1
|
Xu X, Chen X, Zhu W, Zhao J, Liu Y, Duan C, Qi Y. Efficacy and Safety of Ultrasound Guided-Deep Serratus Anterior Plane Blockade With Different Doses of Dexmedetomidine for Women Undergoing Modified Radical Mastectomy: A Randomized Controlled Trial. Front Med (Lausanne) 2022; 9:819239. [PMID: 35198576 PMCID: PMC8860248 DOI: 10.3389/fmed.2022.819239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 01/10/2022] [Indexed: 11/21/2022] Open
Abstract
Background Ultrasound guided-deep serratus anterior plane block (USG-DSAPB) has been used for pain management of patients undergoing modified radical mastectomy (MRM), but evidence supporting their adjuvant analgesic benefits is limited. We explored the efficacy and safety of preemptive use of ropivacaine combined with different doses of dexmedetomidine (DEX) in USG-DSAPB for patients undergoing MRM. Methods Ninety-five female patients undergoing unilateral MRM were allocated randomly to two groups. Group RD1 had 20 mL of 0.5% ropivacaine with 5 mg of dexamethasone and 0.5 μg·kg−1 DEX in USG-DSAPB. Group RD2 had 20 mL of 0.5% ropivacaine with 5 mg of dexamethasone and 1 μg·kg−1 DEX in USG-DSAPB. The primary outcome was sufentanil consumption 72 h after USG-DSAPB. Secondary outcomes were: postoperative pain scores and level of sedation; intraoperative hemodynamics; duration of post-anesthesia care unit (PACU) stay; prevalence of moderate-to-severe pain; one-time puncture success; procedure time of blockade; time to first rescue analgesia; requirement of rescue analgesia; satisfaction scores of patients and surgeons; duration of hospital stay; adverse events; prevalence of chronic pain; quality of postoperative functional recovery. Results Compared with the RD1 group, the visual analog scale score for coughing was significantly lower at 4, 8, 12 h and sufentanil consumption was significantly lower at 4, 8, 12, 24, and 48 h after surgery in the RD2 group (P < 0.05). The time to first rescue analgesia was significantly longer in the RD2 group (P < 0.05). The requirement for rescue analgesia was significantly higher in the RD1 group (P < 0.05). The prevalence of moderate-to-severe pain, number of patients using vasoactive agents, duration of PACU stay, as well as consumption of propofol, remifentanil, and DEX were significantly lower in the RD2 group (P < 0.05). There were no significant differences between the two groups with respect to one-time puncture success, procedure time of blockade, total dermatomal spread, satisfaction scores of patients and surgeons, postoperative complications, duration of hospital stay, 40-item Quality of Recovery questionnaire (QoR-40) score, or prevalence of chronic pain (P > 0.05). Conclusions We discovered that 1 μg·kg−1 (not 0.5 μg·kg−1) DEX combined with 20 mL of 0.5% ropivacaine and 5 mg of dexamethasone in USG-DSAPB could provide superior postoperative analgesia for patients undergoing MRM. However, the quality of postoperative functional recovery and prevalence of chronic pain were similar. Clinical Trial Registration:http://www.chictr.org.cn/showproj.aspx?proj=54929, identifier: ChiCTR2000033685.
Collapse
Affiliation(s)
- Xia Xu
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, China
| | - Xingfang Chen
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, China
| | - Wenchao Zhu
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, China
| | - Jing Zhao
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, China
| | - Yanchao Liu
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, China
| | - Caiping Duan
- Department of Anesthesiology, Ordos Central Hospital, Ordos, China
| | - Yingying Qi
- Department of Anesthesiology, Liaocheng People's Hospital, Liaocheng, China
- *Correspondence: Yingying Qi
| |
Collapse
|
2
|
Hill R, Conibear A, Dewey W, Kelly E, Henderson G. Role of Acetaldehyde in Ethanol Reversal of Tolerance to Morphine-Induced Respiratory Depression in Mice. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 1. [PMID: 35909497 PMCID: PMC7613180 DOI: 10.3389/adar.2021.10143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Background: Opioid users regularly consume other drugs such as alcohol (ethanol). Acute administration of ethanol rapidly reverses tolerance to morphine-induced respiratory depression. However, recent research has suggested that the primary metabolite of ethanol, acetaldehyde, may play a key role in mediating the CNS effects seen after ethanol consumption. This research investigated the role of acetaldehyde in ethanol reversal of tolerance to morphine-induced respiratory depression.Methods: Tolerance was induced in mice by 6-days implantation of a 75 mg morphine pellet with control mice implanted with a placebo pellet. Tolerance was assessed by acute morphine administration on day 6 and respiration measured by plethysmography. Levels of acetaldehyde were inhibited or enhanced by pre-treatments with the acetaldehyde chelator D-penicillamine and the inhibitor of acetaldehyde dehydrogenase disulfiram respectively.Results: Morphine pellet implanted mice displayed tolerance to an acute dose of morphine compared to placebo pellet implanted controls. Acute acetaldehyde administration dose-dependently reversed tolerance to morphine respiratory depression. As previously demonstrated, ethanol reversed morphine tolerance, and this was inhibited by D-penicillamine pre-treatment. An acute, low dose of ethanol that did not significantly reverse morphine tolerance was able to do so following disulfiram pre-treatment.Conclusion: These data suggest that acetaldehyde, the primary metabolite of ethanol, is responsible for the reversal of morphine tolerance observed following ethanol administration.
Collapse
Affiliation(s)
- Rob Hill
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
- Correspondence: Rob Hill,
| | - Alexandra Conibear
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - William Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - Eamonn Kelly
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Graeme Henderson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
3
|
Ram A, Edwards TM, McCarty A, McDermott MV, Bobeck EN. Morphine-induced kinase activation and localization in the periaqueductal gray of male and female mice. J Neurochem 2021; 159:590-602. [PMID: 34499746 DOI: 10.1111/jnc.15506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/17/2022]
Abstract
Morphine is a potent opioid analgesic with high propensity for the development of antinociceptive tolerance. Morphine antinociception and tolerance are partially regulated by the midbrain ventrolateral periaqueductal gray (vlPAG). However, the majority of research evaluating mu-opioid receptor signaling has focused on males. Here, we investigate kinase activation and localization patterns in the vlPAG following acute and chronic morphine treatment in both sexes. Male and female mice developed rapid antinociceptive tolerance to morphine (10 mg/kg i.p.) on the hot plate assay, but tolerance did not develop in males on the tail flick assay. Quantitative fluorescence immunohistochemistry was used to map and evaluate the activation of extracellular signal-regulated kinase 1/2 (ERK 1/2), protein kinase-C (PKC), and protein kinase-A (PKA). We observed significantly greater phosphorylated ERK 1/2 in the vlPAG of chronic morphine-treated animals which co-localized with the endosomal marker, Eea1. We note that pPKC is significantly elevated in the vlPAG of both sexes following chronic morphine treatment. We also observed that although PKA activity is elevated following chronic morphine treatment in both sexes, there is a significant reduction in the nuclear translocation of its phosphorylated substrate. Taken together, this study demonstrates increased activation of ERK 1/2, PKC, and PKA in response to repeated morphine treatment. The study opens avenues to explore the impact of chronic morphine treatment on G-protein signaling and kinase nuclear transport.
Collapse
Affiliation(s)
- Akila Ram
- Department of Biology, Utah State University, Logan, Utah, USA
| | | | - Ashley McCarty
- Department of Biology, Utah State University, Logan, Utah, USA
| | - Max V McDermott
- Department of Biology, Utah State University, Logan, Utah, USA
- Interdisciplinary Neuroscience Program, Utah State University, Logan, Utah, USA
| | - Erin N Bobeck
- Department of Biology, Utah State University, Logan, Utah, USA
- Interdisciplinary Neuroscience Program, Utah State University, Logan, Utah, USA
| |
Collapse
|
4
|
Ho IH, Ng LH, Cheng X, Gin T, Chan CS, Sun W, Xiao L, Zhang L, Chan MT, Wu WK, Liu X. Annexin A2 traps mu-opioid receptors in recycling endosomes upon remifentanil-induced internalization. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 10:100071. [PMID: 34401608 PMCID: PMC8358694 DOI: 10.1016/j.ynpai.2021.100071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/28/2021] [Accepted: 07/29/2021] [Indexed: 06/13/2023]
Abstract
•ANXA2 is a novel MOR1-interacting protein regulating MOR1 sub-cellular localization.•ANXA2 retains MOR1 in late recycling endosomes after remifentanil exposure.
Collapse
Affiliation(s)
- Idy H.T. Ho
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
- Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Lhotse H.L. Ng
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
- Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xiaojie Cheng
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Tony Gin
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
- Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Chee Sam Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Wuping Sun
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People’s Hospital and the 6 Affiliated Hospital of Shenzhen University Health Science Center, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Lizu Xiao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People’s Hospital and the 6 Affiliated Hospital of Shenzhen University Health Science Center, National Key Clinical Pain Medicine of China, Shenzhen 518060, China
| | - Lin Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
- Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Matthew T.V. Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
- Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - William K.K. Wu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
- Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- State Key Laboratory of Digestive Disease, Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
- Peter Hung Pain Research Institute, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
5
|
Gledhill LJ, Babey AM. Synthesis of the Mechanisms of Opioid Tolerance: Do We Still Say NO? Cell Mol Neurobiol 2021; 41:927-948. [PMID: 33704603 DOI: 10.1007/s10571-021-01065-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/12/2021] [Indexed: 10/21/2022]
Abstract
The use of morphine as a first-line agent for moderate-to-severe pain is limited by the development of analgesic tolerance. Initially opioid receptor desensitization in response to repeated stimulation, thought to underpin the establishment of tolerance, was linked to a compensatory increase in adenylate cyclase responsiveness. The subsequent demonstration of cross-talk between N-methyl-D-aspartate (NMDA) glutamate receptors and opioid receptors led to the recognition of a role for nitric oxide (NO), wherein blockade of NO synthesis could prevent tolerance developing. Investigations of the link between NO levels and opioid receptor desensitization implicated a number of events including kinase recruitment and peroxynitrite-mediated protein regulation. Recent experimental advances and the identification of new cellular constituents have expanded the potential signaling candidates to include unexpected, intermediary compounds not previously linked to this process such as zinc, histidine triad nucleotide-binding protein 1 (HINT1), micro-ribonucleic acid (mi-RNA) and regulator of G protein signaling Z (RGSZ). A further complication is a lack of consistency in the protocols used to create tolerance, with some using acute methods measured in minutes to hours and others using days. There is also an emphasis on the cellular changes that are extant only after tolerance has been established. Although a review of the literature demonstrates a lack of spatio-temporal detail, there still appears to be a pivotal role for nitric oxide, as well as both intracellular and intercellular cross-talk. The use of more consistent approaches to verify these underlying mechanism(s) could provide an avenue for targeted drug development to rescue opioid efficacy.
Collapse
Affiliation(s)
- Laura J Gledhill
- CURA Pharmacy, St. John of God Hospital, Bendigo, VIC, 3550, Australia
| | - Anna-Marie Babey
- Faculty of Medicine and Health, University of New England, Armidale, NSW, 2351, Australia.
| |
Collapse
|
6
|
Zakaria ZA, Roosli RAJ, Marmaya NH, Omar MH, Basir R, Somchit MN. Methanol Extract of Dicranopteris linearis Leaves Attenuate Pain via the Modulation of Opioid/NO-Mediated Pathway. Biomolecules 2020; 10:biom10020280. [PMID: 32059475 PMCID: PMC7072186 DOI: 10.3390/biom10020280] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/18/2019] [Accepted: 10/10/2019] [Indexed: 11/25/2022] Open
Abstract
Dicranopteris linearis leaf has been reported to exert antinociceptive activity. The present study elucidates the possible mechanisms of antinociception modulated by the methanol extract of D. linearis leaves (MEDL) using various mouse models. The extract (25, 150, and 300 mg/kg) was administered orally to mice for 30 min priot to subjection to the acetic acid-induced writhing-, hot plate- or formalin-test to establish the antinociceptive profile of MEDL. The most effective dose was then used in the elucidation of possible mechanisms of action stage. The extract was also subjected to the phytochemical analyses. The results confirmed that MEDL exerted significant (p < 0.05) antinociceptive activity in those pain models as well as the capsaicin-, glutamate-, bradykinin- and phorbol 12-myristate 13-acetate (PMA)-induced paw licking model. Pretreatment with naloxone (a non-selective opioid antagonist) significantly (p < 0.05) reversed MEDL effect on thermal nociception. Only l-arginine (a nitric oxide (NO) donor) but not N(ω)-nitro-l-arginine methyl ester (l-NAME; a NO inhibitor) or 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ; a specific soluble guanylyl cyclase inhibitor) significantly (p < 0.05) modified MEDL effect on the writhing test. Several polyphenolics and volatile antinociceptive compounds were detected in MEDL. In conclusion, MEDL exerted the opioid/NO-mediated antinociceptive activity, thus, justify D. linearis as a potential source for new analgesic agents development.
Collapse
Affiliation(s)
- Zainul Amiruddin Zakaria
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia; (R.A.J.R.); (M.N.S.)
- Integrative Pharmacogenomics Institute (iPROMISE), Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam Campus, Bandar Puncak Alam Selangor 42300, Malaysia
- Correspondence: ; Tel.: +60-19-211-7090
| | - Rushduddin Al Jufri Roosli
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia; (R.A.J.R.); (M.N.S.)
| | - Najihah Hanisah Marmaya
- Faculty of Business and Management, Universiti Teknologi MARA, Melaka Campus, Melaka 75300, Malaysia;
| | - Maizatul Hasyima Omar
- Phytochemistry Unit, Herbal Medicine Research Centre, Institute for Medical Research, Jalan Pahang, Kuala Lumpur 50588, Malaysia;
| | - Rusliza Basir
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia;
| | - Muhammad Nazrul Somchit
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia; (R.A.J.R.); (M.N.S.)
| |
Collapse
|
7
|
Protein kinase C-mediated mu-opioid receptor phosphorylation and desensitization in rats, and its prevention during early diabetes. Pain 2017; 157:910-921. [PMID: 26713421 DOI: 10.1097/j.pain.0000000000000459] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Painful diabetic neuropathy is associated with impaired opioid analgesia; however, the precise mechanism in sensory neurons remains unclear. This study aimed to identify putative mechanisms involved in modified opioid responsiveness during early streptozotocin-induced diabetes in rats. In this study, we demonstrate that in diabetic animals, impaired peripheral opioid analgesia is associated with a reduction in functional mu-opioid receptor (MOR) G protein coupling. Mu-opioid receptor immunoreactive neurons colocalized with activated forms of protein kinase C (PKC) and with the receptor for advanced glycation end products (RAGE) during streptozotocin-induced diabetes. Moreover, MOR phosphorylation at Thr370 in sensory neurons of diabetic rats, and thus desensitization, was due to RAGE-dependent PKC activation. Importantly, blocking PKC activation using PKC selective inhibitor, silencing RAGE with intrathecal RAGE siRNA, or inhibiting advanced glycation end product (AGE) formation prevented sensory neuron MOR phosphorylation and, consequently, restored MOR G protein coupling and analgesic efficacy. Thus, our findings give the first in vivo evidence of a RAGE-dependent PKC-mediated heterologous MOR phosphorylation and desensitization in sensory neurons under pathological conditions such as diabetic neuropathy. This may unravel putative mechanisms and suggest possible prevention strategies of impaired opioid responsiveness.
Collapse
|
8
|
Ahmadi S, Miraki F, Rostamzadeh J. Association of morphine-induced analgesic tolerance with changes in gene expression of GluN1 and MOR1 in rat spinal cord and midbrain. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:924-931. [PMID: 27803778 PMCID: PMC5080421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES We aimed to examine association of gene expression of MOR1 and GluN1 at mRNA level in the lumbosacral cord and midbrain with morphine tolerance in male Wistar rats. MATERIALS AND METHODS Analgesic effects of morphine administrated intraperitoneally at doses of 0.1, 1, 5 and 10 mg/kg were examined using a hot plate test in rats with and without a history of 15 days morphine (10 mg/kg) treatment. Morphine-induced analgesic tolerance was also assessed on days 1, 5, 10 and 15 of chronic morphine injections. Two groups with history of 15 days injections of saline or morphine (10 mg/kg) were decapitated on day 15 and their lumbosacral cord and midbrain were dissected for evaluating MOR1 and GluN1 gene expression. RESULTS The results of the hot plate test showed that morphine (5 and 10 mg/kg) induced significant analgesia in naïve rats but its analgesic effects in rats receiving 15 days injections of morphine (10 mg/kg) was decreased, indicating tolerance to morphine analgesia. The results also showed that the GluN1 gene expression in tolerant rats was decreased by 71% in the lumbosacral cord but increased by 110 % in the midbrain compared to the control group. However, no significant change was observed for the MOR1 gene expression in both areas. CONCLUSION It can be concluded that tolerance following administration of morphine (10 mg/kg) for 15 days is associated with site specific changes in the GluN1 gene expression in the spinal cord and midbrain but the MOR1 gene expression is not affected.
Collapse
Affiliation(s)
- Shamseddin Ahmadi
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran,Corresponding author: Shamseddin Ahmadi. Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran. Tel: +98-87-33660075,
| | - Fatemeh Miraki
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Jalal Rostamzadeh
- Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| |
Collapse
|
9
|
Zhao J, Wang H, Song T, Yang Y, Gu K, Ma P, Zhang Z, Shen L, Liu J, Wang W. Thalidomide Promotes Morphine Efficacy and Prevents Morphine-Induced Tolerance in Rats with Diabetic Neuropathy. Neurochem Res 2016; 41:3171-3180. [DOI: 10.1007/s11064-016-2041-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/13/2016] [Accepted: 08/20/2016] [Indexed: 12/24/2022]
|
10
|
Knapman A, Connor M. Cellular signalling of non-synonymous single-nucleotide polymorphisms of the human μ-opioid receptor (OPRM1). Br J Pharmacol 2014; 172:349-63. [PMID: 24527749 DOI: 10.1111/bph.12644] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/21/2014] [Accepted: 02/07/2014] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED There is significant variability in individual responses to opioid drugs, which is likely to have a significant genetic component. A number of non-synonymous single-nucleotide polymorphisms (SNPs) in the coding regions of the μ-opioid receptor gene (OPRM1) have been postulated to contribute to this variability. Although many studies have investigated the clinical influences of these μ-opioid receptor variants, the outcomes are reported in the context of thousands of other genes and environmental factors, and we are no closer to being able to predict individual response to opioids based on genotype. Investigation of how μ-opioid receptor SNPs affect their expression, coupling to second messengers, desensitization and regulation is necessary to understand how subtle changes in receptor structure can impact individual responses to opioids. To date, the few functional studies that have investigated the consequences of SNPs on the signalling profile of the μ-opioid receptor in vitro have shown that the common N40D variant has altered functional responses to some opioids, while other, rarer, variants display altered signalling or agonist-dependent regulation. Here, we review the data available on the effects of μ-opioid receptor polymorphisms on receptor function, expression and regulation in vitro, and discuss the limitations of the studies to date. Whether or not μ-opioid receptor SNPs contribute to individual variability in opioid responses remains an open question, in large part because we have relatively little good data about how the amino acid changes affect μ-opioid receptor function. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
Affiliation(s)
- Alisa Knapman
- Australian School of Advanced Medicine, Macquarie University, Sydney, NSW, Australia
| | | |
Collapse
|
11
|
Vacca V, Marinelli S, Luvisetto S, Pavone F. Botulinum toxin A increases analgesic effects of morphine, counters development of morphine tolerance and modulates glia activation and μ opioid receptor expression in neuropathic mice. Brain Behav Immun 2013; 32:40-50. [PMID: 23402794 DOI: 10.1016/j.bbi.2013.01.088] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/28/2013] [Accepted: 01/31/2013] [Indexed: 02/02/2023] Open
Abstract
The use of botulinum neurotoxin type A (BoNT/A) against pain, with emphasis for its possible use in alleviating chronic pain, still represents an outstanding challenge for experimental research. In this study, we examined the effects of BoNT/A on morphine-induced tolerance during chronic morphine treatment in neuropathic CD1 mice subjected to sciatic nerve lesion according to the Chronic Constriction Injury (CCI) model of neuropathic pain. We measured the effects of BoNT/A on CCI-induced allodynia and hyperalgesia and on the expression of glial fibrillary acidic protein (GFAP, marker of astrocytes), complement receptor 3/cluster of differentiation 11b (CD11b, marker of microglia), and neuronal nuclei (NeuN) at the spinal cord level. We also analyzed the colocalized expression of GFAP, CD11b and NeuN with phosphorylated p-38 mitogen-activated protein kinase and with μ-opioid receptor (MOR). A single intraplantar injection of BoNT/A (15 pg/paw) into the injured hindpaw, the day before the beginning of chronic morphine treatment (9 days of twice daily injections of 40 mg/kg morphine), was able to counteract allodynia and enhancement of astrocytes expression/activation induced by CCI. In addition, BoNT/A increased the analgesic effect of morphine and countered morphine-induced tolerance during chronic morphine treatment. These effects were accompanied, in neurons, by re-expression of MORs that had been reduced by repeated morphine administration. The combinatory effects of BoNT/A and morphine could have relevant therapeutic implications for sufferers of chronic pain who could benefit of pain relief reducing tolerance due to repeated treatment with opiates.
Collapse
Affiliation(s)
- Valentina Vacca
- CNR - National Research Council of Italy, Cell Biology and Neurobiology Institute/IRCCS - Santa Lucia Foundation, Rome, Italy
| | | | | | | |
Collapse
|
12
|
Zhang Y, Donica CL, Standifer KM. Sex differences in the Nociceptin/Orphanin FQ system in rat spinal cord following chronic morphine treatment. Neuropharmacology 2012; 63:427-33. [PMID: 22575074 PMCID: PMC5009626 DOI: 10.1016/j.neuropharm.2012.04.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 04/18/2012] [Accepted: 04/23/2012] [Indexed: 12/24/2022]
Abstract
Nociceptin/Orphanin FQ (N/OFQ) appears to contribute to the development of morphine tolerance, as blockade of its actions will block or reverse the process. To better understand the contribution of N/OFQ to the development of morphine tolerance, this study examined the effect of chronic morphine treatment on levels of N/OFQ and levels and activity of the N/OFQ peptide (NOP) receptor in spinal cord (SC) from male and female rats. Both male and female Wistar rats showed less responsiveness to morphine after subcutaneous injection of escalating doses of morphine (10, 20, 40, 60 and 80 mg/kg, respectively) twice daily for five consecutive days. Male rats were more tolerant to the antinociceptive actions of morphine than females. The N/OFQ content of SC extracts was higher in females than in males, regardless of treatment; following chronic morphine treatment the difference in N/OFQ levels between males and females was more pronounced. N/OFQ content in cerebrospinal fluid (CSF) was reduced 40% in male and 16% in female rats with chronic morphine exposure, but increased in periaqueductal grey of both sexes. Chronic morphine treatment increased NOP receptor levels 173% in males and 137% in females, while decreasing affinity in both. Chronic morphine increased the efficacy of N/OFQ-stimulated [³⁵S]GTPγS binding to SC membranes from male rats, consistent with increased receptor levels. Taken together, these findings demonstrate sex differences in N/OFQ-NOP receptor expression and NOP receptor activity following chronic morphine treatment. They also suggest interplay between endogenous N/OFQ and chronic morphine treatment that results in nociceptive modulation.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA
| | - Courtney L. Donica
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA
| | - Kelly M. Standifer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73126, USA
| |
Collapse
|
13
|
Vacca V, Marinelli S, Eleuteri C, Luvisetto S, Pavone F. Botulinum neurotoxin A enhances the analgesic effects on inflammatory pain and antagonizes tolerance induced by morphine in mice. Brain Behav Immun 2012; 26:489-99. [PMID: 22281280 DOI: 10.1016/j.bbi.2012.01.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 01/05/2012] [Accepted: 01/05/2012] [Indexed: 11/30/2022] Open
Abstract
Over the recent years compelling evidence has accumulated indicating that botulinum neurotoxin serotype A (BoNT/A) results in analgesic effects on neuropathic as well as inflammatory pain, both in humans and in animal models. In the present study, the pharmacological interaction of BoNT/A with morphine in fighting inflammatory pain was investigated in mice using the formalin test. Moreover, the effects of BoNT/A on the tolerance-induced by chronic administration of morphine were tested and the behavioral effects were correlated with immunofluorescence staining of glial fibrillary acidic protein, the specific marker of astrocytes, at the spinal cord level. An ineffective dose of BoNT/A (2 pg/paw) combined with an ineffective dose of morphine (1 mg/kg) exerted a significant analgesic action both during the early and the late phases of formalin test. A single intraplantar injection of BoNT/A (15 pg/paw; i.pl.), administered the day before the beginning of chronic morphine treatment (7 days of s.c. injections of 20 mg/kg), was able to counteract the occurrence of tolerance to morphine. Moreover, BoNT/A reduces the enhancement of the expression of astrocytes induced by inflammatory formalin pain. Side effects of opiates, including the development of tolerance during repeated use, may limit their therapeutic use, the possibility of using BoNT/A for lowering the effective dose of morphine and preventing the development of opioid tolerance would have relevant implications in terms of potential therapeutic perspectives.
Collapse
Affiliation(s)
- Valentina Vacca
- CNR-National Research Council of Italy (Cell Biology and Neurobiology Institute IBCN)/IRCCS Fondazione Santa Lucia, Roma, Italy
| | | | | | | | | |
Collapse
|
14
|
Raehal KM, Schmid CL, Groer CE, Bohn LM. Functional selectivity at the μ-opioid receptor: implications for understanding opioid analgesia and tolerance. Pharmacol Rev 2011; 63:1001-19. [PMID: 21873412 DOI: 10.1124/pr.111.004598] [Citation(s) in RCA: 197] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Opioids are the most effective analgesic drugs for the management of moderate or severe pain, yet their clinical use is often limited because of the onset of adverse side effects. Drugs in this class produce most of their physiological effects through activation of the μ opioid receptor; however, an increasing number of studies demonstrate that different opioids, while presumably acting at this single receptor, can activate distinct downstream responses, a phenomenon termed functional selectivity. Functional selectivity of receptor-mediated events can manifest as a function of the drug used, the cellular or neuronal environment examined, or the signaling or behavioral measure recorded. This review summarizes both in vitro and in vivo work demonstrating functional selectivity at the μ opioid receptor in terms of G protein coupling, receptor phosphorylation, interactions with β-arrestins, receptor desensitization, internalization and signaling, and details on how these differences may relate to the progression of analgesic tolerance after their extended use.
Collapse
Affiliation(s)
- Kirsten M Raehal
- Molecular Therapeutics and Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA
| | | | | | | |
Collapse
|
15
|
Berger AC, Whistler JL. Morphine-induced mu opioid receptor trafficking enhances reward yet prevents compulsive drug use. EMBO Mol Med 2011; 3:385-97. [PMID: 21656686 PMCID: PMC3394511 DOI: 10.1002/emmm.201100144] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 03/29/2011] [Accepted: 03/31/2011] [Indexed: 01/08/2023] Open
Abstract
Morphine, heroin and other commonly abused opioids induce little mu opioid receptor (MOR) trafficking compared to endogenous opioids. We utilized knock-in mice expressing a mutant recycling MOR (RMOR) that desensitizes and is internalized in response to morphine to show that facilitating MOR trafficking not only enhances morphine reward but, despite this, reduces the development of addiction-like behaviours. To demonstrate this, we developed a novel model of the transition from controlled to compulsive drug use that recapitulates many features of human addiction, including persistent drug seeking despite adverse consequences and a decreased preference for alternative rewards. These behaviours emerged spontaneously in wild-type but not RMOR mice, and their intensity predicted the reinstatement of morphine seeking after extended abstinence, while prior morphine intake did not. These results confirm previous findings in the rat that addiction can be dissociated from both reward and consumption. Most importantly, these results demonstrate that one can simultaneously reduce the ‘addictiveness’ of morphine and enhance its desirable effects by promoting agonist-induced MOR trafficking.
Collapse
Affiliation(s)
- Amy Chang Berger
- Neuroscience Program, University of California, San Francisco, Ernest Gallo Clinic and Research Center, 5858 Horton Street, Suite 200, Emeryville, CA 94608, USA
| | | |
Collapse
|
16
|
He L, Whistler JL. Chronic ethanol consumption in rats produces opioid antinociceptive tolerance through inhibition of mu opioid receptor endocytosis. PLoS One 2011; 6:e19372. [PMID: 21602922 PMCID: PMC3094338 DOI: 10.1371/journal.pone.0019372] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 04/01/2011] [Indexed: 02/07/2023] Open
Abstract
It is well known that the mu-opioid receptor (MOR) plays an important role in the rewarding properties of ethanol. However, it is less clear how chronic ethanol consumption affects MOR signaling. Here, we demonstrate that rats with prolonged voluntary ethanol consumption develop antinociceptive tolerance to opioids. Signaling through the MOR is controlled at many levels, including via the process of endocytosis. Importantly, agonists at the MOR that promote receptor endocytosis, such as the endogenous peptides enkephalin and β-endorphin, show a reduced propensity to promote antinociceptive tolerance than do agonists, like morphine, which do not promote receptor endocytosis. These observations led us to examine whether chronic ethanol consumption produced opioid tolerance by interfering with MOR endocytosis. Indeed, here we show that chronic ethanol consumption inhibits the endocytosis of MOR in response to opioid peptide. This loss of endocytosis was accompanied by a dramatic decrease in G protein coupled receptor kinase 2 (GRK2) protein levels after chronic drinking, suggesting that loss of this component of the trafficking machinery could be a mechanism by which endocytosis is lost. We also found that MOR coupling to G-protein was decreased in ethanol-drinking rats, providing a functional explanation for loss of opioid antinociception. Together, these results suggest that chronic ethanol drinking alters the ability of MOR to endocytose in response to opioid peptides, and consequently, promotes tolerance to the effects of opioids.
Collapse
Affiliation(s)
- Li He
- Ernest Gallo Clinic and Research Center, University of California, Emeryville, California, United States of America
| | - Jennifer L. Whistler
- Ernest Gallo Clinic and Research Center, University of California, Emeryville, California, United States of America
- Department of Neurology, University of California, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
17
|
Feng B, Li Z, Wang JB. Protein kinase C-mediated phosphorylation of the μ-opioid receptor and its effects on receptor signaling. Mol Pharmacol 2011; 79:768-75. [PMID: 21212139 DOI: 10.1124/mol.110.069096] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Phosphorylation of the μ opioid receptor (MOPr), mediated by several protein kinases, is a critical process in the regulation of MOPr signaling. Although G protein-coupled receptor kinases are known to play an essential role in the agonist-induced phosphorylation and desensitization of MOPr, evidence suggests that other protein kinases, especially protein kinase C (PKC), also participate in the regulation of MOPr signaling. In this study, we investigated the biochemical nature and downstream effects of PKC-mediated MOPr phosphorylation. We observed in vitro phosphorylation of the MOPr C terminus by purified PKC. Protein mass spectrometry and site-directed mutagenesis implicated Ser363 of MOPr as the primary substrate for PKC, and this was confirmed in Chinese hamster ovary cells stably expressing full-length MOPr using an antibody that specifically recognizes phosphorylated Ser363. Alanine mutation of Ser363 did not affect the affinity of MOPr-ligand binding and the efficiency of receptor G-protein coupling. However, the S363A mutation attenuated the desensitization of receptor G-protein coupling induced by phorbol 12-myristate. Our research thus has identified a specific PKC phosphorylation site in MOPr and demonstrated that PKC-mediated phosphorylation of MOPr induces receptor desensitization at the G protein coupling level.
Collapse
Affiliation(s)
- Bo Feng
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA
| | | | | |
Collapse
|
18
|
Berger AC, Whistler JL. How to design an opioid drug that causes reduced tolerance and dependence. Ann Neurol 2010; 67:559-69. [PMID: 20437553 DOI: 10.1002/ana.22002] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Mu opioid receptor (MOR) agonists such as morphine are extremely effective treatments for acute pain. In the setting of chronic pain, however, their long-term utility is limited by the development of tolerance and physical dependence. Drug companies have tried to overcome these problems by simply "dialing up" signal transduction at the receptor, designing more potent and efficacious agonists and more long-lasting formulations. Neither of these strategies has proven to be successful, however, because the net amount of signal transduction, particularly over extended periods of drug use, is a product of much more than the pharmacokinetic properties of potency, efficacy, half-life, and bioavailability, the mainstays of traditional pharmaceutical screening. Both the quantity and quality of signal transduction are influenced by many regulated processes, including receptor desensitization, trafficking, and oligomerization. Importantly, the efficiency with which an agonist first stimulates signal transduction is not necessarily related to the efficiency with which it stimulates these other processes. Here we describe recent findings that suggest MOR agonists with diminished propensity to cause tolerance and dependence can be identified by screening drugs for the ability to induce MOR desensitization, endocytosis, and recycling. We also discuss preliminary evidence that heteromers of the delta opioid receptor and the MOR are pronociceptive, and that drugs that spare such heteromers may also induce reduced tolerance.
Collapse
Affiliation(s)
- Amy Chang Berger
- Department of Neurology, University of California, San Francisco, Ernest Gallo Clinic and Research Center, Emeryville, CA, USA
| | | |
Collapse
|
19
|
Jin WY, Yu LC. Involvement of protein kinase C in morphine tolerance at spinal levels of rats. ACS Chem Neurosci 2010; 1:122-8. [PMID: 22778820 DOI: 10.1021/cn900005d] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Accepted: 09/16/2009] [Indexed: 11/28/2022] Open
Abstract
The present study was performed to investigate the possible role of protein kinase C (PKC) in morphine tolerance at spinal levels of rats. Intrathecal injection of 10 μg of morphine induced increases in the hindpaw withdrawal latency (HWL) to noxious thermal and mechanical stimulation in rats. After intrathecal injections of 10 μg of morphine (twice a day) lasted for 5 days, the antinociceptive effects induced by intrathecal injections of morphine decreased significantly in rats. Interestingly, we found that there were significant increases in the content of PKC in the dorsal horn of the spinal cord and the dorsal root ganglion, but not in the ventral horn of the spinal cord, in rats with morphine tolerance determined by Western blot, suggesting that PKC is involved in morphine tolerance at spinal levels of rats. Furthermore, our results demonstrated that chronic intrathecal injection of the PKC inhibitor significantly inhibited the development of morphine tolerance. Moreover, we found that the maintenance of morphine tolerance was blocked by intrathecal administration of a PKC inhibitor in rats, and the inhibitory effects of the PKC inhibitor on morphine tolerance lasted for more than two days. Taken together, the present study clearly showed that PKC is involved in morphine tolerance at the spinal level of rats and that intrathecal administration of a PKC inhibitor can block the development and maintenance of morphine tolerance.
Collapse
Affiliation(s)
- Wu-Yang Jin
- Neurobiology Laboratory and National Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, PR China
| | - Long-Chuan Yu
- Neurobiology Laboratory and National Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing 100871, PR China
| |
Collapse
|
20
|
Hamabe W, Yamane H, Harada S, Tokuyama S. Involvement of kappa opioid receptors in the inhibition of receptor desensitization and PKC activation induced by repeated morphine treatment. J Pharm Pharmacol 2008; 60:1183-8. [PMID: 18718122 DOI: 10.1211/jpp.60.9.0010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Analgesic tolerance to morphine can develop from long-term use of this drug for the treatment of pain. Many reports have shown that stimulation of the kappa opioid receptor (KOR) suppresses development of analgesic tolerance to morphine. Here, we studied the KOR-mediated inhibition of morphine tolerance during repeated morphine treatment, with a focus on desensitization of the receptor. The development of analgesic tolerance to morphine during repeated morphine administration (10 mg kg(-1) s.c.) was completely suppressed by U-50488H (2 mg kg(-1) i.p.), a KOR agonist. The decrease in [35S] GTPgammaS binding activity stimulated by the mu opioid receptor (MOR) agonist [D-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin (DAMGO) was also significantly inhibited by U-50488H. These results indicate that stimulation of KOR caused by repeated morphine treatment either inhibits MOR desensitization or accelerates recycling of MOR on the cell surface, thereby suppressing morphine tolerance. Furthermore, we found that activity of protein kinase C (PKC) was significantly decreased in mice treated with both U-50488H and morphine. These results suggest that the mechanisms underlying KOR-mediated inhibition of analgesic tolerance to morphine may be partly due to suppression of PKC activation and prevention of receptor desensitization.
Collapse
Affiliation(s)
- Wakako Hamabe
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe, 650-8586, Japan
| | | | | | | |
Collapse
|
21
|
Berg KA, Patwardhan AM, Sanchez TA, Silva YM, Hargreaves KM, Clarke WP. Rapid modulation of micro-opioid receptor signaling in primary sensory neurons. J Pharmacol Exp Ther 2007; 321:839-47. [PMID: 17347322 DOI: 10.1124/jpet.106.116681] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Management of pain by opioid analgesics is confounded by central adverse effects that limit clinical dosages. Consequently, there is considerable interest to understand peripheral analgesic effects of opioids. The actions of opioids on peripheral sensory neurons have been difficult to study because of a general lack of effect of opioid agonists on nociceptor function in culture despite documented presence of opioid receptors. In this study, the micro-opioid receptor agonist, [D-Ala(2),N-MePhe(4),Gly-ol(5)]-enkephalin (DAMGO), did not alter guanosine 5'-O-(3-[(35)S]thio)-triphosphate (GTPgamma[(35)S]) binding, adenylyl cyclase activity, or neuropeptide release in primary cultures of rat trigeminal ganglion (TG). However, after brief exposure to bradykinin (BK), DAMGO stimulated GTPgamma[(35)S] binding and inhibited both prostaglandin E(2) (PGE(2))-stimulated adenylyl cyclase activity and BK/PGE(2)-stimulated neuropeptide release. The effect of BK was blocked by the B(2) antagonist HOE 140 [D-Arg[Hyp(3),Thi(5),D-Tic(7),Oic(8)]-bradykinin], but not by the B(1) antagonist, Lys-[Leu8]des-Arg9-BK, and was mimicked by the protease-activated receptor-2 agonist, Ser-Leu-Ile-Gly-Arg-Leu-NH(2), and by activation of protein kinase C (PKC) or by administration of arachidonic acid (AA). The enhanced responsiveness of micro-opioid receptor signaling by BK priming was blocked by both cyclooxygenase and PKC inhibitors; however, the effect of AA was blocked only by a cyclooxygenase inhibitor. The results indicate that micro-opioid receptor signaling in primary sensory TG neurons is enhanced by activation of phospholipase C-coupled receptors via a cyclooxygenase-dependent AA metabolite that is downstream of PKC.
Collapse
MESH Headings
- Adenylyl Cyclase Inhibitors
- Adenylyl Cyclases/metabolism
- Adrenergic beta-Antagonists/pharmacology
- Animals
- Arachidonic Acid/pharmacology
- Bradykinin/pharmacology
- Calcitonin Gene-Related Peptide/metabolism
- Capsaicin/pharmacology
- Cells, Cultured
- Cyclic AMP/metabolism
- Cyclooxygenase Inhibitors/pharmacology
- Dinoprostone/pharmacology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Enzyme Inhibitors/pharmacology
- Gene Expression/drug effects
- Inositol Phosphates/metabolism
- Male
- Neurons, Afferent/cytology
- Neurons, Afferent/drug effects
- Neurons, Afferent/metabolism
- Phorbol 12,13-Dibutyrate/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptor, Bradykinin B2/agonists
- Receptor, Bradykinin B2/genetics
- Receptor, Bradykinin B2/metabolism
- Receptor, PAR-2/agonists
- Receptor, PAR-2/metabolism
- Receptors, Opioid, mu/analysis
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Type C Phospholipases/antagonists & inhibitors
- Type C Phospholipases/metabolism
Collapse
Affiliation(s)
- Kelly A Berg
- Department of Pharmacology, MS 7764, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | |
Collapse
|
22
|
Wang ZJ, Wang LX. Phosphorylation: A molecular switch in opioid tolerance. Life Sci 2006; 79:1681-91. [PMID: 16831450 DOI: 10.1016/j.lfs.2006.05.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2005] [Revised: 04/26/2006] [Accepted: 05/24/2006] [Indexed: 02/07/2023]
Abstract
Protein phosphorylation is a key posttranslational modification mechanism controlling the conformation and activity of many proteins. Increasing evidence has implicated an essential role of phosphorylation by several major protein kinases in promoting and maintaining opioid tolerance. We review some of the most recent studies on protein kinase C (PKC), cyclic AMP dependent protein kinase A (PKA), calcium/calmodulin-dependent protein kinase II (CaMKII), protein kinase G (PKG), and G protein receptor kinase (GRK). These kinases act as the molecular switches to modulate opioid tolerance. Pharmacological interventions at one or more of the protein kinases and phosphatases may provide valuable strategies to improve opioid analgesia by attenuating tolerance to these drugs.
Collapse
Affiliation(s)
- Zaijie Jim Wang
- Department of Biopharmaceutical Sciences and Cancer Center, University of Illinois, Chicago, IL 60612, USA.
| | | |
Collapse
|
23
|
Ko SW, Jia Y, Xu H, Yim SJ, Jang DH, Lee YS, Zhao MG, Toyoda H, Wu LJ, Chatila T, Kaang BK, Zhuo M. Evidence for a role of CaMKIV in the development of opioid analgesic tolerance. Eur J Neurosci 2006; 23:2158-68. [PMID: 16630062 DOI: 10.1111/j.1460-9568.2006.04748.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
cAMP response-element binding protein (CREB), a transcription factor involved in learning, memory and drug addiction, is phosphorylated by calcium-calmodulin-dependent protein kinase IV (CaMKIV). Here, we show that CaMKIV-knockout (KO) mice developed less analgesic tolerance after chronic morphine administration with no alteration in physical dependence or acute morphine-induced analgesia. The increase in phosphorylated CREB expression observed in wild-type mice after chronic morphine was absent in CaMKIV-KO mice, while there was no difference in the expression or phosphorylation of the micro-opioid receptor between groups. Morphine-treated CaMKIV-KO mice showed less G-protein uncoupling from the micro-opioid receptor than did wild-type mice, while uncoupling was similar in control wild-type and KO mice. In addition, morphine reduced inhibitory transmission to a greater degree in CaMKIV-KO mice than in controls after chronic morphine exposure. Our results provide novel evidence for the role of CaMKIV in the development of opioid analgesic tolerance but not physical dependence.
Collapse
MESH Headings
- Analgesics, Opioid/administration & dosage
- Animals
- Animals, Newborn
- Behavior, Animal
- Blotting, Western/methods
- Calcium-Calmodulin-Dependent Protein Kinase Type 4
- Calcium-Calmodulin-Dependent Protein Kinases/deficiency
- Calcium-Calmodulin-Dependent Protein Kinases/physiology
- Conditioning, Operant/drug effects
- Conditioning, Operant/physiology
- Cyclic AMP Response Element-Binding Protein/metabolism
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Drug Tolerance
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Exploratory Behavior/physiology
- Guanosine 5'-O-(3-Thiotriphosphate)/pharmacokinetics
- Immunohistochemistry/methods
- Immunoprecipitation/methods
- In Vitro Techniques
- Male
- Membrane Potentials/drug effects
- Membrane Potentials/physiology
- Membrane Potentials/radiation effects
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Morphine/administration & dosage
- Neurons/drug effects
- Neurons/physiology
- Neurons/radiation effects
- Pain Measurement/methods
- Patch-Clamp Techniques/methods
- Radioligand Assay/methods
- Spinal Cord/cytology
- Sulfur Isotopes/pharmacokinetics
- Time Factors
Collapse
Affiliation(s)
- Shanelle W Ko
- Department of Physiology, Faculty of Medicine, University of Toronto, University of Toronto Centre for the Study of Pain, 1 King's College Circle, Medical Sciences Building Rm3342, Toronto, Canada, M5S 1A8
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wijesekara N, Tung A, Thong F, Klip A. Muscle cell depolarization induces a gain in surface GLUT4 via reduced endocytosis independently of AMPK. Am J Physiol Endocrinol Metab 2006; 290:E1276-86. [PMID: 16418206 DOI: 10.1152/ajpendo.00573.2005] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Contracting skeletal muscle increases glucose uptake to sustain energy demand. This is achieved through a gain in GLUT4 at the membrane, but the traffic mechanisms and regulatory signals involved are unknown. Muscle contraction is elicited by membrane depolarization followed by a rise in cytosolic Ca2+ and actomyosin activation, drawing on ATP stores. It is unknown whether one or more of these events triggers the rise in surface GLUT4. Here, we investigate the effect of membrane depolarization on GLUT4 cycling using GLUT4myc-expressing L6 myotubes devoid of sarcomeres and thus unable to contract. K+-induced membrane depolarization elevated surface GLUT4myc, and this effect was additive to that of insulin, was not prevented by inhibiting phosphatidylinositol 3-kinase (PI3K) or actin polymerization, and did not involve Akt activation. Instead, depolarization elevated cytosolic Ca2+, and the surface GLUT4myc elevation was prevented by dantrolene (an inhibitor of Ca2+ release from sarcoplasmic reticulum) and by extracellular Ca2+ chelation. Ca2+-calmodulin-dependent protein kinase-II (CaMKII) was not phosphorylated after 10 min of K+ depolarization, and the CaMK inhibitor KN62 did not prevent the gain in surface GLUT4myc. Interestingly, although 5'-AMP-activated protein kinase (AMPK) was phosphorylated upon depolarization, lowering AMPKalpha via siRNA did not alter the surface GLUT4myc gain. Conversely, the latter response was abolished by the PKC inhibitors bisindolylmaleimide I and calphostin C. Unlike insulin, K+ depolarization caused only a small increase in GLUT4myc exocytosis and a major reduction in its endocytosis. We propose that K+ depolarization reduces GLUT4 internalization through signals and mechanisms distinct from those engaged by insulin. Such a pathway(s) is largely independent of PI3K, Akt, AMPK, and CaMKII but may involve PKC.
Collapse
Affiliation(s)
- Nadeeja Wijesekara
- Programme in Cell Biology, The Hospital for Sick Children, 555 University Ave., Toronto, ON, Canada M5G 1X8
| | | | | | | |
Collapse
|
25
|
Matsumoto T, Nishiyama M, Kobayashi T, Kasuya Y, Kamata K. Effect of phorbol 12,13-dibutyrate on smooth muscle tone in rat stomach fundus. J Smooth Muscle Res 2005; 41:107-16. [PMID: 15988154 DOI: 10.1540/jsmr.41.107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We investigated the effects of phorbol 12,13-dibutyrate (PDBu), a typical protein kinase C (PKC) activator, on smooth muscle tone in the rat stomach fundus. In 5-hydroxytriptamine (5-HT)-precontracted stomach fundus strips, PDBu induced dose-dependent relaxation, but 4alpha-phorbol 12,13-didecanoate, a phorbol ester that does not activate PKC, did not induce relaxation. A PDBu-induced dose-dependent relaxation was also observed in strips precontracted with platelet-activating factor (PAF), carbachol, or 60 mM K+. In stomach fundus strips pretreated with PDBu, the contractile responses to 5-HT and PAF were completely blocked, but those induced by carbachol and endothelin-1 (ET-1) were only partially inhibited. In stomach fundus strips preincubated with carbachol in Ca2+-free medium, the Ca2+-induced contraction was decreased by preincubation with PDBu. In strips preincubated with 5-HT, PAF, or ET-1 in Ca2+-free medium, Ca2+-induced contractions were greatly inhibited by pretreatment with PDBu. These results suggest that in rat stomach fundus strips, PDBu-induced relaxation is mediated by activation of PKC. We speculate that a major factor mediating the relaxant action of PDBu in rat stomach fundus smooth muscle is represented by a reduction in Ca2+ influx via an inhibition of Ca2+ channels.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Tokyo, Japan
| | | | | | | | | |
Collapse
|
26
|
Xiong W, Gao L, Sapra A, Yu LC. Antinociceptive role of galanin in the spinal cord of rats with inflammation, an involvement of opioid systems. ACTA ACUST UNITED AC 2005; 132:85-90. [PMID: 16214241 DOI: 10.1016/j.regpep.2005.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2005] [Accepted: 09/08/2005] [Indexed: 11/17/2022]
Abstract
The present study investigated the role of galanin in the transmission of nociceptive information in the spinal cord of rats with inflammation. Bilateral decreases in hindpaw withdrawal latencies (HWLs) to thermal and mechanical stimulation were observed after acute inflammation induced by injection of carrageenan into the plantar region of the rat left hindpaw. Intrathecal injection of galanin induced significant increases in the HWLs to thermal and mechanical stimulation in rats with inflammation. The galanin-induced antinociceptive effect was more pronounced in rats with inflammation than that in intact rats. The antinociceptive effect of galanin was partly inhibited by intrathecal injection of naloxone. Furthermore, intrathecal administration of galantide, an antagonist of galanin receptor, could attenuate the antinociceptive effect induced by intraperitoneal injection of morphine, suggesting an involvement of opioid systems in the galanin-induced antinociception. The results indicate that galanin plays an important role in the transmission of nociceptive information in the spinal cord of rats with inflammation, and opioid systems are involved in the galanin-induced antinociception.
Collapse
Affiliation(s)
- Wei Xiong
- Laboratory of Neurobiology and National Laboratory of Biomembrane and Membrane Biotechnology, College Of Life Science, Peking University, Beijing 100871, China
| | | | | | | |
Collapse
|
27
|
Carey RM, Balcz BA, Lopez-Coviella I, Slack BE. Inhibition of dynamin-dependent endocytosis increases shedding of the amyloid precursor protein ectodomain and reduces generation of amyloid beta protein. BMC Cell Biol 2005; 6:30. [PMID: 16095541 PMCID: PMC1208872 DOI: 10.1186/1471-2121-6-30] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2005] [Accepted: 08/11/2005] [Indexed: 11/13/2022] Open
Abstract
Background The amyloid precursor protein (APP) is transported via the secretory pathway to the cell surface, where it may be cleaved within its ectodomain by α-secretase, or internalized within clathrin-coated vesicles. An alternative proteolytic pathway occurs within the endocytic compartment, where the sequential action of β- and γ-secretases generates the amyloid β protein (Aβ). In this study, we investigated the effects of modulators of endocytosis on APP processing. Results Human embryonic kidney cells were transfected with a dominant negative mutant of dynamin I, an important mediator of clathrin-dependent endocytosis, and APP proteolysis was analyzed. Overexpression of the mutant dynamin (dyn I K44A) resulted in increased shedding of the APP ectodomain (sAPPα), accumulation of the C-terminal α-secretase product C83, and a reduction in the release of Aβ. Levels of mature APP on the cell surface were increased in cells expressing dyn I K44A, and internalization of surface-immunolabeled APP, assessed by fluorescence microscopy, was inhibited. Dynamin is a substrate for protein kinase C (PKC), and it was hypothesized that activators of PKC, which are known to stimulate α-secretase-mediated cleavage of APP, might exert their effects by inhibiting dynamin-dependent endocytosis. However, the internalization of surface-biotinylated APP was unaffected by treatment of cells with phorbol 12-myristate 13-acetate in the presence of the α-secretase inhibitor TAPI-1. Conclusion The results indicate that APP is internalized by a dynamin-dependent process, and suggest that alterations in the activity of proteins that mediate endocytosis might lead to significant changes in Aβ production.
Collapse
Affiliation(s)
- Robyn M Carey
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, 715 Albany Street, Rm. L808, Boston MA 02118, USA
| | - Brigitte A Balcz
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, 715 Albany Street, Rm. L808, Boston MA 02118, USA
- Gemeinnützige Salzburger Landeskliniken Betriebsgesellschaft mbH, Universitätsklinik für Innere Medizin III, Paracelsus Medizinische Privatuniversität, Müllner Hauptstrasse 48, A-5020 Salzburg, Austria
| | - Ignacio Lopez-Coviella
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, 715 Albany Street, Rm. L808, Boston MA 02118, USA
| | - Barbara E Slack
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, 715 Albany Street, Rm. L808, Boston MA 02118, USA
| |
Collapse
|
28
|
Ueda H. Locus-specific involvement of anti-opioid systems in morphine tolerance and dependence. Ann N Y Acad Sci 2005; 1025:376-82. [PMID: 15542739 DOI: 10.1196/annals.1307.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Opioid tolerance and addiction could be discussed as two types of plasticity or counteradaptation, at the cellular level and through neuronal circuits. Cellular counteradaptation mechanisms include receptor desensitization through phosphorylation and endocytosis and through altered gene expression. The former mechanisms are related to the acute tolerance mechanisms, while the latter to chronic one. From current studies, it is known that various phosphorylation steps, such as protein kinase C (PKC) and G protein-coupled receptor (GPCR) kinase (GRK) regulate endocytosis. Of interest is that there are some differences in the physiological roles between opioid receptor endocytosis and other GPCR ones. Endocytosis of the opioid receptor is conceived as a recycling and resensitization step rather than the desensitization step. PKC phosphorylation inhibits endocytosis (PKC hypothesis). Therefore the PKC inhibitor attenuates acute analgesic tolerance. The agonist, which shows high-endocytosis stimulation, therefore makes less significant tolerance liability (RAVE hypothesis). Chronic tolerance is more likely related to the mechanisms through plastic modulation of neuronal circuits, where anti-opioidergic neurons are involved. The knockout mice lacking the receptors for anti-opioidergic nociceptin/orphanin FQ (N/OFQ) or glutamatergic neurons show weak or no morphine tolerance and dependence. As their gene expression or protein expression increases during chronic morphine treatments, we propose the hypothesis that the enhanced anti-opioid system may cause a counteradaptation to show tolerance and dependence. By a novel electroporation technique to deliver the receptor into the brain of knockout mice, we succeeded in determining the specific locus for the site of anti-opioid (through GluRepsilon1 or NR2A) action. All these results suggest that enhanced anti-opioid systems may contribute to the development of morphine tolerance and dependence, and their contributions could be brain locus specific.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Division of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8521, Japan.
| |
Collapse
|
29
|
Tegeder I, Geisslinger G. Opioids as modulators of cell death and survival--unraveling mechanisms and revealing new indications. Pharmacol Rev 2005; 56:351-69. [PMID: 15317908 DOI: 10.1124/pr.56.3.2] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Opioids are powerful analgesics but also drugs of abuse. Because opioid addicts are susceptible to certain infections, opioids have been suspected to suppress the immune response. This was supported by the finding that various immune-competent cells express opioid receptors and undergo apoptosis when treated with opioid alkaloids. Recent evidence suggests that opioids may also effect neuronal survival and proliferation or migrating properties of tumor cells. A multitude of signaling pathways has been suggested to be involved in these extra-analgesic effects of opioids. Growth-promoting effects were found to be mediated through Akt and Erk signaling cascades. Death-promoting effects have been ascribed to inhibition of nuclear factor-kappaB, increase of Fas expression, p53 stabilization, cytokine and chemokine release, and activation of nitric oxide synthase, p38, and c-Jun-N-terminal kinase. Some of the observed effects were inhibited with opioid receptor antagonists or pertussis toxin; others were unaffected. It is still unclear whether these properties are mediated through typical opioid receptor activation and inhibitory G-protein-signaling. The present review tries to unravel controversial findings and provides a hypothesis that may help to integrate diverse results.
Collapse
Affiliation(s)
- Irmgard Tegeder
- Pharmazentrum Frankfurt, Institut für Klinische Pharmakologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt, Germany.
| | | |
Collapse
|
30
|
Herring D, Huang R, Singh M, Dillon GH, Leidenheimer NJ. PKC modulation of GABAA receptor endocytosis and function is inhibited by mutation of a dileucine motif within the receptor β2 subunit. Neuropharmacology 2005; 48:181-94. [PMID: 15695157 DOI: 10.1016/j.neuropharm.2004.09.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2004] [Revised: 08/04/2004] [Accepted: 09/16/2004] [Indexed: 10/26/2022]
Abstract
The modulation of GABAA receptors by protein kinase C is complex and involves effects on both ion channel function and receptor trafficking. Although PKC regulates receptor cell surface expression the mechanism is not well understood. Using immunofluorescence studies in HEK 293 cells, we demonstrate that activation of PKC by the phorbol ester PMA promotes receptor endocytosis and is dependent on the presence of a gamma subunit. This endocytosis is blocked by the dominant negative dynamin mutant K44A indicating that PKC-induced receptor endocytosis involves the dynamin endocytic pathway. Mutation of a dileucine motif within the receptor beta2 subunit inhibits the effect of PKC activation on receptor endocytosis. Using patch clamp analysis, we show that PKC activation produces a robust inhibition of GABA-gated chloride currents in cells expressing wildtype GABAA receptors, but it is ineffective in modulating receptors lacking the dileucine motif. Furthermore, the introduction into the patch pipette of a 10-amino acid peptide corresponding to the dileucine motif present in the receptor beta2 subunit prevents PKC modulation of wildtype recombinant receptors. Furthermore, in cerebral cortical neuronal slices inclusion of this peptide in the patch pipette prevents PKC modulation of native GABAA receptors. Using limited chymotrypsin digestion assays, we also show that PKC increases receptor internalization in primary cultures of cerebral cortical neurons. Lastly, PKC inhibitors do not block constitutive receptor endocytosis or affect GABA-gated chloride currents suggesting that PKC-dependent phosphorylation is not required for GABAA receptor endocytosis but plays a modulatory role in the process.
Collapse
Affiliation(s)
- Dina Herring
- Department of Pharmacology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, USA
| | | | | | | | | |
Collapse
|
31
|
Endo K, Matsumoto T, Kobayashi T, Kasuya Y, Kamata K. Diabetes-related changes in contractile responses of stomach fundus to endothelin-1 in streptozotocin-induced diabetic rats. J Smooth Muscle Res 2005; 41:35-47. [PMID: 15855738 DOI: 10.1540/jsmr.41.35] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The contractile response of the stomach fundus to endothelin-1 (ET-1) was examined in streptozotocin (STZ)-induced diabetic rats. In STZ-diabetic rats (versus age-matched control rats) (a) ET-1 caused a longer-lasting contraction of stomach fundus strips, and (b) in the dose-response curve, the ET-1-induced contraction was significantly greater for a given concentration (3 x 10(-7) to 10(-7) M). Although repeated application of ET-1 led to desensitization, the desensitization was less pronounced in STZ-diabetic rats than in the controls. The density of the binding sites for [(125)I]-ET-1 was increased in the diabetic stomach fundus (versus the controls), but Kd values were similar between the two groups. The ET(B) receptor mRNA expression level was significantly increased in the diabetic stomach fundus. These results suggest that the diabetes-related enhancement of the ET-1-induced contraction of the stomach fundus may be due to an increase in the ET(B) receptor population.
Collapse
Affiliation(s)
- Kazuki Endo
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Tokyo, Japan
| | | | | | | | | |
Collapse
|
32
|
Li PF, Hao YS, Huang DA, Liu XH, Liu SL, Li G. Morphine-promoted survival of CEMx174 cells in early stages of SIV infection in vitro: involvement of the multiple molecular mechanisms. Toxicol In Vitro 2004; 18:449-56. [PMID: 15130602 DOI: 10.1016/j.tiv.2004.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2003] [Accepted: 01/02/2004] [Indexed: 10/26/2022]
Abstract
Progression of HIV infections to AIDS is a complex process and it differs considerably among individuals infected with HIV, influenced by both genetic and environmental factors. Opiates have been implicated to be a cofactor in HIV infections leading to AIDS. However, little is known about the molecular mechanisms involved in the effects of opioids on HIV infected immune cells. Cell cycle analysis was carried out by flow cytometry, the phosphorylation of mitogen-activated protein kinases ERK1 and ERK2 was detected by Western blotting assay, and changes of calcium concentration were monitored by scanning intracellular fluorescence intensity. In response to the treatment with morphine, SIV-infected cells were accumulated in G1 phase. Morphine increased the content of intracellular calcium in a time-dependent manner. In addition, morphine also elevated the levels of PKC activity and phosphorylated ERK1/2. Therefore, it is implicated that the calcium-PKC-MAPK cascade is involved in morphine-prolonged survival of SIV-infected cells in the early stages of virus infection.
Collapse
Affiliation(s)
- Ping-Feng Li
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Xueyuan Rd 38, Beijing 100083, China
| | | | | | | | | | | |
Collapse
|
33
|
Xu J, Li PF, Liu XH, Li G. Morphine aggravates the apoptosis of simian immunodeficiency virus infected CEM x174 cells in the prolonged culture in vitro. Int Immunopharmacol 2004; 4:1805-16. [PMID: 15531296 DOI: 10.1016/j.intimp.2004.07.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2004] [Revised: 07/19/2004] [Accepted: 07/23/2004] [Indexed: 11/19/2022]
Abstract
This study was designed to assess the in vitro effects of morphine on the lymphocytes infected with SIV. CEM x174 cells were cotreated with morphine and simian immunodeficiency virus (SIVmac239). Cells were cultured for 96 h and the effects of morphine on the viability of infected cells were determined. At the concentration of 1 micromol/l, morphine could inhibit the proliferation of CEM x174 cells at the culture of 72 h. The stronger effect was observed in the case of viral infection. During 72 h SIV loading, the cells were accumulated in S phase in all SIV infected groups. The S arrest was observed in every experimental group and statistically different from normal groups (P<0.05). The results from annexin V binding assay showed that SIV infection resulted in a lower proportion of vital cells and higher mortality compared with corresponding control (P<0.01). Morphine failed to induce detectable alteration in the cell cycle profile of viral infected cells. Western blotting showed that the synthesis of intracellular p53 and bax protein was gradually up-regulated in the virus-loading period of 72 h. Naloxone had an apparent additive rather than antagonistic effect on the morphine-associated enhancement of bax expression. The ratio of bax/bcl-2 proteins appeared to tilt the balance toward apoptosis. At 72 h of infection, 1 micromol/l of morphine significantly elevated the level of caspase-3. These results indicated that the alteration in the balance of intracellular apoptotic and anti-apoptotic elements is one of the reasons of accelerated progression of acquired immunodeficiency syndrome (AIDS) by opioids abuse.
Collapse
Affiliation(s)
- Jin Xu
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100083, China
| | | | | | | |
Collapse
|
34
|
Abstract
Opioid receptors belong to the large superfamily of seven transmembrane-spanning (7TM) G protein-coupled receptors (GPCRs). As a class, GPCRs are of fundamental physiological importance mediating the actions of the majority of known neurotransmitters and hormones. Opioid receptors are particularly intriguing members of this receptor family. They are activated both by endogenously produced opioid peptides and by exogenously administered opiate compounds, some of which are not only among the most effective analgesics known but also highly addictive drugs of abuse. A fundamental question in addiction biology is why exogenous opioid drugs, such as morphine and heroin, have a high liability for inducing tolerance, dependence, and addiction. This review focuses on many aspects of opioid receptors with the aim of gaining a greater insight into mechanisms of opioid tolerance and dependence.
Collapse
Affiliation(s)
- Maria Waldhoer
- Ernest Gallo Clinic and Research Center, University of California, San Francisco, Emeryville, California 94608, USA.
| | | | | |
Collapse
|
35
|
Bailey CP, Kelly E, Henderson G. Protein kinase C activation enhances morphine-induced rapid desensitization of mu-opioid receptors in mature rat locus ceruleus neurons. Mol Pharmacol 2004; 66:1592-8. [PMID: 15361548 DOI: 10.1124/mol.104.004747] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent studies have shown that morphine, in contrast to other agonists at the mu-opioid receptor, causes very little rapid mu-opioid receptor desensitization or internalization in adult rat mammalian neurons, raising important questions about how morphine tolerance is induced. Here we show that morphine can indeed cause marked rapid desensitization of mu-opioid receptors in mature rat locus ceruleus neurons when protein kinase C is also activated. Thus, activation of Gq-coupled M3 muscarinic receptors or application of a phorbol ester enhanced the desensitization of the mu-opioid receptor-evoked potassium current in rat locus ceruleus neurons. The enhancement of desensitization was reversible by the protein kinase C inhibitors chelerythrine and 2-[1-(3-dimethylaminopropyl)-1H-indol-3-yl]-3-(1H-indol-3-yl)-maleimide (GF109203X) and resulted from an effect at the level of the mu-opioid receptor rather than the potassium channel. This is the first finding that morphine can induce rapid mu-opioid receptor desensitization in adult rat neurons, and because reduced protein kinase C activity in vivo attenuates morphine tolerance, we propose that G-protein coupled receptor cross-talk and the level of protein kinase C activity may play critical roles in the desensitization of the mu-opioid receptor and could underlie the development of morphine tolerance.
Collapse
Affiliation(s)
- Christopher P Bailey
- Department of Pharmacology, University of Bristol, University Walk, Bristol, United Kingdom.
| | | | | |
Collapse
|
36
|
Eisinger DA, Schulz R. Extracellular Signal-Regulated Kinase/Mitogen-Activated Protein Kinases Block Internalization of δ-Opioid Receptors. J Pharmacol Exp Ther 2004; 309:776-85. [PMID: 14742744 DOI: 10.1124/jpet.103.061788] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Translocation of G protein-coupled receptors (GPCRs) from the cell membrane to cytosol depends on the kind of ligand activating the receptor. This principle is clearly demonstrated for opioid receptors, because diverse opiate agonists rapidly induce receptor internalization, whereas morphine almost fails. We report here the impact of mitogen-activated protein (MAP) kinase isoforms extracellular signal-regulated kinase (ERK)1/2 on the internalization of delta-opioid receptors (DORs) expressed in human embryonic kidney (HEK)293 cells. Receptor activation by etorphine turned out to transiently phosphorylate ERK/MAP kinases and bring about DOR internalization within 20 min. In contrast, prolonged exposure of HEK293 cells to morphine excited persistent phosphorylation of ERK/MAP kinases, and those cells failed to internalize the opioid receptor. When ERK/MAP kinase phosphorylation was blocked by 2'-Amino-3'-methoxyflavone (PD98059), morphine gained the ability to strongly induce DOR endocytosis. The importance of activated MAP kinases for DOR internalization is further demonstrated by glutamate and paclitaxel because these substances induce phosphorylation of ERK1/2 and concomitantly prevent DOR sequestration by etorphine. In addition, receptor internalization by morphine was facilitated by inhibition of protein kinase C and opioid-mediated transactivation of epidermal growth factor receptor (EGFR), both activating ERK/MAP kinases by opioids. The mechanism affording DOR internalization by PD98059 may relate to arrestin, which uncouples GPCRs and thus triggers receptor internalization. Arrestin considerably translocates toward the cell membrane upon DOR activation by morphine in presence of the MAP kinase blocker, but it fails in the absence of PD98059. We conclude that ERK/MAP kinase activity prevents opioid receptor desensitization and sequestration by blocking arrestin 2 interaction with activated DORs.
Collapse
Affiliation(s)
- Daniela A Eisinger
- Institute of Pharmacology, Toxicology and Pharmacy, University of Munich, Germany.
| | | |
Collapse
|
37
|
Ueda H, Inoue M, Mizuno K. New approaches to study the development of morphine tolerance and dependence. Life Sci 2004; 74:313-20. [PMID: 14607259 DOI: 10.1016/j.lfs.2003.09.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Morphine is now believed not to cause tolerance and dependence when it is appropriately used in clinic. However, in terminal cancer pain, patients' analgesic tolerance to morphine is developed due to the use of high doses of morphine for complete blockade of pain. At higher doses, morphine has more opportunity to show serious side effects, which worsens quality of life (QOL), and leads to the use of potent analgesic adjuvants to reduce the morphine dosage. Here we attempt to summarize recent studies of the molecular basis of morphine tolerance and dependence, and to discuss whether these mechanisms could provide new molecular targets as analgesic adjuvants. They include protein kinase C inhibitor, opioid agonist with low RAVE value, and antagonists of antiopioid receptors (GluRepsilon1 or nociceptin/OFQ receptor). In addition, we demonstrate new approaches to find further candidates of such molecular targets. These approaches include the visualization of neuronal networks in the downstream of opioid neurons by use of the WGA transgene technique and the single cell dissection technique to get new genes involved in plasticity during morphine tolerance and dependence.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Division of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, 852-8521, Nagasaki, Japan.
| | | | | |
Collapse
|
38
|
Li PF, Hao YS, Zhang FX, Liu XH, Liu SL, Li G. Signaling pathway involved in methionine enkephalin-promoted survival of lymphocytes infected by simian immunodeficiency virus in the early stage in vitro. Int Immunopharmacol 2004; 4:79-90. [PMID: 14975362 DOI: 10.1016/j.intimp.2003.10.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2003] [Revised: 10/16/2003] [Accepted: 10/16/2003] [Indexed: 11/24/2022]
Abstract
Methionine enkephalin, the endogenous opioid peptide, has a diversity of effects on the immune system. Although the biological effects of the pentapeptide have been well documented, little is known about the intracellular events involved in the effects of opioids on human immunodeficiency virus (HIV) infected immune cells. In the present investigation, the possible mechanism of apoptosis alleviated by exposure of methionine enkephalin at 1 micromol/l to CEM x 174 cells, the hybrid lymphocytes, infected with simian immunodeficiency virus (SIV) in vitro is elucidated. Apoptosis and cell cycle analysis is carried out by flow cytometry, the phosphorylation of mitogen-activated protein kinases (MAPK) ERK1 and ERK2 is detected by Western blotting assay, and changes of calcium concentration were analyzed using the calcium-sensitive dye Fluo-3 AM. The results exhibit that methionine enkephalin at the concentrations of 1 micromol/l increase remarkably the proportion of vital cells and decrease the apoptotic cells based on annexin V binding assay. In response to the treatment with methionine enkephalin, SIV-infected cells display a prolonged survival and are accumulated in G1 phase. Methionine enkephalin increase obviously the content of intracellular calcium in normal cells within 1-2 min and maintains a high level within monitoring time. However, the intracellular calcium reaches the highest level at 1 min and subsequently decline to background in SIV infected group. In addition, methionine enkephalin also elevates the levels of protein kinase C (PKC) activity and phosphorylated extracellular signal-regulated kinase (ERK) 1/2. It is proposed that calcium-PKC-MAPK cascade is involved in methionine enkephalin-prolonged survival of SIV-infected cells in the early stages of virus infection. The results provide a further evidence for potential use of methionine enkephalin on the therapy of Acquired Immunodeficiency Syndrome (AIDS).
Collapse
Affiliation(s)
- Ping-feng Li
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, Beijing 100083, China
| | | | | | | | | | | |
Collapse
|
39
|
Harlan RE, Kailas SR, Tagoe CEF, Garcia MM. Morphine actions in the rat forebrain: role of protein kinase C. Brain Res Bull 2004; 62:285-95. [PMID: 14709343 DOI: 10.1016/j.brainresbull.2003.09.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Acute administration of morphine induces expression of the immediate-early gene (IEG) c-Fos in dorsomedial striatum, portions of cerebral cortex, and in several midline-intralaminar thalamic nuclei, partly via a trans-synaptic mechanism that involves activation of glutamate receptors. Because activation of protein kinase C (PKC) may occur following the activation of glutamate receptors, we determined whether pharmacological inhibition of PKC would attenuate morphine-induced c-Fos expression, and whether acute administration of morphine would induce translocation of PKC. The selective PKC antagonist NPC 15437 given 30 min prior to morphine significantly decreased morphine-induced c-Fos expression in striatum and cingulate cortex, but not in centrolateral thalamus. In another experiment, rats were given an acute dose of morphine, and immunocytochemical analysis was performed for the betaI and betaII isoforms of PKC. Morphine induced a rapid and transient translocation of PKC betaII, but not betaI, from perinuclear spots to plasma membrane in numerous cortical and striatal neurons. Prior administration of naloxone blocked this response. Ultrastructural studies confirmed translocation from Golgi apparatus to plasma membrane 15 min after morphine injection. Double immunocytochemistry at the light microscopic level demonstrated co-localization of translocated PKC betaII and c-Fos in some cortical neurons 90 min after morphine injection. These results support a role for PKC, especially PKC betaII, in the rapid effects of morphine on the brain.
Collapse
Affiliation(s)
- Richard E Harlan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, LA 70112, USA.
| | | | | | | |
Collapse
|
40
|
RUNG-RUANGKIJKRAI T, FUJIKURA D, KON Y, IWANAGA T. Cellular expression of src-suppressed C kinase substrate (SSeCKS) in the peripheral nervous system and sensory organs. Biomed Res 2004. [DOI: 10.2220/biomedres.25.155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
41
|
Méndez M, Leriche M, Carlos Calva J. Acute ethanol administration transiently decreases [3H]-DAMGO binding to mu opioid receptors in the rat substantia nigra pars reticulata but not in the caudate-putamen. Neurosci Res 2003; 47:153-60. [PMID: 14512140 DOI: 10.1016/s0168-0102(03)00188-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ethanol's actions in brain have been suggested to be partially mediated by a mechanism involving the ethanol-induced activation of the endogenous opioid system. Opioid systems, which are closely linked with dopamine transmission, are thought to be affected by ethanol through alterations in the processing, release, and/or receptor binding of opioid peptides. We studied the effects of a single acute dose of ethanol on rat nigrostriatal mu opioid receptors by quantitative receptor autoradiography, using [3H] [D-Ala(2),MePhe(4),Gly-ol(5)]-enkephalin ([3H]-DAMGO) as radioligand. [3H]-DAMGO binding was significantly decreased in the pars reticulata of the substantia nigra 1 h after ethanol administration. Ethanol exposure did not affect [3H]-DAMGO binding neither in the pars compacta of the substantia nigra nor in the caudate-putamen at any time tested after drug administration. The observed effects may reflect ethanol-induced changes in ligand binding affinity (Kd) or in receptor density (Bmax). Early and transitory ethanol-induced changes of mu receptors in the substantia nigra pars reticulata may be related to regulation of dopaminergic nigrostriatal transmission and contribute to determine brain sensitivity to the drug.
Collapse
Affiliation(s)
- Milagros Méndez
- Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Subdirección de Investigaciones Clínicas, Calzada México Xochimilco 101, Col. San Lorenzo Huipulco, 14370, Mexico D F, México.
| | | | | |
Collapse
|
42
|
Smith FL, Javed RR, Elzey MJ, Dewey WL. The expression of a high level of morphine antinociceptive tolerance in mice involves both PKC and PKA. Brain Res 2003; 985:78-88. [PMID: 12957370 DOI: 10.1016/s0006-8993(03)03170-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We have previously reported that intracerebroventricular (i.c.v.) injection of either a PKC or PKA inhibitor completely reversed the expression of 5- to 8-fold morphine antinociceptive tolerance. We developed a model of 45-fold morphine tolerance that included a 75-mg morphine pellet and twice daily morphine injections. PKC inhibitor doses of bisindolylmaleimide I and Gö-7874 that completely reversed 8-fold tolerance only partly reversed the 45-fold level of antinociceptive tolerance. A component of tolerance was resistant to PKC inhibition, since even higher inhibitor doses failed to further reverse the high level of morphine tolerance. In addition, the 45-fold tolerance was only partly reversed by the PKA inhibitor KT-5720 at a dose previously cited by others to reverse 5-fold tolerance. Another PKA inhibitor 4-cyano-3-methylisoquinoline only partly reversed the morphine tolerance as well. In other experiments PKC and PKA inhibitors were co-administered together to determine their effectiveness for completely reversing the 45-fold level of morphine tolerance. Co-administering either bisindolylmaleimide I with KT-5720, or Gö-7874 with KT-5720, completely reversed the high level of tolerance. The high level of morphine tolerance was also completely reversed by co-administering Gö-7874 with 4-cyano-3-methylisoquinoline. Thus, high levels of morphine tolerance may reflect increases in protein phosphorylation by the terminal kinases of both the adenylyl cyclase and phosphatidylinositol cascades in brain and spinal cord areas critical to the expression of antinociception.
Collapse
Affiliation(s)
- Forrest L Smith
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, P.O. Box 980613, Richmond, VA 23298-0613, USA.
| | | | | | | |
Collapse
|
43
|
Ueda H. [Pain and QOL--morphine-tolerance and morphine-resistant neuropathic pain]. Nihon Yakurigaku Zasshi 2003; 122:192-200. [PMID: 12939537 DOI: 10.1254/fpj.122.192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Morphine is now said to have no problematic side effects such as analgesic tolerance and physical dependence for cancer pain patients in clinic, as far as it is appropriately used. However, sub-sensitivity to morphine might be developed when higher doses of morphine are used for terminal cancer pain patients. Along with the severity of cancer, the nature of pain becomes changed to neuropathic pain, which is resistant to morphine or NSAIDS. In order to safely use morphine in the clinic, we need to know how morphine tolerance and neuropathic pain are developed and what adjuvants could be used to completely suppress the pain. Here I overview the proposed mechanisms for morphine tolerance and neuropathic pain in relation to the availability of analgesic adjuvants.
Collapse
Affiliation(s)
- Hiroshi Ueda
- Division of Molecular Pharmacology and Neuroscience, Nagasaki University, Graduate School of Biomedical Sciences, Japan.
| |
Collapse
|
44
|
Ohsawa M, Mizoguchi H, Narita M, Nagase H, Dun NJ, Tseng LF. Involvement of beta-arrestin-2 in modulation of the spinal antinociception induced by mu-opioid receptor agonists in the mouse. Neurosci Lett 2003; 346:13-6. [PMID: 12850536 DOI: 10.1016/s0304-3940(03)00591-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Beta-arrestins have been suggested to regulate mu-, delta-, and kappa-opioid receptor-mediated responses. In the present study, we examined the effects of pretreatment with beta-arrestin-2 antibody on tail-flick inhibition induced by opioid receptor agonists in the mouse spinal cord. Intrathecal (i.t.) pretreatment with beta-arrestin-2 antibody potentiated the antinociception induced by i.t.-administered mu-opioid receptor agonists [D-Ala(2),NMePhe(4),Gly-ol(5)]enkephalin (DAMGO) and endomorphin-1, but not endomorphin-2, the delta-opioid receptor agonist [D-Ala(2)]deltorphin II or the kappa-opioid receptor agonist U50,488H. The present result suggests that beta-arrestin-2 may tonically down-regulate a selected population of mu-opioid receptors activated by endomorphin-1 or DAMGO in the mouse spinal cord.
Collapse
Affiliation(s)
- Masahiro Ohsawa
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | |
Collapse
|
45
|
Inoue M, Mishina M, Ueda H. Locus-specific rescue of GluRepsilon1 NMDA receptors in mutant mice identifies the brain regions important for morphine tolerance and dependence. J Neurosci 2003; 23:6529-36. [PMID: 12878694 PMCID: PMC6740642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Tolerance and physical dependence caused by chronic treatment of narcotics are good models to study basic neuronal plasticity. Activation of the NMDA subtype of the glutamate receptor has been implicated as an anti-opioid system in the development of morphine analgesic tolerance and dependence. The present study examines the specific role of the epsilon1 subunit of the NMDA receptor using mice lacking the gene encoding epsilon1 subunit of the NMDA receptor (GluRepsilon1-/- mice). GluRepsilon1-/- mice showed significant enhancement and prolongation of morphine anti-nociception, compared with wild-type GluRepsilon1+/+ mice. GluRepsilon1-/- mice also showed a marked loss of the analgesic tolerance after repeated morphine treatments. In C57BL/6J mice treated with chronic morphine after tolerance paradigm, the GluRepsilon1 protein expression significantly increased in periaqueductal gray matter (PAG), ventral tegmental area (VTA) and nucleus accumbens (NAc), but not amygdala or hippocampus. The rescue of GluRepsilon1 protein by electroporation into the PAG and VTA, but not NAc of GluRepsilon1-/- mice significantly reversed morphine analgesic tolerance liability. Similar attempts were also performed in the naloxone-precipitated physical dependence paradigm. GluRepsilon1-/- mice showed marked loss of typical withdrawal abstinence behaviors, and significant enhancement of GluRepsilon1 protein expression was only observed in NAc by chronic morphine treatments after dependence paradigm. The rescue of GluRepsilon1 protein by electroporation into the NAc of GluRepsilon1-/- mice significantly reversed the loss of abstinence behaviors. These findings suggest that GluRepsilon1 has locus-specific roles in the development of morphine analgesic tolerance and physical dependence.
Collapse
Affiliation(s)
- Makoto Inoue
- Division of Molecular Pharmacology and Neuroscience, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8521, Japan
| | | | | |
Collapse
|
46
|
Abstract
Recent discoveries in opioid pharmacology help explain the enormous variability in clinical responses to these powerful analgesics. Although there is only one m opioid receptor gene, splice variants of that gene's expression result in a panoply of different functioning receptors. Other sources of variable response include polymorphisms in the m opioid receptor regulatory region, and pharmacokinetic differences because of cytochrome P-450 mono-oxygenase heterogeneity. Analgesic tolerance is likely the key phenomenon limiting the benefit of opioids. A plethora of intracellular pathways affects this. Among them are the N-methyl-D-aspartate receptor, protein kinase C gamma activity, nitric oxide synthase, and GM1 ganglioside content of the neuronal membrane. Clinical studies undercut the routine use of meperidine in most settings. Other studies have shown better ways to diminish opioid side effects.
Collapse
MESH Headings
- Analgesics, Opioid/adverse effects
- Analgesics, Opioid/therapeutic use
- Animals
- Clinical Trials as Topic
- Drug Tolerance
- Humans
- Pain/drug therapy
- Pain/physiopathology
- Receptors, Opioid/drug effects
- Receptors, Opioid/genetics
- Receptors, Opioid/physiology
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/physiology
- Trans-Activators/genetics
Collapse
Affiliation(s)
- Eric Chevlen
- Cancer Care Center, St. Elizabeth Hospital, 1044 Belmont Avenue, Youngstown, OH 44501, USA.
| |
Collapse
|
47
|
Smith FL, Javed R, Elzey MJ, Welch SP, Selley D, Sim-Selley L, Dewey WL. Prolonged reversal of morphine tolerance with no reversal of dependence by protein kinase C inhibitors. Brain Res 2002; 958:28-35. [PMID: 12468027 DOI: 10.1016/s0006-8993(02)03394-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The phosphatidylinositol (PI) cascade plays a pivotal role in mediating behavioral tolerance to the antinociceptive effects of morphine. Earlier we reported that antinociceptive tolerance was completely reversed 30 min after the administration of inhibitors of each step in the PI cascade. The aim of this study was to determine whether injection of a single dose of protein kinase C (PKC) inhibitor would elicit a prolonged reversal of morphine tolerance for up to 24 h. Three days after implantation of placebo- or 75-mg morphine pellets, mice received intracerebroventricular (i.c.v.) injections of vehicle or PKC inhibitor drug. Morphine challenge doses were then administered 4, 8 and 24 h later to test for tolerance reversal. In non-tolerant mice, Gö-7874 and sangivamycin had no effect on the potency of morphine. However, Gö-7874 and sangivamycin significantly reversed morphine tolerance at 4, 8 and 24 h. In addition, the role of PKC in morphine physical dependence was determined. Gö-7874 and sangivamycin by themselves did not precipitate spontaneous morphine withdrawal. Therefore, experiments were conducted to determine whether the PKC inhibitors would block naloxone-precipitated withdrawal. However, neither a 30-min nor a 24-h pretreatment with Gö-7874 or sangivamycin blocked naloxone withdrawal. Our results along with other publications indicate that PKC is a pivotal kinase essential for maintaining animals in an opioid tolerant state. Finally, the use of persistent PKC inhibitors that lasted for 24 h demonstrated that the neuronal systems in these animals did not adapt by increasing the activity of other protein kinase cascades to re-establish morphine tolerance.
Collapse
Affiliation(s)
- Forrest L Smith
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, PO Box 980613, Richmond, VA 23298-0613, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Ono T, Inoue M, Rashid MH, Sumikawa K, Ueda H. Stimulation of peripheral nociceptor endings by low dose morphine and its signaling mechanism. Neurochem Int 2002; 41:399-407. [PMID: 12213227 DOI: 10.1016/s0197-0186(02)00047-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In this report, we demonstrated that peripheral application of very low dose (amol ranges) of morphine induced flexor response through a substance P (SP) release at the nociceptor endings in mice. The intraplantar (i.pl.) application of morphine produced flexor response in a dose-dependent manner from 0.1 to 1000amol. The mu-opioid receptor (MOP-R) agonist [D-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO) also produced dose-dependent flexor response in same dose ranges. Morphine-induced flexor responses were markedly inhibited by naloxone and D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr amide (CTOP) both MOP-R antagonists and by intrathecal injection of antisense oligodeoxynucleotide (AS-ODN) for MOP-R which is expected to reduce the receptor expression in sensory nerve endings. Prior incubation with capsaicin, a depletor of SP from polymodal C fibers and [(+)-(2S,3S)-(2-methoxybenzylamino)-2-phenylpiperidine] (CP-99994), a tachykinin 1 receptor antagonist, also blocked the morphine-induced flexor responses. Moreover, pertussis toxin (PTX) which inactivates G(alpha)(i/o); [(1-[6-([(17b)-3-methoxyestra-1,3,5(10)-trien-17-yl]amino)hexyl]-1H-pyrrole-2,5-dione)] (U-73122), an inhibitor of phospholipase C (PLC); ethyleneglycol-bis(beta-aminoethyl ether) N,N,N',N'-tetraacetic acid (EGTA), a Ca(2+) chelating agent; xestospongin C, a membrane-permeable inositol trisphosphate (InsP(3)) receptor antagonist inhibited the morphine-flexor responses. However, thapsigargin, a depletor of intracellular Ca(2+) concentration and diphenhydramine, a histamine (His) H1 receptor antagonist, were unable to block the morphine-induced flexor responses. These results suggest that extremely low doses of morphine can stimulate sensory nerve endings through activation of peripheral MOP-R and its downstream mechanisms include activation of PLC through a SP release from polymodal C fibers.
Collapse
Affiliation(s)
- Takeshi Ono
- Department of Anesthesiology, Nagasaki University School of Medicine, Nagasaki, Japan
| | | | | | | | | |
Collapse
|
49
|
Abstract
This paper is the twenty-fourth installment of the annual review of research concerning the opiate system. It summarizes papers published during 2001 that studied the behavioral effects of the opiate peptides and antagonists. The particular topics covered this year include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology(Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, CUNY, 65-30 Kissena Blvd., Flushing, NY 11367, USA.
| | | |
Collapse
|
50
|
Bhattacharyya S, Puri S, Miledi R, Panicker MM. Internalization and recycling of 5-HT2A receptors activated by serotonin and protein kinase C-mediated mechanisms. Proc Natl Acad Sci U S A 2002; 99:14470-5. [PMID: 12388782 PMCID: PMC137907 DOI: 10.1073/pnas.212517999] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Serotonin (5-HT), a major neurotransmitter, has a large number of G protein-coupled receptors in mammals. On activation by exposure to their ligand, 5-HT(2) receptor subtypes increase IP(3) levels and undergo desensitization and internalization. To visualize the receptor in cells during these processes, we have constructed a 5-HT(2A)-enhanced GFP (SR2-GFP) fusion receptor. We show that this fusion receptor undergoes internalization on exposure to its natural ligand, 5-HT. Because 5-HT(2A) receptors activate the phospholipase C pathway, we studied the effect of protein kinase C (PKC) on the internalization process and found that activation of PKC by its specific activator phorbol 12-myristate 13-acetate, in the absence of 5-HT, leads to internalization of the receptor. Moreover, inhibition of PKC by its inhibitor sphingosine in the presence of 5-HT prevents the internalization process, suggesting that activation of PKC is sufficient and necessary for the internalization of 5-HT(2A) receptors. We also show that SR2-GFP recycles back to the plasma membrane after 5-HT-dependent internalization, suggesting a mechanism for resensitization. In addition, receptors that have been internalized on addition of phorbol 12-myristate 13-acetate in the absence of 5-HT also recycle to the surface, with a time course similar to that seen after activation of the receptors by 5-HT. Our study suggests that 5-HT(2A) receptors internalize and return to the surface after both serotonin- and PKC-mediated processes. This study reveals a role for PKC in receptor internalization and also shows that 5-HT(2A) receptors are recycled.
Collapse
Affiliation(s)
- Samarjit Bhattacharyya
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bellary Road, Bangalore 560065, India
| | | | | | | |
Collapse
|