Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Neurol ; 359: 114255, 2023 01.
Article in English | MEDLINE | ID: mdl-36279935

ABSTRACT

Opioids are among the most effective analgesics for the management of pain in the acute phase of a spinal cord injury (SCI), and approximately 80% of patients are treated with morphine in the first 24 h following SCI. We have found that morphine treatment in the first 7 days after SCI increases symptoms of pain at 42 days post-injury and undermines the recovery of locomotor function in a rodent model. Prior research has implicated microglia/macrophages in opioid-induced hyperalgesia and the development of neuropathic pain. We hypothesized that glial activation may also underlie the development of morphine-induced pain and cell death after SCI. Supporting this hypothesis, our previous studies found that intrathecal and intravenous morphine increase the number of activated microglia and macrophages present at the spinal lesion site, and that the adverse effects of intrathecal morphine can be blocked with intrathecal minocycline. Recognizing that the cellular expression of opioid receptors, and the intracellular signaling pathways engaged, can change with repeated administration of opioids, the current study tested whether minocycline was also protective with repeated intravenous morphine administration, more closely simulating clinical treatment. Using a rat model of SCI, we co-administered intravenous morphine and intrathecal minocycline for the first 7 days post injury and monitored sensory and locomotor recovery. Contrary to our hypothesis and previous findings with intrathecal morphine, we found that minocycline did not prevent the negative effects of morphine. Surprisingly, we also found that intrathecal minocycline alone is detrimental for locomotor recovery after SCI. Using ex vivo cell cultures, we investigated how minocycline and morphine altered microglia/macrophage function. Commensurate with published studies, we found that minocycline blocked the effects of morphine on the release of pro-inflammatory cytokines but, like morphine, it increased glial phagocytosis. While phagocytosis is critical for the removal of cellular and extracellular debris at the spinal injury site, increased phagocytosis after injury has been linked to the clearance of stressed but viable neurons and protracted inflammation. In sum, our data suggest that both morphine and minocycline alter the acute immune response, and reduce locomotor recovery after SCI.


Subject(s)
Neuralgia , Spinal Cord Injuries , Rats , Animals , Morphine , Minocycline/therapeutic use , Recovery of Function , Rats, Sprague-Dawley , Spinal Cord Injuries/complications , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Analgesics, Opioid , Neuralgia/metabolism , Spinal Cord/pathology
2.
BMC Neurosci ; 23(1): 58, 2022 10 10.
Article in English | MEDLINE | ID: mdl-36217122

ABSTRACT

BACKGROUND: Opioids are among the most effective and commonly prescribed analgesics for the treatment of acute pain after spinal cord injury (SCI). However, morphine administration in the early phase of SCI undermines locomotor recovery, increases cell death, and decreases overall health in a rodent contusion model. Based on our previous studies we hypothesize that morphine acts on classic opioid receptors to alter the immune response. Indeed, we found that a single dose of intrathecal morphine increases the expression of activated microglia and macrophages at the injury site. Whether similar effects of morphine would be seen with repeated intravenous administration, more closely simulating clinical treatment, is not known. METHODS: To address this, we used flow cytometry to examine changes in the temporal expression of microglia and macrophages after SCI and intravenous morphine. Next, we explored whether morphine changed the function of these cells through the engagement of cell-signaling pathways linked to neurotoxicity using Western blot analysis. RESULTS: Our flow cytometry studies showed that 3 consecutive days of morphine administration after an SCI significantly increased the number of microglia and macrophages around the lesion. Using Western blot analysis, we also found that repeated administration of morphine increases ß-arrestin, ERK-1 and dynorphin (an endogenous kappa opioid receptor agonist) production by microglia and macrophages. CONCLUSIONS: These results suggest that morphine administered immediately after an SCI changes the innate immune response by increasing the number of immune cells and altering neuropeptide synthesis by these cells.


Subject(s)
Morphine , Spinal Cord Injuries , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Dynorphins/metabolism , Dynorphins/pharmacology , Dynorphins/therapeutic use , Macrophages , Microglia/pathology , Morphine/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, kappa/therapeutic use , Recovery of Function , Spinal Cord/metabolism , Spinal Cord Injuries/pathology , beta-Arrestins/metabolism , beta-Arrestins/pharmacology , beta-Arrestins/therapeutic use
3.
Biology (Basel) ; 11(2)2022 Jan 26.
Article in English | MEDLINE | ID: mdl-35205056

ABSTRACT

After spinal cord injury (SCI), 80% of individuals are diagnosed with osteopenia or osteoporosis. The dramatic loss of bone after SCI increases the potential for fractures 100-fold, with post-fracture complications occurring in 54% of cases. With the age of new SCI injuries increasing, we hypothesized that a SCI-induced reduction in weight bearing could further exacerbate age-induced bone loss. To test this, young (2-3 months) and old (20-30 months) male and female mice were given a moderate spinal contusion injury (T9-T10), and recovery was assessed for 28 days (BMS, rearing counts, distance traveled). Tibial trabecular bone volume was measured after 28 days with ex vivo microCT. While BMS scores did not differ across groups, older subjects travelled less in the open field and there was a decrease in rearing with age and SCI. As expected, aging decreased trabecular bone volume and cortical thickness in both old male and female mice. SCI alone also reduced trabecular bone volume in young mice, but did not have an additional effect beyond the age-dependent decrease in trabecular and cortical bone volume seen in both sexes. Interestingly, both rearing and total activity correlated with decreased bone volume. These data underscore the importance of load and use on bone mass. While partial weight-bearing does not stabilize/reverse bone loss in humans, our data suggest that therapies that simulate complete loading may be effective after SCI.

4.
J Neurosci Methods ; 346: 108894, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32771372

ABSTRACT

BACKGROUND: Electrical Stimulation is a traditional tool in neuroscience and is commonly used in vivo to evoke behavior and in vitro to study neural mechanisms. In vivo intracerebral microdialysis, also a traditional technique, is used to assay neurotransmitter release. However, the combination of these techniques is highly limited to studies using anesthetized animals; therefore, evoking and measuring exocytotic neurotransmitter release in awake models is lacking. Combining these techniques in an awake animal preparation is presented here with evidence to support the mechanistic action of electrical stimulation in vivo. NEW METHODS: This report presents converging evidence to validate the combination of intracerebral electrical stimulation with microdialysis as a novel procedure to study exocytotic-like dopamine release in behaving animals. RESULTS: It is shown that electrical stimulation of the medial forebrain bundle can be used to evoke frequency- and intensity-dependent exocytotic-like dopamine overflow and rotational behavior that are sensitive to Na+ channel blockade and Ca++ availability. COMPARISON WITH EXISTING METHODS: Studies using modern techniques to evoke neurotransmitter release, combined with in vivo intracerebral microdialysis, and measured behavioral output are scarce. In contrast, commonly used pharmacological methods often are less precise and inefficient to evoke exocytotic dopamine release and behavior. Here we demonstrate, the combination of in vivo intracerebral microdialysis with electrical stimulation as a simple approach to simultaneously assess physiologically relevant neurotransmitter 'release' and behavior. CONCLUSIONS: Research that aims to understand how dopamine neurotransmission is altered in behavioral disorders can utilize this innovative combination of electrical stimulation with in vivo intracerebral microdialysis.


Subject(s)
Dopamine , Exocytosis , Animals , Electric Stimulation , Microdialysis , Neurotransmitter Agents
5.
Brain Behav Immun ; 79: 125-138, 2019 07.
Article in English | MEDLINE | ID: mdl-30684649

ABSTRACT

Opioids are among the most effective and widely prescribed medications for the treatment of pain following spinal cord injury (SCI). Spinally-injured patients receive opioids within hours of arrival at the emergency room, and prolonged opioid regimens are often employed for the management of post-SCI chronic pain. However, previous studies in our laboratory suggest that the effects of opioids such as morphine may be altered in the pathophysiological context of neurotrauma. Specifically, we have shown that morphine administration in a rodent model of SCI increases mortality and tissue loss at the injury site, and decreases recovery of motor and sensory function, and overall health, even weeks after treatment. The literature suggests that opioids may produce these adverse effects by acting as endotoxins and increasing glial activation and inflammation. To better understand the effects of morphine following SCI, in this study we used flow cytometry to assess immune-competent cells at the lesion site. We observed a morphine-induced increase in the overall number of CD11b+ cells, with marked effects on microglia, in SCI subjects. Next, to investigate whether this increase in the inflammatory profile is necessary to produce morphine's effects, we challenged morphine treatment with minocycline. We found that pre-treatment with minocycline reduced the morphine-induced increase in microglia at the lesion site. More importantly, minocycline also blocked the adverse effects of morphine on recovery of function without disrupting the analgesic efficacy of this opioid. Together, our findings suggest that following SCI, morphine may exacerbate the inflammatory response, increasing cell death at the lesion site and negatively affecting functional recovery.


Subject(s)
Minocycline/metabolism , Minocycline/pharmacology , Spinal Cord Injuries/drug therapy , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacology , Animals , Inflammation/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Microglia/metabolism , Morphine/adverse effects , Morphine/metabolism , Morphine/pharmacology , Pain/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...