator of nitric oxide (NO)) as downstream indicators. We found that LPS treatment did not induce nitrite in human microglia, macrophages or THP-1 cells; however LPS treatment did induce nitrite release in rat microglia and macrophages. Following LPS exposure, propentofylline blocked TNF-α release in rodent microglia with all the doses tested (1-100 µM), and dose-dependently decreased TNF-α release in rodent macrophages. Propentofylline partially decreased TNF-α (35%) at 100 µM in human microglia, macrophages and THP-1 macrophages. Propentofylline blocked nitrite release from LPS stimulated rat microglia and inhibited nitrite in LPS-stimulated rat macrophages. IL-1ß was decreased in LPS-stimulated human microglia following propentofylline at 100 µM. Overall, human microglia were less responsive to LPS stimulation and propentofylline treatment than the other cell types. Our data demonstrate significant functional differences between cell types and species following propentofylline treatment and LPS stimulation. These results may help explain the differential behavioral effects of propentofylline observed between rodent models of pain and the human clinical trial.


Subject(s)
Macrophages/drug effects , Microglia/drug effects , Neuralgia, Postherpetic/drug therapy , Xanthines/pharmacology , Animals , Humans , Macrophages/metabolism , Microglia/metabolism , Neuralgia, Postherpetic/immunology , Neuralgia, Postherpetic/physiopathology , Tumor Necrosis Factor-alpha/metabolism , Xanthines/therapeutic use
7.
PLoS One ; 5(5): e10891, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20531936

ABSTRACT

BACKGROUND: An alarming portion of patients develop persistent or chronic pain following surgical procedures, but the mechanisms underlying the transition from acute to chronic pain states are not fully understood. In general, endocannabinoids (ECBs) inhibit nociceptive processing by stimulating cannabinoid receptors type 1 (CB(1)) and type 2 (CB(2)). We have previously shown that intrathecal administration of a CB(2) receptor agonist reverses both surgical incision-induced behavioral hypersensitivity and associated over-expression of spinal glial markers. We therefore hypothesized that endocannabinoid signaling promotes the resolution of acute postoperative pain by modulating pro-inflammatory signaling in spinal cord glial cells. METHODOLOGY/PRINCIPAL FINDINGS: To test this hypothesis, rats receiving paw incision surgery were used as a model of acute postoperative pain that spontaneously resolves. We first characterized the concentration of ECBs and localization of CB(1) and CB(2) receptors in the spinal cord following paw incision. We then administered concomitant CB(1) and CB(2) receptor antagonists/inverse agonists (AM281 and AM630, 1 mg x kg(-1) each, i.p.) during the acute phase of paw incision-induced mechanical allodynia and evaluated the expression of glial cell markers and phosphorylated p38 (a MAPK associated with inflammation) in the lumbar dorsal horn. Dual blockade of CB(1) and CB(2) receptor signaling prevented the resolution of postoperative allodynia and resulted in persistent over-expression of spinal Glial Fibrillary Acidic Protein (GFAP, an astrocytic marker) and phospho-p38 in astrocytes. We provide evidence for the functional significance of these astrocytic changes by demonstrating that intrathecal administration of propentofylline (50 microg, i.t.) attenuated both persistent behavioral hypersensitivity and over-expression of GFAP and phospho-p38 in antagonist-treated animals. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that endocannabinoid signaling via CB(1) and CB(2) receptors is necessary for the resolution of paw incision-induced behavioral hypersensitivity and for the limitation of pro-inflammatory signaling in astrocytes following surgical insult. Our findings suggest that therapeutic strategies designed to enhance endocannabinoid signaling may prevent patients from developing persistent or chronic pain states following surgery.


Subject(s)
Astrocytes/metabolism , Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Pain, Postoperative/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Astrocytes/drug effects , Astrocytes/pathology , Behavior, Animal/drug effects , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Glial Fibrillary Acidic Protein/metabolism , Male , Microfilament Proteins , Neuroglia/drug effects , Neuroglia/metabolism , Pain, Postoperative/pathology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Xanthines/pharmacology
8.
Mol Pain ; 5: 25, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19476641

ABSTRACT

BACKGROUND: Cannabinoid receptor type 2 (CBR2) inhibits microglial reactivity through a molecular mechanism yet to be elucidated. We hypothesized that CBR2 activation induces an anti-inflammatory phenotype in microglia by inhibiting extracellular signal-regulated kinase (ERK) pathway, via mitogen-activated protein kinase-phosphatase (MKP) induction. MKPs regulate mitogen activated protein kinases, but their role in the modulation of microglial phenotype is not fully understood. RESULTS: JWH015 (a CBR2 agonist) increased MKP-1 and MKP-3 expression, which in turn reduced p-ERK1/2 in LPS-stimulated primary microglia. These effects resulted in a significant reduction of tumor necrosis factor-alpha (TNF) expression and microglial migration. We confirmed the causative link of these findings by using MKP inhibitors. We found that the selective inhibition of MKP-1 by Ro-31-8220 and PSI2106, did not affect p-ERK expression in LPS+JWH015-treated microglia. However, the inhibition of both MKP-1 and MKP-3 by triptolide induced an increase in p-ERK expression and in microglial migration using LPS+JWH015-treated microglia. CONCLUSION: Our results uncover a cellular microglial pathway triggered by CBR2 activation. These data suggest that the reduction of pro-inflammatory factors and microglial migration via MKP-3 induction is part of the mechanism of action of CBR2 agonists. These findings may have clinical implications for further drug development.


Subject(s)
Cell Movement , Extracellular Signal-Regulated MAP Kinases/metabolism , Microglia/cytology , Mitogen-Activated Protein Kinase Phosphatases/physiology , Receptor, Cannabinoid, CB2/physiology , Animals , Animals, Newborn , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/genetics , Dual Specificity Phosphatase 1/physiology , Dual Specificity Phosphatase 6/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation, Enzymologic , Inflammation/prevention & control , Mitogen-Activated Protein Kinase Phosphatases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Phosphatases/genetics , Phenotype , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL