1
|
Rau J, Joseph R, Weise L, Bryan J, Wardeh J, Konda A, Duplessis L, Hook MA. Acute Opioid Administration Undermines Recovery after SCI: Adverse Effects Are Not Restricted to Morphine. J Neurotrauma 2025. [PMID: 39912807 DOI: 10.1089/neu.2024.0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025] Open
Abstract
Previous studies have shown that administration of high doses of morphine in the acute phase of spinal cord injury (SCI) significantly undermines locomotor recovery and increases symptoms of chronic pain in a rat spinal contusion model. Similarly, SCI patients treated with high doses of opioid for the first 24 h postinjury have increased symptoms of chronic pain 1 year later. Whether these adverse effects are driven by morphine only or all opioids compromise recovery after SCI, however, is unknown. Based on our previous findings we hypothesized that activation of the kappa opioid receptor (KOR) is key in the morphine-induced attenuation of locomotor recovery after SCI. Thus, we posited that opioids that engage KOR-mediated signaling pathways (morphine, oxycodone) would undermine recovery, and clinically relevant opioids with less KOR activity (fentanyl and buprenorphine) would not. To test this, we compared the effects of the clinically relevant opioids on locomotor recovery and pain in a male rat spinal contusion model. Rats were given a moderate spinal contusion injury followed by 7 days of intravenous morphine, oxycodone, fentanyl, buprenorphine, or saline, and recovery was assessed for 28 days. All opioids produced analgesia on tests of thermal, mechanical, and incremented shock reactivity. However, tolerance developed rapidly with buprenorphine administration, particularly with daily administrations of 5 morphine milligram equivalent (MME) buprenorphine. Opioid-induced hyperalgesia (OIH) also developed across days following administration of higher doses (10 MME, 20 MME) of morphine and oxycodone. Fentanyl and buprenorphine did not produce OIH. Contrary to our hypothesis, however, we found that high doses of all opioids reduced recovery of locomotor function. Unlike the other opioids, the effects of buprenorphine on locomotor recovery appeared transient, but it also produced chronic pain. Morphine, oxycodone, and buprenorphine decreased reactivity thresholds on tests of mechanical and incremented shock stimulation. In sum, all opioids undermined long-term recovery in the rat model. Further interrogation of the molecular mechanisms driving the adverse effects is essential. This study provides critical insight into pain management strategies in the acute phase of SCI and potential long-term consequences of early opioid administration.
Collapse
Affiliation(s)
- Josephina Rau
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
- Texas A&M Institute for Neuroscience, College Station, Texas, USA
| | - Rose Joseph
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Lara Weise
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Jessica Bryan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
- Texas A&M Institute for Neuroscience, College Station, Texas, USA
| | - Jad Wardeh
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Alekya Konda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Landon Duplessis
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
- Texas A&M Institute for Neuroscience, College Station, Texas, USA
| |
Collapse
|
2
|
Farrokhfar S, Tiraihi T, Movahedin M, Azizi H. The Effect of Antinociceptive Dose of Morphine on Cell Therapy in Rats with Spinal Cord Injury. Mol Neurobiol 2025; 62:1620-1630. [PMID: 39012442 DOI: 10.1007/s12035-024-04350-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024]
Abstract
Spinal cord injury (SCI) is a sensory-motor injury. Today, combined treatments such as cell therapy along with drug therapy and their interactions are of interest. Morphine is an opioid drug used to relieve intolerable pain. This study aims to evaluate the impact of an antinociceptive dose of morphine (with minimal tolerance/dependence but effective pain relief) on cell therapy in SCI. The antinociceptive dose of morphine was determined in rats with SCI through the Hargreaves and naloxone-induced morphine withdrawal tests. The rats were then allocated to 5 groups: laminectomy, SCI, SCI + Morphine, SCI + cell therapy, SCI + Morphine + cell therapy. The antinociceptive dose (5 mg/kg) was administered on days 1, 4, 10, and 13 (i.p.) post-SCI. On day 7, Neural-like stem cells derived from adipose tissue were transplanted intraspinally into the injured animals, and they were monitored for 12 weeks. The outcomes were assessed using the BBB test, somatosensory evoked potential (SSEP), and histology. The BBB test indicated that morphine significantly hindered functional recovery post-cell transplantation compared to animals receiving only cell therapy (p < 0.05). In the SSEP test, the analysis of amplitude and latency of waves did not reveal a significant difference (p > 0.05). The histological results showed that cell therapy reduced the cavity size post-SCI, while morphine had no significant impact on it. Morphine at the antinociceptive dose significantly impairs motor recovery despite cell therapy. Nonetheless, there was no significant difference between groups in terms of sensory pathway outcomes.
Collapse
Affiliation(s)
- Samaneh Farrokhfar
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Anatomical Sciences, Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | - Taki Tiraihi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mansoureh Movahedin
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
3
|
Bourguignon L, Lukas LP, Kondiles BR, Tong B, Lee JJ, Gomes T, Tetzlaff W, Kramer JLK, Walter M, Jutzeler CR. Impact of commonly administered drugs on the progression of spinal cord injury: a systematic review. COMMUNICATIONS MEDICINE 2024; 4:213. [PMID: 39448737 PMCID: PMC11502874 DOI: 10.1038/s43856-024-00638-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Complications arising from acute traumatic spinal cord injury (SCI) are routinely managed by various pharmacological interventions. Despite decades of clinical application, the potential impact on neurological recovery has been largely overlooked. This study aims to highlight commonly administered drugs with potential disease-modifying effects. METHODS This systematic literature review included studies referenced in PubMed, Scopus and Web of Science from inception to March 31st, 2021, which assess disease-modifying properties on neurological and/or functional recovery of drugs routinely administered following spinal cord injury. Drug effects were classified as positive, negative, mixed, no effect, or not (statistically) reported. Risk of bias was assessed separately for animal, randomized clinical trials, and observational human studies. RESULTS We analyzed 394 studies conducting 486 experiments that evaluated 144 unique or combinations of drugs. 195 of the 464 experiments conducted on animals (42%) and one study in humans demonstrate positive disease-modifying properties on neurological and/or functional outcomes. Methylprednisolone, melatonin, estradiol, and atorvastatin are the most common drugs associated with positive effects. Two studies on morphine and ethanol report negative effects on recovery. CONCLUSION Despite a large heterogeneity observed in study protocols, research from bed to bench and back to bedside provides an alternative approach to identify new candidate drugs in the context of SCI. Future research in human populations is warranted to determine if introducing drugs like melatonin, estradiol, or atorvastatin would contribute to enhancing neurological outcomes after acute SCI.
Collapse
Affiliation(s)
- Lucie Bourguignon
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- Schulthess Klinik, Zurich, Switzerland.
| | - Louis P Lukas
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- Schulthess Klinik, Zurich, Switzerland.
| | - Bethany R Kondiles
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Bobo Tong
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
| | - Jaimie J Lee
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
| | - Tomás Gomes
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - John L K Kramer
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Anesthesiology, Pharmacology, and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Matthias Walter
- Department of Urology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Catherine R Jutzeler
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Schulthess Klinik, Zurich, Switzerland
| |
Collapse
|
4
|
Grau JW, Hudson KE, Johnston DT, Partipilo SR. Updating perspectives on spinal cord function: motor coordination, timing, relational processing, and memory below the brain. Front Syst Neurosci 2024; 18:1184597. [PMID: 38444825 PMCID: PMC10912355 DOI: 10.3389/fnsys.2024.1184597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 01/29/2024] [Indexed: 03/07/2024] Open
Abstract
Those studying neural systems within the brain have historically assumed that lower-level processes in the spinal cord act in a mechanical manner, to relay afferent signals and execute motor commands. From this view, abstracting temporal and environmental relations is the province of the brain. Here we review work conducted over the last 50 years that challenges this perspective, demonstrating that mechanisms within the spinal cord can organize coordinated behavior (stepping), induce a lasting change in how pain (nociceptive) signals are processed, abstract stimulus-stimulus (Pavlovian) and response-outcome (instrumental) relations, and infer whether stimuli occur in a random or regular manner. The mechanisms that underlie these processes depend upon signal pathways (e.g., NMDA receptor mediated plasticity) analogous to those implicated in brain-dependent learning and memory. New data show that spinal cord injury (SCI) can enable plasticity within the spinal cord by reducing the inhibitory effect of GABA. It is suggested that the signals relayed to the brain may contain information about environmental relations and that spinal cord systems can coordinate action in response to descending signals from the brain. We further suggest that the study of stimulus processing, learning, memory, and cognitive-like processing in the spinal cord can inform our views of brain function, providing an attractive model system. Most importantly, the work has revealed new avenues of treatment for those that have suffered a SCI.
Collapse
Affiliation(s)
- James W. Grau
- Lab of Dr. James Grau, Department of Psychological and Brain Sciences, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | | | | | | |
Collapse
|
5
|
Jutzeler CR, Bourguignon L, Tong B, Ronca E, Bailey E, Harel NY, Geisler F, Ferguson AR, Kwon BK, Cragg JJ, Grassner L, Kramer JLK. Pharmacological management of acute spinal cord injury: a longitudinal multi-cohort observational study. Sci Rep 2023; 13:5434. [PMID: 37012257 PMCID: PMC10070428 DOI: 10.1038/s41598-023-31773-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 03/16/2023] [Indexed: 04/05/2023] Open
Abstract
Multiple types and classes of medications are administered in the acute management of traumatic spinal cord injury. Prior clinical studies and evidence from animal models suggest that several of these medications could modify (i.e., enhance or impede) neurological recovery. We aimed to systematically determine the types of medications commonly administered, alone or in combination, in the transition from acute to subacute spinal cord injury. For that purpose, type, class, dosage, timing, and reason for administration were extracted from two large spinal cord injury datasets. Descriptive statistics were used to describe the medications administered within the first 60 days after spinal cord injury. Across 2040 individuals with spinal cord injury, 775 unique medications were administered within the two months after injury. On average, patients enrolled in a clinical trial were administered 9.9 ± 4.9 (range 0-34), 14.3 ± 6.3 (range 1-40), 18.6 ± 8.2 (range 0-58), and 21.5 ± 9.7 (range 0-59) medications within the first 7, 14, 30, and 60 days post-injury, respectively. Those enrolled in an observational study were administered on average 1.7 ± 1.7 (range 0-11), 3.7 ± 3.7 (range 0-24), 8.5 ± 6.3 (range 0-42), and 13.5 ± 8.3 (range 0-52) medications within the first 7, 14, 30, and 60 days post-injury, respectively. Polypharmacy was commonplace (up to 43 medications per day per patient). Approximately 10% of medications were administered acutely as prophylaxis (e.g., against the development of pain or infections). To our knowledge, this was the first time acute pharmacological practices have been comprehensively examined after spinal cord injury. Our study revealed a high degree of polypharmacy in the acute stages of spinal cord injury, raising the potential to impact neurological recovery. All results can be interactively explored on the RXSCI web site ( https://jutzelec.shinyapps.io/RxSCI/ ) and GitHub repository ( https://github.com/jutzca/Acute-Pharmacological-Treatment-in-SCI/ ).
Collapse
Affiliation(s)
- Catherine R Jutzeler
- Department of Health Sciences and Technology, ETH Zurich, Lengghalde 2, 8008, Zurich, Switzerland.
| | - Lucie Bourguignon
- Department of Health Sciences and Technology, ETH Zurich, Lengghalde 2, 8008, Zurich, Switzerland
| | - Bobo Tong
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, Canada
| | - Elias Ronca
- Swiss Paraplegic Research, Nottwil, Switzerland
| | - Eric Bailey
- James J Peters Veterans Affairs Medical Center, Bronx, NY, USA
| | - Noam Y Harel
- James J Peters Veterans Affairs Medical Center, Bronx, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fred Geisler
- University of Saskatchewan, Saskatoon, SK, Canada
| | - Adam R Ferguson
- Brain and Spinal Injury Center, Weill Institute for Neurosciences, University of California San Francisco (UCSF), San Francisco, CA, USA
- San Francisco Veteran's Affairs Health Care System, San Francisco, CA, USA
| | - Brian K Kwon
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, Canada
| | - Jacquelyn J Cragg
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, Canada
| | - Lukas Grassner
- Department of Neurosurgery, Medical University Innsbruck, Innsbruck, Austria
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - John L K Kramer
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Anesthesiology, Pharmacology, and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Hugill Centre for Anesthesia, University of British Columbia, Vancouver, Canada
| |
Collapse
|
6
|
Wilkinson RL, Castillo C, Herrity A, Wang D, Sharma M, Dietz N, Adams S, Khattar N, Nuno M, Drazin D, Boakye M, Ugiliweneza B. Opioid Dependence and Associated Health Care Utilization and Cost in Traumatic Spinal Cord Injury Population: Analysis Using Marketscan Database. Top Spinal Cord Inj Rehabil 2023; 29:118-130. [PMID: 36819927 PMCID: PMC9936895 DOI: 10.46292/sci22-00026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Background Postinjury pain is a well-known debilitating complication of spinal cord injury (SCI), often resulting in long-term, high-dose opioid use with the potential for dependence. There is a gap in knowledge about the risk of opioid dependence and the associated health care utilization and cost in SCI. Objectives To evaluate the association of SCI with postinjury opioid use and dependence and evaluate the effect of this opioid dependence on postinjury health care utilization. Methods Using the MarketScan Database, health care utilization claims data were queried to extract 7187 adults with traumatic SCI from 2000 to 2019. Factors associated with post-SCI opioid use and dependence, postinjury health care utilization, and payments were analyzed with generalized linear regression models. Results After SCI, individuals were more likely to become opioid users or transition from nondependent to dependent users (negative change: 31%) than become nonusers or transition from dependent to nondependent users (positive change: 14%, p < .0001). Individuals who were opioid-dependent users pre-SCI had more than 30 times greater odds of becoming dependent after versus not (OR 34; 95% CI, 26-43). Dependent users after injury (regardless of prior use status) had 2 times higher utilization payments and 1.2 to 6 times more health care utilization than nonusers. Conclusion Opioid use and dependence were associated with high health care utilization and cost after SCI. Pre-SCI opioid users were more likely to remain users post-SCI and were heavier consumers of health care. Pre- and postopioid use history should be considered for treatment decision-making in all individuals with SCI.
Collapse
Affiliation(s)
| | - Camilo Castillo
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky
| | - April Herrity
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Lousville, Kentucky
| | - Dengzhi Wang
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Lousville, Kentucky
| | - Mayur Sharma
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky
| | - Nick Dietz
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky
| | - Shawn Adams
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky
| | - Nicholas Khattar
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky
| | - Miriam Nuno
- Department of Public Health Science, University of California, Davis
| | - Doniel Drazin
- College of Medicine Pacific Northwest, Yamika, Washington
| | - Maxwell Boakye
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Lousville, Kentucky
| | - Beatrice Ugiliweneza
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Lousville, Kentucky
- Department of Health Management and Systems Science, University of Louisville, Louisville, Kentucky
| |
Collapse
|
7
|
Rau J, Weise L, Moore R, Terminel M, Brakel K, Cunningham R, Bryan J, Stefanov A, Hook MA. Intrathecal minocycline does not block the adverse effects of repeated, intravenous morphine administration on recovery of function after SCI. Exp Neurol 2023; 359:114255. [PMID: 36279935 DOI: 10.1016/j.expneurol.2022.114255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 09/18/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
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.
Collapse
Affiliation(s)
- Josephina Rau
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA.
| | - Lara Weise
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA.
| | - Robbie Moore
- Department of Microbial Pathogenesis and Immunology, Texas A&M Institute for Neuroscience, Address: 8447 Riverside Parkway, Medical and Research Education Building 2, Bryan, TX 77807, USA.
| | - Mabel Terminel
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA
| | - Kiralyn Brakel
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA
| | - Rachel Cunningham
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA
| | - Jessica Bryan
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA.
| | - Alexander Stefanov
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA.
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Address: 8447 Riverside Parkway, Medical and Research Education Building 1, Bryan, TX 77807, USA; Texas A&M Institute for Neuroscience, Address: 301 Old Main Drive, Interdisciplinary Life Sciences Building, College Station, TX 77843, USA.
| |
Collapse
|
8
|
Rau J, Hemphill A, Araguz K, Cunningham R, Stefanov A, Weise L, Hook MA. Adverse Effects of Repeated, Intravenous Morphine on Recovery after Spinal Cord Injury in Young, Male Rats Are Blocked by a Kappa Opioid Receptor Antagonist. J Neurotrauma 2022; 39:1741-1755. [PMID: 35996351 PMCID: PMC10039279 DOI: 10.1089/neu.2022.0208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Immediately following spinal cord injury (SCI) patients experience pain associated with injury to the spinal cord and nerves as well as with accompanying peripheral injuries. This pain is usually treated with opioids, and most commonly with morphine. However, in a rodent model we have shown that, irrespective of the route of administration, morphine administered in the acute phase of SCI undermines long-term locomotor recovery. Our previous data suggest that activation of kappa opioid receptors (KORs) mediates these negative effects. Blocking KORs with norbinaltorphimine (norBNI), prior to a single dose of epidural morphine, prevented the morphine-induced attenuation of locomotor recovery. Because numerous cellular changes occur with chronic opioid administration compared with a single dose, the current study tested whether norBNI was also effective in a more clinically relevant paradigm of repeated, intravenous morphine administration after SCI. We hypothesized that blocking KOR activation during repeated, intravenous morphine administration would also protect recovery. Supporting this hypothesis, we found that blocking KOR activation in young, male rats prevented the negative effects of morphine on locomotor recovery, although neither norBNI nor morphine had an effect on long-term pain at the doses used. We also found that norBNI treatment blocked the adverse effects of morphine on lesion size. These data suggest that a KOR antagonist given in conjunction with morphine may provide a clinical strategy for effective analgesia without compromising locomotor recovery after SCI.
Collapse
Affiliation(s)
- Josephina Rau
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
- Texas A&M Institute for Neuroscience, Bryan, Texas, USA
| | - Annebel Hemphill
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Kendall Araguz
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Rachel Cunningham
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Alexander Stefanov
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
- Texas A&M Institute for Neuroscience, Bryan, Texas, USA
| | - Lara Weise
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
| | - Michelle A. Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas, USA
- Texas A&M Institute for Neuroscience, Bryan, Texas, USA
| |
Collapse
|
9
|
Terminel MN, Bassil C, Rau J, Trevino A, Ruiz C, Alaniz R, Hook MA. Morphine-induced changes in the function of microglia and macrophages after acute spinal cord injury. BMC Neurosci 2022; 23:58. [PMID: 36217122 PMCID: PMC9552511 DOI: 10.1186/s12868-022-00739-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
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 β-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. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-022-00739-3.
Collapse
Affiliation(s)
- Mabel N Terminel
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, 8447 Riverside Parkway 47, Bryan, TX, 77807, USA.
| | - Carla Bassil
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, 8447 Riverside Parkway 47, Bryan, TX, 77807, USA
| | - Josephina Rau
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, 8447 Riverside Parkway 47, Bryan, TX, 77807, USA
| | - Amanda Trevino
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, 8447 Riverside Parkway 47, Bryan, TX, 77807, USA
| | - Cristina Ruiz
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, 8447 Riverside Parkway 47, Bryan, TX, 77807, USA
| | - Robert Alaniz
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, 8447 Riverside Parkway 47, Bryan, TX, 77807, USA
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, 8447 Riverside Parkway 47, Bryan, TX, 77807, USA
| |
Collapse
|
10
|
Hudson KE, Grau JW. Ionic Plasticity: Common Mechanistic Underpinnings of Pathology in Spinal Cord Injury and the Brain. Cells 2022; 11:2910. [PMID: 36139484 PMCID: PMC9496934 DOI: 10.3390/cells11182910] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
The neurotransmitter GABA is normally characterized as having an inhibitory effect on neural activity in the adult central nervous system (CNS), which quells over-excitation and limits neural plasticity. Spinal cord injury (SCI) can bring about a modification that weakens the inhibitory effect of GABA in the central gray caudal to injury. This change is linked to the downregulation of the potassium/chloride cotransporter (KCC2) and the consequent rise in intracellular Cl- in the postsynaptic neuron. As the intracellular concentration increases, the inward flow of Cl- through an ionotropic GABA-A receptor is reduced, which decreases its hyperpolarizing (inhibitory) effect, a modulatory effect known as ionic plasticity. The loss of GABA-dependent inhibition enables a state of over-excitation within the spinal cord that fosters aberrant motor activity (spasticity) and chronic pain. A downregulation of KCC2 also contributes to the development of a number of brain-dependent pathologies linked to states of neural over-excitation, including epilepsy, addiction, and developmental disorders, along with other diseases such as hypertension, asthma, and irritable bowel syndrome. Pharmacological treatments that target ionic plasticity have been shown to bring therapeutic benefits.
Collapse
Affiliation(s)
- Kelsey E. Hudson
- Neuroscience, Texas A&M University, College Station, TX 77843, USA
| | - James W. Grau
- Psychological & Brain Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
11
|
Kawakami K, Tanaka S, Sugiyama Y, Mochizuki N, Kawamata M. Intrathecal morphine exacerbates paresis with increasing muscle tone of hindlimbs in rats with mild thoracic spinal cord injury but without damage of lumbar α-motoneurons. PLoS One 2022; 17:e0273095. [PMID: 35969631 PMCID: PMC9377624 DOI: 10.1371/journal.pone.0273095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 08/02/2022] [Indexed: 11/25/2022] Open
Abstract
Adverse effects of morphine on locomotor function after moderate to severe spinal cord injury (SCI) have been reported; however, the effects after mild SCI without damage of lumbar α-motoneurons have not been investigated. We investigated the effects of lumbar intrathecal morphine on locomotor function after mild thoracic SCI and the involvement of classic opioid receptor activation. A mild thoracic contusive SCI was induced in adult rats at the T9-T10 spine level under sevoflurane anesthesia. We evaluated the effects of single doses of intrathecal morphine and selective μ-, δ-, and κ-opioid receptor agonists, continuous infusion of intrathecal morphine for 72 hours, and administration of physiological saline on locomotor function and muscle tone in the hindlimbs. The numbers of damaged and total α-motoneurons in the lumbar spinal cord were also investigated. Single doses of morphine aggravated residual locomotor function after SCI but did not affect functional recovery. Single doses of morphine and μ- and δ-opioid receptor agonists significantly aggravated residual locomotor function with increases in muscle tone after SCI, and the effects of the drugs were reversed by naloxone. In contrast, continuous infusion of morphine led to persistent decline in locomotor function with increased muscle tone, which was not reversed by naloxone, but did not increase the number of damaged lumbar α-motoneurons. These results indicate that a single dose of morphine at an analgesic dose transiently increases muscle tone of the hindlimbs via activation of spinal μ- and δ- opioid receptors, resulting in further deterioration of locomotor function in the acute phase of mild SCI. Our results also suggest that an increased dose of morphine with prolonged administration leads to persistent decline in locomotor function with increased muscle tone via mechanisms other than direct activation of classical opioid receptors. Morphine should be used cautiously even after mild SCI.
Collapse
Affiliation(s)
- Katsuhiro Kawakami
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Nagano, Japan
| | - Satoshi Tanaka
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Nagano, Japan
- * E-mail:
| | - Yuki Sugiyama
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Nagano, Japan
| | - Noriaki Mochizuki
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Nagano, Japan
| | - Mikito Kawamata
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Nagano, Japan
| |
Collapse
|
12
|
Lovell S, Simon B, Boudreau EC, Mankin J, Jeffery N. Randomized clinical trial comparing outcomes after fentanyl or ketamine-dexmedetomidine analgesia in thoracolumbar spinal surgery in dogs. J Vet Intern Med 2022; 36:1742-1751. [PMID: 35962706 PMCID: PMC9511085 DOI: 10.1111/jvim.16514] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/28/2022] [Indexed: 11/30/2022] Open
Abstract
Background Opioids are widely used for perioperative pain control in dogs undergoing spinal surgery, but alternatives may be required because data suggest that opioids exacerbate inflammation in the injured spinal cord and veterinary access to opioids may become more restricted in the future. Objectives To compare recovery of ambulation and other functions between spinal cord‐injured dogs receiving peri‐operative fentanyl and those receiving a ketamine‐dexmedetomidine combination. Animals A total of 102 client‐owned dogs undergoing decompressive surgery for thoracolumbar intervertebral disc herniation. Methods Randomized clinical trial. Dogs were randomized 1:1 to fentanyl or a ketamine‐dexmedetomidine combination for intra and postoperative analgesia. Primary outcome was time to recovery of ambulation; secondary outcomes were the postoperative Colorado Acute Pain Scale, the short‐form Glasgow Composite Measure Pain Scale, time to recovery of voluntary urination and time to unassisted eating. Results No difference was found in time to recovery of ambulation between groups (adjusted sub‐hazard ratio, 0.83; 95% confidence interval [CI], 0.55‐1.24; P = .36) or in pain scores (Colorado: χ2 = 14.74; P = .32; Glasgow: χ2 = 6.61; P = .76). Differences in time to recovery of eating and urination were small but favored ketamine‐dexmedetomidine (adjusted odds ratios, 3.31; 95% CI, 1.53‐7.16; P = .002 and 2.43; 95% CI, 1.00‐5.96; P = .05, respectively). Conclusions and Clinical Importance There was no evidence that, at the doses used, fentanyl impaired ambulatory outcome after surgery for thoracolumbar intervertebral disc herniation in dogs. Pain control appeared similar between groups. Secondary outcomes suggested minor benefits associated with ketamine‐dexmedetomidine. The ketamine‐dexmedetomidine combination appears to be a reasonable alternative to peri‐operative opioids.
Collapse
Affiliation(s)
- Stephanie Lovell
- Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| | - Bradley Simon
- Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| | - Elizabeth C Boudreau
- Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| | - Joseph Mankin
- Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| | - Nicholas Jeffery
- Small Animal Clinical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
13
|
Fauss GNK, Hudson KE, Grau JW. Role of Descending Serotonergic Fibers in the Development of Pathophysiology after Spinal Cord Injury (SCI): Contribution to Chronic Pain, Spasticity, and Autonomic Dysreflexia. BIOLOGY 2022; 11:234. [PMID: 35205100 PMCID: PMC8869318 DOI: 10.3390/biology11020234] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/29/2022] [Indexed: 12/12/2022]
Abstract
As the nervous system develops, nerve fibers from the brain form descending tracts that regulate the execution of motor behavior within the spinal cord, incoming sensory signals, and capacity to change (plasticity). How these fibers affect function depends upon the transmitter released, the receptor system engaged, and the pattern of neural innervation. The current review focuses upon the neurotransmitter serotonin (5-HT) and its capacity to dampen (inhibit) neural excitation. A brief review of key anatomical details, receptor types, and pharmacology is provided. The paper then considers how damage to descending serotonergic fibers contributes to pathophysiology after spinal cord injury (SCI). The loss of serotonergic fibers removes an inhibitory brake that enables plasticity and neural excitation. In this state, noxious stimulation can induce a form of over-excitation that sensitizes pain (nociceptive) circuits, a modification that can contribute to the development of chronic pain. Over time, the loss of serotonergic fibers allows prolonged motor drive (spasticity) to develop and removes a regulatory brake on autonomic function, which enables bouts of unregulated sympathetic activity (autonomic dysreflexia). Recent research has shown that the loss of descending serotonergic activity is accompanied by a shift in how the neurotransmitter GABA affects neural activity, reducing its inhibitory effect. Treatments that target the loss of inhibition could have therapeutic benefit.
Collapse
Affiliation(s)
| | | | - James W. Grau
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX 77843, USA; (G.N.K.F.); (K.E.H.)
| |
Collapse
|
14
|
The μ-opioid receptor induces miR-21 expression and is ERK/PKCμ-dependent. J Neuroimmunol 2021; 356:577585. [PMID: 33940234 DOI: 10.1016/j.jneuroim.2021.577585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 11/22/2022]
Abstract
Micro RNA-21 (miR-21) is believed to perform an important role in the transition from inflammation to resolution in the innate immune response. The biochemical basis for the induction of miR-21 remains uncertain. However, the activation of the μ-opioid receptor (MOR) induces the expression of miR-21. Our results show that human monocytes treated with μ-opioid agonists exhibit a significant increase in miR-21 expression. We found that MOR-induction of miR-21 requires the activation of the Ras-Raf-MEK-ERK signaling cascade, and to our surprise, the activation of PKCμ (PKD1). These results are significant given the role of miR-21 in the sensitivity to pain.
Collapse
|
15
|
Dermorphin [D-Arg2, Lys4] (1-4) amide inhibits below-level heat hypersensitivity in mice after contusive thoracic spinal cord injury. Pain 2020; 160:2710-2723. [PMID: 31365470 DOI: 10.1097/j.pain.0000000000001671] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Opioid use for chronic pain is limited by severe central adverse effects. We examined whether activating mu-opioid receptors (MORs) in the peripheral nervous system attenuates spinal cord injury (SCI) pain-like behavior in mice. We produced a contusive SCI at the T10 vertebral level and examined motor and sensory dysfunction for 6 weeks. At 6 weeks, we tested the effect of subcutaneous (s.c.) injection of dermorphin [D-Arg2, Lys4] (1-4) amide (DALDA), a peripherally acting MOR-preferring agonist, on mechanical and heat hypersensitivity. Basso mouse scale score was significantly decreased after SCI, and mice showed hypersensitivity to mechanical and heat stimulation at the hind paw beginning at 2 weeks, as indicated by increased paw withdrawal frequency to mechanical stimulation and decreased paw withdrawal latency to heat stimulation. In wild-type SCI mice, DALDA (1 mg/kg, s.c.) attenuated heat but not mechanical hypersensitivity. The effect was blocked by pretreatment with an intraperitoneal injection of methylnaltrexone (5 mg/kg), a peripherally restricted opioid receptor antagonist, and was also diminished in Pirt-MOR conditional knockout mice. DALDA did not adversely affect exploratory activity or induced preference to drug treatment in SCI mice. In vivo calcium imaging showed that DALDA (1, 10 mg/kg, s.c.) inhibited responses of small dorsal root ganglion neurons to noxious heat stimulation in Pirt-GCaMP6s mice after SCI. Western blot analysis showed upregulation of MOR in the lumbar spinal cord and sciatic nerves at 6 weeks after SCI. Our findings suggest that peripherally acting MOR agonist may inhibit heat hypersensitivity below the injury level with minimal adverse effects.
Collapse
|
16
|
Davis JA, Bopp AC, Henwood MK, Baine RE, Cox CC, Grau JW. Pharmacological Transection of Brain-Spinal Cord Communication Blocks Pain-Induced Hemorrhage and Locomotor Deficits after Spinal Cord Injury in Rats. J Neurotrauma 2020; 37:1729-1739. [PMID: 32368946 DOI: 10.1089/neu.2019.6973] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Spinal cord injury (SCI) is often accompanied by additional tissue damage (polytrauma), which engages pain (nociceptive) fibers. Prior research has shown that nociceptive input can increase cell death, expand the area of hemorrhage, and impair long-term recovery. The current study shows that these adverse effects can be blocked by the sodium channel blocker lidocaine applied rostral to a contusion injury. Rats received a lower thoracic (T12) contusion injury, and noxious electrical stimulation (shock) was applied to the tail 24 h later. Immediately before shock treatment, a pharmacological transection was performed by slowly infusing lidocaine at T2. Long-term locomotor recovery was assessed over the next 21 days. Noxious electrical stimulation impaired locomotor recovery, and this effect was blocked by rostral lidocaine. Next, the acute effect of lidocaine was assessed. Tissue was collected 3 h after noxious stimulation, and the extent of hemorrhage was evaluated by assessing hemoglobin content using Western blotting. Nociceptive stimulation increased the extent of hemorrhage. Lidocaine applied at T2 before, but not immediately after, stimulation blocked this effect. A similar pattern of results was observed when lidocaine was applied at the site of injury by means of a lumbar puncture. The results show that a pharmacological transection blocks nociception-induced hemorrhage and exacerbation of locomotor deficits.
Collapse
Affiliation(s)
- Jacob A Davis
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas, USA
| | - Anne C Bopp
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas, USA
| | - Melissa K Henwood
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas, USA
| | - Rachel E Baine
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas, USA
| | - Carol C Cox
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas, USA
| | - James W Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
17
|
Kartha S, Yan L, Ita ME, Amirshaghaghi A, Luo L, Wei Y, Tsourkas A, Winkelstein BA, Cheng Z. Phospholipase A 2 Inhibitor-Loaded Phospholipid Micelles Abolish Neuropathic Pain. ACS NANO 2020; 14:8103-8115. [PMID: 32484651 PMCID: PMC7438274 DOI: 10.1021/acsnano.0c00999] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Treating persistent neuropathic pain remains a major clinical challenge. Current conventional treatment approaches carry a substantial risk of toxicity and provide only transient pain relief. In this work, we show that the activity and expression of the inflammatory mediator secretory phospholipase-A2 (sPLA2) enzyme increases in the spinal cord after painful nerve root compression. We then develop phospholipid micelle-based nanoparticles that release their payload in response to sPLA2 activity. Using a rodent model of neuropathic pain, phospholipid micelles loaded with the sPLA2 inhibitor, thioetheramide-PC (TEA-PC), are administered either locally or intravenously at the time of painful injury or 1-2 days afterward. Local micelle administration immediately after compression prevents pain for up to 7 days. Delayed intravenous administration of the micelles attenuates existing pain. These findings suggest that sPLA2 inhibitor-loaded micelles can be a promising anti-inflammatory nanotherapeutic for neuropathic pain treatment.
Collapse
Affiliation(s)
- Sonia Kartha
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, 240 Skirkanich Hall, Philadelphia, Pennsylvania 19104, United States
| | - Lesan Yan
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, 240 Skirkanich Hall, Philadelphia, Pennsylvania 19104, United States
| | - Meagan E Ita
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, 240 Skirkanich Hall, Philadelphia, Pennsylvania 19104, United States
| | - Ahmad Amirshaghaghi
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, 240 Skirkanich Hall, Philadelphia, Pennsylvania 19104, United States
| | - Lijun Luo
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, 240 Skirkanich Hall, Philadelphia, Pennsylvania 19104, United States
| | - Yulong Wei
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, 240 Skirkanich Hall, Philadelphia, Pennsylvania 19104, United States
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, 240 Skirkanich Hall, Philadelphia, Pennsylvania 19104, United States
| | - Beth A Winkelstein
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, 240 Skirkanich Hall, Philadelphia, Pennsylvania 19104, United States
- Department of Neurosurgery, University of Pennsylvania, Hospital of the University of Pennsylvania, 3400 Spruce Street, 3 Silverstein, Philadelphia, Pennsylvania 19104, United States
| | - Zhiliang Cheng
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, 240 Skirkanich Hall, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
18
|
The first 24 h: opioid administration in people with spinal cord injury and neurologic recovery. Spinal Cord 2020; 58:1080-1089. [PMID: 32461572 DOI: 10.1038/s41393-020-0483-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 11/09/2022]
Abstract
STUDY DESIGN Retrospective chart review. OBJECTIVES The objective of this study was to characterize opioid administration in people with acute SCI and examine the association between opioid dose and (1) changes in motor/functional scores from hospital to rehabilitation discharge, and (2) pain, depression, and quality of life (QOL) scores 1-year post injury. SETTING Spinal Cord Injury Model System (SCIMS) inpatient acute rehabilitation facility. METHODS Patients included in the SCIMS from 2008 to 2011 were linked to the National Trauma Registry and the electronic medical record. Three opioid dose groups (low, medium, and high) were defined based on the total morphine equivalence in milligrams at 24 h. The associations between opioid dose groups and functional/motor outcomes were assessed, as well as 1-year follow-up pain and QOL surveys. RESULTS In all, 85/180 patients had complete medication records. By 24 h, all patients had received opioids. Patients receiving higher amounts of opioids had higher pain scores 1 year later compared with medium- and low-dose groups (pain levels 5.5 vs. 4 vs. 1, respectively, p = 0.018). There was also an 8× greater risk of depression 1 year later in the high-dose group compared with the low-dose group (OR: 8.1, 95% CI: 1.2-53.7). In analyses of motor scores, we did not find a significant interaction between opioid dose and duration of injury. CONCLUSIONS These preliminary findings suggest that higher doses of opioids administered within 24 h of injury are associated with increased pain in the chronic phase of people with SCI.
Collapse
|
19
|
Strain MM, Hook MA, Reynolds JD, Huang YJ, Henwood MK, Grau JW. A brief period of moderate noxious stimulation induces hemorrhage and impairs locomotor recovery after spinal cord injury. Physiol Behav 2019; 212:112695. [PMID: 31647990 DOI: 10.1016/j.physbeh.2019.112695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 10/25/2022]
Abstract
Spinal cord injury (SCI) is often accompanied by additional tissue damage (polytrauma) that provides a source of pain input. Our studies suggest that this pain input may be detrimental to long-term recovery. In a rodent model, we have shown that engaging pain (nociceptive) fibers caudal to a lower thoracic contusion SCI impairs recovery of locomotor function and increases tissue loss (secondary injury) and hemorrhage at the site of injury. In these studies, nociceptive fibers were activated using intermittent electrical stimulation. The stimulation parameters were derived from earlier studies demonstrating that 6 min of noxious stimulation, at an intensity (1.5 mA) that engages unmyelinated C (pain) fibers, induces a form of maladaptive plasticity within the lumbosacral spinal cord. We hypothesized that both shorter bouts of nociceptive input and lower intensities of stimulation will decrease locomotor function and increase spinal cord hemorrhage when rats have a spinal cord contusion. To test this, the present study exposed rats to electrical stimulation 24 h after a moderate lower thoracic contusion SCI. One group of rats received 1.5 mA stimulation for 0, 14.4, 72, or 180 s. Another group received six minutes of stimulation at 0, 0.17, 0.5, and 1.5 mA. Just 72 s of stimulation induced an acute disruption in motor performance, increased hemorrhage, and undermined the recovery of locomotor function. Likewise, less intense (0.5 mA) stimulation produced an acute disruption in motor performance, fueled hemorrhage, and impaired long-term recovery. The results imply that a brief period of moderate pain input can trigger hemorrhage after SCI and undermine long-term recovery. This highlights the importance of managing nociceptive signals after concurrent peripheral and central nervous system injuries.
Collapse
Affiliation(s)
- Misty M Strain
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA.
| | - Michelle A Hook
- Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Joshua D Reynolds
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - Yung-Jen Huang
- ChemPartner, 998 Halei Rd., Zhangjiang Hi-Tech Park, Pudong New Area, Shanghai, 201203 China
| | - Melissa K Henwood
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| | - James W Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
20
|
Huot-Lavoie M, Ting WKC, Demers M, Mercier C, Ethier C. Impaired Motor Learning Following a Pain Episode in Intact Rats. Front Neurol 2019; 10:927. [PMID: 31507526 PMCID: PMC6718695 DOI: 10.3389/fneur.2019.00927] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/09/2019] [Indexed: 01/22/2023] Open
Abstract
Motor learning and pain are important factors influencing rehabilitation. Despite being mostly studied independently from each other, important interactions exist between them in the context of spinal cord injury, whether to the spinal cord or the body. Ongoing or recent past episodes of nociceptive activity can prevent motor learning in spinalized rats. In intact animals, it has been proposed that supraspinal activity could counter the repressive effect of nociception on motor system plasticity, but this has not yet been verified in behavioral conditions. The aim of this study was to test whether a recent episode of nociception affects subsequent motor learning in intact animals. We trained rodents to walk on a custom-made horizontal ladder. After initial training, the rats underwent a week-long rest, during which they were randomly assigned to a control group, or one out of two pain conditions. Nociceptive stimuli of different durations were induced through capsaicin or Complete Freund's Adjuvant injections and timed so that the mechanical hypersensitivity had entirely subsided by the end of the resting period. Training then resumed on a modified version of the horizontal ladder. We evaluated the animals' ability to adapt to the modified task by measuring their transit time and paw misplacements over 4 days. Our results show that prior pain episodes do affect motor learning in neurologically intact rats. Motor learning deficits also seem to be influenced by the duration of the pain episode. Rats receiving a subcutaneous injection of capsaicin displayed immediate signs of mechanical hypersensitivity, which subsided rapidly. Nonetheless, they still showed learning deficits 24 h after injection. Rats who received a Complete Freund's Adjuvant injection displayed mechanical hypersensitivity for up to 7 days during the resting period. When trained on the modified ladder task upon returning to normal sensitivity levels, these rats exhibited more prolonged motor learning deficits, extending over 3 days. Our results suggest that prior pain episodes can negatively influence motor learning, and that the duration of the impairment relates to the duration of the pain episode. Our results highlight the importance of addressing pain together with motor training after injury.
Collapse
Affiliation(s)
- Maxime Huot-Lavoie
- CERVO Research Center, Psychiatry and Neurosciences Department, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Windsor Kwan-Chun Ting
- CERVO Research Center, Psychiatry and Neurosciences Department, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Maxime Demers
- CERVO Research Center, Psychiatry and Neurosciences Department, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Catherine Mercier
- Centre for Interdisciplinary Research in Rehabilitation and Social Integration, Department of Rehabilitation, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Christian Ethier
- CERVO Research Center, Psychiatry and Neurosciences Department, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
21
|
Aceves M, Terminel MN, Okoreeh A, Aceves AR, Gong YM, Polanco A, Sohrabji F, Hook MA. Morphine increases macrophages at the lesion site following spinal cord injury: Protective effects of minocycline. Brain Behav Immun 2019; 79:125-138. [PMID: 30684649 DOI: 10.1016/j.bbi.2019.01.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 01/05/2019] [Accepted: 01/21/2019] [Indexed: 12/13/2022] Open
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.
Collapse
Affiliation(s)
- Miriam Aceves
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Mabel N Terminel
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Andre Okoreeh
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Alejandro R Aceves
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Yan Ming Gong
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Alan Polanco
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, United States.
| |
Collapse
|
22
|
Emery MA, Eitan S. Members of the same pharmacological family are not alike: Different opioids, different consequences, hope for the opioid crisis? Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:428-449. [PMID: 30790677 DOI: 10.1016/j.pnpbp.2019.02.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 01/14/2023]
Abstract
Pain management is the specialized medical practice of modulating pain perception and thus easing the suffering and improving the life quality of individuals suffering from painful conditions. Since this requires the modulation of the activity of endogenous systems involved in pain perception, and given the large role that the opioidergic system plays in pain perception, opioids are currently the most effective pain treatment available and are likely to remain relevant for the foreseeable future. This contributes to the rise in opioid use, misuse, and overdose death, which is currently characterized by public health officials in the United States as an epidemic. Historically, the majority of preclinical rodent studies were focused on morphine. This has resulted in our understanding of opioids in general being highly biased by our knowledge of morphine specifically. However, recent in vitro studies suggest that direct extrapolation of research findings from morphine to other opioids is likely to be flawed. Notably, these studies suggest that different opioid analgesics (opioid agonists) engage different downstream signaling effects within the cell, despite binding to and activating the same receptors. This recognition implies that, in contrast to the historical status quo, different opioids cannot be made equivalent by merely dose adjustment. Notably, even at equianalgesic doses, different opioids could result in different beneficial and risk outcomes. In order to foster further translational research regarding drug-specific differences among opioids, here we review basic research elucidating differences among opioids in pharmacokinetics, pharmacodynamics, their capacity for second messenger pathway activation, and their interactions with the immune system and the dopamine D2 receptors.
Collapse
Affiliation(s)
- Michael A Emery
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA; Interdisciplinary Program in Neuroscience, Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA; Interdisciplinary Program in Neuroscience, Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, USA.
| |
Collapse
|
23
|
Engaging pain fibers after a spinal cord injury fosters hemorrhage and expands the area of secondary injury. Exp Neurol 2018; 311:115-124. [PMID: 30268767 DOI: 10.1016/j.expneurol.2018.09.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/07/2018] [Accepted: 09/27/2018] [Indexed: 11/24/2022]
Abstract
In humans, spinal cord injury (SCI) is often accompanied by additional tissue damage (polytrauma) that can engage pain (nociceptive) fibers. Prior work has shown that this nociceptive input can expand the area of tissue damage (secondary injury), undermine behavioral recovery, and enhance the development of chronic pain. Here, it is shown that nociceptive input given a day after a lower thoracic contusion injury in rats enhances the infiltration of red blood cells at the site of injury, producing an area of hemorrhage that expands secondary injury. Peripheral nociceptive fibers were engaged 24 h after injury by means of electrical stimulation (shock) applied at an intensity that engages unmyelinated pain (C) fibers or through the application of the irritant capsaicin. Convergent western immunoblot and cyanmethemoglobin colorimetric assays showed that both forms of stimulation increased the concentration of hemoglobin at the site of injury, with a robust effect observed 3-24 h after stimulation. Histopathology confirmed that shock treatment increased the area of hemorrhage and the infiltration of red blood cells. SCI can lead to hemorrhage by engaging the sulfonylurea receptor 1 (SUR1) transient receptor potential melastatin 4 (TRPM4) channel complex in neurovascular endothelial cells, which leads to cell death and capillary fragmentation. Histopathology confirmed that areas of hemorrhage showed capillary fragmentation. Co-immunoprecipitation of the SUR1-TRPM4 complex showed that it was up-regulated by noxious stimulation. Shock-induced hemorrhage was associated with an acute disruption in locomotor performance. These results imply that noxious stimulation impairs long-term recovery because it amplifies the breakdown of the blood spinal cord barrier (BSCB) and the infiltration of red blood cells, which expands the area of secondary injury.
Collapse
|
24
|
Turtle JD, Strain MM, Reynolds JA, Huang YJ, Lee KH, Henwood MK, Garraway SM, Grau JW. Pain Input After Spinal Cord Injury (SCI) Undermines Long-Term Recovery and Engages Signal Pathways That Promote Cell Death. Front Syst Neurosci 2018; 12:27. [PMID: 29977195 PMCID: PMC6021528 DOI: 10.3389/fnsys.2018.00027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/17/2018] [Indexed: 12/19/2022] Open
Abstract
Pain (nociceptive) input caudal to a spinal contusion injury increases tissue loss and impairs long-term recovery. It was hypothesized that noxious stimulation has this effect because it engages unmyelinated pain (C) fibers that produce a state of over-excitation in central pathways. The present article explored this issue by assessing the effect of capsaicin, which activates C-fibers that express the transient receptor potential vanilloid receptor-1 (TRPV1). Rats received a lower thoracic (T11) contusion injury and capsaicin was applied to one hind paw the next day. For comparison, other animals received noxious electrical stimulation at an intensity that engages C fibers. Both forms of stimulation elicited similar levels of c-fos mRNA expression, a cellular marker of nociceptive activation, and impaired long-term behavioral recovery. Cellular assays were then performed to compare the acute effect of shock and capsaicin treatment. Both forms of noxious stimulation increased expression of tumor necrosis factor (TNF) and caspase-3, which promotes apoptotic cell death. Shock, but not capsaicin, enhanced expression of signals related to pyroptotic cell death [caspase-1, inteleukin-1 beta (IL-1ß)]. Pyroptosis has been linked to the activation of the P2X7 receptor and the outward flow of adenosine triphosphate (ATP) through the pannexin-1 channel. Blocking the P2X7 receptor with Brilliant Blue G (BBG) reduced the expression of signals related to pyroptotic cell death in contused rats that had received shock. Blocking the pannexin-1 channel with probenecid paradoxically had the opposite effect. BBG enhanced long-term recovery and lowered reactivity to mechanical stimulation applied to the girdle region (an index of chronic pain), but did not block the adverse effect of nociceptive stimulation. The results suggest that C-fiber input after injury impairs long-term recovery and that this effect may arise because it induces apoptotic cell death.
Collapse
Affiliation(s)
- Joel D Turtle
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | - Misty M Strain
- United States Army Institute of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX, United States
| | - Joshua A Reynolds
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | - Yung-Jen Huang
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | - Kuan H Lee
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | - Melissa K Henwood
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| | - Sandra M Garraway
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - James W Grau
- Lab of Dr. James Grau, Department of Psychology, Cellular and Behavioral Neuroscience, Texas A&M University, College Station, TX, United States
| |
Collapse
|
25
|
Brolin E, Zelleroth S, Jonsson A, Hallberg M, Grönbladh A, Nyberg F. Chronic administration of morphine using mini-osmotic pumps affects spatial memory in the male rat. Pharmacol Biochem Behav 2018; 167:1-8. [DOI: 10.1016/j.pbb.2018.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 12/19/2017] [Accepted: 01/31/2018] [Indexed: 11/30/2022]
|
26
|
Warner FM, Cragg JJ, Jutzeler CR, Röhrich F, Weidner N, Saur M, Maier DD, Schuld C, Curt A, Kramer JK. Early Administration of Gabapentinoids Improves Motor Recovery after Human Spinal Cord Injury. Cell Rep 2017; 18:1614-1618. [PMID: 28199834 DOI: 10.1016/j.celrep.2017.01.048] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 11/24/2016] [Accepted: 01/19/2017] [Indexed: 12/16/2022] Open
Abstract
The anticonvulsant pregabalin promotes neural regeneration in a mouse model of spinal cord injury (SCI). We have also previously observed that anticonvulsants improve motor outcomes following human SCI. The present study examined the optimal timing and type of anticonvulsants administered in a large, prospective, multi-center, cohort study in acute SCI. Mixed-effects regression techniques were used to model total motor scores at 1, 3, 6, and 12 months post injury. We found that early (not late) administration of anticonvulsants significantly improved motor recovery (6.25 points over 1 year). The beneficial effect of anticonvulsants remained significant after adjustment for differences in 1-month motor scores and injury characteristics. A review of a subset of patients revealed that gabapentinoids were the most frequently administrated anticonvulsant. Together with preclinical findings, intervention with anticonvulsants represents a potential pharmacological strategy to improve motor function after SCI.
Collapse
Affiliation(s)
- Freda M Warner
- School of Kinesiology, University of British Columbia, Vancouver, BC V6T 1Z1, Canada; International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Jacquelyn J Cragg
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC V5Z 1M9, Canada; Spinal Cord Injury Center, University Hospital Balgrist, University of Zurich, Zurich 8008, Switzerland
| | - Catherine R Jutzeler
- School of Kinesiology, University of British Columbia, Vancouver, BC V6T 1Z1, Canada; International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC V5Z 1M9, Canada; Spinal Cord Injury Center, University Hospital Balgrist, University of Zurich, Zurich 8008, Switzerland
| | - Frank Röhrich
- Berufsgenossenschaftliche Klinik Bergmanstrost of Halle, Halle 06112, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Marion Saur
- Orthopädische Klinik, Hessisch Lichtenau 37235, Germany
| | - Doris D Maier
- Berufsgenossenschaftliche Unfallklinik Murnau, Murnau 82418, Germany
| | - Christian Schuld
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg 69120, Germany
| | - Armin Curt
- Spinal Cord Injury Center, University Hospital Balgrist, University of Zurich, Zurich 8008, Switzerland; European Multi-centre Study about Spinal Cord Injury (EMSCI) Study Group, University Hospital Balgrist, University of Zurich, Zurich 8008, Switzerland
| | - John K Kramer
- School of Kinesiology, University of British Columbia, Vancouver, BC V6T 1Z1, Canada; International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC V5Z 1M9, Canada.
| |
Collapse
|
27
|
Gaudet AD, Ayala MT, Schleicher WE, Smith EJ, Bateman EM, Maier SF, Watkins LR. Exploring acute-to-chronic neuropathic pain in rats after contusion spinal cord injury. Exp Neurol 2017; 295:46-54. [PMID: 28552717 DOI: 10.1016/j.expneurol.2017.05.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/25/2017] [Accepted: 05/25/2017] [Indexed: 01/25/2023]
Abstract
Spinal cord injury (SCI) causes chronic pain in 65% of individuals. Unfortunately, current pain management is inadequate for many SCI patients. Rodent models could help identify how SCI pain develops, explore new treatment strategies, and reveal whether acute post-SCI morphine worsens chronic pain. However, few studies explore or compare SCI-elicited neuropathic pain in rats. Here, we sought to determine how different clinically relevant contusion SCIs in male and female rats affect neuropathic pain, and whether acute morphine worsens later chronic SCI pain. First, female rats received sham surgery, or 150kDyn or 200kDyn midline T9 contusion SCI. These rats displayed modest mechanical allodynia and long-lasting thermal hyperalgesia. Next, a 150kDyn (1s dwell) midline contusion SCI was performed in male and female rats. Interestingly, males, but not females showed SCI-elicited mechanical allodynia; rats of both sexes had thermal hyperalgesia. In this model, acute morphine treatment had no significant effect on chronic neuropathic pain symptoms. Unilateral SCIs can also elicit neuropathic pain that could be exacerbated by morphine, so male rats received unilateral T13 contusion SCI (100kDyn). These rats exhibited significant, transient mechanical allodynia, but not thermal hyperalgesia. Acute morphine did not exacerbate chronic pain. Our data show that specific rat contusion SCI models cause neuropathic pain. Further, chronic neuropathic pain elicited by these contusion SCIs was not amplified by our course of early post-trauma morphine. Using clinically relevant rat models of SCI could help identify novel pain management strategies.
Collapse
Affiliation(s)
- Andrew D Gaudet
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA.
| | - Monica T Ayala
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA
| | - Wolfgang E Schleicher
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA
| | - Elana J Smith
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA
| | - Emily M Bateman
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA
| | - Linda R Watkins
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA; Center for Neuroscience, University of Colorado Boulder, Muenzinger D244 | 345 UCB, Boulder, CO 80309, USA
| |
Collapse
|
28
|
Grau JW, Huang YJ, Turtle JD, Strain MM, Miranda RC, Garraway SM, Hook MA. When Pain Hurts: Nociceptive Stimulation Induces a State of Maladaptive Plasticity and Impairs Recovery after Spinal Cord Injury. J Neurotrauma 2017; 34:1873-1890. [PMID: 27788626 PMCID: PMC5444485 DOI: 10.1089/neu.2016.4626] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) is often accompanied by other tissue damage (polytrauma) that provides a source of pain (nociceptive) input. Recent findings are reviewed that show SCI places the caudal tissue in a vulnerable state that exaggerates the effects nociceptive stimuli and promotes the development of nociceptive sensitization. Stimulation that is both unpredictable and uncontrollable induces a form of maladaptive plasticity that enhances nociceptive sensitization and impairs spinally mediated learning. In contrast, relational learning induces a form of adaptive plasticity that counters these adverse effects. SCI sets the stage for nociceptive sensitization by disrupting serotonergic (5HT) fibers that quell overexcitation. The loss of 5HT can enhance neural excitability by reducing membrane-bound K+-Cl- cotransporter 2, a cotransporter that regulates the outward flow of Cl-. This increases the intracellular concentration of Cl-, which reduces the hyperpolarizing (inhibitory) effect of gamma-aminobutyric acid. Uncontrollable noxious stimulation also undermines the recovery of locomotor function, and increases behavioral signs of chronic pain, after a contusion injury. Nociceptive stimulation has a greater effect if experienced soon after SCI. This adverse effect has been linked to a downregulation in brain-derived neurotrophic factor and an upregulation in the cytokine, tumor necrosis factor. Noxious input enhances tissue loss at the site of injury by increasing the extent of hemorrhage and apoptotic/pyroptotic cell death. Intrathecal lidocaine blocks nociception-induced hemorrhage, cellular indices of cell death, and its adverse effect on behavioral recovery. Clinical implications are discussed.
Collapse
Affiliation(s)
- James W. Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas
| | - Yung-Jen Huang
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas
| | - Joel D. Turtle
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas
| | - Misty M. Strain
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station, Texas
| | - Rajesh C. Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas
| | - Sandra M. Garraway
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Michelle A. Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, Texas
| |
Collapse
|
29
|
Stampas A, York HS, O'Dell MW. Is the Routine Use of a Functional Electrical Stimulation Cycle for Lower Limb Movement Standard of Care for Acute Spinal Cord Injury Rehabilitation? PM R 2017; 9:521-528. [PMID: 28526124 DOI: 10.1016/j.pmrj.2017.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 03/18/2017] [Indexed: 11/20/2022]
Affiliation(s)
- Argyrios Stampas
- TIRR Memorial Hermann and Department of PM&R, UT Health Science Center at Houston, Houston, TX(∗)
| | - Henry S York
- Department of Neurology, University of Maryland School of Medicine, and Spinal Cord Injury Unit, University of Maryland Rehabilitation & Orthopaedic Institute, Baltimore, MD(†)
| | - Michael W O'Dell
- Weill Cornell Medicine, 525 E. 68th Street, 16th FL, New York, NY 10065(‡).
| |
Collapse
|
30
|
Mercier C, Roosink M, Bouffard J, Bouyer LJ. Promoting Gait Recovery and Limiting Neuropathic Pain After Spinal Cord Injury. Neurorehabil Neural Repair 2016; 31:315-322. [PMID: 27913797 PMCID: PMC5405804 DOI: 10.1177/1545968316680491] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Most persons living with a spinal cord injury experience neuropathic pain in the months following their lesion, at the moment where they receive intensive gait rehabilitation. Based on studies using animal models, it has been proposed that central sensitization in nociceptive pathways (maladaptive plasticity) and plasticity related to motor learning (adaptive plasticity) share common neural mechanisms and compete with each other. This article aims to address the discrepancy between the growing body of basic science literature supporting this hypothesis and the general belief in rehabilitation research that pain and gait rehabilitation represent two independent problems. First, the main findings from basic research showing interactions between nociception and learning in the spinal cord will be summarized, focusing both on evidence demonstrating the impact of nociception on motor learning and of motor learning on central sensitization. Then, the generalizability of these findings in animal models to humans will be discussed. Finally, the way potential interactions between nociception and motor learning are currently taken into account in clinical research in patients with spinal cord injury will be presented. To conclude, recommendations will be proposed to better integrate findings from basic research into future clinical research in persons with spinal cord injury.
Collapse
Affiliation(s)
- Catherine Mercier
- 1 Centre Interdisciplinaire de Recherche en Réadaptation et Intégration Sociale, Quebec City, Quebec, Canada.,2 Laval University, Quebec City, Quebec, Canada
| | - Meyke Roosink
- 1 Centre Interdisciplinaire de Recherche en Réadaptation et Intégration Sociale, Quebec City, Quebec, Canada
| | - Jason Bouffard
- 1 Centre Interdisciplinaire de Recherche en Réadaptation et Intégration Sociale, Quebec City, Quebec, Canada.,2 Laval University, Quebec City, Quebec, Canada
| | - Laurent J Bouyer
- 1 Centre Interdisciplinaire de Recherche en Réadaptation et Intégration Sociale, Quebec City, Quebec, Canada.,2 Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
31
|
Turtle JD, Strain MM, Aceves M, Huang YJ, Reynolds JA, Hook MA, Grau JW. Pain Input Impairs Recovery after Spinal Cord Injury: Treatment with Lidocaine. J Neurotrauma 2016; 34:1200-1208. [PMID: 27912032 DOI: 10.1089/neu.2016.4778] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
More than 90% of spinal cord injuries are caused by traumatic accidents and are often associated with other tissue damage (polytrauma) that can provide a source of continued pain input during recovery. In a clinically relevant spinal cord contusion injury model, prior work has shown that noxious stimulation at an intensity that engages pain (C) fibers soon after injury augments secondary injury and impairs functional recovery. Noxious input increases the expression of pro-inflammatory cytokines (interleukin 1β and 18), cellular signals associated with cell death (caspase 3 and 8), and physiological signs of hemorrhage. Here, it is shown that reducing neural excitability after spinal cord injury (SCI) with the local anesthetic lidocaine (micro-injected by means of a lumbar puncture) blocks these adverse cellular effects. In contrast, treatment with an analgesic dose of morphine had no effect. Contused rats that received nociceptive stimulation soon after injury exhibited poor locomotor recovery, less weight gain, and greater tissue loss at the site of injury. Prophylactic application of lidocaine blocked the adverse effect of nociceptive stimulation on behavioral recovery and reduced tissue loss from secondary injury. The results suggest that quieting neural excitability using lidocaine can reduce the adverse effect of pain input (from polytrauma or surgery) after SCI.
Collapse
Affiliation(s)
- Joel D Turtle
- 1 Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University , College Station, Texas
| | - Misty M Strain
- 1 Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University , College Station, Texas
| | - Miriam Aceves
- 2 Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University , College Station, Texas
| | - Yung-Jen Huang
- 1 Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University , College Station, Texas
| | - Joshua A Reynolds
- 1 Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University , College Station, Texas
| | - Michelle A Hook
- 2 Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University , College Station, Texas
| | - James W Grau
- 1 Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University , College Station, Texas
| |
Collapse
|
32
|
Aceves M, Bancroft EA, Aceves AR, Hook MA. Nor-Binaltorphimine Blocks the Adverse Effects of Morphine after Spinal Cord Injury. J Neurotrauma 2016; 34:1164-1174. [PMID: 27736318 DOI: 10.1089/neu.2016.4601] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Opioids are frequently used for the treatment of pain following spinal cord injury (SCI). Unfortunately, we have shown that morphine administered in the acute phase of SCI results in significant, adverse secondary consequences including compromised locomotor and sensory recovery. Similarly, we showed that selective activation of the κ-opioid receptor (KOR), even at a dose 32-fold lower than morphine, is sufficient to attenuate recovery of locomotor function. In the current study, we tested whether activation of the KOR is necessary to produce morphine's adverse effects using nor-Binaltorphimine (norBNI), a selective KOR antagonist. Rats received a moderate spinal contusion (T12) and 24 h later, baseline locomotor function and nociceptive reactivity were assessed. Rats were then administered norBNI (0, 0.02, 0.08, or 0.32 μmol) followed by morphine (0 or 0.32 μmol). Nociception was reassessed 30 min after drug treatment, and recovery was evaluated for 21 days. The effects of norBNI on morphine-induced attenuation of recovery were dose dependent. At higher doses, norBNI blocked the adverse effects of morphine on locomotor recovery, but analgesia was also significantly decreased. Conversely, at low doses, analgesia was maintained, but the adverse effects on recovery persisted. A moderate dose of norBNI, however, adequately protected against morphine's adverse effects without eliminating its analgesic efficacy. This suggests that activation of the KOR system plays a significant role in the morphine-induced attenuation of recovery. Our research suggests that morphine, and other opioid analgesics, may be contraindicated for the SCI population. Blocking KOR activity may be a viable strategy for improving the safety of clinical opioid use.
Collapse
Affiliation(s)
- Miriam Aceves
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Eric A Bancroft
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Alejandro R Aceves
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| |
Collapse
|
33
|
Hook MA, Woller SA, Bancroft E, Aceves M, Funk MK, Hartman J, Garraway SM. Neurobiological Effects of Morphine after Spinal Cord Injury. J Neurotrauma 2016; 34:632-644. [PMID: 27762659 DOI: 10.1089/neu.2016.4507] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Opioids and non-steroidal anti-inflammatory drugs are used commonly to manage pain in the early phase of spinal cord injury (SCI). Despite its analgesic efficacy, however, our studies suggest that intrathecal morphine undermines locomotor recovery and increases lesion size in a rodent model of SCI. Similarly, intravenous (IV) morphine attenuates locomotor recovery. The current study explores whether IV morphine also increases lesion size after a spinal contusion (T12) injury and quantifies the cell types that are affected by early opioid administration. Using an experimenter-administered escalating dose of IV morphine across the first seven days post-injury, we quantified the expression of neuron, astrocyte, and microglial markers at the injury site. SCI decreased NeuN expression relative to shams. In subjects with SCI treated with IV morphine, virtually no NeuN+ cells remained across the rostral-caudal extent of the lesion. Further, whereas SCI per se increased the expression of astrocyte and microglial markers (glial fibrillary acidic protein and OX-42, respectively), morphine treatment decreased the expression of these markers. These cellular changes were accompanied by attenuation of locomotor recovery (Basso, Beattie, Bresnahan scores), decreased weight gain, and the development of opioid-induced hyperalgesia (increased tactile reactivity) in morphine-treated subjects. These data suggest that morphine use is contraindicated in the acute phase of a spinal injury. Faced with a lifetime of intractable pain, however, simply removing any effective analgesic for the management of SCI pain is not an ideal option. Instead, these data underscore the critical need for further understanding of the molecular pathways engaged by conventional medications within the pathophysiological context of an injury.
Collapse
Affiliation(s)
- Michelle A Hook
- 1 Texas A&M University Institute for Neuroscience, Texas A&M University , College Station, Texas.,2 Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Sarah A Woller
- 3 Department of Anesthesiology, University of California , San Diego, California
| | - Eric Bancroft
- 2 Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Miriam Aceves
- 1 Texas A&M University Institute for Neuroscience, Texas A&M University , College Station, Texas.,2 Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Mary Katherine Funk
- 2 Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - John Hartman
- 2 Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | - Sandra M Garraway
- 4 Department of Physiology, Emory University School of Medicine , Atlanta, Georgia
| |
Collapse
|
34
|
Ellis A, Grace PM, Wieseler J, Favret J, Springer K, Skarda B, Ayala M, Hutchinson MR, Falci S, Rice KC, Maier SF, Watkins LR. Morphine amplifies mechanical allodynia via TLR4 in a rat model of spinal cord injury. Brain Behav Immun 2016; 58:348-356. [PMID: 27519154 PMCID: PMC5067205 DOI: 10.1016/j.bbi.2016.08.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 08/07/2016] [Accepted: 08/07/2016] [Indexed: 01/09/2023] Open
Abstract
Central neuropathic pain (CNP) is a pervasive, debilitating problem that impacts thousands of people living with central nervous system disorders, including spinal cord injury (SCI). Current therapies for treating this type of pain are ineffective and often have dose-limiting side effects. Although opioids are one of the most commonly used CNP treatments, recent animal literature has indicated that administering opioids shortly after a traumatic injury can actually have deleterious effects on long-term health and recovery. In order to study the deleterious effects of administering morphine shortly after trauma, we employed our low thoracic (T13) dorsal root avulsion model (Spinal Neuropathic Avulsion Pain, SNAP). Administering a weeklong course of 10mg/kg/day morphine beginning 24h after SNAP resulted in amplified mechanical allodynia. Co-administering the non-opioid toll-like receptor 4 (TLR4) antagonist (+)-naltrexone throughout the morphine regimen prevented morphine-induced amplification of SNAP. Exploration of changes induced by early post-trauma morphine revealed that this elevated gene expression of TLR4, TNF, IL-1β, and NLRP3, as well as IL-1β protein at the site of spinal cord injury. These data suggest that a short course of morphine administered early after spinal trauma can exacerbate CNP in the long term. TLR4 initiates this phenomenon and, as such, may be potential therapeutic targets for preventing the deleterious effects of administering opioids after traumatic injury.
Collapse
Affiliation(s)
- Amanda Ellis
- University of Colorado Boulder, Department of Psychology and Neuroscience, Center for Neuroscience, Boulder, CO, USA
| | - Peter M Grace
- University of Colorado Boulder, Department of Psychology and Neuroscience, Center for Neuroscience, Boulder, CO, USA; School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Julie Wieseler
- University of Colorado Boulder, Department of Psychology and Neuroscience, Center for Neuroscience, Boulder, CO, USA
| | - Jacob Favret
- University of Colorado Boulder, Department of Psychology and Neuroscience, Center for Neuroscience, Boulder, CO, USA
| | - Kendra Springer
- University of Colorado Boulder, Department of Psychology and Neuroscience, Center for Neuroscience, Boulder, CO, USA
| | - Bryce Skarda
- University of Colorado Boulder, Department of Psychology and Neuroscience, Center for Neuroscience, Boulder, CO, USA
| | - Monica Ayala
- University of Colorado Boulder, Department of Psychology and Neuroscience, Center for Neuroscience, Boulder, CO, USA
| | - Mark R Hutchinson
- School of Medicine, University of Adelaide, Adelaide, SA, Australia; Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Australia
| | | | - Kenner C Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD, USA
| | - Steven F Maier
- University of Colorado Boulder, Department of Psychology and Neuroscience, Center for Neuroscience, Boulder, CO, USA
| | - Linda R Watkins
- University of Colorado Boulder, Department of Psychology and Neuroscience, Center for Neuroscience, Boulder, CO, USA.
| |
Collapse
|
35
|
Kandasamy R, Calsbeek JJ, Morgan MM. Analysis of inflammation-induced depression of home cage wheel running in rats reveals the difference between opioid antinociception and restoration of function. Behav Brain Res 2016; 317:502-507. [PMID: 27746208 DOI: 10.1016/j.bbr.2016.10.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 02/06/2023]
Abstract
Opioids are effective at inhibiting responses to noxious stimuli in rodents, but have limited efficacy and many side effects in chronic pain patients. One reason for this disconnect is that nociception is typically assessed using withdrawal from noxious stimuli in animals, whereas chronic pain patients suffer from abnormal pain that disrupts normal activity. We hypothesized that assessment of home cage wheel running in rats would provide a much more clinically relevant method to assess opioid efficacy to restore normal behavior. Intraplantar injection of Complete Freund's Adjuvant (CFA) into the right hindpaw depressed wheel running and caused mechanical allodynia measured with the von Frey test in both male and female rats. Administration of an ED50 dose of morphine (3.2mg/kg) reversed mechanical allodynia, but did not reverse CFA-induced depression of wheel running. In contrast, administration of a low dose of morphine (1.0mg/kg) restored running for one hour in both sexes, but had no effect on mechanical allodynia. Administration of the atypical opioid buprenorphine had no effect on inflammation-induced depression of wheel running in male or female rats, but attenuated mechanical allodynia in male rats. Administration of buprenorphine and higher doses of morphine depressed wheel running in non-inflamed rats, suggesting that the side effects of opioids interfere with restoration of function. These data indicate that restoration of pain-depressed function requires antinociception in the absence of disruptive side effects. The disruptive side effects of opioids are consistent with the major limitation of opioid use in human pain patients.
Collapse
Affiliation(s)
- Ram Kandasamy
- Graduate Program in Neuroscience, Washington State University, Pullman, WA, United States.
| | - Jonas J Calsbeek
- Department of Psychology, Washington State University Vancouver, Vancouver, WA, United States
| | - Michael M Morgan
- Graduate Program in Neuroscience, Washington State University, Pullman, WA, United States; Department of Psychology, Washington State University Vancouver, Vancouver, WA, United States
| |
Collapse
|
36
|
Experimental Procedures for Demonstration of MicroRNA Mediated Enhancement of Functional Neuroprotective Effects of Estrogen Receptor Agonists. Methods Mol Biol 2016. [PMID: 26585150 DOI: 10.1007/978-1-4939-3127-9_29] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Protection of motoneurons is an important therapeutic goal in the treatment of neurological disorders. Recent reports have suggested that specific microRNAs (miRs) could modulate the expression of particular proteins for significant alterations in the pathogenesis of different neurological disorders. Thus, combination of overexpression of a specific neuroprotective miR and treatment with a neuroprotective agent could be a novel strategy for functional protection of motoneurons. The protocols described herein demonstrate that miR-7-1, a neuroprotective miR, can enhance the functional neuroprotective effects of estrogen receptor agonists such as 1,3,5-tris(4-hydroxyphenyl)-4-propyl-1H-pyrazole (PPT), Way 200070 (WAY), and estrogen (E2) in preventing apoptosis in A23187 calcium ionophore (CI) exposed VSC4.1 motoneurons. This article describes the protocols for the cell viability assay, transfection of VSC4.1 motoneurons with miRs, Annexin V/propidium iodide staining for apoptosis, Western blotting, patch-clamp recording of whole-cell membrane potential, and JC-1 staining for detection of mitochondrial membrane potential. Taken together, these protocols are used to demonstrate that miR-7-1 caused significant enhancement of the efficacy of estrogen receptor agonists for functional neuroprotection in VSC4.1 motoneurons.
Collapse
|
37
|
Hu HZ, Granger N, Jeffery ND. Pathophysiology, Clinical Importance, and Management of Neurogenic Lower Urinary Tract Dysfunction Caused by Suprasacral Spinal Cord Injury. J Vet Intern Med 2016; 30:1575-1588. [PMID: 27527382 PMCID: PMC5032886 DOI: 10.1111/jvim.14557] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/30/2016] [Accepted: 07/06/2016] [Indexed: 12/24/2022] Open
Abstract
Management of persistent lower urinary tract dysfunction resulting from severe thoracolumbar spinal cord injury can be challenging. Severe suprasacral spinal cord injury releases the spinal cord segmental micturition reflex from supraspinal modulation and increases nerve growth factor concentration in the bladder wall, lumbosacral spinal cord, and dorsal root ganglion, which subsequently activates hypermechanosensitive C-fiber bladder wall afferents. Hyperexcitability of bladder afferents and detrusor overactivity can cause urine leaking during the storage phase. During urine voiding, the loss of supraspinal control that normally coordinates detrusor contraction with sphincter relaxation can lead to spinal cord segmental reflex-mediated simultaneous detrusor and sphincter contractions or detrusor-sphincter dyssynergia, resulting in inefficient urine voiding and high residual volume. These disease-associated changes can impact on the quality of life and life expectancy of spinal-injured animals. Here, we discuss the pathophysiology and management considerations of lower urinary tract dysfunction as the result of severe, acute, suprasacral spinal cord injury. In addition, drawing from experimental, preclinical, and clinical medicine, we introduce some treatment options for neurogenic lower urinary tract dysfunction that are designed to: (1) prevent urine leakage arising because of detrusor overactivity during bladder filling, (2) preserve upper urinary tract integrity and function by reducing intravesical pressure and subsequent vesicoureteral reflux, and (3) prevent urinary tract and systemic complications by treating and preventing urinary tract infections.
Collapse
Affiliation(s)
- H Z Hu
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA
| | - N Granger
- School of Veterinary Sciences, University of Bristol, Langford House, Langford, North Somerset, UK
| | - N D Jeffery
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA.
| |
Collapse
|
38
|
Cragg JJ, Haefeli J, Jutzeler CR, Röhrich F, Weidner N, Saur M, Maier DD, Kalke YB, Schuld C, Curt A, Kramer JK. Effects of Pain and Pain Management on Motor Recovery of Spinal Cord–Injured Patients. Neurorehabil Neural Repair 2016; 30:753-61. [DOI: 10.1177/1545968315624777] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background. Approximately 60% of patients suffering from acute spinal cord injury (SCI) develop pain within days to weeks after injury, which ultimately persists into chronic stages. To date, the consequences of pain after SCI have been largely examined in terms of interfering with quality of life. Objective. The objective of this study was to examine the effects of pain and pain management on neurological recovery after SCI. Methods. We analyzed clinical data in a prospective multicenter observational cohort study in patients with SCI. Using mixed effects regression techniques, total motor and sensory scores were modelled at 1, 3, 6, and 12 months postinjury. Results. A total of 225 individuals were included in the study (mean age: 45.8 ± 18 years, 80% male). At 1 month postinjury, 28% of individuals with SCI reported at- or below-level neuropathic pain. While pain classification showed no effect on neurological outcomes, individuals administered anticonvulsant medications at 1 month postinjury showed significant reductions in pain intensity (2 points over 1 year; P < .05) and greater recovery in total motor scores (7.3 points over 1 year; P < .05). This drug effect on motor recovery remained significant after adjustment for injury level and injury severity, pain classification, and pain intensity. Conclusion. While initial pain classification and intensity did not reveal an effect on motor recovery following acute SCI, anticonvulsants conferred a significant beneficial effect on motor outcomes. Early intervention with anticonvulsants may have effects beyond pain management and warrant further studies to evaluate the therapeutic effectiveness in human SCI.
Collapse
Affiliation(s)
- Jacquelyn J. Cragg
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Jenny Haefeli
- Brain and Spinal Injury Center, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | | | - Frank Röhrich
- Berufsgenossenschaftliche Klinik Bergmanstrost of Halle, Halle, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Marion Saur
- Orthopädische Klinik, Hessisch Lichtenau, Germany
| | | | - Yorck B. Kalke
- RKU Universitäts und Rehabilitationskliniken Ulm, Ulm, Germany
| | - Christian Schuld
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Armin Curt
- University Hospital Balgrist, Zurich, Switzerland
| | - John K. Kramer
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
39
|
Morphine paradoxically prolongs neuropathic pain in rats by amplifying spinal NLRP3 inflammasome activation. Proc Natl Acad Sci U S A 2016; 113:E3441-50. [PMID: 27247388 DOI: 10.1073/pnas.1602070113] [Citation(s) in RCA: 283] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Opioid use for pain management has dramatically increased, with little assessment of potential pathophysiological consequences for the primary pain condition. Here, a short course of morphine, starting 10 d after injury in male rats, paradoxically and remarkably doubled the duration of chronic constriction injury (CCI)-allodynia, months after morphine ceased. No such effect of opioids on neuropathic pain has previously been reported. Using pharmacologic and genetic approaches, we discovered that the initiation and maintenance of this multimonth prolongation of neuropathic pain was mediated by a previously unidentified mechanism for spinal cord and pain-namely, morphine-induced spinal NOD-like receptor protein 3 (NLRP3) inflammasomes and associated release of interleukin-1β (IL-1β). As spinal dorsal horn microglia expressed this signaling platform, these cells were selectively inhibited in vivo after transfection with a novel Designer Receptor Exclusively Activated by Designer Drugs (DREADD). Multiday treatment with the DREADD-specific ligand clozapine-N-oxide prevented and enduringly reversed morphine-induced persistent sensitization for weeks to months after cessation of clozapine-N-oxide. These data demonstrate both the critical importance of microglia and that maintenance of chronic pain created by early exposure to opioids can be disrupted, resetting pain to normal. These data also provide strong support for the recent "two-hit hypothesis" of microglial priming, leading to exaggerated reactivity after the second challenge, documented here in the context of nerve injury followed by morphine. This study predicts that prolonged pain is an unrealized and clinically concerning consequence of the abundant use of opioids in chronic pain.
Collapse
|
40
|
Aceves M, Mathai BB, Hook MA. Evaluation of the effects of specific opioid receptor agonists in a rodent model of spinal cord injury. Spinal Cord 2016; 54:767-777. [PMID: 26927293 PMCID: PMC5009008 DOI: 10.1038/sc.2016.28] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 01/07/2016] [Accepted: 01/14/2016] [Indexed: 12/18/2022]
Abstract
Objective The current study aimed to evaluate the contribution(s) of specific
opioid receptor systems to the analgesic and detrimental effects of
morphine, observed after spinal cord injury in prior studies. Study Design We used specific opioid receptor agonists to assess the effects of
µ- (DAMGO), δ- (DPDPE), and κ- (GR89696) opioid
receptor activation on locomotor (BBB, tapered beam, ladder tests) and
sensory (girdle, tactile, and tail-flick tests) recovery in a rodent
contusion model (T12). We also tested the contribution of non-classic opioid
binding using [+]- morphine. Methods First, a dose-response curve for analgesic efficacy was generated for
each opioid agonist. Baseline locomotor and sensory reactivity was assessed
24 h after injury. Subjects were then treated with an intrathecal dose of a
specific agonist and re-tested after 30 min. To evaluate effects on
recovery, subjects were treated with a single dose of an agonist and both
locomotor and sensory function were monitored for 21 d. Results All agonists for the classic opioid receptors, but not the [+]-
morphine enantiomer, produced antinociception at a concentration equivalent
to a dose of morphine previously shown to produce strong analgesic effects
(0.32 μmol). DAMGO and [+]- morphine did not affect long-term
recovery. GR89696, however, significantly undermined recovery of locomotor
function at all doses tested. Conclusions Based on these data, we hypothesize that the analgesic efficacy of
morphine is primarily mediated by binding to the classic μ-opioid
receptor. Conversely, the adverse effects of morphine may be linked to
activation of the κ-opioid receptor. Ultimately, elucidating the
molecular mechanisms underlying the effects of morphine is imperative in
order to develop safe and effective pharmacological interventions in a
clinical setting. Setting USA
Collapse
Affiliation(s)
- M Aceves
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, USA
| | - B B Mathai
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, USA
| | - M A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, USA
| |
Collapse
|
41
|
Bespalov A, Mus L, Zvartau E. Preclinical models of muscle spasticity: valuable tools in the development of novel treatment for neurological diseases and conditions. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:457-66. [PMID: 26861550 DOI: 10.1007/s00210-016-1215-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/01/2016] [Indexed: 01/08/2023]
Abstract
Poor validity of preclinical animal models is one of the most commonly discussed explanations for the failures to develop novel drugs in general and in neuroscience in particular. However, there are several areas of neuroscience such as injury-induced spasticity where etiological factor can be adequately recreated and models can focus on specific pathophysiological mechanisms that likely contribute to spasticity syndrome in humans (such as motoneuron hyperexcitability and spinal hyperreflexia). Methods used to study spasticity in preclinical models are expected to have a high translational value (e.g., electromyogram (EMG)-based electrophysiological tools) and can efficiently assist clinical development programs. However, validation of these models is not complete yet. First, true predictive validity of these models is not established as clinically efficacious drugs have been used to reverse validate preclinical models while newly discovered mechanisms effective in preclinical models are yet to be fully explored in humans (e.g., 5-HT2C receptor inverse agonists, fatty acid amid hydrolase inhibitors). Second, further efforts need to be invested into cross-laboratory validation of study protocols and tools, adherence to the highest quality standards (blinding, randomization, pre-specified study endpoints, etc.), and systematic efforts to replicate key sets of data. These appear to be readily achievable tasks that will enable development not only of symptomatic but also of disease-modifying therapy of spasticity, an area that seems to be currently not in focus of research efforts.
Collapse
Affiliation(s)
- Anton Bespalov
- Institute of Pharmacology, Pavlov Medical University, St. Petersburg, Russia.
- Partnership for Assessment and Accreditation of Scientific Practice, Heidelberg, Germany.
| | - Liudmila Mus
- Institute of Pharmacology, Pavlov Medical University, St. Petersburg, Russia
| | - Edwin Zvartau
- Institute of Pharmacology, Pavlov Medical University, St. Petersburg, Russia
| |
Collapse
|
42
|
IL-1 receptor antagonist improves morphine and buprenorphine efficacy in a rat neuropathic pain model. Eur J Pharmacol 2015; 764:240-248. [DOI: 10.1016/j.ejphar.2015.05.058] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/27/2015] [Accepted: 05/29/2015] [Indexed: 12/11/2022]
|
43
|
Grau JW, Huie JR, Lee KH, Hoy KC, Huang YJ, Turtle JD, Strain MM, Baumbauer KM, Miranda RM, Hook MA, Ferguson AR, Garraway SM. Metaplasticity and behavior: how training and inflammation affect plastic potential within the spinal cord and recovery after injury. Front Neural Circuits 2014; 8:100. [PMID: 25249941 PMCID: PMC4157609 DOI: 10.3389/fncir.2014.00100] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 07/31/2014] [Indexed: 12/30/2022] Open
Abstract
Research has shown that spinal circuits have the capacity to adapt in response to training, nociceptive stimulation and peripheral inflammation. These changes in neural function are mediated by physiological and neurochemical systems analogous to those that support plasticity within the hippocampus (e.g., long-term potentiation and the NMDA receptor). As observed in the hippocampus, engaging spinal circuits can have a lasting impact on plastic potential, enabling or inhibiting the capacity to learn. These effects are related to the concept of metaplasticity. Behavioral paradigms are described that induce metaplastic effects within the spinal cord. Uncontrollable/unpredictable stimulation, and peripheral inflammation, induce a form of maladaptive plasticity that inhibits spinal learning. Conversely, exposure to controllable or predictable stimulation engages a form of adaptive plasticity that counters these maladaptive effects and enables learning. Adaptive plasticity is tied to an up-regulation of brain derived neurotrophic factor (BDNF). Maladaptive plasticity is linked to processes that involve kappa opioids, the metabotropic glutamate (mGlu) receptor, glia, and the cytokine tumor necrosis factor (TNF). Uncontrollable nociceptive stimulation also impairs recovery after a spinal contusion injury and fosters the development of pain (allodynia). These adverse effects are related to an up-regulation of TNF and a down-regulation of BDNF and its receptor (TrkB). In the absence of injury, brain systems quell the sensitization of spinal circuits through descending serotonergic fibers and the serotonin 1A (5HT 1A) receptor. This protective effect is blocked by surgical anesthesia. Disconnected from the brain, intracellular Cl- concentrations increase (due to a down-regulation of the cotransporter KCC2), which causes GABA to have an excitatory effect. It is suggested that BDNF has a restorative effect because it up-regulates KCC2 and re-establishes GABA-mediated inhibition.
Collapse
Affiliation(s)
- James W Grau
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - J Russell Huie
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California San Francisco San Francisco, CA, USA
| | - Kuan H Lee
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - Kevin C Hoy
- Department of Neurosciences, MetroHealth Medical Center and Case Western Reserve University Cleveland, OH, USA
| | - Yung-Jen Huang
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - Joel D Turtle
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | - Misty M Strain
- Cellular and Behavioral Neuroscience, Department of Psychology, Texas A&M University, College Station TX, USA
| | | | - Rajesh M Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center Bryan, TX, USA
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center Bryan, TX, USA
| | - Adam R Ferguson
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California San Francisco San Francisco, CA, USA
| | - Sandra M Garraway
- Department of Physiology, Emory University School of Medicine Atlanta, GA, USA
| |
Collapse
|
44
|
Woller SA, Malik JS, Aceves M, Hook MA. Morphine self-administration following spinal cord injury. J Neurotrauma 2014; 31:1570-83. [PMID: 24827476 DOI: 10.1089/neu.2013.3293] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Neuropathic pain develops in up to two-thirds of people following spinal cord injury (SCI). Opioids are among the most effective treatments for this pain and are commonly prescribed. There is concern surrounding the use of these analgesics, however, because use is often associated with the development of addiction. Previous data suggests that this concern may not be relevant in the presence of neuropathic pain. Yet, despite the common prescription of opioids for the treatment of SCI-related pain, there has been only one previous study examining the addictive potential of morphine following spinal injury. To address this, the present study used a self-administration paradigm to examine the addictive potential of morphine in a rodent model of SCI. Animals were placed into self-administration chambers 24 h, 14 d, or 35 d following a moderate spinal contusion injury. They were placed into the chambers for seven 12-hour sessions with access to 1.5 mg morphine/lever depression (up to 30 mg/d). In the acute phase of SCI, contused animals self-administered significantly less morphine than their sham counterparts, as previously shown. However, contused animals showing signs of neuropathic pain did not self-administer less morphine than their sham counterparts when administration began 14 or 35 d after injury. Instead, these animals administered nearly the full amount of morphine available each session. This amount of morphine did not affect recovery of locomotor function but did cause significant weight loss. We suggest caution is warranted when prescribing opioids for the treatment of neuropathic pain resulting from SCI, as the addictive potential is not reduced in this model.
Collapse
Affiliation(s)
- Sarah A Woller
- 1 Texas A&M University Institute for Neuroscience , Texas A&M Health Science Center, Bryan, Texas
| | | | | | | |
Collapse
|
45
|
Strickland ER, Woller SA, Hook MA, Grau JW, Miranda RC. The association between spinal cord trauma-sensitive miRNAs and pain sensitivity, and their regulation by morphine. Neurochem Int 2014; 77:40-9. [PMID: 24867772 DOI: 10.1016/j.neuint.2014.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/09/2014] [Accepted: 05/19/2014] [Indexed: 01/01/2023]
Abstract
Increased pain sensitivity is a common sequela to spinal cord injury (SCI). Moreover, drugs like morphine, though critical for pain management, elicit pro-inflammatory effects that exacerbate chronic pain symptoms. Previous reports showed that SCI results in the induction and suppression of several microRNAs (miRNAs), both at the site of injury, as well as in segments of the spinal cord distal to the injury site. We hypothesized that morphine would modulate the expression of these miRNAs, and that expression of these SCI-sensitive miRNAs may predict adaptation of distal nociceptive circuitry following SCI. To determine whether morphine treatment further dysregulates SCI-sensitive miRNAs, their expression was examined by qRT-PCR in sham controls and in response to vehicle and morphine treatment following contusion in rats, at either 2 or 15 days post-SCI. Our data indicated that expression of miR1, miR124, and miR129-2 at the injury site predicted the nociceptive response mediated by spinal regions distal to the lesion site, suggesting a molecular mechanism for the interaction of SCI with adaptation of functionally intact distal sensorimotor circuitry. Moreover, the SCI-induced miRNA, miR21 was induced by subsequent morphine administration, representing an alternate, and hitherto unidentified, maladaptive response to morphine exposure. Contrary to predictions, mRNA for the pro-inflammatory interleukin-6 receptor (IL6R), an identified target of SCI-sensitive miRNAs, was also induced following SCI, indicating dissociation between miRNA and target gene expression. Moreover, IL6R mRNA expression was inversely correlated with locomotor function suggesting that inflammation is a predictor of decreased spinal cord function. Collectively, our data indicate that miR21 and other SCI-sensitive miRNAs may constitute therapeutic targets, not only for improving functional recovery following SCI, but also for attenuating the effects of SCI on pain sensitivity.
Collapse
Affiliation(s)
- Eric R Strickland
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807-3260, USA
| | - Sarah A Woller
- Texas A&M University, Department of Psychology, College Station, TX 77843-4235, USA
| | - Michelle A Hook
- Texas A&M University, Department of Psychology, College Station, TX 77843-4235, USA
| | - James W Grau
- Texas A&M University, Department of Psychology, College Station, TX 77843-4235, USA
| | - Rajesh C Miranda
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807-3260, USA.
| |
Collapse
|
46
|
Luedtke K, Bouchard SM, Woller SA, Funk MK, Aceves M, Hook MA. Assessment of depression in a rodent model of spinal cord injury. J Neurotrauma 2014; 31:1107-21. [PMID: 24564232 DOI: 10.1089/neu.2013.3204] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Despite an increased incidence of depression in patients after spinal cord injury (SCI), there is no animal model of depression after SCI. To address this, we used a battery of established tests to assess depression after a rodent contusion injury. Subjects were acclimated to the tasks, and baseline scores were collected before SCI. Testing was conducted on days 9-10 (acute) and 19-20 (chronic) postinjury. To categorize depression, subjects' scores on each behavioral measure were averaged across the acute and chronic stages of injury and subjected to a principal component analysis. This analysis revealed a two-component structure, which explained 72.2% of between-subjects variance. The data were then analyzed with a hierarchical cluster analysis, identifying two clusters that differed significantly on the sucrose preference, open field, social exploration, and burrowing tasks. One cluster (9 of 26 subjects) displayed characteristics of depression. Using these data, a discriminant function analysis was conducted to derive an equation that could classify subjects as "depressed" on days 9-10. The discriminant function was used in a second experiment examining whether the depression-like symptoms could be reversed with the antidepressant, fluoxetine. Fluoxetine significantly decreased immobility in the forced swim test (FST) in depressed subjects identified with the equation. Subjects that were depressed and treated with saline displayed significantly increased immobility on the FST, relative to not depressed, saline-treated controls. These initial experiments validate our tests of depression, generating a powerful model system for further understanding the relationships between molecular changes induced by SCI and the development of depression.
Collapse
Affiliation(s)
- Kelsey Luedtke
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center , Bryan, Texas
| | | | | | | | | | | |
Collapse
|
47
|
Chakrabarti M, Banik NL, Ray SK. MiR-7-1 potentiated estrogen receptor agonists for functional neuroprotection in VSC4.1 motoneurons. Neuroscience 2014; 256:322-33. [PMID: 24157932 PMCID: PMC4378839 DOI: 10.1016/j.neuroscience.2013.10.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 10/11/2013] [Accepted: 10/11/2013] [Indexed: 01/28/2023]
Abstract
Protection of motoneurons is an important goal in the treatment of spinal cord injury (SCI). We tested whether neuroprotective microRNAs (miRs) like miR-206, miR-17, miR-21, miR-7-1, and miR-106a could enhance efficacy of estrogen receptor (ER) agonists such as 1,3,5-tris (4-hydroxyphenyl)-4-propyl-1H-pyrazole (PPT, ERα agonist), Way200070 (WAY, ERβ agonist), and estrogen (EST, ERα and ERβ agonist) in preventing apoptosis in the calcium ionophore (CI)-insulted ventral spinal cord 4.1 (VSC4.1) motoneurons. We determined that 200 nM CI induced 70% cell death. Treatment with 50 nM PPT, 100 nM WAY, and 150 nM EST induced overexpression of ERα, ERβ, and both receptors, respectively, at mRNA and protein levels. Treatment with ER agonists significantly upregulated miR-206, miR-17, and miR-7-1 in the CI-insulted VSC4.1 motoneurons. Transfection with miR-206, miR-17, or miR-7-1 mimic potentiated WAY or EST to inhibit apoptosis in the CI-insulted VSC4.1 motoneurons. Overexpression of miR-7-1 maximally increased efficacy of WAY and EST for down regulation of pro-apoptotic Bax and upregulation of anti-apoptotic Bcl-2. A search using microRNA database (miRDB) indicated that miR-7-1 could inhibit the expression of L-type Ca(2+) channel protein alpha 1C (CPα1C). miR-7-1 overexpression and WAY or EST treatment down regulated CPα1C but upregulated p-Akt to trigger cell survival signaling. The same therapeutic strategy increased expression of the Ca(2+)/calmodulin-dependent protein kinase II beta (CaMKIIβ) and the phosphorylated cAMP response element binding protein (p-CREB) so as to promote Bcl-2 transcription. Whole cell membrane potential and mitochondrial membrane potential studies indicated that miR-7-1 highly potentiated EST to preserve functionality in the CI-insulted VSC4.1 motoneurons. In conclusion, our data indicated that miR-7-1 most significantly potentiated efficacy of EST for functional neuroprotection and this therapeutic strategy could be used in the future to attenuate apoptosis of motoneurons in SCI.
Collapse
Affiliation(s)
- M Chakrabarti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States
| | - N L Banik
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, United States
| | - S K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States.
| |
Collapse
|
48
|
Woller SA, Hook MA. Opioid administration following spinal cord injury: implications for pain and locomotor recovery. Exp Neurol 2013; 247:328-41. [PMID: 23501709 PMCID: PMC3742731 DOI: 10.1016/j.expneurol.2013.03.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 03/04/2013] [Accepted: 03/06/2013] [Indexed: 12/18/2022]
Abstract
Approximately one-third of people with a spinal cord injury (SCI) will experience persistent neuropathic pain following injury. This pain negatively affects quality of life and is difficult to treat. Opioids are among the most effective drug treatments, and are commonly prescribed, but experimental evidence suggests that opioid treatment in the acute phase of injury can attenuate recovery of locomotor function. In fact, spinal cord injury and opioid administration share several common features (e.g. central sensitization, excitotoxicity, aberrant glial activation) that have been linked to impaired recovery of function, as well as the development of pain. Despite these effects, the interactions between opioid use and spinal cord injury have not been fully explored. A review of the literature, described here, suggests that caution is warranted when administering opioids after SCI. Opioid administration may synergistically contribute to the pathology of SCI to increase the development of pain, decrease locomotor recovery, and leave individuals at risk for infection. Considering these negative implications, it is important that guidelines are established for the use of opioids following spinal cord and other central nervous system injuries.
Collapse
Affiliation(s)
- Sarah A Woller
- Texas A&M Institute for Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843-4235, USA.
| | | |
Collapse
|
49
|
Loram LC, Grace PM, Strand KA, Taylor FR, Ellis A, Berkelhammer D, Bowlin M, Skarda B, Maier SF, Watkins LR. Prior exposure to repeated morphine potentiates mechanical allodynia induced by peripheral inflammation and neuropathy. Brain Behav Immun 2012; 26:1256-64. [PMID: 22902523 PMCID: PMC3491069 DOI: 10.1016/j.bbi.2012.08.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 08/02/2012] [Accepted: 08/03/2012] [Indexed: 01/09/2023] Open
Abstract
Opioids, such as morphine, induce potent analgesia and are the gold standard for the treatment of acute pain. However, opioids also activate glia, inducing pro-inflammatory cytokine and chemokine production, which counter-regulates the analgesic properties of classical opioid receptor activation. It is not known how long these adverse pro-inflammatory effects last or whether prior morphine could sensitize the central nervous system (CNS) such that responses to a subsequent injury/inflammation would be exacerbated. Here, multiple models of inflammation or injury were induced two days after morphine (5mg/kg b.i.d., five days , s.c.) to test the generality of morphine sensitization of later pain. Prior repeated morphine potentiated the duration of allodynia from peripheral inflammatory challenges (complete Freund's adjuvant (CFA) into either hind paw skin or masseter muscle) and from peripheral neuropathy (mild chronic constriction injury (CCI) of the sciatic nerve). Spinal cord and trigeminal nucleus caudalis mRNAs were analyzed to identify whether repeated morphine was sufficient to alter CNS expression of pro-inflammatory response genes, measured two days after cessation of treatment. Prior morphine elevated IL-1β mRNA at both sites, MHC-II and TLR4 in the trigeminal nucleus caudalis but not spinal cord, but not glial activation markers at either site. Finally, in order to identify whether morphine sensitized pro-inflammatory cytokine release, spinal cord was isolated two days after morphine dosing for five days , and slices stimulated ex vivo with lipopolysaccharide. The morphine significantly induced TNFα protein release. Therefore, repeated morphine is able to sensitize subsequent CNS responses to immune challenges.
Collapse
Affiliation(s)
- Lisa C. Loram
- Corresponding author: Lisa Loram, PhD, Department of Psychology and Neuroscience, UCB 345, University of Colorado at Boulder, Boulder, Colorado, 80309 USA, , Fax: 303-492-2967, Phone: 720-224-2860
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Temporal regularity determines the impact of electrical stimulation on tactile reactivity and response to capsaicin in spinally transected rats. Neuroscience 2012; 227:119-33. [PMID: 23036621 DOI: 10.1016/j.neuroscience.2012.09.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 09/21/2012] [Accepted: 09/22/2012] [Indexed: 01/20/2023]
Abstract
Nociceptive plasticity and central sensitization within the spinal cord depend on neurobiological mechanisms implicated in learning and memory in higher neural systems, suggesting that the factors that impact brain-mediated learning and memory could modulate how stimulation affects spinal systems. One such factor is temporal regularity (predictability). The present paper shows that intermittent hindleg shock has opposing effects in spinally transected rats depending upon whether shock is presented in a regular or irregular (variable) manner. Variable intermittent legshock (900 shocks) enhanced mechanical reactivity to von Frey stimuli (hyperreactivity), whereas 900 fixed-spaced legshocks produced hyporeactivity. The impact of fixed-spaced shock depended upon the duration of exposure; a brief exposure (36 shocks) induced hyperreactivity whereas an extended exposure (900 shocks) produced hyporeactivity. The enhanced reactivity observed after variable shock was most evident 60-180 min after treatment. Fixed and variable intermittent stimulation applied to the sciatic nerve, or the tail, yielded a similar pattern of results. Stimulation had no effect on thermal reactivity. Exposure to fixed-spaced shock, but not variable shock, attenuated the enhanced mechanical reactivity (EMR) produced by treatment with hindpaw capsaicin. The effect of fixed-spaced stimulation lasted 24h. Treatment with fixed-spaced shock also attenuated the maintenance of capsaicin-induced EMR. The results show that variable intermittent shock enhances mechanical reactivity, while an extended exposure to fixed-spaced shock has the opposite effect on mechanical reactivity and attenuates capsaicin-induced EMR.
Collapse
|