51
|
The mitochondrial calcium uniporter contributes to morphine tolerance through pCREB and CPEB1 in rat spinal cord dorsal horn. Br J Anaesth 2019; 123:e226-e238. [PMID: 31253357 DOI: 10.1016/j.bja.2019.05.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The long-term use of opioid analgesics is limited by the development of unwanted side-effects, such as tolerance. The molecular mechanisms of morphine anti-nociceptive tolerance are still unclear. The mitochondrial calcium uniporter (MCU) is involved in painful hyperalgesia, but the role of MCU in morphine tolerance has not been uncharacterised. METHODS Rats received intrathecal injection of morphine for 7 days to induce morphine tolerance. The mechanical withdrawal threshold was measured using von Frey filaments, and thermal latency using the hotplate test. The effects of an MCU inhibitor, antisense oligodeoxynucleotide against cyclic adenosine monophosphate response element (CRE)-binding protein (CREB) or cytoplasmic polyadenylation element-binding protein 1 (CPEB1) in morphine tolerance were examined. RESULTS Spinal morphine tolerance was associated with an increased expression of neuronal MCU, phospho-CREB (pCREB), and CPEB1 in the spinal cord dorsal horn. MCU inhibition increased the mechanical threshold and thermal latency, and reduced the accumulation of mitochondrial calcium in morphine tolerance. Intrathecal antisense oligodeoxynucleotide against CREB or CPEB1 restored the anti-nociceptive effects of morphine compared with mismatch oligodeoxynucleotide in von Frey test and hotplate test. Chromatin immunoprecipitation with quantitative PCR assay showed that CREB knockdown reduced the interaction of pCREB with the ccdc109a gene (encoding MCU expression) promoter and decreased the MCU mRNA transcription. RNA immunoprecipitation assay suggested that CPEB1 binds to the MCU mRNA 3' untranslated region. CPEB1 knockdown decreased the expression of MCU protein. CONCLUSIONS These findings suggest that spinal MCU is regulated by pCREB and CPEB1 in morphine tolerance, and that inhibition of MCU, pCREB, or CPEB1 may be useful in preventing the development of opioid tolerance.
Collapse
|
52
|
Sellas MN, Kyllonen KC, Lepak MR, Rodriguez RJ. Dexmedetomidine for the Management of Postoperative Pain and Sedation in Newborns. J Pediatr Pharmacol Ther 2019; 24:227-233. [PMID: 31093022 PMCID: PMC6510519 DOI: 10.5863/1551-6776-24.3.227] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2018] [Indexed: 11/11/2022]
Abstract
BACKGROUND Opioids and benzodiazepines have been the mainstay of neonatal analgesia and sedation. However, based on evidence in neonatal animals, these drugs may be deleterious for the developing brain. Dexmedetomidine (DEX), a central alpha-2 agonist, has sedative and analgesic effects and has been shown to be neuroprotective in animal models. Despite increasing use of DEX in newborns, there is a paucity of data regarding its safety and efficacy in this population. OBJECTIVES The impact of using DEX in postsurgical neonates, either alone or with opioid infusions, for sedation/analgesia was evaluated. The cumulative dose of opioids among patients who did or did not receive DEX was calculated to examine the hypothesis that the addition of DEX can reduce the patient exposure to opioids without significantly increasing side effects and providing adequate sedation and pain control. METHODOLOGY This was a retrospective cohort study in which patients were matched by postnatal age and surgical procedure into 2 groups. One group received DEX in the regimen for treatment of pain or sedation after a surgical procedure, and the other group received no DEX. Episodes of bradycardia, respiratory depression and hypotension, and the cumulative dose of opioids and number of supplemental doses administered in both groups were documented. RESULTS Although there was no difference in gestational age or weight at birth between the DEX and no-DEX groups, the DEX group's median postconceptional date was older at the time of surgery (39.6 vs 37.4 weeks; p = 0.003). Patients in the DEX group experienced more episodes of bradycardia (12.8% vs 5.1%; p = 0.01). There was no difference between groups in episodes of hypotension or respiratory depression. The cumulative dose of opioids was significantly lower in the DEX group compared with the no-DEX group (1155 mcg/kg vs 1841 mcg/kg; p = 0.01). There was no difference in the number of supplemental doses of opioids given between the groups. CONCLUSIONS The addition of DEX to opioid infusions resulted in a significant decrease in the cumulative dose of opioids but was associated with more episodes of bradycardia than opioids alone.
Collapse
|
53
|
Spinal bromodomain-containing protein 4 contributes to neuropathic pain induced by HIV glycoprotein 120 with morphine in rats. Neuroreport 2019; 29:441-446. [PMID: 29465625 DOI: 10.1097/wnr.0000000000000992] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The symptoms of HIV-sensory neuropathy are dominated by neuropathic pain. Recent data show that repeated use of opiates enhances the chronic pain states in HIV patients. Limited attention has so far been devoted to exploring the exact pathogenesis of HIV painful disorder and opiate abuse in vivo, for which there is no effective treatment. Bromodomain-containing protein 4 (Brd4) is a member of the bromodomain and extraterminal domain protein (BET) family and functions as a chromatin 'reader' that binds acetylated lysines in histones in brain neurons to mediate the transcriptional regulation underlying learning and memory. Here, we established a neuropathic pain model of interaction of intrathecal HIV envelope glycoprotein 120 (gp120) and chronic morphine in rats. The combination of gp120 and morphine (gp120/M, for 5 days) induced persistent mechanical allodynia compared with either gp120 or morphine alone. Mechanical allodynia reached the lowest values at day 10 from gp120/M application, beginning to recover from day 21. In the model, gp120/M induced overexpression of Brd4 mRNA and protein at day 10 using RT-qPCR and western blots, respectively. Immunohistochemical studies showed that Brd4 at day 10 was expressed in the neurons of spinal cord dorsal horn. BET inhibitor I-BET762 dose-dependently increased the mechanical threshold in the gp120/M pain state. The present study provides preclinical evidence for treating HIV neuropathic pain with opioids using the BET inhibitor.
Collapse
|
54
|
Chau CMY, Ross CJD, Chau V, Synnes AR, Miller SP, Carleton B, Grunau RE. Morphine biotransformation genes and neonatal clinical factors predicted behaviour problems in very preterm children at 18 months. EBioMedicine 2019; 40:655-662. [PMID: 30709768 PMCID: PMC6413679 DOI: 10.1016/j.ebiom.2019.01.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/21/2019] [Accepted: 01/22/2019] [Indexed: 01/18/2023] Open
Abstract
Background Behaviour problems are prevalent among children born very preterm (≤ 32 weeks gestation), and have been associated with morphine exposure. Morphine accumulation in the brain is determined by genetic variations related to morphine biotransformation. The objective of the study was to investigate whether morphine-biotransformation genotypes contribute to individual differences in long-term effects of morphine on behaviour at 18 months corrected age (CA). Methods 198 children born very preterm (24–32 weeks gestation) were followed from birth and seen at 18 months CA. Relationships between child behavior (Internalizing, Externalizing on the Child Behavior Checklist), morphine exposure, neonatal clinical variables, and morphine biotransformation gene variants in ABCB1, UGT1A9, UGT 2B7*2, ABCC2, ABCC3, SLCO1B1, CYP3A4, COMT were examined. Findings Neonatal clinical predictors and genotypes accounted for 39% of the overall variance in behaviour. In children with the minor allele of UGT1A9 rs17863783 (marker of UGT1A6*4, UDP-glucuronosyltransferase), greater morphine exposure (p = ·0011) was associated with more Internalizing behaviour. More Externalizing behaviour was predicted by greater morphine exposure in children with the COMT rs4680 Met/Met genotype (p = ·0006). Interpretation Genetic variations that affect relative accumulation of morphine in the brain, together with neonatal clinical factors, are differentially related to anxiety and depressive symptoms (internalizing) and to acting out (externalizing) behaviours at 18 months CA in children born very preterm. Fund NIH/NICHD HD039783 (REG); CIHR MOP86489 (REG), MOP68898 (SPM), MOP79262 (SPM, REG).
Collapse
Affiliation(s)
- Cecil M Y Chau
- BC Children's Hospital Research Institute, Vancouver, Canada; Pediatrics, University of British Columbia, Vancouver, Canada
| | - Colin J D Ross
- BC Children's Hospital Research Institute, Vancouver, Canada; Pediatrics, University of British Columbia, Vancouver, Canada
| | - Vann Chau
- Neurology, The Hospital for Sick Children, Toronto, Canada; Paediatrics, University of Toronto, Toronto, Canada
| | - Anne R Synnes
- BC Children's Hospital Research Institute, Vancouver, Canada; Pediatrics, University of British Columbia, Vancouver, Canada
| | - Steven P Miller
- Neurology, The Hospital for Sick Children, Toronto, Canada; Paediatrics, University of Toronto, Toronto, Canada
| | - Bruce Carleton
- BC Children's Hospital Research Institute, Vancouver, Canada; Pediatrics, University of British Columbia, Vancouver, Canada
| | - Ruth E Grunau
- BC Children's Hospital Research Institute, Vancouver, Canada; Pediatrics, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
55
|
Butler PM, Barash JA, Casaletto KB, Cotter DL, Joie RL, Geschwind MD, Rosen HJ, Kramer JH, Miller BL. An Opioid-Related Amnestic Syndrome With Persistent Effects on Hippocampal Structure and Function. J Neuropsychiatry Clin Neurosci 2019; 31:392-396. [PMID: 31177905 PMCID: PMC7469957 DOI: 10.1176/appi.neuropsych.19010017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- P. Monroe Butler
- The Department of Neurology, University of California at San Francisco
| | - Jed A. Barash
- The Department of Neurology, University of California at San Francisco and Soldiers’ Home, Chelsea, Mass
| | | | - Devyn L. Cotter
- The Department of Neurology, University of California at San Francisco
| | - Renaud La Joie
- The Department of Neurology, University of California at San Francisco
| | | | - Howie J. Rosen
- The Department of Neurology, University of California at San Francisco
| | - Joel H. Kramer
- The Department of Neurology, University of California at San Francisco
| | - Bruce L. Miller
- The Department of Neurology, University of California at San Francisco
| |
Collapse
|
56
|
The Protective and Restorative Effects of Growth Hormone and Insulin-Like Growth Factor-1 on Methadone-Induced Toxicity In Vitro. Int J Mol Sci 2018; 19:ijms19113627. [PMID: 30453639 PMCID: PMC6274959 DOI: 10.3390/ijms19113627] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 11/17/2022] Open
Abstract
Evidence to date suggests that opioids such as methadone may be associated with cognitive impairment. Growth hormone (GH) and insulin-like growth factor-1 (IGF-1) are suggested to be neuroprotective and procognitive in the brain and may therefore counteract these effects. This study aims to explore the protective and restorative effects of GH and IGF-1 in methadone-treated cell cultures. Primary cortical cell cultures were harvested from rat fetuses and grown for seven days in vitro. To examine the protective effects, methadone was co-treated with or without GH or IGF-1 for three consecutive days. To examine the restorative effects, methadone was added for the first 24 h, washed, and later treated with GH or IGF-1 for 48 h. At the end of each experiment, mitochondrial function and membrane integrity were evaluated. The results revealed that GH had protective effects in the membrane integrity assay and that both GH and IGF-1 effectively recovered mitochondrial function and membrane integrity in cells pretreated with methadone. The overall conclusion of the present study is that GH, but not IGF-1, protects primary cortical cells against methadone-induced toxicity, and that both GH and IGF-1 have a restorative effect on cells pretreated with methadone.
Collapse
|
57
|
Knutsen HK, Alexander J, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Hoogenboom LR, Nebbia CS, Oswald IP, Petersen A, Rose M, Roudot AC, Schwerdtle T, Vollmer G, Wallace H, Benford D, Calò G, Dahan A, Dusemund B, Mulder P, Németh-Zámboriné É, Arcella D, Baert K, Cascio C, Levorato S, Schutte M, Vleminckx C. Update of the Scientific Opinion on opium alkaloids in poppy seeds. EFSA J 2018; 16:e05243. [PMID: 32625895 PMCID: PMC7009406 DOI: 10.2903/j.efsa.2018.5243] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Poppy seeds are obtained from the opium poppy (Papaver somniferum L.). They are used as food and to produce edible oil. The opium poppy plant contains narcotic alkaloids such as morphine and codeine. Poppy seeds do not contain the opium alkaloids, but can become contaminated with alkaloids as a result of pest damage and during harvesting. The European Commission asked EFSA to provide an update of the Scientific Opinion on opium alkaloids in poppy seeds. The assessment is based on data on morphine, codeine, thebaine, oripavine, noscapine and papaverine in poppy seed samples. The CONTAM Panel confirms the acute reference dose (ARfD) of 10 μg morphine/kg body weight (bw) and concluded that the concentration of codeine in the poppy seed samples should be taken into account by converting codeine to morphine equivalents, using a factor of 0.2. The ARfD is therefore a group ARfD for morphine and codeine, expressed in morphine equivalents. Mean and high levels of dietary exposure to morphine equivalents from poppy seeds considered to have high levels of opium alkaloids (i.e. poppy seeds from varieties primarily grown for pharmaceutical use) exceed the ARfD in most age groups. For poppy seeds considered to have relatively low concentrations of opium alkaloids (i.e. primarily varieties for food use), some exceedance of the ARfD is also seen at high levels of dietary exposure in most surveys. For noscapine and papaverine, the available data do not allow making a hazard characterisation. However, comparison of the dietary exposure to the recommended therapeutical doses does not suggest a health concern for these alkaloids. For thebaine and oripavine, no risk characterisation was done due to insufficient data. However, for thebaine, limited evidence indicates a higher acute lethality than for morphine and the estimated exposure could present a health risk.
Collapse
|
58
|
Adedayo AD, Aderinola AA, Adekilekun TA, Olaolu OO, Olanike AM, Olayemi IK. Morphine-alcohol treatment impairs cognitive functions and increases neuro-inflammatory responses in the medial prefrontal cortex of juvenile male rats. Anat Cell Biol 2018; 51:41-51. [PMID: 29644109 PMCID: PMC5890016 DOI: 10.5115/acb.2018.51.1.41] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/04/2017] [Accepted: 12/13/2017] [Indexed: 02/01/2023] Open
Abstract
In the developed and developing world, opioid consumption in combination with alcohol has become one of the substances abused. In this experiment, we examined the effects of alcohol, morphine, and morphine+alcohol combination on cognitive functions and neuroinflammatory responses in the medial prefrontal cortex (mPFC) of juvenile male rats. Alcohol (1.0 ml of 15% v/v ethanol twice daily, subcutaneously, 7 hours apart), morphine (0.5 ml/kg of 0.4 mg/kg morphine chlorate twice daily, subcutaneously, 7 hours apart), morphine+alcohol co-treatment (0.5 ml/kg of 0.4 mg/kg morphine chlorate+1.0 ml of 15% v/v ethanol twice daily, subcutaneously, 7 hours apart) were administered for 21 days. Treatment with morphine+alcohol significantly impairs cognition functions in the Morris water maze, passive avoidance, and novel object recognition tests, furthermore, the treatment significantly increased the quantitative count of astrocytic cells and also conferred marked neuronal cell death in the mPFC, which were studied by glial fibrillary acidic protein immunochemistry for astrocytes and Cresyl violet for Nissl's substance distribution in neurons respectively. These results suggest that alcohol, morphine, and morphine+alcohol co-treatment may trigger cognitive deficits and neuroinflammatory responses in the brain.
Collapse
Affiliation(s)
| | | | | | - Olaniyan Olayinka Olaolu
- Department of Medical Biochemistry (Chemical Pathology Unit), Osun State University (Osogbo Campus), Osogbo, Nigeria
| | | | - Ijomone Kafilat Olayemi
- Department of Anatomy, Faculty of Basic Medical Science, University of Medical Sciences, Ondo, Nigeria
| |
Collapse
|
59
|
Fan R, Schrott LM, Arnold T, Snelling S, Rao M, Graham D, Cornelius A, Korneeva NL. Chronic oxycodone induces axonal degeneration in rat brain. BMC Neurosci 2018; 19:15. [PMID: 29571287 PMCID: PMC5865283 DOI: 10.1186/s12868-018-0417-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 03/17/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Chronic opioid therapy for non-malignant pain conditions has significantly increased over the last 15 years. Recently, the correlation between opioid analgesics and alternations in brain structure, such as leukoencephalopathy, axon demyelination, and white matter lesions, has been demonstrated in patients with a history of long-term use of prescription opioids. The exact mechanisms underlying the neurotoxic effect of opioids on the central nervous system are still not fully understood. We investigated the effect of chronic opioids using an animal model in which female rats were orally gavaged with 15 mg/kg of oxycodone every 24 h for 30 days. In addition we tested oxycodone, morphine and DAMGO in breast adenocarcinoma MCF7 cells, which are known to express the μ-opioid receptor. RESULTS We observed several changes in the white matter of animals treated with oxycodone: deformation of axonal tracks, reduction in size of axonal fascicles, loss of myelin basic protein and accumulation of amyloid precursor protein beta (β-APP), suggesting axonal damages by chronic oxycodone. Moreover, we demonstrated activation of pro-apoptotic machinery amid suppression of anti-apoptotic signaling in axonal tracks that correlated with activation of biomarkers of the integrated stress response (ISR) in these structures after oxycodone exposure. Using MCF7 cells, we observed induction of the ISR and pro-apoptotic signaling after opioid treatment. We showed that the ISR inhibitor, ISRIB, suppresses opioid-induced Bax and CHOP expression in MCF7 cells. CONCLUSIONS Altogether, our data suggest that chronic opioid administration may cause neuronal degeneration by activation of the integrated stress response leading to induction of apoptotic signaling in neurons and also by promoting demyelination in CNS.
Collapse
Affiliation(s)
- Ruping Fan
- Department of Emergency Medicine, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, USA
| | - Lisa M. Schrott
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, USA
| | - Thomas Arnold
- Department of Emergency Medicine, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, USA
| | - Stephen Snelling
- Department of Emergency Medicine, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, USA
| | - Meghana Rao
- Department of Emergency Medicine, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, USA
| | - Derrel Graham
- Department of Emergency Medicine, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, USA
| | - Angela Cornelius
- Department of Emergency Medicine, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, USA
| | - Nadejda L. Korneeva
- Department of Emergency Medicine, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, USA
- Department of Pharmacology, Toxicology, and Neuroscience, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, USA
| |
Collapse
|
60
|
Karkhah A, Ataee R, Ataie A. Morphine pre- and post-conditioning exacerbates apoptosis in rat hippocampus cells in a model of homocysteine-induced oxidative stress. Biomed Rep 2017; 7:309-313. [PMID: 28928969 DOI: 10.3892/br.2017.962] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/23/2017] [Indexed: 11/06/2022] Open
Abstract
Recent investigations indicated that morphine has protective effects in different ischemia/reperfusion models and may protect against neuronal cell death, while other evidence showed that morphine induces apoptosis in neurons. Therefore, the current study was conducted to investigate pre- and post-conditioning effects of morphine on hippocampal cell apoptosis in a rat model of homocysteine (Hcy)-induced oxidative stress. In the present study, 0.5 µmol/µl Hcy was injected into bilateral intrahipocampal in the rat brain and morphine at a therapeutic dose of 10 mg/kg was injected intraperitoneally 5 days before and after Hcy injection in rats. The left and right rat hippocampus were removed for biochemical and histopathological analysis. In addition, hippocampal cell apoptosis was assayed by the TUNEL kit. Our results indicated that malondialdehyde (MDA) and superoxide anion (SOA) levels in the Hcy group were increased significantly compared to the control group (P<0.001). In addition, morphine pre- and post-treatment increased the MDA and SOA levels significantly in rat hippocampus compared with other groups (P<0.001). It was found that Hcy alone induced apoptosis in hippocampus cells and significantly increased the number of TUNEL-positive cells in rat hippocampus compared to the other group (P<0.001). Notably, our results indicated that pre- and post-treatment by morphine increased apoptosis in hippocampus cells compared with the other group (P<0.001). In conclusion, morphine neuroprotection and neurotoxicity needs to be further investigated to determine morphine side-effects in medical applications and to identify new targets for potential therapies.
Collapse
Affiliation(s)
- Ahmad Karkhah
- Neuroscience Research Center, Babol University of Medical Sciences, Babol, Mazandaran, Iran.,Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Mazandaran, Iran
| | - Ramin Ataee
- Pharmaceutical Sciences Research Center, Department of Pharmacology and Toxicology, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran
| | - Amin Ataie
- Neuroscience Research Center, Babol University of Medical Sciences, Babol, Mazandaran, Iran
| |
Collapse
|
61
|
Lack of association between two genetic polymorphisms of SOD2 (rs2758339 and rs5746136) and the risk of opium dependency. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.poamed.2017.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
62
|
Bodzon-Kulakowska A, Antolak A, Drabik A, Marszalek-Grabska M, Kotlińska J, Suder P. Brain lipidomic changes after morphine, cocaine and amphetamine administration — DESI — MS imaging study. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:686-691. [DOI: 10.1016/j.bbalip.2017.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 12/28/2022]
|
63
|
Hauser KF, Knapp PE. Opiate Drugs with Abuse Liability Hijack the Endogenous Opioid System to Disrupt Neuronal and Glial Maturation in the Central Nervous System. Front Pediatr 2017; 5:294. [PMID: 29410949 PMCID: PMC5787058 DOI: 10.3389/fped.2017.00294] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/20/2017] [Indexed: 01/19/2023] Open
Abstract
The endogenous opioid system, comprised of multiple opioid neuropeptide and receptor gene families, is highly expressed by developing neural cells and can significantly influence neuronal and glial maturation. In many central nervous system (CNS) regions, the expression of opioid peptides and receptors occurs only transiently during development, effectively disappearing with subsequent maturation only to reemerge under pathologic conditions, such as with inflammation or injury. Opiate drugs with abuse liability act to modify growth and development by mimicking the actions of endogenous opioids. Although typically mediated by μ-opioid receptors, opiate drugs can also act through δ- and κ-opioid receptors to modulate growth in a cell-type, region-specific, and developmentally regulated manner. Opioids act as biological response modifiers and their actions are highly contextual, plastic, modifiable, and influenced by other physiological processes or pathophysiological conditions, such as neuro-acquired immunodeficiency syndrome. To date, most studies have considered the acute effects of opiates on cellular maturation. For example, activating opioid receptors typically results in acute growth inhibition in both neurons and glia. However, with sustained opioid exposure, compensatory factors become operative, a concept that has been largely overlooked during CNS maturation. Accordingly, this article surveys prior studies on the effects of opiates on CNS maturation, and also suggests new directions for future research in this area. Identifying the cellular and molecular mechanisms underlying the adaptive responses to chronic opiate exposure (e.g., tolerance) during maturation is crucial toward understanding the consequences of perinatal opiate exposure on the CNS.
Collapse
Affiliation(s)
- Kurt F Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.,Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.,Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Pamela E Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.,Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.,Institute for Drug and Alcohol Studies, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| |
Collapse
|
64
|
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
|
65
|
Zhou J, Lin W, Chen H, Fan Y, Yang C. TRESK contributes to pain threshold changes by mediating apoptosis via MAPK pathway in the spinal cord. Neuroscience 2016; 339:622-633. [PMID: 27789381 DOI: 10.1016/j.neuroscience.2016.10.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/10/2016] [Accepted: 10/17/2016] [Indexed: 01/05/2023]
Abstract
The mechanism underlying neuropathic pain (NP) is complex and has not been fully elucidated. The TWIK-related spinal cord K+ (TRESK) is the major background potassium current in dorsal root ganglia (DRG), we found that mitogen-activated protein kinase (MAPK) signal pathway were activated in spinal cord accompanied by TRESK down regulation in response to NP. Therefore, we investigated whether TRESK mediates inflammation and apoptosis by MAPK pathway in the spinal cord of NP rats. SNI rats exhibited reduced TRESK expression in DRG and spinal cord and higher sensitivity to mechanical stimuli but no effect on thermal stimuli. Intrathecal injections of TRESK overexpressing adenovirus alleviated mechanical allodynia, inhibited phosphorylation of extracellular signal-regulated kinase (ERK) and p38, and decreased inflammatory reactions and apoptosis in the spinal cords of SNI rats. Down regulation of TRESK in DRG and spinal cord was detected in normal rats after intrathecal TRESK shRNA lentivirus injection, which induced mechanical allodynia but had no effect on pain thresholds for heat stimulation. Phosphorylated ERK and p38 were increased in the spinal cord. Intrathecal injection of an ERK antagonist (PD98059) and p38 antagonist (SB203580) prevented ERK and p38 activation in the spinal cord and mechanical allodynia induced by TRESK shRNA lentivirus. In conclusion, our study clearly demonstrated an important role for TRESK in NP and that TRESK regulation contributes to pain sensitivity mediates inflammation and apoptosis by ERK and p38 MAPK signaling in the spinal cord.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Anesthesiology, First People's Hospital of Foshan, Foshan, Guangdong Province 528000, China.
| | - Wenjing Lin
- Department of Anesthesiology, First People's Hospital of Foshan, Foshan, Guangdong Province 528000, China
| | - Hongtao Chen
- Department of Anesthesiology, Eighth People's Hospital of Guangzhou, Guangzhou, Guangdong Province 510060, China
| | - Youling Fan
- Department of Anesthesiology, Central Hospital of Panyu District, Guangzhou 511400, Guangdong Province, China
| | - Chengxiang Yang
- Department of Anesthesiology, First People's Hospital of Foshan, Foshan, Guangdong Province 528000, China
| |
Collapse
|
66
|
Nylander E, Grönbladh A, Zelleroth S, Diwakarla S, Nyberg F, Hallberg M. Growth hormone is protective against acute methadone-induced toxicity by modulating the NMDA receptor complex. Neuroscience 2016; 339:538-547. [PMID: 27746341 DOI: 10.1016/j.neuroscience.2016.10.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/12/2016] [Accepted: 10/03/2016] [Indexed: 01/13/2023]
Abstract
Human growth hormone (GH) displays promising protective effects in the central nervous system after damage caused by various insults. Current evidence suggests that these effects may involve N-methyl-d-aspartate (NMDA) receptor function, a receptor that also is believed to play a role in opioid-induced neurotoxicity. The aims of the present study were to examine the acute toxic effects of methadone, an opioid receptor agonist and NMDA receptor antagonist, as well as to evaluate the protective properties of recombinant human GH (rhGH) on methadone-induced toxicity. Primary cortical cell cultures from embryonic day 17 rats were grown for 7days in vitro. Cells were treated with methadone for 24h and the 50% lethal dose was calculated and later used for protection studies with rhGH. Cellular toxicity was determined by measuring mitochondrial activity, lactate dehydrogenase release, and caspase activation. Furthermore, the mRNA expression levels of NMDA receptor subunits were investigated following methadone and rhGH treatment using quantitative PCR (qPCR) analysis. A significant protective effect was observed with rhGH treatment on methadone-induced mitochondrial dysfunction and in methadone-induced LDH release. Furthermore, methadone significantly increased caspase-3 and -7 activation but rhGH was unable to inhibit this effect. The mRNA expression of the NMDA receptor subunit GluN1, GluN2a, and GluN2b increased following methadone treatment, as assessed by qPCR, and rhGH treatment effectively normalized this expression to control levels. We have demonstrated that rhGH can rescue cells from methadone-induced toxicity by maintaining mitochondrial function, cellular integrity, and NMDA receptor complex expression.
Collapse
Affiliation(s)
- Erik Nylander
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, SE-751 24, Uppsala University, Sweden.
| | - Alfhild Grönbladh
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, SE-751 24, Uppsala University, Sweden
| | - Sofia Zelleroth
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, SE-751 24, Uppsala University, Sweden
| | - Shanti Diwakarla
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, SE-751 24, Uppsala University, Sweden
| | - Fred Nyberg
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, SE-751 24, Uppsala University, Sweden
| | - Mathias Hallberg
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, SE-751 24, Uppsala University, Sweden
| |
Collapse
|
67
|
Abstract
Commonly used medications can have neuropsychiatric and behavioral effects that may be idiosyncratic or metabolic in nature, or a function of interactions with other drugs, toxicity, or withdrawal. This article explores an approach to the patient with central nervous system toxicity, depending on presentation of sedation versus agitation and accompanying physical signs and symptoms. The effects of antihypertensives, opioids, antibiotics, antiepileptic agents, steroids, Parkinson's disease medications, antipsychotics, medications for human immunodeficiency virus infection, cancer chemotherapeutics, and immunotherapies are discussed. A look at the prevalence of adverse reactions to medications and the errors underlying such occurrences is included.
Collapse
Affiliation(s)
- Sai Krishna J Munjampalli
- Department of Neurology, Louisiana State University Health Sciences Center - Shreveport, 1501 Kings Highway, Shreveport, LA 71103, USA
| | - Debra E Davis
- Department of Neurology, Louisiana State University Health Sciences Center - Shreveport, 1501 Kings Highway, Shreveport, LA 71103, USA.
| |
Collapse
|
68
|
Cao L, Walker MP, Vaidya NK, Fu M, Kumar S, Kumar A. Cocaine-Mediated Autophagy in Astrocytes Involves Sigma 1 Receptor, PI3K, mTOR, Atg5/7, Beclin-1 and Induces Type II Programed Cell Death. Mol Neurobiol 2016; 53:4417-30. [PMID: 26243186 PMCID: PMC4744147 DOI: 10.1007/s12035-015-9377-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/27/2015] [Indexed: 12/15/2022]
Abstract
Cocaine, a commonly used drug of abuse, has been shown to cause neuropathological dysfunction and damage in the human brain. However, the role of autophagy in this process is not defined. Autophagy, generally protective in nature, can also be destructive leading to autophagic cell death. This study was designed to investigate whether cocaine induces autophagy in the cells of CNS origin. We employed astrocyte, the most abundant cell in the CNS, to define the effects of cocaine on autophagy. We measured levels of the autophagic marker protein LC3II in SVGA astrocytes after exposure with cocaine. The results showed that cocaine caused an increase in LC3II level in a dose- and time-dependent manner, with the peak observed at 1 mM cocaine after 6-h exposure. This result was also confirmed by detecting LC3II in SVGA astrocytes using confocal microscopy and transmission electron microscopy. Next, we sought to explore the mechanism by which cocaine induces the autophagic response. We found that cocaine-induced autophagy was mediated by sigma 1 receptor, and autophagy signaling proteins p-mTOR, Atg5, Atg7, and p-Bcl-2/Beclin-1 were also involved, and this was confirmed by using selective inhibitors and small interfering RNAs (siRNAs). In addition, we found that chronic treatment with cocaine resulted in cell death, which is caspase-3 independent and can be ameliorated by autophagy inhibitor. Therefore, this study demonstrated that cocaine induces autophagy in astrocytes and is associated with autophagic cell death.
Collapse
Affiliation(s)
- Lu Cao
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA
| | - Mary P Walker
- Department of Oral and Craniofacial Sciences, School of Dentistry Center of Excellence in Musculoskeletal and Dental Tissues, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| | - Naveen K Vaidya
- Department of Mathematics and Statistics, University of Missouri, Kansas City, MO, 64110, USA
| | - Mingui Fu
- Department of Basic Medical Science, School of Medicine, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Anil Kumar
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, 64108, USA.
| |
Collapse
|
69
|
Hua Z, Liu L, Shen J, Cheng K, Liu A, Yang J, Wang L, Qu T, Yang H, Li Y, Wu H, Narouze J, Yin Y, Cheng J. Mesenchymal Stem Cells Reversed Morphine Tolerance and Opioid-induced Hyperalgesia. Sci Rep 2016; 6:32096. [PMID: 27554341 PMCID: PMC4995471 DOI: 10.1038/srep32096] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 08/02/2016] [Indexed: 01/27/2023] Open
Abstract
More than 240 million opioid prescriptions are dispensed annually to treat pain in the US. The use of opioids is commonly associated with opioid tolerance (OT) and opioid-induced hyperalgesia (OIH), which limit efficacy and compromise safety. The dearth of effective way to prevent or treat OT and OIH is a major medical challenge. We hypothesized that mesenchymal stem cells (MSCs) attenuate OT and OIH in rats and mice based on the understanding that MSCs possess remarkable anti-inflammatory properties and that both OT and chronic pain are associated with neuroinflammation in the spinal cord. We found that the development of OT and OIH was effectively prevented by either intravenous or intrathecal MSC transplantation (MSC-TP), which was performed before morphine treatment. Remarkably, established OT and OIH were significantly reversed by either intravenous or intrathecal MSCs when cells were transplanted after repeated morphine injections. The animals did not show any abnormality in vital organs or functions. Immunohistochemistry revealed that the treatments significantly reduced activation level of microglia and astrocytes in the spinal cord. We have thus demonstrated that MSC-TP promises to be a potentially safe and effective way to prevent and reverse two of the major problems of opioid therapy.
Collapse
Affiliation(s)
- Zhen Hua
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
- Department of Anesthesiology, Beijing Hospital, No. 1 Dahua Road, Beijing 100730, China
| | - LiPing Liu
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Jun Shen
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Katherine Cheng
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Aijun Liu
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Jing Yang
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Lina Wang
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Tingyu Qu
- Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - HongNa Yang
- Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Yan Li
- Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Haiyan Wu
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - John Narouze
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Yan Yin
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Jianguo Cheng
- Departments of Pain Management and Neurosciences, Lerner Research Institute and Anaesthesiology Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| |
Collapse
|
70
|
Creeley CE. From Drug-Induced Developmental Neuroapoptosis to Pediatric Anesthetic Neurotoxicity-Where Are We Now? Brain Sci 2016; 6:brainsci6030032. [PMID: 27537919 PMCID: PMC5039461 DOI: 10.3390/brainsci6030032] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/05/2016] [Accepted: 08/12/2016] [Indexed: 01/02/2023] Open
Abstract
The fetal and neonatal periods are critical and sensitive periods for neurodevelopment, and involve rapid brain growth in addition to natural programmed cell death (i.e., apoptosis) and synaptic pruning. Apoptosis is an important process for neurodevelopment, preventing redundant, faulty, or unused neurons from cluttering the developing brain. However, animal studies have shown massive neuronal cell death by apoptosis can also be caused by exposure to several classes of drugs, namely gamma-aminobutyric acid (GABA) agonists and N-methyl-d-aspartate (NMDA) antagonists that are commonly used in pediatric anesthesia. This form of neurotoxic insult could cause a major disruption in brain development with the potential to permanently shape behavior and cognitive ability. Evidence does suggest that psychoactive drugs alter neurodevelopment and synaptic plasticity in the animal brain, which, in the human brain, may translate to permanent neurodevelopmental changes associated with long-term intellectual disability. This paper reviews the seminal animal research on drug-induced developmental apoptosis and the subsequent clinical studies that have been conducted thus far. In humans, there is growing evidence that suggests anesthetics have the potential to harm the developing brain, but the long-term outcome is not definitive and causality has not been determined. The consensus is that there is more work to be done using both animal models and human clinical studies.
Collapse
Affiliation(s)
- Catherine E Creeley
- Department of Psychology, State University of New York at Fredonia, Fredonia, NY 14063, USA.
| |
Collapse
|
71
|
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
|
72
|
Liu B, Liu X, Tang SJ. Interactions of Opioids and HIV Infection in the Pathogenesis of Chronic Pain. Front Microbiol 2016; 7:103. [PMID: 26903982 PMCID: PMC4748029 DOI: 10.3389/fmicb.2016.00103] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/19/2016] [Indexed: 12/30/2022] Open
Abstract
Over 50% of HIV-1/AIDS patients suffer chronic pain. Currently, opioids are the cornerstone medications for treating severe pain in these patients. Ironically, emerging clinical data indicates that repeated use of opiate pain medicines might in fact heighten the chronic pain states in HIV patients. Both laboratory-based and clinical studies strongly suggest that opioids exacerbate the detrimental effects of HIV-1 infection on the nervous system, both on neurons and glia. The combination of opioids and HIV-1infection may promote the damage of neurons, including those in the pain sensory and transmission pathway, by activating both caspase-dependent and caspase-independent pro-apoptotic pathways. In addition, the opiate-HIV-1 interaction may also cause widespread disturbance of glial function and elicit glial-derived pro-inflammatory responses that dysregulate neuronal function. The deregulation of neuron-glia cross-talk that occurs with the combination of HIV-1 and opioids appears to play an important role in the development of the pathological pain state. In this article, we wish to provide an overview of the potential molecular and cellular mechanisms by which opioids may interact with HIV-1 to cause neurological problems, especially in the context of HIV-associated pathological pain. Elucidating the underlying mechanisms will help researchers and clinicians to understand how chronic use of opioids for analgesia enhances HIV-associated pain. It will also assist in optimizing therapeutic approaches to prevent or minimize this significant side effect of opiate analgesics in pain management for HIV patients.
Collapse
Affiliation(s)
- Bolong Liu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, GalvestonTX, USA; Department of Urology, Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, China
| | - Xin Liu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston TX, USA
| | - Shao-Jun Tang
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston TX, USA
| |
Collapse
|
73
|
Morphine Inhibited the Rat Neural Stem Cell Proliferation Rate by Increasing Neuro Steroid Genesis. Neurochem Res 2016; 41:1410-9. [DOI: 10.1007/s11064-016-1847-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 12/28/2015] [Accepted: 01/22/2016] [Indexed: 11/26/2022]
|
74
|
Morphine, a potential antagonist of cisplatin cytotoxicity, inhibits cisplatin-induced apoptosis and suppression of tumor growth in nasopharyngeal carcinoma xenografts. Sci Rep 2016; 6:18706. [PMID: 26729257 PMCID: PMC4700493 DOI: 10.1038/srep18706] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/24/2015] [Indexed: 12/21/2022] Open
Abstract
Morphine is an opioid analgesic drug often used for pain relief in cancer patients. However, there is growing evidence that morphine may modulate tumor growth, progression and metastasis. In this study, we evaluated whether morphine modulates cisplatin-induced apoptosis in human nasopharyngeal carcinoma CNE-2 cells and whether morphine affects the antitumor activity of cisplatin on tumor growth in human nasopharyngeal carcinoma CNE-2 xenografts in nude mice. We showed that a pretreatment with morphine (1 μg/ml) inhibited the sensitivity of CNE-2 cells to cisplatin by inhibiting cisplatin-induced CNE-2 cell apoptosis, decreasing caspase-3 activity and increasing the Bcl-2/Bax ratio. However, a high dose of morphine (1000 μg/ml) had the opposite effect. We also showed that at a low dose, morphine enhances chemoresistance in an in vivo nasopharyngeal carcinoma (NPC) model by inhibiting cisplatin-induced apoptosis and decreasing neovascularization. Taken together, our results indicate that a low dose of morphine may lead to chemoresistance of cisplatin in NPC models in vitro and in vivo by inhibiting cisplatin-induced apoptosis and decreasing neovascularization.
Collapse
|
75
|
Saify K, Saadat I, Saadat M. Down-regulation of antioxidant genes in human SH-SY5Y cells after treatment with morphine. Life Sci 2016; 144:26-29. [DOI: 10.1016/j.lfs.2015.11.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 11/11/2015] [Accepted: 11/16/2015] [Indexed: 10/22/2022]
|
76
|
Zhou P, Jiang J, Dong Z, Yan H, You Z, Su R, Gong Z. The proteins interacting with C-terminal of μ receptor are identified by bacterial two-hybrid system from brain cDNA library in morphine-dependent rats. Life Sci 2015; 143:156-67. [DOI: 10.1016/j.lfs.2015.10.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 10/21/2015] [Accepted: 10/23/2015] [Indexed: 10/22/2022]
|
77
|
Neuroprotection of donepezil against morphine-induced apoptosis is mediated through Toll-like receptors. Eur J Pharmacol 2015; 764:292-297. [DOI: 10.1016/j.ejphar.2015.07.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/08/2015] [Accepted: 07/10/2015] [Indexed: 11/23/2022]
|
78
|
Mohammad Ahmadi Soleimani S, Ekhtiari H, Cadet JL. Drug-induced neurotoxicity in addiction medicine: From prevention to harm reduction. PROGRESS IN BRAIN RESEARCH 2015; 223:19-41. [PMID: 26806769 DOI: 10.1016/bs.pbr.2015.07.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neurotoxicity is considered as a major cause of neurodegenerative disorders. Most drugs of abuse have nonnegligible neurotoxic effects many of which are primarily mediated by several dopaminergic and glutamatergic neurotransmitter systems. Although many researchers have investigated the medical and cognitive consequences of drug abuse, the neurotoxicity induced by these drugs still requires comprehensive attention. The science of neurotoxicity promises to improve preventive and therapeutic strategies for brain disorders such as Alzheimer disease and Parkinson's disease. However, its clinical applications for addiction medicine remain to be defined adequately. This chapter reviews the most commonly discussed mechanisms underlying neurotoxicity induced by common drugs of abuse including amphetamines, cocaine, opiates, and alcohol. In addition, the known factors that trigger and/or predispose to drug-induced neurotoxicity are discussed. These factors include drug-related, individual-related, and environmental insults. Moreover, we introduce some of the potential pharmacological antineurotoxic interventions deduced from experimental animal studies. These interventions involve various targets such as dopaminergic system, mitochondria, cell death signaling, and NMDA receptors, among others. We conclude the chapter with a discussion of addicted patients who might benefit from such interventions.
Collapse
Affiliation(s)
- S Mohammad Ahmadi Soleimani
- Neurocognitive Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran; Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamed Ekhtiari
- Neurocognitive Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran; Translational Neuroscience Program, Institute for Cognitive Science Studies (ICSS), Tehran, Iran; Research Center for Molecular and Cellular Imaging (RCMCI), Tehran University of Medical Sciences, Tehran, Iran
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
79
|
Reversible Inactivation of the Dorsal Raphe Nucleus Increases Morphine-Induced Antinociception in Tolerated but Not in Nontolerated Rats. NEUROPHYSIOLOGY+ 2015. [DOI: 10.1007/s11062-015-9522-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
80
|
Vousooghi N, Shirazi MSS, Goodarzi A, Abharian PH, Zarrindast MR. X Chromosome Inactivation in Opioid Addicted Women. Basic Clin Neurosci 2015; 6:179-84. [PMID: 26904175 PMCID: PMC4656991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
INTRODUCTION X chromosome inactivation (XCI) is a process during which one of the two X chromosomes in female human is silenced leading to equal gene expression with males who have only one X chromosome. Here we have investigated XCI ratio in females with opioid addiction to see whether XCI skewness in women could be a risk factor for opioid addiction. METHODS 30 adult females meeting DSM IV criteria for opioid addiction and 30 control females with no known history of addiction were included in the study. Digested and undigested DNA samples which were extracted from blood were analyzed after amplification of the polymorphic androgen receptor (AR) gene located on the X chromosome. XCI skewness was studied in 3 ranges: 50:50-64:36 (random inactivation), 65:35-80:20 (moderately skewed) and >80:20 (highly skewed). RESULTS XCI from informative females in control group was 63% (N=19) random, 27% (N=8) moderately skewed and 10% (N=3) highly skewed. Addicted women showed 57%, 23% and 20%, respectively. The distribution and frequency of XCI status in women with opioid addiction was not significantly different from control group (P=0.55). DISCUSSION Our data did not approve our hypothesis of increased XCI skewness among women with opioid addiction or unbalanced (non-random) expression of genes associated with X chromosome in female opioid addicted subjects.
Collapse
Affiliation(s)
- Nasim Vousooghi
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Iranian National Center for Addiction Studies (INCAS), Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra-Sadat Sadat Shirazi
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Iranian National Center for Addiction Studies (INCAS), Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Goodarzi
- Iranian National Center for Addiction Studies (INCAS), Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyman Hassani Abharian
- Iranian National Center for Addiction Studies (INCAS), Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Iranian National Center for Addiction Studies (INCAS), Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Cognitive Neuroscience, Institute for Cognitive Science Studies, Tehran, Iran.,Genomics Center, School of Advanced Sciences, Tehran Medical Branch, Islamic Azad University, Tehran, Iran.,School of Cognitive Sciences, Institute for Studies in Theoretical Physics and Mathematics, Tehran, Iran.,Corresponding Author: Mohammad Reza Zarrindast, PhD, Address: Iranian National Center for Addiction Studies (INCAS), Iranian Institute for Reduction of High-Risk Behaviors, Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. Tel.: +98 (21) 88991118 Fax: +98 (21) 88991117 E-mail:
| |
Collapse
|
81
|
Abstract
Abstract
Opioid-induced hyperalgesia (OIH) is a relatively new paradigm that has added to the already growing uncertainty surrounding long-term opioid treatment. OIH is the oversensitization to stimuli in the nervous system resulting from opioid exposure and subsequent neuroplastic changes. Because of its novelty and difficulty in identification, the true prevalence of OIH is unknown. Several mechanisms have been proposed for its development. These include changes in the N-methyl-D-aspartate system, descending pathway modulation, dynorphin activity, inflammatory changes mediated by cyclooxygenase, and increased sensitivity to excitatory neurochemicals. The clinical controversy regarding the management of OIH is due largely to the lack of guidance in diagnosis and lack of quality evidence to direct treatment. As a diagnosis of exclusion, several alternative causes of antianalgesia must be ruled out before OIH can be declared. Pharmacodynamic phenomena such as opioid tolerance share overlapping mechanisms with OIH and may present similarly. Pharmacokinetic changes such as drug-induced or disease-induced alterations to the cytochrome P450 or P-glycoprotein systems should also be excluded as causes of increased opioid demand that may be seen as OIH. Certain pharmacologic agents, such as N-methyl-D-aspartate receptor antagonists, alpha2 receptor agonists, and cyclooxygenase inhibitors, have been identified as possible treatments to reverse the effects of OIH. Opioid rotation and dose reductions have also been used with some degree of success. Pharmacist involvement in the identification and management of OIH will be central to success because of the unique expertise they offer. The quality of these studies is limited by study design, small sample sizes, and lack of generalizability to chronic pain patients with long-standing opioid use.
Collapse
|
82
|
Ghasemi F, Moradi A, Izadpanah E, Moloudi MR, Hassanzadeh K, Rahimmi A, Hassanzadeh K. Simvastatin prevents morphine antinociceptive tolerance and withdrawal symptoms in rats. J Formos Med Assoc 2015; 114:399-406. [DOI: 10.1016/j.jfma.2014.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 07/28/2014] [Accepted: 07/29/2014] [Indexed: 01/06/2023] Open
|
83
|
Lee HJ, Yeomans DC. Opioid induced hyperalgesia in anesthetic settings. Korean J Anesthesiol 2014; 67:299-304. [PMID: 25473457 PMCID: PMC4252340 DOI: 10.4097/kjae.2014.67.5.299] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 10/04/2014] [Accepted: 10/10/2014] [Indexed: 11/10/2022] Open
Abstract
Pain is difficult to investigate and difficult to treat, in part, because of problems in quantification and assessment. The use of opioids, combined with classic anesthetics to maintain hemodynamic stability by controlling responses to intraoperative painful events has gained significant popularity in the anesthetic field. However, several side effects profiles concerning perioperative use of opioid have been published. Over the past two decades, many concerns have arisen with respect to opioid-induced hyperalgesia (OIH), which is the paradoxical effect wherein opioid usage may decrease pain thresholds and increase atypical pain unrelated to the original, preexisting pain. This brief review focuses on the evidence, mechanisms, and modulatory and pharmacologic management of OIH in order to elaborate on the clinical implication of OIH.
Collapse
Affiliation(s)
- Hyeon Jeong Lee
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA. ; Department of Anesthesia, Pusan National University School of Medicine, Busan, Korea
| | - David C Yeomans
- Department of Anesthesia, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
84
|
Autophagy in superficial spinal dorsal horn accelerates the cathepsin B-dependent morphine antinociceptive tolerance. Neuroscience 2014; 275:384-94. [DOI: 10.1016/j.neuroscience.2014.06.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 05/27/2014] [Accepted: 06/03/2014] [Indexed: 01/06/2023]
|
85
|
Boules M, Shimizu H, Zelisko A, El-Hayek K, Rizk MK, Kroh M. Pre-operative opioid analgesia reduces clinical success of laparoscopic gastric electrical stimulation placement in patients with gastroparesis. Surg Endosc 2014; 29:805-9. [PMID: 25115865 DOI: 10.1007/s00464-014-3754-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 07/10/2014] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Gastroparesis is a common chronic and costly disorder for which medical therapy is often unsuccessful. Gastric electrical stimulation (GES) has been used to treat refractory cases, however, response is variable and difficult to predict. This study aims to assess whether pre-operative opioid analgesics (OA) use affects clinical success of GES. METHODS Records of 128 patients who underwent laparoscopic GES placement from March 2001 to September 2012 were analyzed retrospectively. Data collected included demographics, surgical outcomes, and clinical parameters. Pre- and post-operative opioid analgesic dosing (No = 0 morphine equivalents (ME)/day, Low = 0-40 ME/day, Mid = 41-80 ME/day, High >80 ME/day), as well as clinical symptom assessment was collected for up to 3 years post-operatively. Clinical success was defined as (1) OA reduction of >50 %, (2) maintenance of weight, or (3) symptom improvement. Descriptive statistics were computed for all factors. A p < 0.05 was considered statistically significant. RESULTS Fifty-three patients were on OA pre-operatively compared to 69 patients who were not. Patients not on OA pre-operatively were less likely than those on OA pre-op group to be on OA post-operatively (p = 0.005); however, there were no differences in weight or symptom improvement. Sub-group analysis of the 53 patients on OA demonstrated significant improvement in clinical symptoms in the low-morphine cohort compared to the mid-morphine cohort (p = 0.02), and OA dosing post-operatively in the low-morphine cohort diminished significantly compared to mid- and high-morphine cohort (p = 0.032). There was no significant difference in weight. CONCLUSION OA dosing pre-operatively significantly affects clinical success of GES placement. Criteria for offering GES implantation may need to take OA dosing into consideration.
Collapse
Affiliation(s)
- Mena Boules
- Digestive Disease Institute, Cleveland Clinic, 9500 Euclid Avenue, A100, Cleveland, OH, 44195, USA,
| | | | | | | | | | | |
Collapse
|
86
|
Shahidi S, Hashemi-Firouzi N. The effects of a 5-HT7 receptor agonist and antagonist on morphine withdrawal syndrome in mice. Neurosci Lett 2014; 578:27-32. [DOI: 10.1016/j.neulet.2014.06.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 05/30/2014] [Accepted: 06/11/2014] [Indexed: 12/16/2022]
|
87
|
RHODIN A, EHREN M, SKOTTHEIM B, GRÖNBLADH A, ORTIZ‐NIETO F, RAININKO R, GORDH T, NYBERG F. Recombinant human growth hormone improves cognitive capacity in a pain patient exposed to chronic opioids. Acta Anaesthesiol Scand 2014; 58:759-65. [PMID: 24712862 PMCID: PMC4265204 DOI: 10.1111/aas.12309] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2014] [Indexed: 01/09/2023]
Abstract
During recent decades, the increasing use of opioids for chronic non-cancer pain has raised concerns regarding tolerance, addiction, and importantly cognitive dysfunction. Current research suggests that the somatotrophic axis could play an important role in cognitive function. Administration of growth hormone (GH) to GH-deficient humans and experimental animals has been shown to result in significant improvements in cognitive capacity. In this report, a patient with cognitive disabilities resulting from chronic treatment with opioids for neuropathic pain received recombinant human growth hormone (rhGH) replacement therapy. A 61-year-old man presented with severe cognitive dysfunction after long-term methadone treatment for intercostal neuralgia and was diagnosed with GH insufficiency by GH releasing hormone-arginine testing. The effect of rhGH replacement therapy on his cognitive capacity and quality of life was investigated. The hippocampal volume was measured using magnetic resonance imaging, and the ratios of the major metabolites were calculated using proton magnetic resonance spectroscopy. Cognitive testing revealed significant improvements in visuospatial cognitive function after rhGH. The hippocampal volume remained unchanged. In the right hippocampus, the N-acetylaspartate/creatine ratio (reflecting nerve cell function) was initially low but increased significantly during rhGH treatment, as did subjective cognitive, physical and emotional functioning. This case report indicates that rhGH replacement therapy could improve cognitive behaviour and well-being, as well as hippocampal metabolism and functioning in opioid-treated patients with chronic pain. The idea that GH could affect brain function and repair disabilities induced by long-term exposure to opioid analgesia is supported.
Collapse
Affiliation(s)
- A. RHODIN
- Department of Surgical Sciences, Anaesthesia/Pain Research Uppsala University Uppsala Sweden
| | - M. EHREN
- Department of Radiology Uppsala University Uppsala Sweden
| | - B. SKOTTHEIM
- Department of Neuroscience Uppsala University Uppsala Sweden
| | - A. GRÖNBLADH
- Department of Pharmaceutical Biosciences Uppsala University Uppsala Sweden
| | - F. ORTIZ‐NIETO
- Department of Radiology Uppsala University Uppsala Sweden
| | - R. RAININKO
- Department of Radiology Uppsala University Uppsala Sweden
| | - T. GORDH
- Department of Surgical Sciences, Anaesthesia/Pain Research Uppsala University Uppsala Sweden
| | - F. NYBERG
- Department of Pharmaceutical Biosciences Uppsala University Uppsala Sweden
| |
Collapse
|
88
|
Liu X, Wang G, Pu H, Jing H. Abnormal intracellular calcium homeostasis associated with vulnerability in the nerve cells from heroin-dependent rat. Brain Res 2014; 1572:40-9. [DOI: 10.1016/j.brainres.2014.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 04/11/2014] [Accepted: 05/12/2014] [Indexed: 11/26/2022]
|
89
|
Singh R, Visintainer PF. Adjunctive therapy for neonatal abstinence syndrome: why not personalize it for each infant based on their in-utero exposure? J Perinatol 2014; 34:575-6. [PMID: 24968905 DOI: 10.1038/jp.2014.99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- R Singh
- 1] Division of Neonatology, Department of Pediatrics, Baystate Children's Hospital, Springfield, MA, USA [2] Department of Pediatrics, Tufts University School of Medicine, Boston, MA, USA
| | - P F Visintainer
- 1] Department of Epidemiology, Baystate Medical Center, Springfield, MA, USA [2] Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
90
|
Landais A. Severe memory impairment following acute morphine intoxication. J Neurol Sci 2014; 343:242-4. [PMID: 24948561 DOI: 10.1016/j.jns.2014.05.061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/25/2014] [Accepted: 05/29/2014] [Indexed: 11/16/2022]
Affiliation(s)
- Anne Landais
- University Hospital of Pointe-à-Pitre/CHU de Pointe-à-Pitre, Neurology Unit, Route de Chauvel, 97139 Abymes, France.
| |
Collapse
|
91
|
Chai E, Yu F, Xie M, Wang J. Neurotoxic effect of chronic heroin administration on the expression of c-Fos and Bax and glial cells in rat prefrontal cortex. TOXIN REV 2014. [DOI: 10.3109/15569543.2013.867884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
92
|
Razavi Y, Alamdary SZ, Katebi SN, Khodagholi F, Haghparast A. Morphine-induced apoptosis in the ventral tegmental area and hippocampus after the development but not extinction of reward-related behaviors in rats. Cell Mol Neurobiol 2014; 34:235-45. [PMID: 24281942 PMCID: PMC11488956 DOI: 10.1007/s10571-013-0007-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 11/11/2013] [Indexed: 10/26/2022]
Abstract
Some data suggest that morphine induces apoptosis in neurons, while other evidences show that morphine could have protective effects against cell death. In this study, we suggested that there is a parallel role of morphine in reward circuitry and apoptosis processing. Therefore, we investigated the effect of morphine on modifications of apoptotic factors in the ventral tegmental area (VTA) and hippocampus (HPC) which are involved in the reward circuitry after the acquisition and extinction periods of conditioned place preference (CPP). In behavioral experiments, different doses of morphine (0.5, 5, and 10 mg/kg) and saline were examined in the CPP paradigm. Conditioning score and locomotor activity were recorded by Ethovision software after acquisition on the post-conditioning day, and days 4 and 8 of extinction periods. In order to investigate the molecular mechanisms in each group, we then dissected the brains and measured the expression of apoptotic factors in the VTA and HPC by western blotting analysis. All of the morphine-treated groups showed an increase of apoptotic factors in these regions during acquisition but not in extinction period. In the HPC, morphine significantly increased the ratio of Bax/Bcl-2, caspases-3, and PARP by the lowest dose (0.5 mg/kg), but, in the VTA, a considerable increase was seen in the dose of 5 mg/kg; promotion of apoptotic factors in the HPC and VTA insinuates that morphine can affect the molecular mechanisms that interfere with apoptosis through different receptors. Our findings suggest that a specific opioid receptor involves in modification of apoptotic factors expression in these areas. It seems that the reduction of cell death in response to high dose of morphine in the VTA and HPC may be due to activation of low affinity opioid receptors which are involved in neuroprotective features of morphine.
Collapse
Affiliation(s)
- Yasaman Razavi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box 19615-1178, Tehran, Iran
| | - Shabnam Zeighamy Alamdary
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box 19615-1178, Tehran, Iran
| | - Seyedeh-Najmeh Katebi
- Physiology-Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box 19615-1178, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box 19615-1178, Tehran, Iran
| |
Collapse
|
93
|
|
94
|
Ghafari S, Golalipour MJ. Prenatal morphine exposure reduces pyramidal neurons in CA1, CA2 and CA3 subfields of mice hippocampus. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2014; 17:155-61. [PMID: 24847417 PMCID: PMC4016685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 10/16/2013] [Indexed: 11/23/2022]
Abstract
OBJECTIVE(S) This study was carried out to evaluate the effect of maternal morphine exposure during gestational and lactation period on pyramidal neurons of hippocampus in 18 and 32 day mice offspring. MATERIALS AND METHODS Thirty female mice were randomly allocated into cases and controls. In case group, animals received morphine sulfate 10 mg/kg.body weight intraperitoneally during 7 days before mating, gestational period (GD 0-21), 18 and 32 days after delivery in the experimental groups. The control animals received an equivalent volume of normal saline. Cerebrum of six offsprings in each group was removed and stained with cresyl violet and a monoclonal antibody NeuN for immunohistochemical detection of surviving pyramidal neurons. Quantitative computer-assisted morphometric study was done on hippocampus. RESULTS The number of pyramidal neurons in CA1, CA2 and CA3 in treated groups was significantly reduced in postnatal day 18 and 32 (P18, P32) compared to control groups (P<0.05). The mean thickness of the stratum pyramidal layer was decreased in the treated groups in comparison with controls (P<0.05), whereas the mean thickness of the stratum oriens, stratum radiatum and stratum lacunosum-moleculare in CA1 field and stratum oriens, stratum lucidum, stratum radiatum and stratum lacunosum-moleculare in CA3 were significantly increased in morphine treated group in comparison with controls (P<0.05). CONCLUSION Morphine administration before and during pregnancy and during lactation period causes pyramidal neurons loss in 18 and 32 days old infant mice.
Collapse
Affiliation(s)
- Soraya Ghafari
- Department of Anatomical Sciences, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohammad Jafar Golalipour
- Gorgan Congenital Malformations Research Center, Department of Anatomical Sciences, Golestan University of Medical Sciences, Gorgan, Iran,Corresponding author: Mohammad Jafar Golalipour.Gorgan Congenital Malformations Research Center, Golestan University of Medical Sciences, Gorgan, Iran. P.O. Box: 49175-1141; Tel/Fax: +98-171-4425165, 4421660;
| |
Collapse
|
95
|
Analgesia-sedation in PICU and neurological outcome: a secondary analysis of long-term neuropsychological follow-up in meningococcal septic shock survivors*. Pediatr Crit Care Med 2014; 15:189-96. [PMID: 24366510 DOI: 10.1097/pcc.0000000000000044] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVES To investigate whether analgesic and sedative drug use during PICU treatment is associated with long-term neurodevelopmental outcome in children who survived meningococcal septic shock. DESIGN This study concerned a secondary analysis of data from medical and psychological follow-up of a cross-sectional cohort of all consecutive surviving patients with septic shock and purpura requiring intensive care treatment between 1988 and 2001 at the Erasmus MC-Sophia Children's Hospital. At least 4 years after PICU admission, these children showed impairments on several domains of neuropsychological functioning. In the present study, type, number, and dose of sedatives and analgesics were retrospectively evaluated. SETTING Tertiary care university hospital. PATIENTS Seventy-seven meningococcal septic shock survivors (median age, 2.1 yr). INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Forty-five patients (58%) received one or more analgesic and/or sedative drugs during PICU admission, most commonly benzodiazepines (n = 39; 51%), followed by opioids (n = 23; 30%). In total, 12 different kinds of analgesic or sedative drugs were given. The use and dose of opioids were significantly associated with poor test outcome on full-scale intelligence quotient (p = 0.02; Z = -2.28), verbal intelligence quotient (p = 0.02; Z = -2.32), verbal intelligence quotient subtests (verbal comprehension [p = 0.01; Z = -2.56] and vocabulary [p = 0.01; Z = -2.45]), and visual attention/executive functioning (Trial Making Test part B) (p = 0.03; Z = -2.17). In multivariate analysis adjusting for patient and disease characteristics, the use of opioids remained significant on most neuropsychological tests. CONCLUSIONS The use of opioids during PICU admission was significantly associated with long-term adverse neuropsychological outcome independent of severity of illness scores in meningococcal septic shock survivors.
Collapse
|
96
|
Sharifipour M, Izadpanah E, Nikkhoo B, Zare S, Abdolmaleki A, Hassanzadeh K, Moradi F, Hassanzadeh K. A new pharmacological role for donepezil: attenuation of morphine-induced tolerance and apoptosis in rat central nervous system. J Biomed Sci 2014; 21:6. [PMID: 24455992 PMCID: PMC3906771 DOI: 10.1186/1423-0127-21-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 01/20/2014] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Tolerance to the analgesic effect of opioids is a pharmacological phenomenon that occurs after their prolonged administration. It has been shown that morphine-induced tolerance is associated with apoptosis in the central nervous system and neuroprotective agents which prevented apoptosis signaling could attenuate tolerance to the analgesic effects. On the other hand donepezil, an acetylcholinesterase inhibitor, has been reported to have neuroprotective effects. Therefore in this study, the effect of systemic administration of donepezil on morphine-induced tolerance and apoptosis in the rat cerebral cortex and lumbar spinal cord was evaluated. Various groups of rats received morphine (ip) and different doses of donepezil (0, 0.5, 1, 1.5 mg/kg/day). Nociception was assessed using tail flick apparatus. Tail flick latency was recorded when the rat shook its tail. For apoptosis assay other groups of rats received the above treatment and apoptosis was evaluated by in situ terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling (TUNEL) method. RESULTS The results showed that administration of donepezil (0.5, 1, 1.5 mg/kg, ip) delayed the morphine tolerance for 9, 12 and 17 days, respectively. Furthermore pretreatment injection of donepezil attenuated the number of apoptotic cells in the cerebral cortex and lumbar spinal cord compared to the control group. CONCLUSION In conclusion, we found that systemic administration of donepezil attenuated morphine-induced tolerance and apoptosis in the rat cerebral cortex and lumbar spinal cord.
Collapse
Affiliation(s)
- Mozhdeh Sharifipour
- Department of Biology, Faculty of Basic Science, Urmia University, Urmia, Iran
| | - Esmaeal Izadpanah
- Cellular and Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Department of Physiology and Pharmacology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Bahram Nikkhoo
- Department of Pathology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Samad Zare
- Department of Biology, Faculty of Basic Science, Urmia University, Urmia, Iran
| | - Ali Abdolmaleki
- Science and Research Branch, Islamic Azad University, Hamedan, Iran
| | - Katayoun Hassanzadeh
- Department of Physiology and Pharmacology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Farshid Moradi
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Kambiz Hassanzadeh
- Cellular and Molecular Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Department of Physiology and Pharmacology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
97
|
Manchikanti L, Benyamin R, Datta S, Vallejo R, Smith H. Opioids in chronic noncancer pain. Expert Rev Neurother 2014; 10:775-89. [DOI: 10.1586/ern.10.37] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
98
|
Katebi SN, Razavi Y, Zeighamy Alamdary S, Khodagholi F, Haghparast A. Morphine could increase apoptotic factors in the nucleus accumbens and prefrontal cortex of rat brain's reward circuitry. Brain Res 2013; 1540:1-8. [DOI: 10.1016/j.brainres.2013.09.045] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 09/06/2013] [Accepted: 09/27/2013] [Indexed: 01/10/2023]
|
99
|
Harada S, Nakamoto K, Tokuyama S. The involvement of midbrain astrocyte in the development of morphine tolerance. Life Sci 2013; 93:573-8. [DOI: 10.1016/j.lfs.2013.08.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 08/02/2013] [Accepted: 08/13/2013] [Indexed: 01/28/2023]
|
100
|
Rafati A, Noorafshan A, Torabi N. Stereological study of the effects of morphine consumption and abstinence on the number of the neurons and oligodendrocytes in medial prefrontal cortex of rats. Anat Cell Biol 2013; 46:191-7. [PMID: 24179694 PMCID: PMC3811856 DOI: 10.5115/acb.2013.46.3.191] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/26/2013] [Accepted: 06/04/2013] [Indexed: 01/25/2023] Open
Abstract
Quantitative studies to date on the effects of opioid consumption and abstinence on the nervous system using modern stereological methods have not received enough attention. In addition, they have yielded controversial results. The present study was conducted to investigate the effects of morphine, with or without abstinence, on the neurons and oligodendrocytes of the medial prefrontal cortex (MPFC) in rats using quantitative stereological methods. The male rats were divided into four groups: the first (saline [SAL]) and second (morphine [MOR]) groups were treated with saline and an escalating dose of morphine (5-20 mg/kg) for 30 days, respectively; the third (SAL+abstinence [ABS]) and fourth (MOR+ABS) groups were treated in the same manner as the previous groups plus they had a 30-day abstinence period. The results showed that the volume of the MPFC and its subdivisions decreased by approximately 15% in the MOR group compared with that in the SAL group (P<0.05). In addition, the volume decreased by approximately 24% in the MOR+ABS group compared with that in the SAL+ABS group (P<0.05). The number of neurons in the MOR and MOR+ABS groups decreased by approximately 44% and 35%, respectively, compared with that in their corresponding control groups. Moreover, the number of the oligodendrocytes in the MOR and MOR+ABS groups decreased by approximately 41% and 37%, respectively. No significant difference was noted in the number of cells in the MOR and MOR+ABS groups. In conclusion, morphine consumption leads to a permanent reduction in the number of neurons and oligodendrocytes, and no additional neuron and oligodendrocyte loss occurs after abstinence.
Collapse
Affiliation(s)
- Ali Rafati
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran. ; Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | |
Collapse
|