1
|
Pereira-Silva R, Teixeira-Pinto A, Neto FL, Martins I. μ-Opioid Receptor Activation at the Dorsal Reticular Nucleus Shifts Diffuse Noxious Inhibitory Controls to Hyperalgesia in Chronic Joint Pain in Male Rats. Anesthesiology 2024; 140:1176-1191. [PMID: 38381969 DOI: 10.1097/aln.0000000000004956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
BACKGROUND The dorsal reticular nucleus is a pain facilitatory area involved in diffuse noxious inhibitory control (DNIC) through opioidergic mechanisms that are poorly understood. The hypothesis was that signaling of μ-opioid receptors is altered in this area with prolonged chronic inflammatory pain and that this accounts for the loss of DNICs occurring in this condition. METHODS Monoarthritis was induced in male Wistar rats (n = 5 to 9/group) by tibiotarsal injection of complete Freund's adjuvant. The immunolabeling of µ-opioid receptors and the phosphorylated forms of µ-opioid receptors and cAMP response element binding protein was quantified. Pharmacologic manipulation of μ-opioid receptors at the dorsal reticular nucleus was assessed in DNIC using the Randall-Selitto test. RESULTS At 42 days of monoarthritis, μ-opioid receptor labeling decreased at the dorsal reticular nucleus, while its phosphorylated form and the phosphorylated cAMP response element binding protein increased. [d-Ala2, N-Me-Phe4, Gly5-ol]-enkephalin acetate (DAMGO) enhanced DNIC analgesia in normal animals (means ± SD: pre-DNIC: 126.9 ± 7.0 g; DNIC - DAMGO: 147.5 ± 8.0 g vs. DNIC + DAMGO: 198.1 ± 19.3 g; P < 0.001), whereas it produced hyperalgesia in monoarthritis (pre-DNIC: 67.8 ± 7.5 g; DNIC - DAMGO: 70.6 ± 7.7 g vs. DNIC + DAMGO: 32.2 ± 2.6 g; P < 0.001). An ultra-low dose of naloxone, which prevents the excitatory signaling of the μ-opioid receptor, restored DNIC analgesia in monoarthritis (DNIC - naloxone: 60.0 ± 6.1 g vs. DNIC + naloxone: 98.0 ± 13.5 g; P < 0.001), compared to saline (DNIC - saline: 62.5 ± 5.2 g vs. DNIC + saline: 64.2 ± 3.8 g). When injected before DAMGO, it restored DNIC analgesia and decreased the phosphorylated cAMP response element binding protein in monoarthritis. CONCLUSIONS The dorsal reticular nucleus is likely involved in a facilitatory pathway responsible for DNIC hyperalgesia. The shift of μ-opioid receptor signaling to excitatory in this pathway likely accounts for the loss of DNIC analgesia in monoarthritis. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Raquel Pereira-Silva
- Institute for Research and Innovation in Health (i3S) of the University of Porto, Porto, Portugal; Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine - Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Armando Teixeira-Pinto
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia; Centre for Kidney Research, Kids Research Institute, Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Fani L Neto
- Institute for Research and Innovation in Health (i3S) of the University of Porto, Porto, Portugal; Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine - Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Isabel Martins
- Institute for Research and Innovation in Health (i3S) of the University of Porto, Porto, Portugal; Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine - Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
2
|
Xie W, He H, Hong J, Feng C, Li W, Li Y. Effect of Preadministration of Nalmefene on Sufentanil-Induced Cough During Induction of General Anesthesia in Patients Undergoing Breast Surgery: A Double-Blind Randomized Controlled Trial. Drug Des Devel Ther 2024; 18:1865-1874. [PMID: 38828019 PMCID: PMC11144414 DOI: 10.2147/dddt.s462710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
Purpose This study was designed to investigate the effects of preadministration of nalmefene before general anesthesia induction on sufentanil-induced cough (SIC) in patients undergoing breast surgery. Patients and Methods A total of 105 patients scheduled for elective breast surgery under general anesthesia were selected and randomly assigned into three groups: normal saline (Group C), low-dose nalmefene 0.1 μg·kg-1 (Group LN), and high-dose nalmefene 0.25 μg·kg-1 (Group HN). Sufentanil 0.5 μg·kg-1 was injected intravenously within 2 s after 5 min of intervention. The count and severity of cough within 2 min after sufentanil injection, as well as the time to first cough, were recorded. In addition, we also collected intraoperative hemodynamic data, postoperative pain scores, the incidence of receiving rescue analgesics, and side effects up to 24 h after surgery. Results Compared to Group C, the incidence of SIC was significantly lower in Group LN and HN (64.7% vs 30.3% and 14.7%, respectively; P < 0.001), but no significant difference was observed between the two groups (P=0.126). Compared to Group C, the risk factors decreased by 53.4% (95% confidence interval [CI] =0.181-0.735, P=0.008) in Group LN and by 75.9% (95% CI=0.432-0.898, P=0.001) in Group HN. Of the patients with SIC, less frequent SIC within 2 min after induction and a lower proportion of severe coughs were observed than Group C (P < 0.05), and no difference was detected between Group LN and HN. Additionally, the onset time to the first SIC did not differ significantly between the groups. Intraoperative hemodynamic data, postoperative pain scores, and side effects in the first 24 h did not differ among the groups. Conclusion Preadministration of nalmefene prior to induction of general anesthesia effectively suppressed SIC in patients undergoing breast surgery, without affecting intraoperative hemodynamic fluctuation and postoperative pain intensity.
Collapse
Affiliation(s)
- Weiji Xie
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Honglian He
- Department of Anesthesiology, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, 510060, People’s Republic of China
| | - Jishuang Hong
- Department of Anesthesiology, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, 510060, People’s Republic of China
| | - Chengfei Feng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Wei Li
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Yongchun Li
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| |
Collapse
|
3
|
Wang W, Ma X, Du W, Lin R, Li Z, Jiang W, Wang LY, Worley PF, Xu T. Small G-Protein Rheb Gates Mammalian Target of Rapamycin Signaling to Regulate Morphine Tolerance in Mice. Anesthesiology 2024; 140:786-802. [PMID: 38147625 DOI: 10.1097/aln.0000000000004885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
BACKGROUND Analgesic tolerance due to long-term use of morphine remains a challenge for pain management. Morphine acts on μ-opioid receptors and downstream of the phosphatidylinositol 3-kinase signaling pathway to activate the mammalian target of rapamycin (mTOR) pathway. Rheb is an important regulator of growth and cell-cycle progression in the central nervous system owing to its critical role in the activation of mTOR. The hypothesis was that signaling via the GTP-binding protein Rheb in the dorsal horn of the spinal cord is involved in morphine-induced tolerance. METHODS Male and female wild-type C57BL/6J mice or transgenic mice (6 to 8 weeks old) were injected intrathecally with saline or morphine twice daily at 12-h intervals for 5 consecutive days to establish a tolerance model. Analgesia was assessed 60 min later using the tail-flick assay. After 5 days, the spine was harvested for Western blot or immunofluorescence analysis. RESULTS Chronic morphine administration resulted in the upregulation of spinal Rheb by 4.27 ± 0.195-fold (P = 0.0036, n = 6), in turn activating mTOR by targeting rapamycin complex 1 (mTORC1). Genetic overexpression of Rheb impaired morphine analgesia, resulting in a tail-flick latency of 4.65 ± 1.10 s (P < 0.0001, n = 7) in Rheb knock-in mice compared to 10 s in control mice (10 ± 0 s). Additionally, Rheb overexpression in spinal excitatory neurons led to mTORC1 signaling overactivation. Genetic knockout of Rheb or inhibition of mTORC1 signaling by rapamycin potentiated morphine-induced tolerance (maximum possible effect, 52.60 ± 9.56% in the morphine + rapamycin group vs. 16.60 ± 8.54% in the morphine group; P < 0.0001). Moreover, activation of endogenous adenosine 5'-monophosphate-activated protein kinase inhibited Rheb upregulation and retarded the development of morphine-dependent tolerance (maximum possible effect, 39.51 ± 7.40% in morphine + metformin group vs. 15.58 ± 5.79% in morphine group; P < 0.0001). CONCLUSIONS This study suggests spinal Rheb as a key molecular factor for regulating mammalian target of rapamycin signaling. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Wenying Wang
- Department of Anesthesiology, Sixth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaqing Ma
- Department of Anesthesiology, Sixth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjie Du
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Raozhou Lin
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zhongping Li
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Wei Jiang
- Department of Anesthesiology, Sixth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu-Yang Wang
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada; and Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Paul F Worley
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tao Xu
- Department of Anesthesiology, Sixth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China; and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
4
|
Costa AR, Tavares I, Martins I. How do opioids control pain circuits in the brainstem during opioid-induced disorders and in chronic pain? Implications for the treatment of chronic pain. Pain 2024; 165:324-336. [PMID: 37578500 DOI: 10.1097/j.pain.0000000000003026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 07/07/2023] [Indexed: 08/15/2023]
Abstract
ABSTRACT Brainstem areas involved in descending pain modulation are crucial for the analgesic actions of opioids. However, the role of opioids in these areas during tolerance, opioid-induced hyperalgesia (OIH), and in chronic pain settings remains underappreciated. We conducted a revision of the recent studies performed in the main brainstem areas devoted to descending pain modulation with a special focus on the medullary dorsal reticular nucleus (DRt), as a distinctive pain facilitatory area and a key player in the diffuse noxious inhibitory control paradigm. We show that maladaptive processes within the signaling of the µ-opioid receptor (MOR), which entail desensitization and a switch to excitatory signaling, occur in the brainstem, contributing to tolerance and OIH. In the context of chronic pain, the alterations found are complex and depend on the area and model of chronic pain. For example, the downregulation of MOR and δ-opioid receptor (DOR) in some areas, including the DRt, during neuropathic pain likely contributes to the inefficacy of opioids. However, the upregulation of MOR and DOR, at the rostral ventromedial medulla, in inflammatory pain models, suggests therapeutic avenues to explore. Mechanistically, the rationale for the diversity and complexity of alterations in the brainstem is likely provided by the alternative splicing of opioid receptors and the heteromerization of MOR. In conclusion, this review emphasizes how important it is to consider the effects of opioids at these circuits when using opioids for the treatment of chronic pain and for the development of safer and effective opioids.
Collapse
Affiliation(s)
- Ana Rita Costa
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
- IBMC-Institute of Molecular and Cell Biology, University of Porto, Porto, Portugal
- I3S- Institute of Investigation and Innovation in Health, University of Porto, Porto, Portugal. Costa is now with the Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden and Science for Life Laboratory, Solna, Sweden
| | - Isaura Tavares
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
- IBMC-Institute of Molecular and Cell Biology, University of Porto, Porto, Portugal
- I3S- Institute of Investigation and Innovation in Health, University of Porto, Porto, Portugal. Costa is now with the Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden and Science for Life Laboratory, Solna, Sweden
| | - Isabel Martins
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Porto, Portugal
- IBMC-Institute of Molecular and Cell Biology, University of Porto, Porto, Portugal
- I3S- Institute of Investigation and Innovation in Health, University of Porto, Porto, Portugal. Costa is now with the Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden and Science for Life Laboratory, Solna, Sweden
| |
Collapse
|
5
|
Rupp A, Young E, Chadwick AL. Low-dose naltrexone's utility for non-cancer centralized pain conditions: a scoping review. PAIN MEDICINE (MALDEN, MASS.) 2023; 24:1270-1281. [PMID: 37302106 PMCID: PMC10628981 DOI: 10.1093/pm/pnad074] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/24/2023] [Accepted: 06/02/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND At low doses, naltrexone (LDN) has been shown to modulate inflammation through the interruption of microglial cell activation within the central nervous system. One of the most likely contributors to centralized pain is changes in microglial cell processing. Therefore, it has been postulated that LDN can be used to manage patients with pain resulting from central sensitization due to this relationship. This scoping review aims to synthesize the relevant study data for LDN as a novel treatment strategy for various centralized pain conditions. METHODS A comprehensive literature search was conducted in PubMed, Embase, and Google Scholar, guided by the Scale for Assessment of Narrative Review Articles (SANRA) criteria. RESULTS Forty-seven studies related to centralized pain conditions were identified. Many of the studies were case reports/series and narrative reviews, but a few randomized control trials have been conducted. Overall, the body of evidence revealed improvement in patient-reported pain severity and in outcomes related to hyperalgesia, physical function, quality of life, and sleep. Variability in dosing paradigms and the time to patient response was present in the reviewed studies. CONCLUSIONS Evidence synthesized for this scoping review supports the ongoing use of LDN for the treatment of refractory pain in various centralized chronic pain conditions. Upon review of the currently available published studies, it is apparent that further high-quality, well-powered randomized control trials need to be conducted to establish efficacy, standardization for dosing, and response times. In summary, LDN continues to offer promising results in the management of pain and other distressing symptoms in patients with chronic centralized pain conditions.
Collapse
Affiliation(s)
- Adam Rupp
- Department of Physical Medicine and Rehabilitation, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Erin Young
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Andrea L Chadwick
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, KS 66160, United States
| |
Collapse
|
6
|
Baratzadeh M, Danialy S, Abtin S, Manaheji H. Naloxone could limit morphine hypersensitivity: Considering the molecular mechanisms. Neuropeptides 2023; 100:102345. [PMID: 37172403 DOI: 10.1016/j.npep.2023.102345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/18/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Naloxone has been used as an opioid antagonist to prevent multiple adverse side effects of opioid-like tolerance and hyperalgesia. This study has investigated naloxone combined with morphine to limit pain hypersensitivity. In addition, the expression of brain-derived neurotrophic factor (BDNF) and K+ Cl- cotransporter2 (KCC2) were also studied. METHODS Forty-eight adult male Wistar rats (180-220 g) were divided into eight groups, with six rats in each group. Rats were divided into two tolerance and hyperalgesia groups; the sham group, the morphine group, the treatment group (naloxone along with morphine), and the sham group (naloxone along with saline) for eight consecutive days. Tail-flick test was performed on days 1, 5, and 8, and the plantar test on days 1 and 10. On days 8 and 10, the lumbar segments of the spinal cord were collected, and BDNF and KCC2 expression were analyzed using western blotting and immunohistochemistry, respectively. RESULTS Results showed that tolerance and hyperalgesia developed following eight days of repeated morphine injection. BDNF expression significantly increased, but KCC2 was downregulated. Co-administration of naloxone and morphine decreased tolerance and hyperalgesia by decreasing BDNF and increasing KCC2 expression, respectively. CONCLUSION This study suggests that BDNF and KCC2 may be candidate molecules for decreased morphine tolerance and hyperalgesia.
Collapse
Affiliation(s)
- Mojgan Baratzadeh
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Evin, Tehran, Iran
| | - Samira Danialy
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Evin, Tehran, Iran
| | - Shima Abtin
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Evin, Tehran, Iran
| | - Homa Manaheji
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Evin, Tehran, Iran.
| |
Collapse
|
7
|
Wang J, Zheng K, Wen Q, Sun J. Sufentanil combined with nalmefene reduce the adverse events in recovery period of patients undergoing uvulopalatopharyngoplasty - A randomized controlled trial. Heliyon 2023; 9:e13241. [PMID: 36755590 PMCID: PMC9900260 DOI: 10.1016/j.heliyon.2023.e13241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Background: The opioid receptors related to analgesia are mainly μ recipient, the μ receptor has a two-way mode of action: manifested by opioid analgesics and μ1 Receptor binding produces the desired analgesic effect, and μ2 Receptor binding may cause respiratory depression, nausea and vomiting and other adverse reactions. Nalmefene is an opioid receptor antagonist. Low dose Nalmefene has priority over μ2 Receptor binding makes opioid analgesics turn more to μ1 receptor binding, which is reversed μ2 Receptor mediated nausea, vomiting, respiratory depression and other adverse reactions, but does not reverse the analgesic effect. We assume that sufentanil combined with nalmefene could reduce respiratory adverse events in the patients who underwent uvulopalatopharyngoplasty during recovery. Methods: Patients with UPPP under general anesthesia were selected, and divided into control group (group C) and nalmefene group (group N) randomly. Patients in group N received sufentanil 0.1 μg/kg and nalmefene hydrochloride 0.25μg/kg at the end of the operation. Additionally, patients in group C received sufentanil 0.1 μg/kg at the end of the operation. The heart rate (HR) and mean arterial pressure (MAP) were observed and recorded before operation, immediately after extubation, and 5 min after extubation. The breathing recovery time, tracheal extubation time, consciousness score (Ramsay score、sedation-agitation scale (SAS)) and visual analog score (VAS) were carefully recorded at 5 min after extubation, and the various adverse reactions were monitored during the recovery period. Results: Ninety-six patients were finished our study finally. The breathing recovery and extubation time of group N was significantly shorter than those of group C (P < 0.05). The Ramsay score after extubation in group N was significantly lower as compared to that in group C (P < 0.05). The sedation-agitation scale of group N was observed to be significantly higher than that of group C (P < 0.05). Moreover, the incidences of respiratory depression, nausea and vomiting during the recovery period was significantly less than those in group C (P < 0.05). Conclusion: Nalmefene combined with sufentanil can significantly reduce adverse reactions in the cases of patients after UPPP, which may be beneficial to improve the quality and safety of emergence after general anesthesia.
Collapse
Affiliation(s)
- Jin Wang
- Department of Anesthesiology, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Kang Zheng
- Department of Anesthesiology, Nanjing Pukou District Hospital of Chinese Medicine, Nanjing 210000, China
| | - Quan Wen
- Department of Anesthesiology, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Jie Sun
- Department of Anesthesiology, Zhongda Hospital, Southeast University, Nanjing 210009, China,Corresponding author.
| |
Collapse
|
8
|
Jia Z, Chen Y, Gao T, Yuan Y, Zheng Y, Xie Y, Wang G, Yu Y, Zhang L. Nalmefene vs. dexmedetomidine for prevention of postoperative hyperalgesia in patients undergoing laparoscopic gynecological surgery with remifentanil infusion: A randomized double-blind controlled trial. Front Pharmacol 2023; 14:1131812. [PMID: 36762101 PMCID: PMC9905252 DOI: 10.3389/fphar.2023.1131812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Intraoperative remifentanil infusion may paradoxically induce post-surgical hyperalgesia. Dexmedetomidine reportedly reduces opioid-induced hyperalgesia. Nalmefene selectively reverses several side-effects of opioids without impairing analgesia. Herein, this randomized, double-blind controlled trial investigated whether nalmefene, dexmedetomidine, and both drugs combined prevent remifentanil-induced hyperalgesia. One hundred and fifty patients undergoing elective laparoscopic gynecological surgery under desflurane anesthesia randomly received either intraoperative sufentanil 0.20 μg kg-1 (Group S), or remifentanil 0.20 μg kg-1 min-1 (Group R), or remifentanil and pre-anesthesia nalmefene 0.20 μg kg-1 (Group N), or remifentanil and pre-anesthesia dexmedetomidine 0.50 μg kg-1 (Group D), or remifentanil and the combination of dexmedetomidine 0.25 μg kg-1 and nalmefene 0.10 μg kg-1 (Group DN). The threshold of postoperative mechanical hyperalgesia (primary outcome) was measured with von Frey filaments. We also recorded pain intensity, analgesic consumptions, hyperalgesic area, and side-effects for 24 h postoperatively. Compared with Group S, remifentanil reduced hyperalgesic threshold on the forearm [mean 89.4 (SD 13.7) vs. 62.2 (10.7) g, p < 0.001] at postoperative 24 h. Pain threshold on the forearm at postoperative 24 h was significantly lower in Group R than in Groups N, D and DN [62.2 (10.7) vs. 71.1 (12.3), 72.4 (12.9) and 78.0 (13.8) g]. Compared with Group R, Postoperative pain intensity, analgesic consumption and hyperalgesic area were lower likewise in Groups D and DN. However, the incidence of intraoperative bradycardia was lower and post-anesthesia recovery time was shorter in Group DN than Group D. Preoperative therapy of dexmedetomidine and nalmefene combined attenuates postoperative hyperalgesia in patients undergoing laparoscopic gynecological surgery under desflurane-remifentanil anesthesia.
Collapse
Affiliation(s)
- Zhen Jia
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yi Chen
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Tianyu Gao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yuan Yuan
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yuxin Zheng
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yegong Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Research Institute of Anesthesiology, Tianjin, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Research Institute of Anesthesiology, Tianjin, China,*Correspondence: Linlin Zhang, ; Yonghao Yu,
| | - Linlin Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China,Tianjin Research Institute of Anesthesiology, Tianjin, China,*Correspondence: Linlin Zhang, ; Yonghao Yu,
| |
Collapse
|
9
|
Hummig W, Baggio DF, Lopes RV, dos Santos SMD, Ferreira LEN, Chichorro JG. Antinociceptive effect of ultra-low dose naltrexone in a pre-clinical model of postoperative orofacial pain. Brain Res 2023; 1798:148154. [DOI: 10.1016/j.brainres.2022.148154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/10/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
10
|
Hamad AS, Edward EA, Sheta E, Aboushleib HM, Bahey-El-Din M. Iron Acquisition Proteins of Pseudomonas aeruginosa as Potential Vaccine Targets: In Silico Analysis and In Vivo Evaluation of Protective Efficacy of the Hemophore HasAp. Vaccines (Basel) 2022; 11:vaccines11010028. [PMID: 36679873 PMCID: PMC9864456 DOI: 10.3390/vaccines11010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/12/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Pseudomonas aeruginosa (PA) is a Gram-negative pathogen responsible for fatal nosocomial infections worldwide. Iron is essential for Gram-negative bacteria to establish an infection. Therefore, iron acquisition proteins (IAPs) of bacteria are attractive vaccine targets. METHODOLOGY A "Reverse Vaccinology" approach was employed in the current study. Expression levels of 37 IAPs in various types of PA infections were analyzed in seven previously published studies. The IAP vaccine candidate was selected based on multiple criteria, including a high level of expression, high antigenicity, solubility, and conservation among PA strains, utilizing suitable bioinformatics analysis tools. The selected IAP candidate was recombinantly expressed in Escherichia coli and purified using metal affinity chromatography. It was further evaluated in vivo for protection efficacy. The novel immune adjuvant, naloxone (NAL), was used. RESULTS AND DISCUSSION HasAp antigen met all the in silico selection criteria, being highly antigenic, soluble, and conserved. In addition, it was the most highly expressed IAP in terms of average fold change compared to control. Although HasAp did excel in the in silico evaluation, subcutaneous immunization with recombinant HasAp alone or recombinant HasAp plus NAL (HasAP-NAL) did not provide the expected protection compared to controls. Immunized mice showed a low IgG2a/IgG1 ratio, indicating a T-helper type 2 (Th2)-oriented immune response that is suboptimal for protection against PA infections. Surprisingly, the bacterial count in livers of both NAL- and HasAp-NAL-immunized mice was significantly lower than the count in the HasAp and saline groups. The same trend was observed in kidneys and lungs obtained from these groups, although the difference was not significant. Such protection could be attributed to the enhancement of innate immunity by NAL. CONCLUSIONS We provided a detailed in silico analysis of IAPs of PA followed by in vivo evaluation of the best IAP, HasAp. Despite the promising in silico results, HasAp did not provide the anticipated vaccine efficacy. HasAp should be further evaluated as a vaccine candidate through varying the immunization regimens, models of infection, and immunoadjuvants. Combination with other IAPs might also improve vaccination efficacy. We also shed light on several highly expressed promising IAPs whose efficacy as vaccine candidates is worthy of further investigation.
Collapse
Affiliation(s)
- Abdelrahman S. Hamad
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria P.O. Box 25435, Egypt
| | - Eva A. Edward
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria P.O. Box 25435, Egypt
| | - Eman Sheta
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria P.O. Box 21131, Egypt
| | - Hamida M. Aboushleib
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria P.O. Box 25435, Egypt
| | - Mohammed Bahey-El-Din
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria P.O. Box 25435, Egypt
- Correspondence:
| |
Collapse
|
11
|
Marwari S, Kowalski C, Martemyanov KA. Exploring pharmacological inhibition of G q/11 as an analgesic strategy. Br J Pharmacol 2022; 179:5196-5208. [PMID: 35900909 PMCID: PMC9633401 DOI: 10.1111/bph.15935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 07/06/2022] [Accepted: 07/14/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Misuse of opioids has greatly affected our society. One potential solution is to develop analgesics that act at targets other than opioid receptors. These can be used either as stand-alone therapeutics or to improve the safety profile of opioid drugs. Previous research showed that activation of Gq/11 proteins by G-protein coupled receptors has pro-nociceptive properties, suggesting that blockade of Gq/11 signalling could be beneficial for pain control. The aim of this study was to test this hypothesis pharmacologically by using potent and selective Gq/11 inhibitor YM-254890. EXPERIMENTAL APPROACH We used a series of behavioural assays to evaluate the acute responses of mice to painful thermal stimulation while administering YM-254890 alone and in combination with morphine. We then used electrophysiological recordings to evaluate the effects of YM-254890 on the excitability of dorsal root ganglion (DRG) nociceptor neurons. KEY RESULTS We found that systemic administration of YM-254890 produced anti-nociceptive effects and also augmented morphine analgesia in both hotplate and tail flick paradigms. However, it also caused substantial inhibition of locomotion, which may limit its therapeutic utility. To circumvent these issues, we explored the local administration of YM-254890. Intrathecal injections of YM-254890 produced lasting analgesia in a tail flick test and greatly augmented the anti-nociceptive effects of morphine without any significant effects on locomotor behaviour. Electrophysiological studies showed that YM-254890 reduced the excitability of DRG nociceptors and augmented their opioid-induced inhibition. CONCLUSION AND IMPLICATIONS These findings indicate that pharmacological inhibition of Gq/11 could be explored as an analgesic strategy.
Collapse
Affiliation(s)
- Subhi Marwari
- Department of NeuroscienceThe Scripps Research InstituteJupiterFloridaUSA
| | - Cody Kowalski
- Department of NeuroscienceThe Scripps Research InstituteJupiterFloridaUSA
| | | |
Collapse
|
12
|
Polzin BJ, Maksimoski AN, Stevenson SA, Zhao C, Riters LV. Mu opioid receptor stimulation in the medial preoptic area or nucleus accumbens facilitates song and reward in flocking European starlings. Front Physiol 2022; 13:970920. [PMID: 36171974 PMCID: PMC9510710 DOI: 10.3389/fphys.2022.970920] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/22/2022] [Indexed: 11/14/2022] Open
Abstract
It has been proposed that social cohesion in gregarious animals is reinforced both by a positive affective state induced by social interactions and by the prevention of a negative state that would be caused by social separation. Opioids that bind to mu opioid receptors (MORs) act in numerous brain regions to induce positive and to reduce negative affective states. Here we explored a potential role for MORs in affective states that may impact flocking behavior in mixed-sex flocks of nonbreeding European starlings, Sturnus vulgaris. Singing behavior, which is considered central to flock cohesion, and other social behaviors were quantified after infusions of the MOR agonist D-Ala2, N-Me-Phe4, glycinol5-ENK (DAMGO) into either the medial preoptic area (POM) or the nucleus accumbens (NAC), regions previously implicated in affective state and flock cohesion. We focused on beak wiping, a potential sign of stress or redirected aggression in this species, to provide insight into a presumed negative state. We also used conditioned place preference (CPP) tests to provide insight into the extent to which infusions of DAMGO into POM or NAC that stimulated song might be rewarding. We found that MOR stimulation in either POM or NAC dose-dependently promoted singing behavior, reduced beak wiping, and induced a CPP. Subtle differences in responses to MOR stimulation between NAC and POM also suggest potential functional differences in the roles of these two regions. Finally, because the location of NAC has only recently been identified in songbirds, we additionally performed a tract tracing study that confirmed the presence of dopaminergic projections from the ventral tegmental area to NAC, suggesting homology with mammalian NAC. These findings support the possibility that MORs in POM and NAC play a dual role in reinforcing social cohesion in flocks by facilitating positive and reducing negative affective states.
Collapse
|
13
|
Yang H, Gu X, Xu M, Yang G, Rao Y, Gao L, Gong G, He S. Preventing nausea and vomiting after gynecological laparoscopic surgery by patient-controlled intravenous analgesia with a naloxone admixture: A randomized controlled trial. Medicine (Baltimore) 2022; 101:e29584. [PMID: 35866767 PMCID: PMC9302326 DOI: 10.1097/md.0000000000029584] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Opioid-induced nausea and vomiting are common side effects of patient-controlled intravenous analgesia (PCIA). This study aimed to explore the inhibitory effect of a naloxone admixture on the incidence of sufentanil-induced postoperative nausea and vomiting (PONV). METHODS A total of 132 Uyghur American Society of Anesthesiologists I and II patients scheduled to undergo elective gynecological laparoscopic surgery were recruited; among these, 120 patients were enrolled and randomly allocated into 4 groups: patients receiving PCIA but no naloxone were included in the control group (group A); patients receiving PCIA with a low-dose naloxone admixture at 0.2 μg·kg-1·h-1 were included in group B; patients receiving PCIA with naloxone admixture at 0.4 μg·kg-1·h-1 were included in group C; patients receiving PCIA with naloxone admixture at 0.6 μg·kg-1·h-1 were included in group D. All patients were administered sufentanil at 0.04 kg-1·h-1, butorphanol at 2 kg-1·h-1, and dexmedetomidine at 0.08 kg-1·h-1 using a PCIA device within 2 days of surgery. The occurrence of nausea and vomiting, visual analogue scores for pain intensity, mean arterial pressure, heart rate, oxygen saturation, pruritus, lethargy, respiratory depression, etc, was recorded at 2, 8, 12, 24, and 48 hours postoperatively. RESULTS There was a significant difference in the PONV scores between the groups at 8, 12, and 24 hours after surgery (P < 0.01). At 8 and 12 hours, the score of group C/D was significantly lower than that of group A/B (P < 0.01). At 24 hours after surgery, the PONV score of group B/C/D was significantly lower than that of group A (P < 0.01). No significant difference was observed in the general data and visual analogue scores for postoperative pain between the 4 groups. CONCLUSION Naloxone admixture administered at 0.4 to 0.6 μg·kg-1·h-1 can exert an effective inhibitory effect on the incidence and intensity of PONV in gynecological laparoscopic surgery.
Collapse
Affiliation(s)
- Haihong Yang
- Department of Anesthesiology, The General Hospital of Western Theater Command, Sichuan, China
- Department of Anesthesiology, No. 950 Hospital of PLA, Yecheng, China
| | - Xuedong Gu
- Department of Anesthesiology, The General Hospital of Western Theater Command, Sichuan, China
| | - Meiling Xu
- Department of Geriatrics, The General Hospital of Western Theater Command, Sichuan, China
| | - Guan Yang
- Department of Anesthesiology, The General Hospital of Western Theater Command, Sichuan, China
| | - Yunju Rao
- School of Clinical Medicine, Southwest Medical University, Sichuan, China
| | - Liang Gao
- Military Prevention and Treatment Center for Mountain Sickness, No. 950 Hospital of PLA, Yecheng, China
| | - Gu Gong
- Department of Anesthesiology, The General Hospital of Western Theater Command, Sichuan, China
| | - Siyi He
- Department of Cardiovascular Surgery, The General Hospital of Western Theater Command, Sichuan, China
- * Correspondence: Siyi He, Department of Cardiovascular Surgery, The General Hospital of Western Theater Command, Rongdu Avenue No. 270, Jinniu District, Chengdu City, Sichuan Province 610083, China (e-mail: )
| |
Collapse
|
14
|
He L, Xu W, Zhang C, Ding Z, Guo Q, Zou W, Wang J. Dysregulation of Vesicular Glutamate Transporter VGluT2 via BDNF/TrkB Pathway Contributes to Morphine Tolerance in Mice. Front Pharmacol 2022; 13:861786. [PMID: 35559256 PMCID: PMC9086316 DOI: 10.3389/fphar.2022.861786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
Morphine is widely used in the treatment of moderate to severe pain. Long-term use of morphine leads to various adverse effects, such as tolerance and hyperalgesia. Vesicular glutamate transporter 2 (VGluT2) accumulates glutamate into synaptic vesicles and plays multiple roles in the central nervous system. However, the specific role of VGluT2 in morphine tolerance has not been fully elucidated. Here, we investigated the regulatory role of VGluT2 in morphine tolerance and assessed the potential role of the brain-derived neurotrophic factor (BDNF)/tyrosine kinase B (TrkB) pathway in VGluT2 mediated morphine antinociceptive tolerance in mice. In the present study, we found that VGluT2 is upregulated in the spinal cord after the development of morphine tolerance. Furthermore, inhibition of VGluT2 with its antagonist (Chicago sky blue 6 B, CSB6B) or knockdown of VGluT2 by lentivirus restored the analgesic effect of morphine, suppressed the activation of astrocytes and microglia, and decreased glial-derived pro-inflammatory cytokines. Overexpression of VGluT2 by lentivirus facilitated morphine tolerance and mechanical hyperalgesia. In addition, we found the expression of BDNF is correlated with VGluT2 expression in the spinal cord after chronic morphine administration. Intrathecal injection of the BDNF/TrkB pathway antagonist K252a attenuated the development of morphine tolerance and decreased the expression of VGluT2 in the spinal cord, which suggested the BDNF/TrkB pathway participates in the regulation of VGluT2 in morphine tolerance. This study elucidates the functional capability of VGluT2 in modulating morphine tolerance and identifies a novel mechanism and promising therapeutic target for morphine tolerance.
Collapse
Affiliation(s)
- Liqiong He
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xu
- Department of Anesthesiology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Chengliang Zhang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhuofeng Ding
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wangyuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Barker KE, Lecznar AJ, Schumacher JM, Morris JS, Gutstein HB. Subanalgesic morphine doses augment fentanyl analgesia by interacting with delta opioid receptors in male rats. J Neurosci Res 2021; 100:149-164. [PMID: 34520585 DOI: 10.1002/jnr.24944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022]
Abstract
Opioids are commonly used for the treatment of postoperative and post-traumatic pain; however, their therapeutic effectiveness is limited by undesirable and life-threatening side effects. Researchers have long attempted to develop opioid co-administration therapies that enhance analgesia, but the complexity of opioid analgesia and our incomplete mechanistic understanding has made this a daunting task. We discovered that subanalgesic morphine doses (100 ng/kg-10 µg/kg) augmented the acute analgesic effect of fentanyl (20 µg/kg) following subcutaneous drug co-administration to male rats. In addition, administration of equivalent drug ratios to naïve rat spinal cord membranes induced a twofold increase in G protein activation. The rate of GTP hydrolysis remained unchanged. We demonstrated that these behavioral and biochemical effects were mediated by the delta opioid receptor (DOP). Subanalgesic doses of the DOP-selective agonist SNC80 also augmented the acute analgesic effect of fentanyl. Furthermore, co-administration of the DOP antagonist naltrindole with both fentanyl-morphine and fentanyl-SNC80 combinations prevented augmentation of both analgesia and G protein activation. The mu opioid receptor (MOP) antagonist cyprodime did not block augmentation. Confocal microscopy of the substantia gelatinosa of rats treated with fentanyl, subanalgesic morphine, or this combination showed that changes in MOP internalization did not account for augmentation effects. Together, these findings suggest that augmentation of fentanyl analgesia by subanalgesic morphine is mediated by increased G protein activation resulting from a synergistic interaction between or heterodimerization of MOPs and DOPs. This finding is of great therapeutic significance because it suggests a strategy for the development of DOP-selective ligands that can enhance the therapeutic index of clinically used MOP drugs.
Collapse
Affiliation(s)
- Katherine E Barker
- Department of Anesthesiology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Alynn J Lecznar
- Department of Anesthesiology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Jill M Schumacher
- Department of Genetics, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey S Morris
- Biostatistics Division, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| | - Howard B Gutstein
- Anesthesiology Institute, Allegheny Health Network, Pittsburgh, PA, USA
| |
Collapse
|
16
|
Muralidharan A, Samoshkin A, Convertino M, Piltonen MH, Gris P, Wang J, Jiang C, Klares R, Linton A, Ji RR, Maixner W, Dokholyan NV, Mogil JS, Diatchenko L. Identification and characterization of novel candidate compounds targeting 6- and 7-transmembrane μ-opioid receptor isoforms. Br J Pharmacol 2021; 178:2709-2726. [PMID: 33782947 PMCID: PMC10697213 DOI: 10.1111/bph.15463] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE The μ-opioid receptor (μ receptor) is the primary target for opioid analgesics. The 7-transmembrane (TM) and 6TM μ receptor isoforms mediate inhibitory and excitatory cellular effects. Here, we developed compounds selective for 6TM- or 7TM-μ receptors to further our understanding of the pharmacodynamic properties of μ receptors. EXPERIMENTAL APPROACH We performed virtual screening of the ZINC Drug Now library of compounds using in silico 7TM- and 6TM-μ receptor structural models and identified potential compounds that are selective for 6TM- and/or 7TM-μ receptors. Subsequently, we characterized the most promising candidate compounds in functional in vitro studies using Be2C neuroblastoma transfected cells, behavioural in vivo pain assays using various knockout mice and in ex vivo electrophysiology studies. KEY RESULTS Our virtual screen identified 30 potential candidate compounds. Subsequent functional in vitro cellular assays shortlisted four compounds (#5, 10, 11 and 25) that demonstrated 6TM- or 7TM-μ receptor-dependent NO release. In in vivo pain assays these compounds also produced dose-dependent hyperalgesic responses. Studies using mice that lack specific opioid receptors further established the μ receptor-dependent nature of identified novel ligands. Ex vivo electrophysiological studies on spontaneous excitatory postsynaptic currents in isolated spinal cord slices also validated the hyperalgesic properties of the most potent 6TM- (#10) and 7TM-μ receptor (#5) ligands. CONCLUSION AND IMPLICATIONS Our novel compounds represent a new class of ligands for μ receptors and will serve as valuable research tools to facilitate the development of opioids with significant analgesic efficacy and fewer side-effects.
Collapse
Affiliation(s)
- Arjun Muralidharan
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
- Department of Psychology, McGill University, Montreal, Quebec, Canada
| | - Alexander Samoshkin
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
- Department of Anesthesia, McGill University, Montreal, Quebec, Canada
| | - Marino Convertino
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Marjo Hannele Piltonen
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | - Pavel Gris
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | - Jian Wang
- Department of Pharmacology, and Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Changyu Jiang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Richard Klares
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
- Department of Anesthesia, McGill University, Montreal, Quebec, Canada
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | - Alexander Linton
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
- Department of Anesthesia, McGill University, Montreal, Quebec, Canada
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - William Maixner
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Nikolay V. Dokholyan
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Pharmacology, and Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Jeffrey S. Mogil
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
- Department of Psychology, McGill University, Montreal, Quebec, Canada
- Department of Anesthesia, McGill University, Montreal, Quebec, Canada
| | - Luda Diatchenko
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
- Department of Anesthesia, McGill University, Montreal, Quebec, Canada
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Sources of lumbar back pain during aging and potential therapeutic targets. VITAMINS AND HORMONES 2021; 115:571-583. [PMID: 33706962 DOI: 10.1016/bs.vh.2020.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Lumbar back pain during aging is a major clinical problem, the origins and underlying mechanisms of which are challenging to study. Degenerative changes occur in various parts of the functional spinal unit, such the vertebral endplate and intervertebral disc. The homeostasis of these structural components is regulated by signaling molecules, such as transforming growth factor-β and parathyroid hormone. Previous efforts to understand sources of lumbar back pain focused on sensory innervation in the degenerative intervertebral disc, but intervertebral disc degeneration is frequently asymptomatic. An in vivo mouse model of lumbar spine aging and degeneration, combined with genetic technology, has identified endplate innervation as a major source of lumbar back pain and a potential therapeutic target. In this review, we consider how each structural component of the functional spinal unit contributes to lumbar back pain, how the homeostasis of each component is regulated, and how these findings can be used to develop potential therapies.
Collapse
|
18
|
A Low Dose of Naloxone Added to Ropivacaine Prolongs Femoral Nerve Blockade: A Randomized Clinical Trial. Pain Res Manag 2021; 2021:6639009. [PMID: 33603939 PMCID: PMC7868154 DOI: 10.1155/2021/6639009] [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: 11/06/2020] [Revised: 12/24/2020] [Accepted: 01/20/2021] [Indexed: 11/18/2022]
Abstract
Femoral nerve blocks (FNBs) are used as safe and useful procedures to control severe postoperative pain from total knee arthroplasty (TKA). Various adjuvants have been used to prolong the duration of the local anesthetic blockade. This study evaluated whether a low dose of naloxone administered with local anesthetics prolongs the duration of FNB. A prospective, randomized double-blind controlled study was conducted with 74 patients undergoing unilateral TKA. Through a single-bolus administration guided by ultrasound, the control group (group C) received 20 mL of 0.375% ropivacaine, while the naloxone group (group N) received 20 mL of 0.375% ropivacaine with 100 ng of naloxone. The time elapsed before the first analgesia request, the total amount of opioids consumed at 24 h postoperatively, the onset time of the sensory blockade, the visual analog pain scale (VAS) scores after arriving at the recovery room, after 6, 12, 18, and 24 h at rest and after 12, 18, and 24 h of activity, the quadricep strength before the FNB procedure and at 12 and 24 h postoperatively, the quality of sleep on the first night after surgery, the satisfaction score, and the incidence of postoperative complications were recorded. The time elapsed before the first analgesia request was significantly longer in group N (735.5 ± 187.2 min) than that in group C (602.6 ± 210.4 min) (P=0.003). The total dose of supplementary opioids consumed at 24 h postoperatively was significantly lower in group N (312.4 ± 141.7 μg) than that in group C (456.5 ± 279.5 μg) (P=0.007). Lower VAS scores were recorded in group N than that in group C at rest and during knee activity (rest, 12 h, P=0.001, 18 h, P=0.043; activity, 12 h, P=0.001). The addition of a low dose of naloxone to ropivacaine for FNB significantly delayed the first request for rescue analgesia and decreased the opioid consumption within 24 h, without significant complications.
Collapse
|
19
|
Chen R, Tang LH, Sun T, Zeng Z, Zhang YY, Ding K, Meng QT. Mechanism and Management of Fentanyl-Induced Cough. Front Pharmacol 2020; 11:584177. [PMID: 33324214 PMCID: PMC7723435 DOI: 10.3389/fphar.2020.584177] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/28/2020] [Indexed: 12/19/2022] Open
Abstract
Fentanyl-induced cough (FIC) often occurs after intravenous bolus administration of fentanyl analogs during induction of general anesthesia and analgesia procedure. The cough is generally benign, but sometimes it causes undesirable side effects, including elevated intra-abdominal, intracranial or intraocular pressure. Therefore, understanding the related mechanisms and influencing factors are of great significance to prevent and treat the cough. This paper reviews the molecular mechanism, influencing factors and preventive administration of FIC, focusing on the efficacy and side effects of various drugs in inhibiting FIC to provide some medical reference for anesthesiologists.
Collapse
Affiliation(s)
- Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ling-Hua Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Sun
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zi Zeng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yun-Yan Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing-Tao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
20
|
Abstract
Understanding the molecular biology of opioid analgesia is essential for its proper implementation and mechanistic approach to its modulation in order to maximize analgesia and minimize undesired effects. By appreciating the molecular mechanisms intrinsic to opioid analgesia, one can manipulate a molecular target to augment or diminish a specific effect using adjuvant drugs, select an appropriate opioid for opioid rotation or define a molecular target for new opioid drug development. In this review, we present the cellular and molecular mechanisms of opioid analgesia and that of the associated phenomena of tolerance, dependence, and hyperalgesia. The specific mechanisms highlighted are those that presently can be clinically addressed.
Collapse
|
21
|
Micheli L, Di Cesare Mannelli L, Lucarini E, Parisio C, Toti A, Fiorentino B, Rigamonti MA, Calosi L, Ghelardini C. Intranasal Low-Dose Naltrexone Against Opioid Side Effects: A Preclinical Study. Front Pharmacol 2020; 11:576624. [PMID: 33071790 PMCID: PMC7531600 DOI: 10.3389/fphar.2020.576624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 08/20/2020] [Indexed: 12/20/2022] Open
Abstract
Opioids are broad spectrum analgesics that are an integral part of the therapeutic armamentarium to combat pain in the clinical practice. Unfortunately, together with analgesia, a number of adverse effects can occur such as nausea, vomiting, constipation, gastrointestinal alterations and cognitive impairments. Naltrexone is a competitive antagonist of opioid receptors commonly used to treat opioid addiction; its oral use against agonists side effects is limited by the decrease of opioids-therapeutic efficacy and own adverse effects. The intranasal delivery of naltrexone could offer a quick and effective achievement of CNS based on extracellular mechanisms including perineural and perivascular transport. The aim of the study was to test the efficacy of intranasal low-dose naltrexone in reducing intraperitoneal morphine and oxycodone side effects in rodents. In mice, 1 μg naltrexone intranasally administered 30 min before opioids reduced cognitive impairments and motor alteration induced by 10 mg kg-1 morphine and 60 mg kg-1 oxycodone in the Passive avoidance and Rota rod tests, respectively. Moreover, naltrexone rebalanced opioid-induced reduction of the intestinal transit and latency of feces expulsion as well as food intake inhibition. Importantly, 1 μg naltrexone instillation did not block analgesia as demonstrated by the Hot plate test. In rats, intranasal naltrexone counteracted the opioid-induced pica phenomenon related to emesis and increased water and palatable food intake. The effects were comparable to that achieved by metoclopramide used as reference drug. Treatments did not influence body weight. Lastly, the safety of the intranasal delivery has been checked by hematoxylin-eosin staining that did not show histological alterations of the nasal cavity. In conclusion, intranasal low-dose naltrexone counteracted morphine and oxycodone induced gastrointestinal and CNS side effects without impairing opioid analgesia. It is a candidate to be a valid clinical strategy deserving deep analysis.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Carmen Parisio
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Alessandra Toti
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | | | | | - Laura Calosi
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology & Research Unit of Histology & Embryology, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| |
Collapse
|
22
|
Paton KF, Atigari DV, Kaska S, Prisinzano T, Kivell BM. Strategies for Developing κ Opioid Receptor Agonists for the Treatment of Pain with Fewer Side Effects. J Pharmacol Exp Ther 2020; 375:332-348. [PMID: 32913006 DOI: 10.1124/jpet.120.000134] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/27/2020] [Indexed: 12/21/2022] Open
Abstract
There is significant need to find effective, nonaddictive pain medications. κ Opioid receptor (KOPr) agonists have been studied for decades but have recently received increased attention because of their analgesic effects and lack of abuse potential. However, a range of side effects have limited the clinical development of these drugs. There are several strategies currently used to develop safer and more effective KOPr agonists. These strategies include identifying G-protein-biased agonists, developing peripherally restricted KOPr agonists without centrally mediated side effects, and developing mixed opioid agonists, which target multiple receptors at specific ratios to balance side-effect profiles and reduce tolerance. Here, we review the latest developments in research related to KOPr agonists for the treatment of pain. SIGNIFICANCE STATEMENT: This review discusses strategies for developing safer κ opioid receptor (KOPr) agonists with therapeutic potential for the treatment of pain. Although one strategy is to modify selective KOPr agonists to create peripherally restricted or G-protein-biased structures, another approach is to combine KOPr agonists with μ, δ, or nociceptin opioid receptor activation to obtain mixed opioid receptor agonists, therefore negating the adverse effects and retaining the therapeutic effect.
Collapse
Affiliation(s)
- Kelly F Paton
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Diana V Atigari
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Sophia Kaska
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Thomas Prisinzano
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| | - Bronwyn M Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand (K.P., D.V.A., B.M.K.) and Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky (S.K., T.P.)
| |
Collapse
|
23
|
Epidural Naloxone Attenuates Fentanyl Induced PONV in Patients Undergoing Lower Limb Orthopaedic Surgeries. a Prospective Randomized Double-Blind Comparative Study. Rom J Anaesth Intensive Care 2020; 27:23-28. [PMID: 34056121 PMCID: PMC8158304 DOI: 10.2478/rjaic-2020-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background and aim Epidural administration of opioids with local anaesthetics is a popular choice for perioperative pain relief. But opioid induced side effects limit their use for postoperative analgesia. Hence, this study was designed to evaluate the effectiveness of epidural naloxone, an opioid receptor antagonist, in reducing PONV in patients receiving epidural fentanyl. Methods After obtaining the Institutional Ethics Committee approval and written informed consent, 46 patients, between 18–80 years, of either sex, with ASA physical status 1–3, undergoing lower limb orthopaedic surgeries were enlisted for this prospective, randomized, double blind comparative study. Subjects were allocated to one of the two groups and received epidurally, either fentanyl with bupivacaine (Group C, n = 23) or fentanyl with bupivacaine and naloxone 2 mcg (Group N, n = 25), for reducing postoperative pain. PONV score and Wong Bakers Scale (WBS) for pain score were recorded at 6, 12 and 18hrs, postoperatively. Results All patients were comparable with respect to age, gender, ASA PS, height, body weight as well as duration of surgery. A statistically significant decrease in PONV score was observed in Group N at 6 and 12 hours, postoperatively. The patients who required rescue antiemetic were also significantly lower in Group N at 6 and 12 hours. The mean WBS score for pain also showed significant reduction in Group N at 6 hours, postoperatively. Conclusion Concomitant use of low dose epidural naloxone and fentanyl is effective in attenuating PONV, besides enhancing analgesia in the earlypostoperative period.
Collapse
|
24
|
Shift of µ-opioid Receptor Signaling in the Dorsal Reticular Nucleus Is Implicated in Morphine-induced Hyperalgesia in Male Rats. Anesthesiology 2020; 133:628-644. [PMID: 32568844 DOI: 10.1097/aln.0000000000003412] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Increased descending pain facilitation accounts for opioid-induced hyperalgesia, but the underlying mechanisms remain elusive. Given the role of µ-opioid receptors in opioid-induced hyperalgesia in animals, the authors hypothesized that the dorsal reticular nucleus, a medullary pain facilitatory area, is involved in opioid-induced hyperalgesia through altered µ-opioid receptor signaling. METHODS The authors used male Wistar rats (n = 5 to 8 per group), chronically infused with morphine, to evaluate in the dorsal reticular nucleus the expressions of the µ-opioid receptor and phosphorylated cAMP response element-binding, a downstream marker of excitatory µ-opioid receptor signaling. The authors used pharmacologic and gene-mediated approaches. Nociceptive behaviors were evaluated by the von Frey and hot-plates tests. RESULTS Lidocaine fully reversed mechanical and thermal hypersensitivity induced by chronic morphine. Morphine-infusion increased µ-opioid receptor, without concomitant messenger RNA changes, and phosphorylated cAMP response element-binding levels at the dorsal reticular nucleus. µ-opioid receptor knockdown in morphine-infused animals attenuated the decrease of mechanical thresholds and heat-evoked withdrawal latencies compared with the control vector (von Frey [mean ± SD]: -17 ± 8% vs. -40 ± 9.0%; P < 0.001; hot-plate: -10 ± 5% vs. -32 ± 10%; P = 0.001). µ-opioid receptor knockdown in control animals induced the opposite (von Frey: -31 ± 8% vs. -17 ± 8%; P = 0.053; hotplate: -24 ± 6% vs. -3 ± 10%; P = 0.001). The µ-opioid receptor agonist (D-ALA2,N-ME-PHE4,GLY5-OL)-enkephalin acetate (DAMGO) decreased mechanical thresholds and did not affect heat-evoked withdrawal latencies in morphine-infused animals. In control animals, DAMGO increased both mechanical thresholds and heat-evoked withdrawal latencies. Ultra-low-dose naloxone, which prevents the excitatory signaling of the µ-opioid receptor, administered alone, attenuated mechanical and thermal hypersensitivities, and coadministered with DAMGO, restored DAMGO analgesic effects and decreased phosphorylated cAMP response element-binding levels. CONCLUSIONS Chronic morphine shifted µ-opioid receptor signaling from inhibitory to excitatory at the dorsal reticular nucleus, likely enhancing descending facilitation during opioid-induced hyperalgesia in the rat.
Collapse
|
25
|
Busserolles J, Lolignier S, Kerckhove N, Bertin C, Authier N, Eschalier A. Replacement of current opioid drugs focusing on MOR-related strategies. Pharmacol Ther 2020; 210:107519. [PMID: 32165137 DOI: 10.1016/j.pharmthera.2020.107519] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022]
Abstract
The scarcity and limited risk/benefit ratio of painkillers available on the market, in addition to the opioid crisis, warrant reflection on new innovation strategies. The pharmacopoeia of analgesics is based on products that are often old and derived from clinical empiricism, with limited efficacy or spectrum of action, or resulting in an unsatisfactory tolerability profile. Although they are reference analgesics for nociceptive pain, opioids are subject to the same criticism. The use of opium as an analgesic is historical. Morphine was synthesized at the beginning of the 19th century. The efficacy of opioids is limited in certain painful contexts and these drugs can induce potentially serious and fatal adverse effects. The current North American opioid crisis, with an ever-rising number of deaths by opioid overdose, is a tragic illustration of this. It is therefore legitimate to develop research into molecules likely to maintain or increase opioid efficacy while improving their tolerability. Several avenues are being explored including targeting of the mu opioid receptor (MOR) splice variants, developing biased agonists or targeting of other receptors such as heteromers with MOR. Ion channels acting as MOR effectors, are also targeted in order to offer compounds without MOR-dependent adverse effects. Another route is to develop opioid analgesics with peripheral action or limited central nervous system (CNS) access. Finally, endogenous opioids used as drugs or compounds that modify the metabolism of endogenous opioids (Dual ENKephalinase Inhibitors) are being developed. The aim of the present review is to present these various targets/strategies with reference to current indications for opioids, concerns about their widespread use, particularly in chronic non-cancer pains, and ways of limiting the risk of opioid abuse and misuse.
Collapse
Affiliation(s)
- Jérôme Busserolles
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | - Stéphane Lolignier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France
| | - Nicolas Kerckhove
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Célian Bertin
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Nicolas Authier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France; Observatoire Français des Médicaments Antalgiques (OFMA), French monitoring centre for analgesic drugs, CHU, F-63000 Clermont-Ferrand, France
| | - Alain Eschalier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Pharmacologie Fondamentale et Clinique de la douleur, F-63000 Clermont-Ferrand, France; Institut ANALGESIA, Faculté de Médecine, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
26
|
Effect of low dose naloxone on the immune system function of a patient undergoing video-assisted thoracoscopic resection of lung cancer with sufentanil controlled analgesia - a randomized controlled trial. BMC Anesthesiol 2019; 19:236. [PMID: 31856760 PMCID: PMC6923917 DOI: 10.1186/s12871-019-0912-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/09/2019] [Indexed: 01/29/2023] Open
Abstract
Background Perioperative immune function plays an important role in the prognosis of patients. Several studies have indicated that low-dose opioid receptor blockers can improve immune function. Methods Sixty-nine patients undergoing video-assisted thoracoscopic resection of the lung cancer were randomly assigned to either the naloxone group (n = 35) or the non-naloxone group (n = 34) for postoperative analgesia during the first 48 h after the operation. Both groups received sufentanil and palonosetron via postoperative analgesia pump, while 0.05 μg·kg− 1·h− 1 naloxone was added in naloxone group. The primary outcomes were the level of opioid growth factor (OGF) and immune function assessed by natural killer cells and CD4+/CD8+ T-cell ratio. Second outcomes were assessed by the intensity of postoperative pain, postoperative rescue analgesia dose, postoperative nausea and vomiting (PONV). Results The level of OGF in the naloxone group increased significantly at 24 h (p<0.001) and 48 h after the operation (P < 0.01). The natural killer cells (P < 0.05) and CD4+/CD8+ T-cell ratio (P < 0.01) in the naloxone group increased significantly at 48 h after the operation. The rest VAS scores were better with naloxone at 12 and 24 h after operation(P < 0.05), and the coughing VAS scores were better with naloxone at 48 h after the operation(P < 0.05). The consumption of postoperative rescue analgesics in the naloxone group was lower (0.00(0.00–0.00) vs 25.00(0.00–62.50)), P < 0.05). Postoperative nausea scores at 24 h after operation decreased in naloxone group(0.00 (0.00–0.00) vs 1.00 (0.00–2.00), P < 0.01). Conclusion Infusion of 0.05 μg·kg− 1·h− 1 naloxone for patients undergoing sufentanil-controlled analgesia for postoperative pain can significantly increase the level of OGF, natural killer cells, and CD4+/CD8+ T-cell ratio compared with non-naloxone group, and postoperative pain intensity, request for rescue analgesics, and opioid-related side effects can also be reduced. Trial registration The trial was registered at the Chinese Clinical Trial Registry on January 26, 2019 (ChiCTR1900021043).
Collapse
|
27
|
Zhu C, Han Q, Samoshkin A, Convertino M, Linton A, Faison EM, Ji RR, Diatchenko L, Dokholyan NV. Stabilization of μ-opioid receptor facilitates its cellular translocation and signaling. Proteins 2019; 87:878-884. [PMID: 31141214 DOI: 10.1002/prot.25751] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/09/2019] [Accepted: 05/26/2019] [Indexed: 01/01/2023]
Abstract
The G protein-coupled μ-opioid receptor (μ-OR) mediates the majority of analgesia effects for morphine and other pain relievers. Despite extensive studies of its structure and activation mechanisms, the inherently low maturation efficiency of μ-OR represents a major hurdle to understanding its function. Here we computationally designed μ-OR mutants with altered stability to probe the relationship between cell-surface targeting, signal transduction, and agonist efficacy. The stabilizing mutation T315Y enhanced μ-OR trafficking to the plasma membrane and significantly promoted the morphine-mediated inhibition of downstream signaling. In contrast, the destabilizing mutation R165Y led to intracellular retention of μ-OR and reduced the response to morphine stimulation. These findings suggest that μ-OR stability is an important factor in regulating receptor signaling and provide a viable avenue to improve the efficacy of analgesics.
Collapse
Affiliation(s)
- Cheng Zhu
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Qingjian Han
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina.,Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Alexander Samoshkin
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada.,School of the Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Marino Convertino
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alexander Linton
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - Edgar M Faison
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina.,Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Luda Diatchenko
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada
| | - Nikolay V Dokholyan
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania.,Department of Biochemistry & Molecular Biology, Penn State University College of Medicine, Hershey, Pennsylvania.,Department of Biomedical Engineering, Penn State University, Pennsylvania.,Department of Chemistry, Penn State University, Pennsylvania
| |
Collapse
|
28
|
Liu DQ, Zhou YQ, Gao F. Targeting Cytokines for Morphine Tolerance: A Narrative Review. Curr Neuropharmacol 2019; 17:366-376. [PMID: 29189168 PMCID: PMC6482476 DOI: 10.2174/1570159x15666171128144441] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/06/2017] [Accepted: 11/23/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Despite its various side effects, morphine has been widely used in clinics for decades due to its powerful analgesic effect. Morphine tolerance is one of the major side effects, hindering its long-term usage for pain therapy. Currently, the thorough cellular and molecular mechanisms underlying morphine tolerance remain largely uncertain. METHODS We searched the PubMed database with Medical subject headings (MeSH) including 'morphine tolerance', 'cytokines', 'interleukin 1', 'interleukin 1 beta', 'interleukin 6', 'tumor necrosis factor alpha', 'interleukin 10', 'chemokines'. Manual searching was carried out by reviewing the reference lists of relevant studies obtained from the primary search. The searches covered the period from inception to November 1, 2017. RESULTS The expression levels of certain chemokines and pro-inflammatory cytokines were significantly increased in animal models of morphine tolerance. Cytokines and cytokine receptor antagonist showed potent effect of alleviating the development of morphine tolerance. CONCLUSION Cytokines play a fundamental role in the development of morphine tolerance. Therapeutics targeting cytokines may become alternative strategies for the management of morphine tolerance.
Collapse
Affiliation(s)
| | | | - Feng Gao
- Address correspondence to this author at the Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China; Tel: +86 27 83662853; E-mail:
| |
Collapse
|
29
|
Martínez-Navarro M, Maldonado R, Baños JE. Why mu-opioid agonists have less analgesic efficacy in neuropathic pain? Eur J Pain 2018; 23:435-454. [PMID: 30318675 DOI: 10.1002/ejp.1328] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/06/2018] [Accepted: 10/10/2018] [Indexed: 12/29/2022]
Abstract
Injury to peripheral nerves often leads to abnormal pain states (hyperalgesia, allodynia and spontaneous pain), which can remain long after the injury heals. Although opioid agonists remain the gold standard for the treatment of moderate to severe pain, they show reduced efficacy against neuropathic pain. In addition to analgesia, opioid use is also associated with hyperalgesia and analgesia tolerance, whose underlying mechanisms share some commonalities with nerve injury-induced hypersensitivity. Here, we reviewed up-to-day research exploring the contribution of mu-opioid receptor (MOR) on the pathophysiology of neuropathic pain and on analgesic opioid actions under these conditions. We focused on the specific contributions of MOR populations at peripheral, spinal and supraspinal level. Moreover, evidences of neuroplastic changes that may underlie the low efficacy of MOR agonists under neuropathic pain conditions are reviewed and discussed. Sensitization processes leading to pain hypersensitivity, molecular changes in signalling pathways triggered by MOR and glial activation are some of these mechanisms elicited by both nerve injury and opioid exposure. Nerve injury-induced pain hypersensitivity might be masking the initial analgesic effects of opioid agonists, and alternatively, sustained opioid treatment to individuals already suffering from neuropathic pain could aggravate their pathophysiological state. Finally, some combined therapies that can increase opioid analgesic effectiveness in neuropathic pain treatment are highlighted. SIGNIFICANCE: This review provides evidence of the low benefit of opioid monotherapy in neuropathic pain and analyses the reasons of this reduced effectiveness. Opioid agonists along with drugs targeted to block the sensitization processes induced by MOR stimulation might result in a better management of neuropathic pain.
Collapse
Affiliation(s)
- Miriam Martínez-Navarro
- Department of Experimental and Health Sciences, Laboratory of Neuropharmacology, Universitat Pompeu Fabra, Barcelona, Spain
| | - Rafael Maldonado
- Department of Experimental and Health Sciences, Laboratory of Neuropharmacology, Universitat Pompeu Fabra, Barcelona, Spain
| | - Josep-E Baños
- Department of Experimental and Health Sciences, Laboratory of Neuropharmacology, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
30
|
Szigethy E, Knisely M, Drossman D. Opioid misuse in gastroenterology and non-opioid management of abdominal pain. Nat Rev Gastroenterol Hepatol 2018; 15:168-180. [PMID: 29139482 PMCID: PMC6421506 DOI: 10.1038/nrgastro.2017.141] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Opioids were one of the earliest classes of medications used for pain across a variety of conditions, but morbidity and mortality have been increasingly associated with their chronic use. Despite these negative consequences, chronic opioid use is increasing worldwide, with the USA and Canada having the highest rates. Chronic opioid use for noncancer pain can have particularly negative effects in the gastrointestinal and central nervous systems, including opioid-induced constipation, narcotic bowel syndrome, worsening psychopathology and addiction. This Review summarizes the evidence of opioid misuse in gastroenterology, including the lack of evidence of a benefit from these drugs, as well as the risk of harm and negative consequences of opioid use relative to the brain-gut axis. Guidelines for opioid management and alternative pharmacological and nonpharmacological strategies for pain management in patients with gastrointestinal disorders are also discussed. As chronic pain is complex and involves emotional and social factors, a multimodal approach targeting both pain intensity and quality of life is best.
Collapse
Affiliation(s)
- Eva Szigethy
- Departments of Psychiatry and Medicine, University of Pittsburgh, 3708 Fifth Avenue, Pittsburgh, Pennsylvania 15213, USA
| | - Mitchell Knisely
- School of Nursing, University of Pittsburgh, 3500 Victoria Street, Pittsburgh, Pennsylvania 15261, USA
| | - Douglas Drossman
- Center for Functional GI & Motility Disorders, University of North Carolina, Bioinformatics Building, 130 Mason Farm Road, Chapel Hill, North Carolina 27599, USA
- Drossman Gastroenterology PLLC, 901 Kings Mill Road, Chapel Hill, North Carolina 27517, USA
| |
Collapse
|
31
|
Abstract
OBJECTIVE Studies have consistently shown that long-term meditation practice is associated with reduced pain, but the neural mechanisms by which long-term meditation practice reduces pain remain unclear. This study tested endogenous opioid involvement in meditation analgesia associated with long-term meditation practice. METHODS Electrical pain was induced with randomized, double-blind, cross-over administration of the opioid antagonist naloxone (0.15-mg/kg bolus dose, then 0.2-mg/kg per hour infusion dose) with 32 healthy, experienced meditation practitioners and a standardized open monitoring meditation. RESULTS Under saline, pain ratings were significantly lower during meditation (pain intensity: 6.41 ± 1.32; pain unpleasantness: 3.98 ± 2.17) than at baseline (pain intensity: 6.86 ±1.04, t(31) = 2.476, p = .019, Cohen's d = 0.46; pain unpleasantness: 4.96 ±1.75, t(31) = 3.746, p = .001, Cohen's d = 0.68), confirming the presence of meditation analgesia. Comparing saline and naloxone revealed significantly lower pain intensity (t(31) = 3.12, p = .004, d = 0.56), and pain unpleasantness (t(31) = 3.47, p = .002, d = 0.62), during meditation under naloxone (pain intensity: 5.53 ± 1.54; pain unpleasantness: 2.95 ± 1.88) than under saline (pain intensity: 6.41 ± 1.32; pain unpleasantness: 3.98 ± 2.17). Naloxone not only failed to eliminate meditation analgesia but also made meditation analgesia stronger. CONCLUSIONS Long-term meditation practice does not rely on endogenous opioids to reduce pain. Naloxone's blockade of opioid receptors enhanced meditation analgesia; pain ratings during meditation were significantly lower under naloxone than under saline. Possible biological mechanisms by which naloxone-induced opioid receptor blockade enhances meditation analgesia are discussed.
Collapse
|
32
|
Anderson RI, Moorman DE, Becker HC. Contribution of Dynorphin and Orexin Neuropeptide Systems to the Motivational Effects of Alcohol. Handb Exp Pharmacol 2018. [PMID: 29526023 DOI: 10.1007/164_2018_100] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Understanding the neural systems that drive alcohol motivation and are disrupted in alcohol use disorders is of critical importance in developing novel treatments. The dynorphin and orexin/hypocretin neuropeptide systems are particularly relevant with respect to alcohol use and misuse. Both systems are strongly associated with alcohol-seeking behaviors, particularly in cases of high levels of alcohol use as seen in dependence. Furthermore, both systems also play a role in stress and anxiety, indicating that disruption of these systems may underlie long-term homeostatic dysregulation seen in alcohol use disorders. These systems are also closely interrelated with one another - dynorphin/kappa opioid receptors and orexin/hypocretin receptors are found in similar regions and hypocretin/orexin neurons also express dynorphin - suggesting that these two systems may work together in the regulation of alcohol seeking and may be mutually disrupted in alcohol use disorders. This chapter reviews studies demonstrating a role for each of these systems in motivated behavior, with a focus on their roles in regulating alcohol-seeking and self-administration behaviors. Consideration is also given to evidence indicating that these neuropeptide systems may be viable targets for the development of potential treatments for alcohol use disorders.
Collapse
Affiliation(s)
- Rachel I Anderson
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.,Science and Technology Policy Fellowships, American Association for the Advancement of Science, Washington, DC, USA
| | - David E Moorman
- Department of Psychological and Brain Sciences, Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, USA
| | - Howard C Becker
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA. .,Charleston Alcohol Research Center, Medical University of South Carolina, Charleston, SC, USA. .,Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA. .,Department of Veterans Affairs, Ralph H. Johnson VA Medical Center, Charleston, SC, USA.
| |
Collapse
|
33
|
Langer TM, Neumueller SE, Crumley E, Burgraff NJ, Talwar S, Hodges MR, Pan L, Forster HV. Ventilation and neurochemical changes during µ-opioid receptor activation or blockade of excitatory receptors in the hypoglossal motor nucleus of goats. J Appl Physiol (1985) 2017; 123:1532-1544. [PMID: 28839004 DOI: 10.1152/japplphysiol.00592.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Neuromodulator interdependence posits that changes in one or more neuromodulators are compensated by changes in other modulators to maintain stability in the respiratory control network. Herein, we studied compensatory neuromodulation in the hypoglossal motor nucleus (HMN) after chronic implantation of microtubules unilaterally ( n = 5) or bilaterally ( n = 5) into the HMN. After recovery, receptor agonists or antagonists in mock cerebrospinal fluid (mCSF) were dialyzed during the awake and non-rapid eye movement (NREM) sleep states. During day studies, dialysis of the µ-opioid inhibitory receptor agonist [d-Ala2, N-MePhe4, Gly-ol]enkephalin (DAMGO; 100 µM) decreased pulmonary ventilation (V̇i), breathing frequency ( f), and genioglossus (GG) muscle activity but did not alter neuromodulators measured in the effluent mCSF. However, neither unilateral dialysis of a broad spectrum muscarinic receptor antagonist (atropine; 50 mM) nor unilateral or bilateral dialysis of a mixture of excitatory receptor antagonists altered V̇i or GG activity, but all of these did increase HMN serotonin (5-HT) levels. Finally, during night studies, DAMGO and excitatory receptor antagonist decreased ventilatory variables during NREM sleep but not during wakefulness. These findings contrast with previous dialysis studies in the ventral respiratory column (VRC) where unilateral DAMGO or atropine dialysis had no effects on breathing and bilateral DAMGO or unilateral atropine increased V̇i and f and decreased GABA or increased 5-HT, respectively. Thus we conclude that the mechanisms of compensatory neuromodulation are less robust in the HMN than in the VRC under physiological conditions in adult goats, possibly because of site differences in the underlying mechanisms governing neuromodulator release and consequently neuronal activity, and/or responsiveness of receptors to compensatory neuromodulators. NEW & NOTEWORTHY Activation of inhibitory µ-opioid receptors in the hypoglossal motor nucleus decreased ventilation under physiological conditions and did not affect neurochemicals in effluent dialyzed mock cerebral spinal fluid. These findings contrast with studies in the ventral respiratory column where unilateral [d-Ala2, N-MePhe4, Gly-ol]enkephalin (DAMGO) had no effects on ventilation and bilateral DAMGO or unilateral atropine increased ventilation and decreased GABA or increased serotonin, respectively. Our data support the hypothesis that mechanisms that govern local compensatory neuromodulation within the brain stem are site specific under physiological conditions.
Collapse
Affiliation(s)
- Thomas M Langer
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | | | - Emma Crumley
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Nicholas J Burgraff
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Sawan Talwar
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Neuroscience Research Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Lawrence Pan
- Department of Physical Therapy, Marquette University , Milwaukee, Wisconsin
| | - Hubert V Forster
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Neuroscience Research Center, Medical College of Wisconsin , Milwaukee, Wisconsin.,Zablocki Veterans Affairs Medical Center , Milwaukee, Wisconsin
| |
Collapse
|
34
|
Chen YW, Shieh JP, Liu KS, Wang JJ, Hung CH. Naloxone prolongs cutaneous nociceptive block by lidocaine in rats. Fundam Clin Pharmacol 2017; 31:636-642. [PMID: 28677297 DOI: 10.1111/fcp.12302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/08/2017] [Accepted: 06/30/2017] [Indexed: 11/26/2022]
Abstract
We aimed to investigate the local anesthetic properties of naloxone alone or as an adjunct for the local anesthetic lidocaine. After the block of the cutaneous trunci muscle reflex (CTMR) with drugs delivery by subcutaneous infiltration, cutaneous nociceptive block was tested on the ratsꞌ backs. We demonstrated that naloxone, as well as lidocaine, elicited cutaneous analgesia dose-dependently. The relative potency in inducing cutaneous analgesia was lidocaine [22.6 (20.1 - 25.4) μmol/kg] > naloxone [43.2 (40.3 - 46.4) μmol/kg] (P < 0.05). On an equianesthetic basis [50% effective dose (ED50 ), ED25 , and ED75 ], naloxone displayed a greater duration of cutaneous analgesic action than lidocaine (P < 0.01). Coadministration of lidocaine (ED95 or ED50 ) and ineffective-dose naloxone (13.3 μmol/kg) intensifies sensory block (P < 0.01) with prolonged duration of action (P < 0.001) compared with lidocaine (ED95 or ED50 ) alone or naloxone (13.3 μmol/kg) alone on infiltrative cutaneous analgesia. The preclinical data showed that naloxone is less potent than lidocaine as an infiltrative anesthetic, but its analgesic duration was longer than that of lidocaine. Furthermore, naloxone prolongs lidocaine analgesia, acting synergistically for nociceptive block.
Collapse
Affiliation(s)
- Yu-Wen Chen
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan.,Department of Physical Therapy, College of Health Care, China Medical University, Taichung, Taiwan
| | - Ja-Ping Shieh
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan.,Center for General Education, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Kuo-Sheng Liu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Jhi-Joung Wang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Ching-Hsia Hung
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Allied Health Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
35
|
Barrons RW, Woods JA. Low-Dose Naloxone for Prophylaxis of Postoperative Nausea and Vomiting: A Systematic Review and Meta-analysis. Pharmacotherapy 2017; 37:546-554. [DOI: 10.1002/phar.1930] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Robert W. Barrons
- Department of Pharmacy; Wingate University School of Pharmacy; Wingate North Carolina
| | - Joseph Andrew Woods
- Department of Pharmacy; Wingate University School of Pharmacy; Wingate North Carolina
| |
Collapse
|
36
|
Zhao Q, Zhang L, Shu R, Wang C, Yu Y, Wang H, Wang G. Involvement of Spinal PKMζ Expression and Phosphorylation in Remifentanil-Induced Long-Term Hyperalgesia in Rats. Cell Mol Neurobiol 2017; 37:643-653. [PMID: 27380044 DOI: 10.1007/s10571-016-0401-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/27/2016] [Indexed: 10/21/2022]
Abstract
Up-regulation of GluN2B-containing N-methyl-D-aspartate receptors (NMDARs) expression and trafficking is the key mechanism for remifentanil-induced hyperalgesia (RIH), nevertheless, the signaling pathway and pivotal proteins involved in RIH remain equivocal. PKMζ, an isoform of protein kinase C (PKC), maintains pain memory storage in neuropathic pain and inflammatory pain, which plays a parallel role regulated by NMDARs in long-term memory trace. In the present study, Zeta Inhibitory Peptide (ZIP), a PKMζ inhibitor, and a selective GluN2B antagonist Ro-256981 are injected intrathecally before remifentanil infusion (1 μg kg-1 min-1 for 1 h, iv) in order to detect whether GluN2B contributes to RIH through affecting synthesis and activity of PKMζ in spinal dorsal horn. Nociceptive tests are measured by Paw withdrawal mechanical threshold (PWT) and paw withdrawal thermal latency (PWL). The L4-L6 segments of dorsal horn taken from rats with RIH are for determining expression of PKMζ and pPKMζ by Western blot and immunohistochemistry. Our data suggest that remifentanil infusion causes an increase of PKMζ in expression and phosphorylation in rats with nociceptive sensitization, beginning at 2 h, peaked at 2 days, and returned to basal level at 7 days. ZIP (10 ng) could block behavioral sensitization induced by remifentanil. Ro25-6981 dosage-dependently attenuated mechanical and thermal hyperalgesia and reversed expression of PKMζ and pPKMζ, indicating that GluN2B-containing NMDA receptor facilitates development of RIH through mediating expression and activity of spinal PKMζ in rats. Although detailed mechanisms require further comprehensive study, the preventive role of Ro25-6981 and ZIP provide novel options for the effective precaution of RIH in clinics.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Linlin Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Ruichen Shu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Chunyan Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Haiyun Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Tianjin Research Institute of Anesthesiology, Tianjin, 300052, China.
| |
Collapse
|
37
|
Farmer AD, Gallagher J, Bruckner-Holt C, Aziz Q. Narcotic bowel syndrome. Lancet Gastroenterol Hepatol 2017; 2:361-368. [DOI: 10.1016/s2468-1253(16)30217-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/12/2016] [Accepted: 12/12/2016] [Indexed: 02/06/2023]
|
38
|
Ultralow Dose of Naloxone as an Adjuvant to Intrathecal Morphine Infusion Improves Perceived Quality of Sleep but Fails to Alter Persistent Pain: A Randomized, Double-blind, Controlled Study. Clin J Pain 2016; 31:968-75. [PMID: 25629634 PMCID: PMC4894772 DOI: 10.1097/ajp.0000000000000200] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Supplemental Digital Content is available in the text. Introduction: This randomized, cross-over, double-blind, controlled study of continuous intrathecal morphine administration in patients with severe, long-term pain addresses whether the supplementation of low doses of naloxone in this setting is associated with beneficial clinical effects. Methods: All of the study subjects (n=11) provided informed consent and were recruited from a subset of patients who were already undergoing long-term treatment with continuous intrathecal morphine because of difficult-to-treat pain. The patients were (in a randomized order) also given intrathecal naloxone (40 ng/24 h or 400 ng/24 h). As control, the patients’ ordinary dose of morphine without any additions was used. The pain (Numeric Rating Scale, NRS) during activity, perceived quality of sleep, level of activity, and quality of life as well as the levels of several proinflammatory and anti-inflammatory cytokines in the blood were assessed. The prestudy pain (NRS during activity) in the study group ranged from 3 to 10. Results: A total of 64% of the subjects reported improved quality of sleep during treatment with naloxone at a dose of 40 ng per 24 hours as compared with 9% with sham treatment (P=0.024). Although not statistically significant, pain was reduced by 2 NRS steps or more during supplemental treatment with naloxone in 36% of subjects when using the 40 ng per 24 hours dose and in 18% of the subjects when using naloxone 400 ng per 24 hours dose. The corresponding percentage among patients receiving unaltered treatment was 27%. Conclusions: To conclude, the addition of an ultralow dose of intrathecal naloxone (40 ng/24 h) to intrathecal morphine infusion in patients with severe, persistent pain improved perceived quality of sleep. We were not able to show any statistically significant effects of naloxone on pain relief, level of activity, or quality of life.
Collapse
|
39
|
Vergara AV, Pernites RB, Tiu BDB, de Leon ACC, Mangadlao JD, Binag CA, Advincula RC. Capacitive Detection of Morphine via Cathodically Electropolymerized, Molecularly Imprinted Poly(
p
‐aminostyrene) Films. MACROMOL CHEM PHYS 2016. [DOI: 10.1002/macp.201600127] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Aileen V. Vergara
- Graduate School Department of Chemistry University of Santo Tomas Espana St Manila 1105 Philippines
- Department of Chemistry University of Houston 112 Fleming Bldg Houston TX 77204‐5003 USA
| | - Roderick B. Pernites
- Department of Chemistry University of Houston 112 Fleming Bldg Houston TX 77204‐5003 USA
| | - Brylee David B. Tiu
- Department of Macromolecular Science and Engineering Case Western Reserve University Cleveland OH 44106 USA
| | - Al Christopher C. de Leon
- Department of Macromolecular Science and Engineering Case Western Reserve University Cleveland OH 44106 USA
| | - Joey D. Mangadlao
- Department of Macromolecular Science and Engineering Case Western Reserve University Cleveland OH 44106 USA
| | - Christina A. Binag
- Graduate School Department of Chemistry University of Santo Tomas Espana St Manila 1105 Philippines
| | - Rigoberto C. Advincula
- Department of Macromolecular Science and Engineering Case Western Reserve University Cleveland OH 44106 USA
| |
Collapse
|
40
|
Abstract
OBJECTIVES The paradoxical development of chronic abdominal pain is an underrecognized side effect of opioid use. Narcotic bowel syndrome (NBS), occurring in a small proportion of chronic opioid users, consists of chronic or intermittent abdominal pain, which often increases in severity despite continued or escalating dosages of opioids prescribed to relieve pain. METHODS A PubMed search was conducted using terms such as "narcotic bowel syndrome" and "opioid hyperalgesia" through January 2014. RESULTS Abdominal pain is the defining symptom of NBS and is thought to be mediated by central nervous system dysfunction; it should be distinguished from the peripheral side effects of opioids, such as nausea, bloating, intermittent vomiting, abdominal distension, and constipation. This latter cluster of symptoms is called opioid bowel dysfunction, although it may co-occur with NBS. Hypothesized mechanisms of the central effects of opioids on nociception in NBS include spinal cord inflammation and dysfunction in opioid receptor activity and related neuroanatomical substrates. With continued use, ∼6% of patients taking narcotics chronically will develop NBS, with profound consequences in terms of daily function. The primary management paradigm for NBS is a structured opioid withdrawal program accompanied by centrally acting adjunctive therapy comprising antidepressants, benzodiazepines, and clonidine to target pain, anxiety, and depression, and prevent withdrawal effects, in addition to peripherally acting agents such as laxatives (e.g., osmotic laxatives and chloride channel activators) to control transient constipation. Such structured withdrawal programs have been prospectively evaluated in small clinical trials and have met with considerable success in the short term. CONCLUSIONS Because rates of NBS are likely to rise, integrated intensive pharmacotherapy and psychosocial interventions are needed to help patients with NBS go off and stay off opioids. These programs will likely also reduce comorbid psychopathology and lead to adequate pain control and improved quality of life.
Collapse
|
41
|
Drewes AM, Munkholm P, Simrén M, Breivik H, Kongsgaard UE, Hatlebakk JG, Agreus L, Friedrichsen M, Christrup LL. Definition, diagnosis and treatment strategies for opioid-induced bowel dysfunction–Recommendations of the Nordic Working Group. Scand J Pain 2016; 11:111-122. [DOI: 10.1016/j.sjpain.2015.12.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/12/2015] [Indexed: 02/07/2023]
Abstract
Abstract
Background and aims
Opioid-induced bowel dysfunction (OIBD) is an increasing problem due to the common use of opioids for pain worldwide. It manifests with different symptoms, such as dry mouth, gastro-oesophageal reflux, vomiting, bloating, abdominal pain, anorexia, hard stools, constipation and incomplete evacuation. Opioid-induced constipation (OIC) is one of its many symptoms and probably the most prevalent. The current review describes the pathophysiology, clinical implications and treatment of OIBD.
Methods
The Nordic Working Group was formed to provide input for Scandinavian specialists in multiple, relevant areas. Seven main topics with associated statements were defined. The working plan provided a structured format for systematic reviews and included instructions on how to evaluate the level of evidence according to the GRADE guidelines. The quality of evidence supporting the different statements was rated as high, moderate or low. At a second meeting, the group discussed and voted on each section with recommendations (weak and strong) for the statements.
Results
The literature review supported the fact that opioid receptors are expressed throughout the gastrointestinal tract. When blocked by exogenous opioids, there are changes in motility, secretion and absorption of fluids, and sphincter function that are reflected in clinical symptoms. The group supported a recent consensus statement for OIC, which takes into account the change in bowel habits for at least one week rather than focusing on the frequency of bowel movements. Many patients with pain receive opioid therapy and concomitant constipation is associated with increased morbidity and utilization of healthcare resources. Opioid treatment for acute postoperative pain will prolong the postoperative ileus and should also be considered in this context. There are no available tools to assess OIBD, but many rating scales have been developed to assess constipation, and a few specifically address OIC. A clinical treatment strategy for OIBD/OIC was proposed and presented in a flowchart. First-line treatment of OIC is conventional laxatives, lifestyle changes, tapering the opioid dosage and alternative analgesics. Whilst opioid rotation may also improve symptoms, these remain unalleviated in a substantial proportion of patients. Should conventional treatment fail, mechanism-based treatment with opioid antagonists should be considered, and they show advantages over laxatives. It should not be overlooked that many reasons for constipation other than OIBD exist, which should be taken into consideration in the individual patient.
Conclusion and implications
It is the belief of this Nordic Working Group that increased awareness of adverse effects and OIBD, particularly OIC, will lead to better pain treatment in patients on opioid therapy. Subsequently, optimised therapy will improve quality of life and, from a socio-economic perspective, may also reduce costs associated with hospitalisation, sick leave and early retirement in these patients.
Collapse
Affiliation(s)
- Asbjørn M. Drewes
- Mech-Sense, Department of Gastroenterology and Hepatology , Aalborg University Hospital , Hobrovej Denmark
| | - Pia Munkholm
- NOH (Nordsjællands Hospital) Gastroenterology , Hillerød Denmark
| | - Magnus Simrén
- Department of Internal Medicine & Clinical Nutrition , Institute of Medicine, Sahlgrenska Academy, University of Gothenburg , Göteborg Sweden
| | - Harald Breivik
- Department of Pain Management and Research , Oslo University Hospital and University of Oslo , Rikshospitalet Norway
| | - Ulf E. Kongsgaard
- Department of Anaesthesiology, Division of Emergencies and Critical Care , Oslo University Hospital, Norway and Medical Faculty, University of Oslo , Rikshospitalet Norway
| | - Jan G. Hatlebakk
- Department of Clinical Medicine , Haukeland University Hospital , Bergen , Norway
| | - Lars Agreus
- Division of Family Medicine , Karolinska Institute , Stockholm , Sweden
| | - Maria Friedrichsen
- Department of Social and Welfare Studies , Faculty of Medicine and Health Sciences , Norrköping , Sweden
| | - Lona L. Christrup
- Department of Drug Design and Pharmacology , Faculty of Health Sciences, University of Copenhagen , københavn Denmark
| |
Collapse
|
42
|
A randomized controlled study of the effects of adding ultra-low dose naloxone to lidocaine for intravenous regional anesthesia. EGYPTIAN JOURNAL OF ANAESTHESIA 2016. [DOI: 10.1016/j.egja.2015.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
43
|
Wang D, Zeng J, Li Q, Huang J, Couture R, Hong Y. Contribution of adrenomedullin to the switch of G protein-coupled μ-opioid receptors from Gi to Gs in the spinal dorsal horn following chronic morphine exposure in rats. Br J Pharmacol 2016; 173:1196-207. [PMID: 26750148 DOI: 10.1111/bph.13419] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 12/30/2015] [Accepted: 01/07/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Chronic exposure to morphine increases spinal adrenomedullin (AM) bioactivity resulting in the development and maintenance of morphine tolerance. This study investigated the possible involvement of AM in morphine-evoked alteration in μ-opioid receptor-coupled G proteins. EXPERIMENTAL APPROACH Agents were administered intrathecally (i.t.) in rats. Nociceptive behaviours and cumulative dose-response of morphine analgesia were assessed. Neurochemicals in the spinal dorsal horn were assayed by immunoprecipitation, Western blot analysis and ELISA. KEY RESULTS Intrathecal injection of AM (8 μg) for 9 days decreased and increased the levels of μ receptor-coupled Gi and Gs proteins respectively. Morphine stimulation (5 μg) after chronic treatment with AM also induced an increase in cAMP production in the spinal dorsal horn. Co-administration of the selective AM receptor antagonist AM22-52 inhibited chronic morphine-evoked switch of G protein-coupled μ receptor from Gi to Gs. Chronic exposure to AM increased the phosphorylation of cAMP-responsive element-binding protein (CREB) and ERK. Co-administration of the PKA inhibitor H-89 (5 μg) or MEK1 inhibitor PD98059 (1 μg) reversed the AM-induced thermal/mechanical hypersensitivity, decline in morphine analgesic potency, switch of G protein-coupled μ receptor and increase in cAMP. CONCLUSIONS AND IMPLICATIONS The present study supports the hypothesis that an increase in AM activity in the spinal dorsal horn contributes to the switch of the μ receptor-coupled G protein from Gi to Gs protein via the activation of cAMP/PKA/CREB and ERK signalling pathways in chronic morphine use.
Collapse
Affiliation(s)
- Dongmei Wang
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian, China
| | - Juan Zeng
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian, China
| | - Qi Li
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian, China
| | - Jianzhong Huang
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian, China
| | - Réjean Couture
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Yanguo Hong
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian, China
| |
Collapse
|
44
|
Keefer L, Drossman DA, Guthrie E, Simrén M, Tillisch K, Olden K, Whorwell PJ. Centrally Mediated Disorders of Gastrointestinal Pain. Gastroenterology 2016; 150:S0016-5085(16)00225-0. [PMID: 27144628 DOI: 10.1053/j.gastro.2016.02.034] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 02/11/2016] [Indexed: 02/07/2023]
Abstract
Centrally Mediated Abdominal Pain Syndrome (CAPS), formerly known as Functional Abdominal Pain Syndrome, can be distinguished from other functional GI disorders by its strong central component and relative independence from motility disturbances. CAPS is a result of central sensitization with disinhibition of pain signals rather than increased peripheral afferent excitability. A newly described condition, Narcotic Bowel Syndrome (NBS)/Opioid-Induced GI Hyperalgesia, is characterized by the paradoxical development of or increases in abdominal pain associated with continuous or increasing dosages of opioids. Patients only have relief when opioids are withdrawn. We define both conditions in the context of epidemiology, pathophysiology, clinical evaluation and treatment, emphasizing the importance of a physician-patient relationship in all aspects of care.
Collapse
Affiliation(s)
- Laurie Keefer
- Division of Gastroenterology, Ichan School of Medicine at Mount Sinai, 17 East 102nd Street - 5th Floor, New York, NY 10029.
| | - Douglas A Drossman
- Center for Functional GI and Motility Disorders, University of North Carolina and Center for Education and Practice of Biopsychosocial Care LLC, Drossman Gastroenterology PLLC, Chapel Hill NC USA
| | - Elspeth Guthrie
- Mental Health and Social Care Trust, Manchester Royal Infirmary, Manchester UK
| | - Magnus Simrén
- Dept. of Internal Medicine & Clinical Nutrition, Institute of Medicine Sahlgrenska Academy, University of Gothenburg, Gothenburg Sweden
| | - Kirsten Tillisch
- Oppenheimer Family Center for Neurobiology of Stress Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles CA USA
| | - Kevin Olden
- SJHMC Internal Medicine Department, Phoenix AZ USA
| | - Peter J Whorwell
- Education and Research Centre Wythenshawe Hospital, Manchester UK
| |
Collapse
|
45
|
Morphine promotes cancer stem cell properties, contributing to chemoresistance in breast cancer. Oncotarget 2016; 6:3963-76. [PMID: 25686831 PMCID: PMC4414166 DOI: 10.18632/oncotarget.2894] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 12/11/2014] [Indexed: 01/24/2023] Open
Abstract
Morphine is an opioid analgesic drug commonly used for pain relief in cancer patients. Here, we report that morphine enhances the mammosphere forming capacity and increases the expression of stemness-related transcription factors Oct4, Sox2 and Nanog. Treatment with morphine leads to enrichment of a side population fraction in MCF-7 cells and the CD44+/CD24−/low population in BT549 cells. Consistently, morphine activates Wnt/β-catenin signaling to induce epithelial to mesenchymal transition and promotes metastasis. Moreover, morphine decreases the sensitivity of traditional anti-cancer drugs in breast cancer cells. Nalmefene, an antagonist of morphine, reverses morphine-induced cancer stem cell properties and chemoresistance in breast cancer. In addition, nalmefene abolishes morphine enhancing tumorigenesis in a NOD/SCID mouse model. In conclusion, our findings demonstrate that morphine contributes to chemoresistance via expanding the population of cancer stem cells and promotes tumor growth, thereby revealing a novel role of morphine and providing some new guides in clinical use of morphine.
Collapse
|
46
|
Komatsu T, Katsuyama S, Nagase H, Mizoguchi H, Sakurada C, Tsuzuki M, Sakurada S, Sakurada T. Intrathecal morphine-3-glucuronide-induced nociceptive behavior via Delta-2 opioid receptors in the spinal cord. Pharmacol Biochem Behav 2016; 140:68-74. [DOI: 10.1016/j.pbb.2015.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 10/11/2015] [Accepted: 10/13/2015] [Indexed: 12/30/2022]
|
47
|
Block L. Glial dysfunction and persistent neuropathic postsurgical pain. Scand J Pain 2016; 10:74-81. [PMID: 28361776 DOI: 10.1016/j.sjpain.2015.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/10/2015] [Accepted: 10/13/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Acute pain in response to injury is an important mechanism that serves to protect living beings from harm. However, persistent pain remaining long after the injury has healed serves no useful purpose and is a disabling condition. Persistent postsurgical pain, which is pain that lasts more than 3 months after surgery, affects 10-50% of patients undergoing elective surgery. Many of these patients are affected by neuropathic pain which is characterised as a pain caused by lesion or disease in the somatosensory nervous system. When established, this type of pain is difficult to treat and new approaches for prevention and treatment are needed. A possible contributing mechanism for the transition from acute physiological pain to persistent pain involves low-grade inflammation in the central nervous system (CNS), glial dysfunction and subsequently an imbalance in the neuron-glial interaction that causes enhanced and prolonged pain transmission. AIM This topical review aims to highlight the contribution that inflammatory activated glial cell dysfunction may have for the development of persistent pain. METHOD Relevant literature was searched for in PubMed. RESULTS Immediately after an injury to a nerve ending in the periphery such as in surgery, the inflammatory cascade is activated and immunocompetent cells migrate to the site of injury. Macrophages infiltrate the injured nerve and cause an inflammatory reaction in the nerve cell. This reaction leads to microglia activation in the central nervous system and the release of pro-inflammatory cytokines that activate and alter astrocyte function. Once the astrocytes and microglia have become activated, they participate in the development, spread, and potentiation of low-grade neuroinflammation. The inflammatory activated glial cells exhibit cellular changes, and their communication to each other and to neurons is altered. This renders neurons more excitable and pain transmission is enhanced and prolonged. Astrocyte dysfunction can be experimentally restored using the combined actions of a μ-opioid receptor agonist, a μ-opioid receptor antagonist, and an anti-epileptic agent. To find these agents we searched the literature for substances with possible anti-inflammatory properties that are usually used for other purposes in medicine. Inflammatory induced glial cell dysfunction is restorable in vitro by a combination of endomorphine-1, ultralow doses of naloxone and levetiracetam. Restoring inflammatory-activated glial cells, thereby restoring astrocyte-neuron interaction has the potential to affect pain transmission in neurons. CONCLUSION Surgery causes inflammation at the site of injury. Peripheral nerve injury can cause low-grade inflammation in the CNS known as neuroinflammation. Low-grade neuroinflammation can cause an imbalance in the glial-neuron interaction and communication. This renders neurons more excitable and pain transmission is enhanced and prolonged. Astrocytic dysfunction can be restored in vitro by a combination of endomorphin-1, ultralow doses of naloxone and levetiracetam. This restoration is essential for the interaction between astrocytes and neurons and hence also for modulation of synaptic pain transmission. IMPLICATIONS Larger studies in clinical settings are needed before these findings can be applied in a clinical context. Potentially, by targeting inflammatory activated glial cells and not only neurons, a new arena for development of pharmacological agents for persistent pain is opened.
Collapse
Affiliation(s)
- Linda Block
- Institute of Clinical Sciences at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Anesthesiology and Intensive Care, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
48
|
Structural and functional interactions between six-transmembrane μ-opioid receptors and β2-adrenoreceptors modulate opioid signaling. Sci Rep 2015; 5:18198. [PMID: 26657998 PMCID: PMC4676002 DOI: 10.1038/srep18198] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/11/2015] [Indexed: 12/20/2022] Open
Abstract
The primary molecular target for clinically used opioids is the μ-opioid receptor (MOR). Besides the major seven-transmembrane (7TM) receptors, the MOR gene codes for alternatively spliced six-transmembrane (6TM) isoforms, the biological and clinical significance of which remains unclear. Here, we show that the otherwise exclusively intracellular localized 6TM-MOR translocates to the plasma membrane upon coexpression with β2-adrenergic receptors (β2-ARs) through an interaction with the fifth and sixth helices of β2-AR. Coexpression of the two receptors in BE(2)-C neuroblastoma cells potentiates calcium responses to a 6TM-MOR ligand, and this calcium response is completely blocked by a selective β2-antagonist in BE(2)-C cells, and in trigeminal and dorsal root ganglia. Co-administration of 6TM-MOR and β2-AR ligands leads to substantial analgesic synergy and completely reverses opioid-induced hyperalgesia in rodent behavioral models. Together, our results provide evidence that the heterodimerization of 6TM-MOR with β2-AR underlies a molecular mechanism for 6TM cellular signaling, presenting a unique functional responses to opioids. This signaling pathway may contribute to the hyperalgesic effects of opioids that can be efficiently blocked by β2-AR antagonists, providing a new avenue for opioid therapy.
Collapse
|
49
|
Convertino M, Samoshkin A, Viet CT, Gauthier J, Li Fraine SP, Sharif-Naeini R, Schmidt BL, Maixner W, Diatchenko L, Dokholyan NV. Differential Regulation of 6- and 7-Transmembrane Helix Variants of μ-Opioid Receptor in Response to Morphine Stimulation. PLoS One 2015; 10:e0142826. [PMID: 26554831 PMCID: PMC4640872 DOI: 10.1371/journal.pone.0142826] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/27/2015] [Indexed: 11/18/2022] Open
Abstract
The pharmacological effect of opioids originates, at the cellular level, by their interaction with the μ-opioid receptor (mOR) resulting in the regulation of voltage-gated Ca2+ channels and inwardly rectifying K+ channels that ultimately modulate the synaptic transmission. Recently, an alternative six trans-membrane helix isoform of mOR, (6TM-mOR) has been identified, but its function and signaling are still largely unknown. Here, we present the structural and functional mechanisms of 6TM-mOR signaling activity upon binding to morphine. Our data suggest that despite the similarity of binding modes of the alternative 6TM-mOR and the dominant seven trans-membrane helix variant (7TM-mOR), the interaction with morphine generates different dynamic responses in the two receptors, thus, promoting the activation of different mOR-specific signaling pathways. We characterize a series of 6TM-mOR-specific cellular responses, and observed that they are significantly different from those for 7TM-mOR. Morphine stimulation of 6TM-mOR does not promote a cellular cAMP response, while it increases the intracellular Ca2+ concentration and reduces the cellular K+ conductance. Our findings indicate that 6TM-mOR has a unique contribution to the cellular opioid responses. Therefore, it should be considered as a relevant target for the development of novel pharmacological tools and medical protocols involving the use of opioids.
Collapse
Affiliation(s)
- Marino Convertino
- Biochemistry and Biophysics Department, University of North Carolina, 120 Mason Farm Road, Chapel Hill, NC, United States of America, 27599
| | - Alexander Samoshkin
- Alan Edwards Centre for Research on Pain, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada, H3A 0G1
| | - Chi T Viet
- Bluestone Center for Clinic Research, New York University, New York, NY, United States of America, 10010
| | - Josee Gauthier
- Center for Pain Research and Innovation, University of North Carolina, 385 S. Columbia Street, Chapel Hill, NC, United States of America, 27599
| | - Steven P Li Fraine
- Department of Physiology and Cell Information Systems, McGill University, 3649 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 0B1
| | - Reza Sharif-Naeini
- Department of Physiology and Cell Information Systems, McGill University, 3649 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 0B1
| | - Brian L Schmidt
- Bluestone Center for Clinic Research, New York University, New York, NY, United States of America, 10010
| | - William Maixner
- Center for Pain Research and Innovation, University of North Carolina, 385 S. Columbia Street, Chapel Hill, NC, United States of America, 27599
| | - Luda Diatchenko
- Alan Edwards Centre for Research on Pain, McGill University, 740 Dr. Penfield Avenue, Montreal, Quebec, Canada, H3A 0G1
| | - Nikolay V Dokholyan
- Biochemistry and Biophysics Department, University of North Carolina, 120 Mason Farm Road, Chapel Hill, NC, United States of America, 27599.,Center for Pain Research and Innovation, University of North Carolina, 385 S. Columbia Street, Chapel Hill, NC, United States of America, 27599
| |
Collapse
|
50
|
Chan P, Lutfy K. Molecular Changes in Opioid Addiction: The Role of Adenylyl Cyclase and cAMP/PKA System. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 137:203-27. [PMID: 26810003 DOI: 10.1016/bs.pmbts.2015.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
For centuries, opiate analgesics have had a considerable presence in the treatment of moderate to severe pain. While effective in providing analgesia, opiates are notorious in exerting many undesirable adverse reactions. The receptor targets and the intracellular effectors of opioids have largely been identified. Furthermore, much of the mechanisms underlying the development of tolerance, dependence, and withdrawal have been delineated. Thus, there is a focus on developing novel compounds or strategies in mitigating or avoiding the development of tolerance, dependence, and withdrawal. This review focuses on the adenylyl cyclase and cyclic adenosine 3,5-monophosphate (cAMP)/protein kinase A (AC/cAMP/PKA) system as the central player in mediating the acute and chronic effects of opioids. This chapter also reviews the neuronal adaptive changes in the locus coeruleus, amygdala, periaqueductal gray, and ventral tegmental area induced by acute and chronic actions of opioid because these neuronal adaptive changes in these regions may underlie the behavioral changes observed in opiate users and abusers.
Collapse
Affiliation(s)
- Patrick Chan
- Department of Pharmacy and Pharmacy Administration, Western University of Health Sciences, College of Pharmacy, Pomona, California, USA.
| | - Kabirullah Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|