1
|
Yu YM, Xia SH, Xu Z, Zhao WN, Song L, Pan X, Zhong CC, Wang D, Gao YH, Yang JX, Wu P, Zhang H, An S, Cao JL, Ding HL. An accumbal microcircuit for the transition from acute to chronic pain. Curr Biol 2025; 35:1730-1749.e5. [PMID: 40112811 DOI: 10.1016/j.cub.2025.02.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Persistent nociceptive inputs arising from peripheral tissues or/and nerve injuries cause maladaptive changes in neurons or neural circuits in the central nervous system, which further confer acute injury into chronic pain transitions (pain chronification) even after the injury is resolved. However, the critical brain regions and their neural mechanisms involved in this transition have not yet been elucidated. Here, we reveal an accumbal microcircuit that is essential for pain chronification. Notably, the increase of neuronal activity in the nucleus accumbens shell (NAcS) in the acute phase (<7 days) and in core (NAcC) in the chronic phase (14-21 days) was detected in a neuropathic pain mouse model. Importantly, we demonstrated that the NAcS neuronal activation in the acute phase of injury was necessary and sufficient for the development of chronic neuropathic pain. This process was mediated by the accumbal dopamine D2 receptor-expressing neuronal microcircuit from NAcS to NAcC. Thus, our findings reveal an accumbal microcircuit mechanism for pain chronification and suggest that the early intervention targeting this microcircuit may provide a therapeutic approach to pain chronification.
Collapse
Affiliation(s)
- Yu-Mei Yu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Sun-Hui Xia
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Zheng Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Wei-Nan Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lingzhen Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Xiangyu Pan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Chao-Chao Zhong
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Di Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yi-Hong Gao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Peng Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Shuming An
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China.
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
2
|
Fu C, Wu P, Wu F, He W, Luo Q, Liang H, Wang H, Li Y. Enhanced Pain Sensitization Induced by Chronic Sleep Deprivation: The Role of Dopamine D2 Receptors-Dependent Homer1a Protein. ACS Chem Neurosci 2025; 16:1043-1054. [PMID: 40008853 DOI: 10.1021/acschemneuro.4c00640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
Numerous studies have demonstrated a positive correlation between sleep disorders and hyperalgesia. These sleep disorders adversely affect the descending pain regulatory system. Researchers have extensively studied the midbrain dopamine system in relation to pain associated with sleep disturbances. Our study shows that chronic sleep deprivation decreases dopamine responses to noxious stimuli within the mouse nucleus accumbens, regulated by dopamine release and intracellular signals. Furthermore, we confirmed that the dopamine D2 receptors play a critical role in the pain associated with chronic sleep deprivation. Importantly, we revealed that homer1a in D2 receptor neurons enhances AMPA receptors expression.
Collapse
Affiliation(s)
- Chao Fu
- Department of Anesthesiology, Jinan University First Affiliated Hospital, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou 510630, Guangdong, China
| | - Peng Wu
- Department of Anesthesiology, Jinan University First Affiliated Hospital, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou 510630, Guangdong, China
| | - Fancan Wu
- Department of Anesthesiology, The First People's Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| | - Wanyou He
- Department of Anesthesiology, The First People's Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| | - Qichen Luo
- Department of Anesthesiology, Jinan University First Affiliated Hospital, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou 510630, Guangdong, China
| | - Hongbin Liang
- Department of Anesthesiology, The First People's Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| | - Hanbing Wang
- Department of Anesthesiology, The First People's Hospital of Foshan, 81# North of Ling Nan Road, Foshan 528000, Guangdong, China
| | - Yalan Li
- Department of Anesthesiology, Jinan University First Affiliated Hospital, Number 613, The West of Huangpu Avenue, Tianhe Region, Guangzhou 510630, Guangdong, China
| |
Collapse
|
3
|
Dopheide JA, Roth WR, Chu MKL. Insomnia in ambulatory care: A clinical review. Am J Health Syst Pharm 2025; 82:265-284. [PMID: 39324566 DOI: 10.1093/ajhp/zxae255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Indexed: 09/27/2024] Open
Abstract
PURPOSE One-third to one-half of ambulatory care patients report insomnia. The objective of this clinical review is to detail the many causes and evidence-based treatment options for insomnia in outpatients and to recommend screening and summarize evidence for the place in therapy of prescription and nonprescription treatments. SUMMARY This work provides an overview of the literature on insomnia regarding causes, patient assessment, and nonpharmacological and pharmacological treatments. Patients who present with insomnia should be assessed for sleep apnea, restless legs syndrome, narcolepsy, and all contributing medications as well as medical, psychiatric, and substance use disorder diagnoses. The type of insomnia, namely difficulty falling asleep, difficulty maintaining sleep, and early morning awakening with resulting functional impairment, should be documented in addition to whether insomnia is short term or persistent. Cognitive behavioral therapy for insomnia (CBT-I) or digital CBT-I is first-line treatment for all patients with insomnia irrespective of the cause or type. Nonprescription treatments such as antihistamines or melatonin are for select populations. Prescription hypnotics are best utilized on an as-needed basis or for nightly use for less than 6 weeks. Z-hypnotics are safe and effective for insomnia in persons with depression or an anxiety disorder but should be avoided in older individuals or if there is respiratory or cognitive impairment. Orexin receptor antagonists are effective for sleep initiation and maintenance in healthy persons or if there is mild cognitive impairment, but they require further study in individuals with psychiatric and medical diagnoses. Trazodone is the most prescribed off-label treatment due to its efficacy for sleep initiation and maintenance and its lack of abuse potential. CONCLUSION Insomnia treatment should be guided by patient age, diagnoses, and type of insomnia. Pharmacological treatments should be used at the lowest effective dose for the shortest duration of time.
Collapse
Affiliation(s)
- Julie A Dopheide
- USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Winter R Roth
- USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA
| | - Michelle K L Chu
- USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
4
|
Niklasson A, Finan PH, Smith MT, Forsberg A, Dietz N, Kander T, Werner MU, Irwin MR, Kosek E, Bjurström MF. The relationship between preoperative sleep disturbance and acute postoperative pain control: A systematic review and meta-analysis. Sleep Med Rev 2025; 79:102014. [PMID: 39504912 DOI: 10.1016/j.smrv.2024.102014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/02/2024] [Accepted: 09/27/2024] [Indexed: 11/08/2024]
Abstract
Poor preoperative sleep quality and impaired sleep continuity may heighten acute postoperative pain intensity and increase analgesic consumption, with negative implications for recovery, mental and physical health. The main objective of the current review was to investigate the relationship between preoperative sleep disturbance and acute postoperative pain control. Four electronic databases were systematically searched from inception to December 2023. Two reviewers screened articles, extracted data, and assessed risk of bias for each included study. The search identified 26 prospective cohort studies and 3 retrospective cohort studies (16104 participants). Of the 29 included studies, 23 focused on preoperative insomnia symptoms, and three studies each focused on preoperative objective sleep continuity or sleep-disordered breathing. Meta-analysis, based on five studies with 1226 participants, showed that clinically significant preoperative insomnia symptoms were associated with moderate to severe pain intensity on the first postoperative day (odds ratio 2.69 (95 % confidence interval 2.03-3.57), p < 0.0001). Qualitative analysis showed relatively robust associations between preoperative insomnia symptoms, impaired sleep continuity and poorer acute, as well as subacute, postoperative pain control. Findings related to obstructive sleep apnea syndrome were mixed. Given that insomnia is a potentially modifiable risk factor, interventions targeting sleep prior to surgery may improve postoperative pain control.
Collapse
Affiliation(s)
- Andrea Niklasson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Patrick H Finan
- Department of Anesthesiology, University of Virginia School of Medicine, VA, USA
| | - Michael T Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | - Thomas Kander
- Department of Anesthesiology and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Mads U Werner
- Multidisciplinary Pain Center, Neuroscience Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Michael R Irwin
- Department of Psychiatry and Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Eva Kosek
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin F Bjurström
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Department of Psychiatry and Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA.
| |
Collapse
|
5
|
Yin XS, Chen BR, Ye XC, Wang Y. Modulating the Pronociceptive Effect of Sleep Deprivation: A Possible Role for Cholinergic Neurons in the Medial Habenula. Neurosci Bull 2024; 40:1811-1825. [PMID: 39158824 PMCID: PMC11625038 DOI: 10.1007/s12264-024-01281-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 05/22/2024] [Indexed: 08/20/2024] Open
Abstract
Sleep deprivation has been shown to exacerbate pain sensitivity and may contribute to the onset of chronic pain, yet the precise neural mechanisms underlying this association remain elusive. In our study, we explored the contribution of cholinergic neurons within the medial habenula (MHb) to hyperalgesia induced by sleep deprivation in rats. Our findings indicate that the activity of MHb cholinergic neurons diminishes during sleep deprivation and that chemogenetic stimulation of these neurons can mitigate the results. Interestingly, we did not find a direct response of MHb cholinergic neurons to pain stimulation. Further investigation identified the interpeduncular nucleus (IPN) and the paraventricular nucleus of the thalamus (PVT) as key players in the pro-nociceptive effect of sleep deprivation. Stimulating the pathways connecting the MHb to the IPN and PVT alleviated the hyperalgesia. These results underscore the important role of MHb cholinergic neurons in modulating pain sensitivity linked to sleep deprivation, highlighting potential neural targets for mitigating sleep deprivation-induced hyperalgesia.
Collapse
Affiliation(s)
- Xiang-Sha Yin
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100083, China
- Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Beijing, 100730, China
| | - Bai-Rong Chen
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100083, China
- Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
| | - Xi-Chun Ye
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100083, China
- Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China
| | - Yun Wang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100083, China.
- Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, China.
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
6
|
Gholami-Zanjanbar M, Soleimanian F, Reyhani N, Hajizamani S, Sajadi AE, Ghofrani-Jahromi Z, Vaseghi S. Synaptophysin and GSK-3beta activity in the prefrontal cortex may underlie the effects of REM sleep deprivation and lithium on behavioral functions and memory performance in male rats. Pharmacol Biochem Behav 2024; 245:173894. [PMID: 39413852 DOI: 10.1016/j.pbb.2024.173894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/05/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Rapid-eye movement (REM) stage of sleep serves a critical role in processing cognitive and behavioral functions. Evidence shows that REM sleep deprivation (REM SD) strongly affects the mood state and cognitive abilities. However, there are many inconsistent reports. Although the exact molecular mechanisms underlying REM SD effects have not well been discovered, however, molecular factors including those affected synaptic plasticity and mood state may be involved. There are two important molecular factors that have not been well studied: synaptophysin and glycogen synthase kinase-3 beta (GSK-3beta). The present study aimed to investigate the role of synaptophysin and GSK-3beta in the modulation of memory and behavioral changes induced by REM SD and lithium (as a potent GSK-3beta inhibitor and mood stabilizer). Multiple platform apparatus was used to induce REM SD for 48 h. Lithium was injected at the dose of 50 mg/kg, intraperitoneal (i.p.). Locomotor activity, anxiety-like behavior, pain threshold, novel object recognition memory, and synaptophysin and GSK-3beta level in the prefrontal cortex were evaluated. Results showed REM SD increased locomotor activity, decreased pain threshold, impaired novel object recognition memory, decreased synaptophysin and increased GSK-3beta levels. Lithium reversed these effects. Anxiety-like behavior was unaffected. For the first time, the present study showed that GSK-3beta and synaptophysin may be involved in the modulation of behavior and cognition induced by REM SD and lithium. In conclusion, we suggested that GSK-3beta upregulation and synaptophysin downregulation may underlie the deleterious effects of REM SD, while lithium may counteract REM SD effects via restoring the level of both.
Collapse
Affiliation(s)
| | | | - Niloufar Reyhani
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Shadi Hajizamani
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir-Ehsan Sajadi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Zahra Ghofrani-Jahromi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Salar Vaseghi
- Cognitive Neuroscience Lab, Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
| |
Collapse
|
7
|
Sharma R, Parikh M, Chischolm A, Kempuraj D, Thakkar M. Dopamine D2 receptors in the accumbal core region mediates the effects of fentanyl on sleep-wakefulness. Neuroscience 2024; 560:11-19. [PMID: 39276843 DOI: 10.1016/j.neuroscience.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024]
Abstract
Fentanyl, a potent analgesic and addictive substance, significantly impacts sleep-wakefulness (S-W). Acutely, it promotes wake, whereas chronic abuse leads to severe sleep disruptions, including insomnia, which contributes to opioid use disorders (OUD), a chronic brain disease characterized by compulsive opioid use and harmful consequences. Although the critical association between sleep disruptions and fentanyl addiction is acknowledged, the precise mechanisms through which fentanyl influences sleep remain elusive. Recent studies highlight the role of the dopaminergic system of the nucleus accumbens (NAc) in S-W regulation, but its specific involvement in mediating fentanyl's effects on S-W remains unexplored. We hypothesized that dopamine D2 receptors mediate fentanyl-induced effects on S-W. To test this hypothesis, male C57BL/6J mice, instrumented with sleep recording electrodes and bilateral guide cannulas above the accumbal core region (NAcC), were utilized in this study. At dark onset, animals were bilaterally administered sulpiride (D2 receptors antagonist; 250 ng/side) in the NAcC followed by an intraperitoneal injection of fentanyl (1.2 mg/Kg). S-W was examined for the next 12 h. We found that systemic administration of fentanyl significantly increased wakefulness during the first 6 h of the dark which was followed by a significant increase in NREM and REM sleep during the second 6 h of the dark period. D2-receptor blockade significantly reduced this effect as evidenced by a significant reduction in fentanyl-induced wakefulness during first 6 h of dark period and sleep rebound during the second 6 h. Our findings suggest that D2 receptors in the NAcC plays a vital role in mediating the fentanyl-induced changes in S-W.
Collapse
Affiliation(s)
- Rishi Sharma
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO 65201, United States.
| | - Meet Parikh
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO 65201, United States
| | - Abigail Chischolm
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO 65201, United States
| | - Deepak Kempuraj
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO 65201, United States
| | - Mahesh Thakkar
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, University of Missouri, Columbia, MO 65201, United States
| |
Collapse
|
8
|
Sardi NF, Pescador AC, Torres-Chavez KE, Fischer L. Revealing a role of brainstem monoaminergic nuclei on the pronociceptive effect of sleep restriction. Neuropharmacology 2024; 258:110055. [PMID: 38950692 DOI: 10.1016/j.neuropharm.2024.110055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/03/2024]
Abstract
Sleep disturbances and persistent pain conditions are public health challenges worldwide. Although it is well-known that sleep deficit increases pain sensitivity, the underlying mechanisms remain elusive. We have recently demonstrated the involvement of nucleus accumbens (NAc) and anterior cingulate cortex (ACC) in the pronociceptive effect of sleep restriction. In this study, we found that sleep restriction increases c-Fos expression in NAc and ACC, suggesting hyperactivation of these regions during prolonged wakefulness in male Wistar rats. Blocking adenosine A2A receptors in the NAc or GABAA receptors in the ventral tegmental area (VTA), dorsal raphe nucleus (DRN), or locus coeruleus (LC) effectively mitigated the pronociceptive effect of sleep restriction. In contrast, the blockade of GABAA receptors in each of these nuclei only transiently reduced carrageenan-induced hyperalgesia. Pharmacological activation of dopamine D2, serotonin 5-HT1A and noradrenaline alpha-2 receptors within the ACC also prevented the pronociceptive effect of sleep restriction. While pharmacological inhibition of these same monoaminergic receptors in the ACC restored the pronociceptive effect which had been prevented by the GABAergic disinhibition of the of the VTA, DRN or LC. Overall, these findings suggest that the pronociceptive effect of sleep restriction relies on increased adenosinergic activity on NAc, heightened GABAergic activity in VTA, DRN, and LC, and reduced inhibitory monoaminergic activity on ACC. These findings advance our understanding of the interplay between sleep and pain, shedding light on potential NAc-brainstem-ACC mechanisms that could mediate increased pain sensitivity under conditions of sleep impairment.
Collapse
Affiliation(s)
- Natalia F Sardi
- Laboratory of Neurophysiology of Pain, Department of Physiology, Division of Biological Sciences, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - Ana C Pescador
- Laboratory of Neurophysiology of Pain, Department of Physiology, Division of Biological Sciences, Federal University of Paraná, Curitiba, Paraná, Brazil; Department of Health Sciences, Federal University of Santa Catarina, Araranguá, Santa Catarina, Brazil
| | - Karla E Torres-Chavez
- Laboratory of Physiology, School of Medicine, Catholic University of Santa María, Arequipa, Peru
| | - Luana Fischer
- Laboratory of Neurophysiology of Pain, Department of Physiology, Division of Biological Sciences, Federal University of Paraná, Curitiba, Paraná, Brazil.
| |
Collapse
|
9
|
Neverdahl JP, Uglem M, Matre D, Nilsen KB, Hagen K, Gravdahl GB, Sand T, Omland PM. Endogenous pain modulation after sleep restriction in migraine: a blinded crossover study. J Headache Pain 2024; 25:166. [PMID: 39363172 PMCID: PMC11448287 DOI: 10.1186/s10194-024-01879-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Patients with migraine are vulnerable to insufficient sleep, but the impact of sleep restriction is largely unknown. In addition, the importance of sleep may be different in patients with migraine who mostly have attack onsets during sleep, so called sleep-related migraine, compared to patients with non-sleep-related migraine. In this study we investigate the effect of sleep restriction on endogenous pain modulation in patients with migraine and healthy controls. We also compared the effect of sleep restriction in sleep-related and in non-sleep-related migraine. METHODS Measurements were conducted in 39 patients with migraine between attacks and 31 controls, once after habitual sleep and once after two consecutive nights of partial sleep restriction. There were 29 and 10 patients with non-sleep-related and sleep-related migraine respectively. Test stimulus was 2-min tonic noxious heat to the left volar forearm. Temporal summation was calculated as the regression coefficient for rated pain in the late part of this 2-min stimulation. Conditioning stimulus was right hand-immersion in 7 °C water. Conditioned pain modulation was defined as the difference in rated pain with and without the conditioning stimulus and was calculated for temporal summation and mean rated pain for the test stimulus. The effect of sleep restriction on temporal summation and conditioned pain modulation was compared in migraine subjects and controls using two-level models with recordings nested in subjects. RESULTS Conditioned pain modulation for temporal summation of heat pain tended to be reduced after sleep restriction in patients with migraine compared to controls (p = 0.060) and, in an exploratory analysis, was reduced more after sleep restriction in sleep-related than in non-sleep-related migraine (p = 0.017). No other differences between groups after sleep restriction were found for temporal summation or conditioned pain modulation. CONCLUSION Patients with migraine may have a subtly altered endogenous pain modulation system. Sleep restriction may have an increased pronociceptive effect on this system, suggesting a mechanism for vulnerability to insufficient sleep in migraine. This effect seems to be larger in sleep-related migraine than in non-sleep-related migraine.
Collapse
Affiliation(s)
- Jan Petter Neverdahl
- Department of Neuromedicine and Movement Sciences, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, 7491, Norway.
- NorHEAD - Norwegian Centre for Headache Research, NTNU, Trondheim, Norway.
- Section for Clinical Psychosis Research, Department of Research and Innovation, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway.
| | - Martin Uglem
- Department of Neuromedicine and Movement Sciences, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, 7491, Norway
- NorHEAD - Norwegian Centre for Headache Research, NTNU, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway
| | - Dagfinn Matre
- National Institute of Occupational Health, Oslo, Norway
| | - Kristian Bernhard Nilsen
- Section for Clinical Neurophysiology, Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Knut Hagen
- Department of Neuromedicine and Movement Sciences, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, 7491, Norway
- NorHEAD - Norwegian Centre for Headache Research, NTNU, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway
- Clinical Research Unit, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Gøril Bruvik Gravdahl
- Department of Neuromedicine and Movement Sciences, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, 7491, Norway
- NorHEAD - Norwegian Centre for Headache Research, NTNU, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway
| | - Trond Sand
- Department of Neuromedicine and Movement Sciences, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, 7491, Norway
- NorHEAD - Norwegian Centre for Headache Research, NTNU, Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway
| | - Petter Moe Omland
- Department of Neuromedicine and Movement Sciences, Faculty of Medicine and Health Sciences, NTNU, Norwegian University of Science and Technology, Trondheim, 7491, Norway.
- NorHEAD - Norwegian Centre for Headache Research, NTNU, Trondheim, Norway.
- Department of Neurology and Clinical Neurophysiology, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway.
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
| |
Collapse
|
10
|
Lou Q, Wei HR, Chen D, Zhang Y, Dong WY, Qun S, Wang D, Luo Y, Zhang Z, Jin Y. A noradrenergic pathway for the induction of pain by sleep loss. Curr Biol 2024; 34:2644-2656.e7. [PMID: 38810638 DOI: 10.1016/j.cub.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/31/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024]
Abstract
An epidemic of sleep loss currently affects modern societies worldwide and is implicated in numerous physiological disorders, including pain sensitization, although few studies have explored the brain pathways affected by active sleep deprivation (ASD; e.g., due to recreation). Here, we describe a neural circuit responsible for pain sensitization in mice treated with 9-h non-stress ASD. Using a combination of advanced neuroscience methods, we found that ASD stimulates noradrenergic inputs from locus coeruleus (LCNA) to glutamatergic neurons of the hindlimb primary somatosensory cortex (S1HLGlu). Moreover, artificial inhibition of this LCNA→S1HLGlu pathway alleviates ASD-induced pain sensitization in mice, while chemogenetic activation of this pathway recapitulates the pain sensitization observed following ASD. Our study thus implicates activation of the LCNA→S1HLGlu pathway in ASD-induced pain sensitization, expanding our fundamental understanding of the multisystem interplay involved in pain processing.
Collapse
Affiliation(s)
- Qianqian Lou
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Hong-Rui Wei
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Danyang Chen
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yuzhuo Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230022, China
| | - Wan-Ying Dong
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Sen Qun
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Di Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China.
| | - Yanli Luo
- Department of Psychological Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Zhi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; The Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Department of Biophysics and Neurobiology, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei 230026, China.
| | - Yan Jin
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Department of Biophysics and Neurobiology, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
11
|
Zhu K, Chen S, Qin X, Bai W, Hao J, Xu X, Guo H, Bai H, Yang Z, Wang S, Zhao Z, Ji T, Kong D, Zhang W. Exploring the therapeutic potential of cannabidiol for sleep deprivation-induced hyperalgesia. Neuropharmacology 2024; 249:109893. [PMID: 38428482 DOI: 10.1016/j.neuropharm.2024.109893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024]
Abstract
Hyperalgesia resulting from sleep deprivation (SD) poses a significant a global public health challenge with limited treatment options. The nucleus accumbens (NAc) plays a crucial role in the modulation of pain and sleep, with its activity regulated by two distinct types of medium spiny neurons (MSNs) expressing dopamine 1 or dopamine 2 (D1-or D2) receptors (referred to as D1-MSNs and D2-MSNs, respectively). However, the specific involvement of the NAc in SD-induced hyperalgesia remains uncertain. Cannabidiol (CBD), a nonpsychoactive phytocannabinoid, has demonstrated analgesic effects in clinical and preclinical studies. Nevertheless, its potency in addressing this particular issue remains to be determined. Here, we report that SD induced a pronounced pronociceptive effect attributed to the heightened intrinsic excitability of D2-MSNs within the NAc in Male C57BL/6N mice. CBD (30 mg/kg, i.p.) exhibited an anti-hyperalgesic effect. CBD significantly improved the thresholds for thermal and mechanical pain and increased wakefulness by reducing delta power. Additionally, CBD inhibited the intrinsic excitability of D2-MSNs both in vitro and in vivo. Bilateral microinjection of the selective D2 receptor antagonist raclopride into the NAc partially reversed the antinociceptive effect of CBD. Thus, these findings strongly suggested that SD activates NAc D2-MSNs, contributing heightened to pain sensitivity. CBD exhibits antinociceptive effects by activating D2R, thereby inhibiting the excitability of D2-MSNs and promoting wakefulness under SD conditions.
Collapse
Affiliation(s)
- Kangsheng Zhu
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, School of Chinese Integrative Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei, 050017, China; Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
| | - Siruan Chen
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, School of Chinese Integrative Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei, 050017, China
| | - Xia Qin
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, School of Chinese Integrative Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei, 050017, China
| | - Wanjun Bai
- Department of Pharmacy, Hebei General Hospital, Shijiazhuang, Hebei, 050051, China
| | - Jie Hao
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, School of Chinese Integrative Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei, 050017, China
| | - Xiaolei Xu
- School of Nursing, Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Han Guo
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, School of Chinese Integrative Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei, 050017, China
| | - Hui Bai
- Department of Cardiac Ultrasound, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Zuxiao Yang
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, School of Chinese Integrative Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei, 050017, China
| | - Sheng Wang
- Hebei Key Laboratory of Neurophysiology, Shijiazhuang, Hebei, 050017, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050011, China
| | - Tengfei Ji
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Dezhi Kong
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, School of Chinese Integrative Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei, 050017, China.
| | - Wei Zhang
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, School of Chinese Integrative Medicine, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, Hebei, 050017, China.
| |
Collapse
|
12
|
Sardi NF, Pescador AC, Azevedo EM, Pochapski JA, Kukolj C, Spercoski KM, Andrade AJM, da Cunha C, Fischer L. Sleep and Pain: A Role for the Anterior Cingulate Cortex, Nucleus Accumbens, and Dopamine in the Increased Pain Sensitivity Following Sleep Restriction. THE JOURNAL OF PAIN 2024; 25:331-349. [PMID: 37673193 DOI: 10.1016/j.jpain.2023.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/08/2023]
Abstract
Persistent pain conditions and sleep disorders are public health problems worldwide. It is widely accepted that sleep disruption increases pain sensitivity; however, the underlying mechanisms are poorly understood. In this study, we used a protocol of 6 hours a day of total sleep deprivation for 3 days in rats to advance the understanding of these mechanisms. We focused on gender differences and the dopaminergic mesocorticolimbic system. The findings demonstrated that sleep restriction (SR) increased pain sensitivity in a similar way in males and females, without inducing a significant stress response. This pronociceptive effect depends on a nucleus accumbens (NAc) neuronal ensemble recruited during SR and on the integrity of the anterior cingulate cortex (ACC). Data on indirect dopaminergic parameters, dopamine transporter glycosylation, and dopamine and cyclic adenosine monophosphate (AMP)-regulated phosphoprotein-32 phosphorylation, as well as dopamine, serotonin, and norepinephrine levels, suggest that dopaminergic function decreases in the NAc and ACC after SR. Complementarily, pharmacological activation of dopamine D2, but not D1 receptors either in the ACC or in the NAc prevents SR from increasing pain sensitivity. The ACC and NAc are the main targets of dopaminergic mesocorticolimbic projections with a key role in pain modulation. This study showed their integrative role in the pronociceptive effect of SR, pointing to dopamine D2 receptors as a potential target for pain management in patients with sleep disorders. These findings narrow the focus of future studies on the mechanisms by which sleep impairment increases pain sensitivity. PERSPECTIVE: This study demonstrates that the pronociceptive effect of SR affects similarly males and females and depends on a NAc neuronal ensemble recruited during SR and on the integrity of the ACC. Findings on dopaminergic function support dopamine D2 receptors as targets for pain management in sleep disorders patients.
Collapse
Affiliation(s)
- Natalia F Sardi
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Ana C Pescador
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Evellyn M Azevedo
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - José A Pochapski
- Department of Pharmacology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil; Department of Biochemistry, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Caroline Kukolj
- Department of Biochemistry, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Katherinne M Spercoski
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil; Division of Biosciences, Federal University of Parana, Palotina, Parana, Brazil
| | - Anderson J M Andrade
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Claudio da Cunha
- Department of Pharmacology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Luana Fischer
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| |
Collapse
|
13
|
Ding W, Yang L, Shi E, Kim B, Low S, Hu K, Gao L, Chen P, Ding W, Borsook D, Luo A, Choi JH, Wang C, Akeju O, Yang J, Ran C, Schreiber KL, Mao J, Chen Q, Feng G, Shen S. The endocannabinoid N-arachidonoyl dopamine is critical for hyperalgesia induced by chronic sleep disruption. Nat Commun 2023; 14:6696. [PMID: 37880241 PMCID: PMC10600211 DOI: 10.1038/s41467-023-42283-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 10/05/2023] [Indexed: 10/27/2023] Open
Abstract
Chronic pain is highly prevalent and is linked to a broad range of comorbidities, including sleep disorders. Epidemiological and clinical evidence suggests that chronic sleep disruption (CSD) leads to heightened pain sensitivity, referred to as CSD-induced hyperalgesia. However, the underlying mechanisms are unclear. The thalamic reticular nucleus (TRN) has unique integrative functions in sensory processing, attention/arousal and sleep spindle generation. We report that the TRN played an important role in CSD-induced hyperalgesia in mice, through its projections to the ventroposterior region of the thalamus. Metabolomics revealed that the level of N-arachidonoyl dopamine (NADA), an endocannabinoid, was decreased in the TRN after CSD. Using a recently developed CB1 receptor (cannabinoid receptor 1) activity sensor with spatiotemporal resolution, CB1 receptor activity in the TRN was found to be decreased after CSD. Moreover, CSD-induced hyperalgesia was attenuated by local NADA administration to the TRN. Taken together, these results suggest that TRN NADA signaling is critical for CSD-induced hyperalgesia.
Collapse
Affiliation(s)
- Weihua Ding
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Liuyue Yang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eleanor Shi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bowon Kim
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah Low
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kun Hu
- Department of Pathology, Tuft University School of Medicine, Boston, MA, USA
| | - Lei Gao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ping Chen
- College of Science and Mathematics, University of Massachusetts Boston, Boston, MA, USA
| | - Wei Ding
- College of Science and Mathematics, University of Massachusetts Boston, Boston, MA, USA
| | - David Borsook
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew Luo
- Summer Intern Program of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, currently at Brandeis University, Boston, MA, USA
| | - Jee Hyun Choi
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, South Korea
| | - Changning Wang
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jun Yang
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chongzhao Ran
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristin L Schreiber
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jianren Mao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Qian Chen
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Guoping Feng
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Shiqian Shen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Sun H, Li Z, Qiu Z, Shen Y, Guo Q, Hu SW, Ding HL, An S, Cao JL. A common neuronal ensemble in nucleus accumbens regulates pain-like behaviour and sleep. Nat Commun 2023; 14:4700. [PMID: 37543693 PMCID: PMC10404280 DOI: 10.1038/s41467-023-40450-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 07/28/2023] [Indexed: 08/07/2023] Open
Abstract
A comorbidity of chronic pain is sleep disturbance. Here, we identify a dual-functional ensemble that regulates both pain-like behaviour induced by chronic constrictive injury or complete Freund's adjuvant, and sleep wakefulness, in the nucleus accumbens (NAc) in mice. Specifically, a select population of NAc neurons exhibits increased activity either upon nociceptive stimulation or during wakefulness. Experimental activation of the ensemble neurons exacerbates pain-like (nociceptive) responses and reduces NREM sleep, while inactivation of these neurons produces the opposite effects. Furthermore, NAc ensemble primarily consists of D1 neurons and projects divergently to the ventral tegmental area (VTA) and preoptic area (POA). Silencing an ensemble innervating VTA neurons selectively increases nociceptive responses without affecting sleep, whereas inhibiting ensemble-innervating POA neurons decreases NREM sleep without affecting nociception. These results suggest a common NAc ensemble that encodes chronic pain and controls sleep, and achieves the modality specificity through its divergent downstream circuit targets.
Collapse
Affiliation(s)
- Haiyan Sun
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
- Department of Pediatrics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, China
| | - Zhilin Li
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Zhentong Qiu
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Yu Shen
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Qingchen Guo
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Su-Wan Hu
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China
| | - Shuming An
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, 221004, China.
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China.
| |
Collapse
|
15
|
Zhu M, Huang H. The Underlying Mechanisms of Sleep Deprivation Exacerbating Neuropathic Pain. Nat Sci Sleep 2023; 15:579-591. [PMID: 37533626 PMCID: PMC10392808 DOI: 10.2147/nss.s414174] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
Pain disrupts sleep, and sleep deprivation or interference can alter pain perception in animals and humans, for example by increasing sensitivity to pain. However, the mechanism by which sleep affects neuropathic pain remains unclear. In this review, we discuss the available evidence from the epidemiologic, clinical, and human, as well as laboratory studies. In previous studies, we have found that sleep deprivation affects various injurious systems, including opioids, dopaminergic, immune, orexins, hypothalamic-pituitary-adrenal axis, and adenosine. At the same time, these systems play a crucial role in neuropathic pain regulation. In the complex interactions between these neurobiological systems, there may be potential regulatory pathways through which sleep deprivation amplifies neuropathic pain. Because of the impact sleep problems and neuropathic pain can have on the patients' quality of life, studying the link between sleep and neuropathic pain is important for neuropathic pain prevention and public health.
Collapse
Affiliation(s)
- Manmin Zhu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Guizhou, People’s Republic of China
| | - Hao Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Guizhou, People’s Republic of China
| |
Collapse
|
16
|
Sleep Deprivation Induces Dopamine System Maladaptation and Escalated Corticotrophin-Releasing Factor Signaling in Adolescent Mice. Mol Neurobiol 2023; 60:3190-3209. [PMID: 36813955 DOI: 10.1007/s12035-023-03258-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 02/07/2023] [Indexed: 02/24/2023]
Abstract
Sleep disruption is highly associated with the pathogenesis and progression of a wild range of psychiatric disorders. Furthermore, appreciable evidence shows that experimental sleep deprivation (SD) on humans and rodents evokes anomalies in the dopaminergic (DA) signaling, which are also implicated in the development of psychiatric illnesses such as schizophrenia or substance abuse. Since adolescence is a vital period for the maturation of the DA system as well as the occurrence of mental disorders, the present studies aimed to investigate the impacts of SD on the DA system of adolescent mice. We found that 72 h SD elicited a hyperdopaminergic status, with increased sensitivity to the novel environment and amphetamine (Amph) challenge. Also, altered neuronal activity and expression of striatal DA receptors were noticed in the SD mice. Moreover, 72 h SD influenced the immune status in the striatum, with reduced microglial phagocytic capacity, primed microglial activation, and neuroinflammation. The abnormal neuronal and microglial activity were putatively provoked by the enhanced corticotrophin-releasing factor (CRF) signaling and sensitivity during the SD period. Together, our findings demonstrated the consequences of SD in adolescents including aberrant neuroendocrine, DA system, and inflammatory status. Sleep insufficiency is a risk factor for the aberration and neuropathology of psychiatric disorders.
Collapse
|
17
|
Kourbanova K, Alexandre C, Latremoliere A. Effect of sleep loss on pain-New conceptual and mechanistic avenues. Front Neurosci 2022; 16:1009902. [PMID: 36605555 PMCID: PMC9807925 DOI: 10.3389/fnins.2022.1009902] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Sleep disturbances increase pain sensitivity in clinical and preclinical settings, but the precise mechanisms are unknown. This represents a major public health issue because of the growing sleep deficiency epidemic fueled by modern lifestyle. To understand the neural pathways at the intersection between sleep and pain processes, it is critical to determine the precise nature of the sleep disruptions that increase pain and the specific component of the pain response that is targeted. Methods We performed a review of the literature about sleep disturbances and pain sensitivity in humans and rodents by taking into consideration the targeted sleep stage (REMS, non-NREMS, or both), the amount of sleep lost, and the different types of sleep disruptions (partial or total sleep loss, duration, sleep fragmentation or interruptions), and how these differences might affect distinct components of the pain response. Results We find that the effects of sleep disturbances on pain are highly conserved among species. The major driver for pain hypersensitivity appears to be the total amount of sleep lost, while REMS loss by itself does not seem to have a direct effect on pain sensitivity. Sleep loss caused by extended wakefulness preferentially increases pain perception, whereas interrupted and limited sleep strongly dysregulates descending controls such as DNIC, especially in women. Discussion We discuss the possible mechanisms involved, including an increase in inflammatory processes, a loss of nociceptive inhibitory pathways, and a defect in the cognitive processing of noxious input.
Collapse
Affiliation(s)
- Kamila Kourbanova
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Chloe Alexandre
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Alban Latremoliere
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
18
|
Guo M, Wu Y, Zheng D, Chen L, Xiong B, Wu J, Li K, Wang L, Lin K, Zhang Z, Manyande A, Xu F, Wang J, Peng M. Preoperative Acute Sleep Deprivation Causes Postoperative Pain Hypersensitivity and Abnormal Cerebral Function. Neurosci Bull 2022; 38:1491-1507. [PMID: 36282466 PMCID: PMC9723009 DOI: 10.1007/s12264-022-00955-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 09/17/2022] [Indexed: 11/30/2022] Open
Abstract
Preoperative sleep loss can amplify post-operative mechanical hyperalgesia. However, the underlying mechanisms are still largely unknown. In the current study, rats were randomly allocated to a control group and an acute sleep deprivation (ASD) group which experienced 6 h ASD before surgery. Then the variations in cerebral function and activity were investigated with multi-modal techniques, such as nuclear magnetic resonance, functional magnetic resonance imaging, c-Fos immunofluorescence, and electrophysiology. The results indicated that ASD induced hyperalgesia, and the metabolic kinetics were remarkably decreased in the striatum and midbrain. The functional connectivity (FC) between the nucleus accumbens (NAc, a subregion of the ventral striatum) and the ventrolateral periaqueductal gray (vLPAG) was significantly reduced, and the c-Fos expression in the NAc and the vLPAG was suppressed. Furthermore, the electrophysiological recordings demonstrated that both the neuronal activity in the NAc and the vLPAG, and the coherence of the NAc-vLPAG were suppressed in both resting and task states. This study showed that neuronal activity in the NAc and the vLPAG were weakened and the FC between the NAc and the vLPAG was also suppressed in rats with ASD-induced hyperalgesia. This study highlights the importance of preoperative sleep management for surgical patients.
Collapse
Affiliation(s)
- Meimei Guo
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yuxiang Wu
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, 430056, China
| | - Danhao Zheng
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China
| | - Lei Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Department of Anesthesiology, Peking University Third Hospital, Beijing, 100191, China
| | - Bingrui Xiong
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jinfeng Wu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China
| | - Ke Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Li Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China
| | - Kangguang Lin
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, 510000, China
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, W1S 3PR, UK
| | - Fuqiang Xu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China.
- Institute of Neuroscience and Brain Disease; Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Mian Peng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
19
|
The Sleep-Reward-Pain Pathway Model: an Integrative Review. CURRENT SLEEP MEDICINE REPORTS 2022. [DOI: 10.1007/s40675-022-00232-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Morris EE, Howell MJ, Pickup E, Iber C, Wang SG. Pediatric sleep and pain: etiologies, consequences, and clinical considerations. J Clin Sleep Med 2022; 18:2281-2289. [PMID: 35499282 PMCID: PMC9435332 DOI: 10.5664/jcsm.10008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/31/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022]
Abstract
STUDY OBJECTIVES To examine current evidence of the relationship between sleep and pain from the neonatal period through adolescence. This review serves as a critical review of the literature and of the needs for future research on pediatric sleep and pain. METHODS The PubMed online database was queried from January 1, 1960, to March 1, 2020, producing 149 articles applicable to pain and sleep in the pediatric population. Of those, 97 articles were cited in this review with the key articles including over 3800 participants. RESULTS The pediatric literature supports the relationship between poor sleep (both sleep efficiency and nighttime awakenings) and subsequent risk for pain, especially among children with chronic disease. The reverse effect of pain on sleep is not yet well delineated. The key moderating factors explored in the literature are pharmacologic and nonpharmacologic therapies, psychologic health, and the etiology of pain. There is evidence that both altered sleep and pain early in life impact neurodevelopment, as seen by changes in sleep structure in clinical studies and alterations in brain development in animal models. CONCLUSIONS The complicated relationship between sleep and pain is critically important during pediatric development when alterations to a normal sleep structure can have a lifelong impact. It is becoming clear that sleep deprivation and poor sleep quality exacerbate pain. Further research is needed into the complex alterations of sleep in chronic pain conditions as well as treatments to improve sleep in pediatric care. CITATION Morris EE, Howell MJ, Pickup E, Iber C, Wang SG. Pediatric sleep and pain: etiologies, consequences, and clinical considerations. J Clin Sleep Med. 2022;18(9):2281-2289.
Collapse
Affiliation(s)
- Erin E. Morris
- Department of Pediatrics, University of Minnesota–Twin Cities, Minneapolis, Minnesota
| | - Michael J. Howell
- Department of Neurology, University of Minnesota–Twin Cities, Minneapolis, Minnesota
| | - Elizabeth Pickup
- Pediatric Neurology, Children’s National Hospital, Washington, DC
| | - Conrad Iber
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Minnesota–Twin Cities, Minneapolis, Minnesota
| | - Sonya G. Wang
- Department of Pediatrics, University of Minnesota–Twin Cities, Minneapolis, Minnesota
| |
Collapse
|
21
|
Selvanathan J, Tang NKY, Peng PWH, Chung F. Sleep and pain: relationship, mechanisms, and managing sleep disturbance in the chronic pain population. Int Anesthesiol Clin 2022; 60:27-34. [PMID: 35261343 DOI: 10.1097/aia.0000000000000346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Janannii Selvanathan
- Department of Anesthesia and Pain Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Nicole K Y Tang
- Department of Psychology, University of Warwick, Coventry, UK
| | - Philip W H Peng
- Department of Anesthesia and Pain Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Frances Chung
- Department of Anesthesia and Pain Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Shu P, Ji L, Ping Z, Sun Z, Liu W. Association of insomnia and daytime sleepiness with low back pain: A bidirectional mendelian randomization analysis. Front Genet 2022; 13:938334. [PMID: 36267398 PMCID: PMC9577110 DOI: 10.3389/fgene.2022.938334] [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: 05/07/2022] [Accepted: 09/06/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose: Observational research has indicated the presence of a causal relationship between sleep disturbances and low back pain (LBP). However, the link may have been biased by confounding factors. The purpose of this study was to examine the potential causal association of insomnia and daytime sleepiness with LBP by using mendelian randomization (MR). Methods: Genome-wide association study (GWAS) summary statistics of insomnia were obtained from a large-scale GWAS meta-analysis (n = 1,331,010; individuals from UK Biobank and 23andMe) or UK Biobank alone (n = 453,379). The summary statistics of daytime sleepiness were from UK Biobank (n = 452,071) and LBP were provided by the FinnGen Release 6 (210,645 individuals with 16,356 LBP cases and 194,289 controls) or UK Biobank (5,423 cases versus 355,771 controls). Linkage disequilibrium score (LDSC) regression and bidirectional MR analysis was employed to estimate genetic correlation and causal relationship. In the MR analysis, the inverse variance weighted method (IVW) was utilized as the main analysis procedure, while MR-Egger, Weighted median and Robust adjusted profile score (RAPS) were utilized for supplementary analyses. Results: LDSC analysis showed that LBP were significantly genetically correlated with insomnia (rg = 0.57, p = 2.26e-25) and daytime sleepiness (rg = 0.18, p = 0.001). The MR analysis revealed that genetically predicted insomnia was significantly associated with an increased risk of LBP (OR = 1.250, 95% CI: 1.186-1.318; p = 1.69e-16). However, the reverse causality was not confirmed. No evidence was identified supporting causality of daytime sleepiness and LBP. Conclusion: This study demonstrates a putative causal link of insomnia on LBP and a null causal effect of LBP on insomnia. Furthermore, a causal link between daytime sleepiness and LBP were not reported. This finding may stimulate new strategies for patient management in clinical practice, benefiting public health.
Collapse
Affiliation(s)
- Peng Shu
- Department of Orthopedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Lixian Ji
- Department of Rheumatology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Zichuan Ping
- Department of Orthopedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Zhibo Sun
- Department of Orthopedics, Renmin Hospital, Wuhan University, Wuhan, China
| | - Wei Liu
- Department of Orthopedic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| |
Collapse
|
23
|
Iacovides S, Kamerman P, Baker FC, Mitchell D. Why It Is Important to Consider the Effects of Analgesics on Sleep: A Critical Review. Compr Physiol 2021; 11:2589-2619. [PMID: 34558668 DOI: 10.1002/cphy.c210006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We review the known physiological mechanisms underpinning all of pain processing, sleep regulation, and pharmacology of analgesics prescribed for chronic pain. In particular, we describe how commonly prescribed analgesics act in sleep-wake neural pathways, with potential unintended impact on sleep and/or wake function. Sleep disruption, whether pain- or drug-induced, negatively impacts quality of life, mental and physical health. In the context of chronic pain, poor sleep quality heightens pain sensitivity and may affect analgesic function, potentially resulting in further analgesic need. Clinicians already have to consider factors including efficacy, abuse potential, and likely side effects when making analgesic prescribing choices. We propose that analgesic-related sleep disruption should also be considered. The neurochemical mechanisms underlying the reciprocal relationship between pain and sleep are poorly understood, and studies investigating sleep in those with specific chronic pain conditions (including those with comorbidities) are lacking. We emphasize the importance of further work to clarify the effects (intended and unintended) of each analgesic class to inform personalized treatment decisions in patients with chronic pain. © 2021 American Physiological Society. Compr Physiol 11:1-31, 2021.
Collapse
Affiliation(s)
- Stella Iacovides
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Peter Kamerman
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fiona C Baker
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Human Sleep Research Program, SRI International, Menlo Park, California, USA
| | - Duncan Mitchell
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
24
|
Planchuelo-Gómez Á, García-Azorín D, Guerrero ÁL, Rodríguez M, Aja-Fernández S, de Luis-García R. Gray Matter Structural Alterations in Chronic and Episodic Migraine: A Morphometric Magnetic Resonance Imaging Study. PAIN MEDICINE 2021; 21:2997-3011. [PMID: 33040149 DOI: 10.1093/pm/pnaa271] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE This study evaluates different parameters describing the gray matter structure to analyze differences between healthy controls, patients with episodic migraine, and patients with chronic migraine. DESIGN Cohort study. SETTING Spanish community. SUBJECTS Fifty-two healthy controls, 57 episodic migraine patients, and 57 chronic migraine patients were included in the study and underwent T1-weighted magnetic resonance imaging acquisition. METHODS Eighty-four cortical and subcortical gray matter regions were extracted, and gray matter volume, cortical curvature, thickness, and surface area values were computed (where applicable). Correlation analysis between clinical features and structural parameters was performed. RESULTS Statistically significant differences were found between all three groups, generally consisting of increases in cortical curvature and decreases in gray matter volume, cortical thickness, and surface area in migraineurs with respect to healthy controls. Furthermore, differences were also found between chronic and episodic migraine. Significant correlations were found between duration of migraine history and several structural parameters. CONCLUSIONS Migraine is associated with structural alterations in widespread gray matter regions of the brain. Moreover, the results suggest that the pattern of differences between healthy controls and episodic migraine patients is qualitatively different from that occurring between episodic and chronic migraine patients.
Collapse
Affiliation(s)
| | - David García-Azorín
- Headache Unit, Department of Neurology, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Ángel L Guerrero
- Headache Unit, Department of Neurology, Hospital Clínico Universitario de Valladolid, Valladolid, Spain.,Institute for Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Margarita Rodríguez
- Department of Radiology, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | | | | |
Collapse
|
25
|
Sex Differences, Sleep Disturbance and Risk of Persistent Pain Associated With Groin Hernia Surgery: A Nationwide Register-Based Cohort Study. THE JOURNAL OF PAIN 2021; 22:1360-1370. [PMID: 33964413 DOI: 10.1016/j.jpain.2021.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/30/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023]
Abstract
Persistent pain after groin hernia repair is a major health problem. Sleep disturbance is associated with heightened pain sensitivity. The main objective of this study was to examine the role of sleep disturbance in the development and long-term maintenance of chronic postherniorrhaphy inguinal pain (CPIP), with exploration of sex differences. From 2012 to 2017, a national cohort of patients with prior groin hernia repair (n = 2084;45.8% females) were assessed for the development of CPIP 12 months after surgery. Patients then underwent long-term (median 5.0 years) follow-up to evaluate the contribution of sex and sleep disturbance on the maintenance of CPIP. Associations between pre- and postoperative sleep problems (assessed at long-term follow-up) and CPIP were tested using logistic regression. Females had higher rates of CPIP with negative impact on daily activities 12 months after surgery as compared to males (14.6 vs 9.2%, P < .0005), and were more likely to have moderate-severe CPIP in the long-term (3.1 vs 1.2%, P = .003). Preoperative sleep problems predicted development of CPIP 12 months after surgery (adjusted odds ratio [aOR] 1.76 [95%CI 1.26-2.46], P = .001) and CPIP in the long-term (aOR 2.20 [1.61-3.00] , P < .0001). CPIP was associated with insomnia and depression. Sleep disturbance may increase the risk for CPIP, and contribute to maintenance of postsurgical pain. PERSPECTIVE: Females are at heightened risk for CPIP as compared to males. Increased severity of pain symptoms are linked to poorer sleep and psychiatric morbidity. Given the robust associations between sleep disturbance and CPIP, interventions which consolidate and promote sleep, especially in females, may improve long-term pain control.
Collapse
|
26
|
Bjurström MF, Irwin MR, Bodelsson M, Smith MT, Mattsson-Carlgren N. Preoperative sleep quality and adverse pain outcomes after total hip arthroplasty. Eur J Pain 2021; 25:1482-1492. [PMID: 33682177 DOI: 10.1002/ejp.1761] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/03/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Sleep disturbance is thought to aggravate acute postoperative pain. The influence of preoperative sleep problems on pain control in the long-term and development of chronic postsurgical pain is largely unknown. METHODS This prospective, observational study aimed to examine the links between preoperative sleep disturbance (Pittsburgh Sleep Quality Index, PSQI) and pain severity (Brief Pain Inventory, BPI) 6 months postoperative (primary outcome), objective measures of pain and postoperative pain control variables (secondary outcomes). Patients (n = 52) with disabling osteoarthritis (OA) pain undergoing total hip arthroplasty (THA) were included. Quantitative sensory testing (QST) was performed preoperatively on the day of surgery to evaluate pain objectively. Clinical data, as well as measures of sleep quality and pain, were obtained preoperatively and longitudinally over a 6-month period. RESULTS Preoperatively, sleep disturbance (i.e., PSQI score >5) occurred in 73.1% (n = 38) of THA patients, and pain severity was high (BPI pain severity 5.4 ± 1.3). Regression models, adjusting for relevant covariates, showed that preoperative PSQI score predicted pain severity 6 months postoperative (β = 0.091 (95% CI 0.001-0.181), p = .048, R2 = 0.35). Poor sleep quality was associated with increased pressure pain sensitivity and impaired endogenous pain inhibitory capacity (R2 range 0.14-0.33, all p's < 0.04). Moreover, preoperative sleep disturbance predicted increased opioid treatment during the first 24 hr after surgery (unadjusted β = 0.009 (95% CI 0.002-0.015) mg/kg, p = .007, R2 = 0.15). CONCLUSIONS Preoperative sleep disturbance is prevalent in THA patients, is associated with objective measures of pain severity, and independently predicts immediate postoperative opioid treatment and poorer long-term pain control in patients who have undergone THA. SIGNIFICANCE Poor sleep quality and impaired sleep continuity are associated with heightened pain sensitivity, but previous work has not evaluated whether preoperative sleep problems impact long-term postoperative pain outcomes. Here, we show that sleep difficulties prior to total hip arthroplasty adversely predict postoperative pain control 6 months after surgery. Given sleep difficulties robustly predict pain outcomes, targeting and improving sleep may have salutary effects on postoperative pain reports and management.
Collapse
Affiliation(s)
- Martin F Bjurström
- Department of Anesthesiology and Intensive Care, Skåne University Hospital, Lund, Sweden.,Department of Clinical Sciences Lund, Lund University, Lund, Sweden.,Norman Cousins Center for Psychoneuroimmunology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Michael R Irwin
- Norman Cousins Center for Psychoneuroimmunology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Mikael Bodelsson
- Department of Anesthesiology and Intensive Care, Skåne University Hospital, Lund, Sweden.,Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Michael T Smith
- Behavioral Medicine Division, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Faculty of Medicine, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
27
|
Hamilton LJ, Walker M, Pattabiraman M, Zhong HA, Luedtke B, Chandra S. Novel curcumin analog (cis-trans curcumin) as ligand to adenosine receptors A 2A and A 2B: potential for therapeutics. Pharmacol Res 2021; 165:105410. [PMID: 33401004 PMCID: PMC7979524 DOI: 10.1016/j.phrs.2020.105410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023]
Abstract
All four of the adenosine receptor (AR) subtypes mediate pain and have been targeted by pharmacologists to generate new therapeutics for chronic pain. The vanilloid phytochemicals, which include curcumin, capsaicin, and gingerol, have been shown to alleviate pain. However, there is little to no literature on the interaction of vanilloid phytochemicals with ARs. In this study, photochemical methods were used to generate a novel isomer of curcumin (cis-trans curcumin or CTCUR), and the interactions of both curcumin and CTCUR with the two Gs-linked AR subtypes were studied. Competitive binding assays, docking analysis, and confocal fluorescence microscopy were performed to measure binding affinity; cell survival assays were used to measure toxicity; and cAMP assays were performed to measure receptor activation. Competitive binding results indicated that CTCUR binds to both AR A2A and AR A2B with Ki values of 5 μM and 7 μM, respectively, which is consistent with our docking results. Fluorescence microscopy data also shows binding for A2B and A2A. Cell survival results show that CTCUR and CUR are nontoxic at the tested concentrations in these cell lines. Overall, our results suggest that vanilloid phytochemicals may be slightly modified to increase interaction with Gs-ARs, and thereby can be further explored to provide a novel class of non-opioid antinociceptives.
Collapse
Affiliation(s)
- Luke J Hamilton
- Department of Biology, University of Nebraska-Kearney, United States
| | - Michaela Walker
- Department of Biology, University of Nebraska-Kearney, United States
| | | | - Haizhen A Zhong
- Department of Chemistry, University of Nebraska-Omaha, United States
| | - Brandon Luedtke
- Department of Biology, University of Nebraska-Kearney, United States
| | - Surabhi Chandra
- Department of Biology, University of Nebraska-Kearney, United States.
| |
Collapse
|
28
|
Pain impairs consolidation, but not acquisition or retrieval of a declarative memory. Behav Pharmacol 2020; 31:707-715. [PMID: 32925225 DOI: 10.1097/fbp.0000000000000576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Among the physical conditions that impair memory performance, pain is one of the most prevalent. However, the mechanisms by which pain impairs memory are largely unknown. In this study, we asked whether pain affects memory acquisition, consolidation and retrieval as well as whether memory impairment depends on pain intensity. Wistar rats received a hind paw injection of formalin (1%) at different phases of object recognition test. The injection of formalin after training but not before training or testing impaired object recognition memory. We concluded that pain impairs the consolidation but not acquisition or retrieval of object recognition memory, which is a subtype of declarative memory. Morphine, at a dose that did not affect object recognition memory in control rats, drastically reduced formalin-induced nociceptive behavior without reversing memory impairment. A lower dose of formalin (0.25%) induced less nociceptive behavior, but similar memory impairment. There is no statistical correlation between the intensity of nociceptive response and the performance in object recognition test. However, when formalin-induced nociceptive response was blocked by a local anesthetic, memory impairment was prevented. These findings suggest that pain-induced impairment in the consolidation of object recognition memory does not directly depend on the intensity of nociceptive activity.
Collapse
|
29
|
Chen L, Li S, Zhou Y, Liu T, Cai A, Zhang Z, Xu F, Manyande A, Wang J, Peng M. Neuronal mechanisms of adenosine A 2A receptors in the loss of consciousness induced by propofol general anesthesia with functional magnetic resonance imaging. J Neurochem 2020; 156:1020-1032. [PMID: 32785947 DOI: 10.1111/jnc.15146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/20/2020] [Accepted: 07/30/2020] [Indexed: 01/04/2023]
Abstract
Propofol is the most common intravenous anesthetic agent for induction and maintenance of anesthesia, and has been used clinically for more than 30 years. However, the mechanism by which propofol induces loss of consciousness (LOC) remains largely unknown. The adenosine A2A receptor (A2A R) has been extensively proven to have an effect on physiological sleep. It is, therefore, important to investigate the role of A2A R in the induction of LOC using propofol. In the present study, the administration of the highly selective A2A R agonist (CGS21680) and antagonist (SCH58261) was utilized to investigate the function of A2A R under general anesthesia induced by propofol by means of animal behavior studies, resting-state magnetic resonance imaging and c-Fos immunofluorescence staining approaches. Our results show that CGS21680 significantly prolonged the duration of LOC induced by propofol, increased the c-Fos expression in nucleus accumbens (NAc) and suppressed the functional connectivity of NAc-dorsal raphe nucleus (DR) and NAc-cingulate cortex (CG). However, SCH58261 significantly shortened the duration of LOC induced by propofol, decreased the c-Fos expression in NAc, increased the c-Fos expression in DR, and elevated the functional connectivity of NAc-DR and NAc-CG. Collectively, our findings demonstrate the important roles played by A2A R in the LOC induced by propofol and suggest that the neural circuit between NAc-DR maybe controlled by A2A R in the mechanism of anesthesia induced by propofol.
Collapse
Affiliation(s)
- Lei Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China.,Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Shuang Li
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Ying Zhou
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Aoling Cai
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, PR China
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Fuqiang Xu
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, PR China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, P. R. China.,Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, UK
| | - Jie Wang
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, PR China
| | - Mian Peng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| |
Collapse
|
30
|
Abstract
There is a complex interplay between sleep disturbance and patients in pain. There is an increasing appreciation of the direct effects of analgesic drugs and sleep quality. This review provides an overview of the effects of different analgesic drugs and their effects on phases of sleep. The effects of different pain conditions and their direct effects on sleep physiology are also discussed. A structured search of the scientific literature using MEDLINE and PubMed databases. Original human and animal studies were included. A multi-search term strategy was employed. An appreciation of the physiological effects of these drugs will allow a more considered prescription of them to better manage sleep disturbance.
Collapse
Affiliation(s)
- Adam Woo
- Consultant Anaesthetist & Pain Physician, King's College Hospital, London, UK
| | | |
Collapse
|
31
|
Wang Y, Liu Z, Cai L, Guo R, Dong Y, Huang YH. A Critical Role of Basolateral Amygdala-to-Nucleus Accumbens Projection in Sleep Regulation of Reward Seeking. Biol Psychiatry 2020; 87:954-966. [PMID: 31924324 PMCID: PMC7210061 DOI: 10.1016/j.biopsych.2019.10.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/09/2019] [Accepted: 10/27/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Sleep impacts reward-motivated behaviors partly by retuning the brain reward circuits. The nucleus accumbens (NAc) is a reward processing hub sensitive to acute sleep deprivation. Glutamatergic transmission carrying reward-associated signals converges in the NAc and regulates various aspects of reward-motivated behaviors. The basolateral amygdala projection (BLAp) innervates broad regions of the NAc and critically regulates reward seeking. METHODS Using slice electrophysiology, we measured how acute sleep deprivation alters transmission at BLAp-NAc synapses in male C57BL/6 mice. Moreover, using SSFO (stabilized step function opsin) and DREADDs (designer receptors exclusively activated by designer drugs) (Gi) to amplify and reduce transmission, respectively, we tested behavioral consequences following bidirectional manipulations of BLAp-NAc transmission. RESULTS Acute sleep deprivation increased sucrose self-administration in mice and altered the BLAp-NAc transmission in a topographically specific manner. It selectively reduced glutamate release at the rostral BLAp (rBLAp) onto ventral and lateral NAc (vlNAc) synapses, but spared caudal BLAp onto medial NAc synapses. Furthermore, experimentally facilitating glutamate release at rBLAp-vlNAc synapses suppressed sucrose reward seeking. Conversely, mimicking sleep deprivation-induced reduction of rBLAp-vlNAc transmission increased sucrose reward seeking. Finally, facilitating rBLAp-vlNAc transmission per se did not promote either approach motivation or aversion. CONCLUSIONS Sleep acts on rBLAp-vINAc transmission gain control to regulate established reward seeking but does not convey approach motivation or aversion on its own.
Collapse
Affiliation(s)
- Yao Wang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA,These authors contributed equally to this work
| | - Zheng Liu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,These authors contributed equally to this work
| | - Li Cai
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Rong Guo
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Yan Dong
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA,Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA
| | - Yanhua H. Huang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
32
|
Stroemel-Scheder C, Kundermann B, Lautenbacher S. The effects of recovery sleep on pain perception: A systematic review. Neurosci Biobehav Rev 2020; 113:408-425. [PMID: 32275917 DOI: 10.1016/j.neubiorev.2020.03.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 03/13/2020] [Accepted: 03/29/2020] [Indexed: 01/04/2023]
Abstract
Experimental studies highlight profound effects of sleep disruptions on pain, showing that sleep deprivation (SD) leads to hyperalgesic pain changes. On the other hand, given that sleep helps normalizing bodily functions, a crucial role of restorative sleep in the overnight restoration of the pain system seems likely. Thus, a systematic review of experimental studies on effects of recovery sleep (RS; subsequently to SD) on pain was performed with the aim to check whether RS resets hyperalgesic pain changes occurring due to SD. Empirical animal and human studies including SD-paradigms, RS and pain assessments were searched in three databases (PubMed, Web of Science, PsycINFO) using a predefined algorithm. 29 studies were included in this review. Most results indicated a reset of enhanced pain sensitivity and vulnerability following RS, especially when total SD was implemented and pressure pain or painful symptoms (human studies) were assessed. Further research should focus on whether and how recovery is altered in chronic pain patients, as this yields implications for pain treatment by enhancing or stabilizing RS.
Collapse
Affiliation(s)
| | - Bernd Kundermann
- Vitos Clinic for Psychiatry and Psychotherapy Giessen, Giessen, Germany; Department of Psychiatry and Psychotherapy, Philipps-University of Marburg, Marburg, Germany.
| | | |
Collapse
|
33
|
Attenuated dopamine receptor signaling in nucleus accumbens core in a rat model of chemically-induced neuropathy. Neuropharmacology 2020; 166:107935. [PMID: 31917153 DOI: 10.1016/j.neuropharm.2020.107935] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/10/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023]
Abstract
Neuropathy is major source of chronic pain that can be caused by mechanically or chemically induced nerve injury. Intraplantar formalin injection produces local necrosis over a two-week period and has been used to model neuropathy in rats. To determine whether neuropathy alters dopamine (DA) receptor responsiveness in mesolimbic brain regions, we examined dopamine D1-like and D2-like receptor (D1/2R) signaling and expression in male rats 14 days after bilateral intraplantar formalin injections into both rear paws. D2R-mediated G-protein activation and expression of the D2R long, but not short, isoform were reduced in nucleus accumbens (NAc) core, but not in NAc shell, caudate-putamen or ventral tegmental area of formalin- compared to saline-treated rats. In addition, D1R-stimulated adenylyl cyclase activity was also reduced in NAc core, but not in NAc shell or prefrontal cortex, of formalin-treated rats, whereas D1R expression was unaffected. Other proteins involved in dopamine neurotransmission, including dopamine uptake transporter and tyrosine hydroxylase, were unaffected by formalin treatment. In behavioral tests, the potency of a D2R agonist to suppress intracranial self-stimulation (ICSS) was decreased in formalin-treated rats, whereas D1R agonist effects were not altered. The combination of reduced D2R expression and signaling in NAc core with reduced suppression of ICSS responding by a D2R agonist suggest a reduction in D2 autoreceptor function. Altogether, these results indicate that intraplantar formalin produces attenuation of highly specific DA receptor signaling processes in NAc core of male rats and suggest the development of a neuropathy-induced allostatic state in both pre- and post-synaptic DA receptor function.
Collapse
|
34
|
Sleep deficiency and chronic pain: potential underlying mechanisms and clinical implications. Neuropsychopharmacology 2020; 45:205-216. [PMID: 31207606 PMCID: PMC6879497 DOI: 10.1038/s41386-019-0439-z] [Citation(s) in RCA: 273] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/10/2019] [Accepted: 06/05/2019] [Indexed: 11/09/2022]
Abstract
Pain can be both a cause and a consequence of sleep deficiency. This bidirectional relationship between sleep and pain has important implications for clinical management of patients, but also for chronic pain prevention and public health more broadly. The review that follows will provide an overview of the neurobiological evidence of mechanisms thought to be involved in the modulation of pain by sleep deficiency, including the opioid, monoaminergic, orexinergic, immune, melatonin, and endocannabinoid systems; the hypothalamus-pituitary-adrenal axis; and adenosine and nitric oxide signaling. In addition, it will provide a broad overview of pharmacological and non-pharmacological approaches for the management of chronic pain comorbid with sleep disturbances and for the management of postoperative pain, as well as discuss the effects of sleep-disturbing medications on pain amplification.
Collapse
|
35
|
Li Q, Zhu ZY, Lu J, Chao YC, Zhou XX, Huang Y, Chen XM, Su DS, Yu WF, Gu XY. Sleep deprivation of rats increases postsurgical expression and activity of L-type calcium channel in the dorsal root ganglion and slows recovery from postsurgical pain. Acta Neuropathol Commun 2019; 7:217. [PMID: 31870460 PMCID: PMC6929318 DOI: 10.1186/s40478-019-0868-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023] Open
Abstract
Perioperative sleep disturbance is a risk factor for persistent pain after surgery. Clinical studies have shown that patients with insufficient sleep before and after surgery experience more intense and long-lasting postoperative pain. We hypothesize that sleep deprivation alters L-type calcium channels in the dorsal root ganglia (DRG), thus delaying the recovery from post-surgical pain. To verify this hypothesis, and to identify new predictors and therapeutic targets for persistent postoperative pain, we first established a model of postsurgical pain with perioperative sleep deprivation (SD) by administering hind paw plantar incision to sleep deprivation rats. Then we conducted behavioral tests, including tests with von Frey filaments and a laser heat test, to verify sensory pain, measured the expression of L-type calcium channels using western blotting and immunofluorescence of dorsal root ganglia (an important neural target for peripheral nociception), and examined the activity of L-type calcium channels and neuron excitability using electrophysiological measurements. We validated the findings by performing intraperitoneal injections of calcium channel blockers and microinjections of dorsal root ganglion cells with adeno-associated virus. We found that short-term sleep deprivation before and after surgery increased expression and activity of L-type calcium channels in the lumbar dorsal root ganglia, and delayed recovery from postsurgical pain. Blocking these channels reduced impact of sleep deprivation. We conclude that the increased expression and activity of L-type calcium channels is associated with the sleep deprivation-mediated prolongation of postoperative pain. L-type calcium channels are thus a potential target for management of postoperative pain.
Collapse
|
36
|
Herrero Babiloni A, De Koninck BP, Beetz G, De Beaumont L, Martel MO, Lavigne GJ. Sleep and pain: recent insights, mechanisms, and future directions in the investigation of this relationship. J Neural Transm (Vienna) 2019; 127:647-660. [DOI: 10.1007/s00702-019-02067-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 08/16/2019] [Indexed: 12/13/2022]
|
37
|
Abstract
The nucleus accumbens (NAc) has been implicated in sleep, reward, and pain modulation, but the relationship between these functional roles is unclear. This study aimed to determine whether NAc function at the onset and offset of a noxious thermal stimulus is enhanced by rewarding music, and whether that effect is reversed by experimental sleep disruption. Twenty-one healthy subjects underwent functional magnetic resonance imaging scans on 2 separate days after both uninterrupted sleep and experimental sleep disruption. During functional magnetic resonance imaging scans, participants experienced noxious stimulation while listening to individualized rewarding or neutral music. Behavioral results revealed that rewarding music significantly reduced pain intensity compared with neutral music, and disrupted sleep was associated with decreased pain intensity in the context of listening to music. In whole-brain family-wise error cluster-corrected analysis, the NAc was activated at pain onset, but not during tonic pain or at pain offset. Sleep disruption attenuated NAc activation at pain onset and during tonic pain. Rewarding music altered NAc connectivity with key nodes of the corticostriatal circuits during pain onset. Sleep disruption increased reward-related connectivity between the NAc and the anterior midcingulate cortex at pain onset. This study thus indicates that experimental sleep disruption modulates NAc function during the onset of pain in a manner that may be conditional on the presence of competing reward-related stimuli. These findings point to potential mechanisms for the interaction between sleep, reward, and pain, and suggest that sleep disruption affects both the detection and processing of aversive stimuli that may have important implications for chronic pain.
Collapse
|
38
|
Bjurström MF, Irwin MR. Perioperative Pharmacological Sleep‐Promotion and Pain Control: A Systematic Review. Pain Pract 2019; 19:552-569. [DOI: 10.1111/papr.12776] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/21/2019] [Accepted: 02/07/2019] [Indexed: 01/15/2023]
Affiliation(s)
- Martin F. Bjurström
- Department of Anesthesiology and Intensive Care Skåne University Hospital LundSweden
- Department of Clinical Sciences Lund University Lund Sweden
- Cousins Center for Psychoneuroimmunology University of California, Los Angeles (UCLA) Los Angeles California U.S.A
| | - Michael R. Irwin
- Cousins Center for Psychoneuroimmunology University of California, Los Angeles (UCLA) Los Angeles California U.S.A
| |
Collapse
|
39
|
The Pain of Sleep Loss: A Brain Characterization in Humans. J Neurosci 2019; 39:2291-2300. [PMID: 30692228 DOI: 10.1523/jneurosci.2408-18.2018] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/06/2018] [Accepted: 12/07/2018] [Indexed: 12/24/2022] Open
Abstract
Sleep loss increases the experience of pain. However, the brain mechanisms underlying altered pain processing following sleep deprivation are unknown. Moreover, it remains unclear whether ecologically modest night-to-night changes in sleep, within an individual, confer consequential day-to-day changes in experienced pain. Here, we demonstrate that acute sleep deprivation amplifies pain reactivity within human (male and female) primary somatosensory cortex yet blunts pain reactivity in higher-order valuation and decision-making regions of the striatum and insula cortex. Consistent with this altered neural signature, we further show that sleep deprivation expands the temperature range for classifying a stimulus as painful, specifically through a lowering of pain thresholds. Moreover, the degree of amplified reactivity within somatosensory cortex following sleep deprivation significantly predicts this expansion of experienced pain across individuals. Finally, outside of the laboratory setting, we similarly show that even modest nightly changes in sleep quality (increases and decreases) within an individual determine consequential day-to-day changes in experienced pain (decreases and increases, respectively). Together, these data provide a novel framework underlying the impact of sleep loss on pain and, furthermore, establish that the association between sleep and pain is expressed in a night-to-day, bidirectional relationship within a sample of the general population. More broadly, our findings highlight sleep as a novel therapeutic target for pain management within and outside the clinic, including circumstances where sleep is frequently short yet pain is abundant (e.g., the hospital setting).SIGNIFICANCE STATEMENT Are you experiencing pain? Did you have a bad night of sleep? This study provides underlying brain and behavioral mechanisms explaining this common co-occurrence. We show that sleep deprivation enhances pain responsivity within the primary sensing regions of the brain's cortex yet blunts activity in other regions that modulate pain processing, the striatum and insula. We further establish that even subtle night-to-night changes in sleep in a sample of the general population predict consequential day-to-day changes in pain (bidirectionally). Considering the societal rise in chronic pain conditions in lock-step with the decline in sleep time through the industrial world, our data support the hypothesis that these two trends may not simply be co-occurring but are significantly interrelated.
Collapse
|
40
|
Hirotsu C, Pedroni MN, Berro LF, Tufik S, Andersen ML. Nicotine and sleep deprivation: impact on pain sensitivity and immune modulation in rats. Sci Rep 2018; 8:13837. [PMID: 30218019 PMCID: PMC6138689 DOI: 10.1038/s41598-018-32276-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 06/01/2018] [Indexed: 11/09/2022] Open
Abstract
Repeated nicotine administration has been associated with increased paradoxical sleep in rats and antinociceptive properties, whereas paradoxical sleep deprivation (PSD) elicits pronociceptive and inflammatory responses. Thus, we aimed to evaluate the effect of repeated nicotine administration and its withdrawal combined with PSD on pain sensitivity and inflammatory markers. Sixty adult male Wistar rats were subjected to repeated injections of saline (SAL) or nicotine (NIC) for 12 days or 7 days of nicotine followed by acute mecamylamine administration on day 8 to precipitate nicotine abstinence (ABST). On day 9, the animals were submitted to PSD for 72 h or remained in control condition (CTRL); on day 12, thermal pain threshold was assessed by the hot plate test. PSD significantly decreased the latency to paw withdrawal in all groups compared to their respective controls. ABST-PSD animals presented higher levels of interleukin (IL)-6 compared to all groups, except ABST-CTRL. After adjustment for weight loss, IL-6, IL-4 and tumor necrosis factor alpha, ABST-PSD was associated with the lowest pain threshold. Nicotine and IL-4 levels were predictors of higher pain threshold. Hyperalgesia induced by PSD prevailed over the antinociceptive action of nicotine, while the association between PSD and ABST synergistically increased IL-6 concentrations and decreased pain threshold.
Collapse
Affiliation(s)
- Camila Hirotsu
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Laís Fernanda Berro
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, USA
| | - Sergio Tufik
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Monica Levy Andersen
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil.
| |
Collapse
|
41
|
Sardi NF, Lazzarim MK, Guilhen VA, Marcílio RS, Natume PS, Watanabe TC, Lima MMS, Tobaldini G, Fischer L. Chronic sleep restriction increases pain sensitivity over time in a periaqueductal gray and nucleus accumbens dependent manner. Neuropharmacology 2018; 139:52-60. [PMID: 29928886 DOI: 10.1016/j.neuropharm.2018.06.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/29/2018] [Accepted: 06/16/2018] [Indexed: 11/29/2022]
Abstract
Painful conditions and sleep disturbances are major public health problems worldwide and one directly affects the other. Sleep loss increases pain prevalence and severity; while pain disturbs sleep. However, the underlying mechanisms are largely unknown. Here we asked whether chronic sleep restriction for 6 h daily progressively increases pain sensitivity and if this increase is reversed after two days of free sleep. Also, whether the pronociceptive effect of chronic sleep restriction depends on the periaqueductal grey and on the nucleus accumbens, two key regions involved in the modulation of pain and sleep-wake cycle. We showed that sleep restriction induces a pronociceptive effect characterized by a significant decrease in the mechanical paw withdrawal threshold in rats. Such effect increases progressively from day 3 to day 12 remaining stable thereafter until day 26. Two consecutive days of free sleep were not enough to reverse the effect, not even to attenuate it. This pronociceptive effect depends on the periaqueductal grey and on the nucleus accumbens, since it was prevented by their excitotoxic lesion. Complementarily, chronic sleep restriction significantly increased c-Fos protein expression within the periaqueductal grey and the nucleus accumbens and this correlates with the intensity of the pronociceptive effect, suggesting that the greater the neural activity in this regions, the greater the effect. These findings may contribute not only to understand why painful conditions are more prevalent and severe among people who sleep poorly, but also to develop therapeutic strategies to prevent this, increasing the effectiveness of pain management in this population.
Collapse
Affiliation(s)
- Natalia F Sardi
- Neurophysiology Laboratory, Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Mayla K Lazzarim
- Neurophysiology Laboratory, Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Vinicius A Guilhen
- Neurophysiology Laboratory, Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Renata S Marcílio
- Neurophysiology Laboratory, Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Priscila S Natume
- Neurophysiology Laboratory, Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Thainá C Watanabe
- Neurophysiology Laboratory, Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Marcelo M S Lima
- Neurophysiology Laboratory, Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Glaucia Tobaldini
- Neurophysiology Laboratory, Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Luana Fischer
- Neurophysiology Laboratory, Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil.
| |
Collapse
|
42
|
Tobaldini G, Sardi NF, Guilhen VA, Fischer L. Pain Inhibits Pain: an Ascending-Descending Pain Modulation Pathway Linking Mesolimbic and Classical Descending Mechanisms. Mol Neurobiol 2018; 56:1000-1013. [DOI: 10.1007/s12035-018-1116-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 05/11/2018] [Indexed: 10/14/2022]
|
43
|
Reply. Pain 2018; 159:997-999. [DOI: 10.1097/j.pain.0000000000001178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
44
|
Ruffle JK, Aziz Q, Farmer AD. Pronociceptive effects mediated by adenosinergic A2A activity at the nucleus accumbens, but what about the autonomic nervous system? Pain 2018; 159:997. [PMID: 29672454 DOI: 10.1097/j.pain.0000000000001179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- James K Ruffle
- Centre for Neuroscience and Trauma, Blizard Institute, Wingate Institute of Neurogastroenterology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Qasim Aziz
- Centre for Neuroscience and Trauma, Blizard Institute, Wingate Institute of Neurogastroenterology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Adam D Farmer
- Centre for Neuroscience and Trauma, Blizard Institute, Wingate Institute of Neurogastroenterology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Gastroenterology, University Hospitals Midlands NHS Trust, Stoke on Trent, Staffordshire, United Kingdom
| |
Collapse
|
45
|
Effects of intra-accumbal or intra-prefrontal cortex microinjections of adenosine 2A receptor ligands on responses to cocaine reward and seeking in rats. Psychopharmacology (Berl) 2018; 235:3509-3523. [PMID: 30426181 PMCID: PMC6267142 DOI: 10.1007/s00213-018-5072-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022]
Abstract
RATIONALE AND OBJECTIVES Many studies indicated that adenosine via its A2A receptors influences the behavioral effects of cocaine by modulating dopamine neurotransmission. The hypothesis was tested that A2A receptors in the nucleus accumbens (NAc) or the prefrontral cortex (PFc) may modulate cocaine reward and/or cocaine seeking behavior in rats. METHODS The effects of local bilateral microinjections of the selective A2A receptor agonist CGS 21680 or the A2A receptor antagonists KW 6002 and SCH 58261 were investigated on cocaine self-administration on reinstatement of cocaine seeking. RESULTS The intra-NAc shell, but not intra-infralimbic PFc, administration of CGS 21680 significantly reduced the number of active lever presses and the number of cocaine (0.25 mg/kg) infusions. However, tonic activation of A2A receptors located in the NAc or PFc did not play a role in modulating the rewarding actions of cocaine since neither KW 6002 nor SCH 58261 microinjections altered the cocaine (0.5 mg/kg) infusions. The intra-NAc but not intra-PFc microinjections of CGS 21680 dose- dependently attenuated the reinstatement of active lever presses induced by cocaine (10 mg/kg, i.p.) and the drug-associated combined conditioned stimuli using the subthreshold dose of cocaine (2.5 mg/kg, i.p.). On the other hand, the intra-NAc pretreatment with SCH 58261, but not with KW 6002, given alone evoked reinstatement of cocaine seeking behavior. CONCLUSION The results strongly support the involvement of accumbal shell A2A receptors as a target, the activation of which exerts an inhibitory control over cocaine reward and cocaine seeking.
Collapse
|
46
|
Nucleus accumbens: a novel forebrain mechanism underlying the increase in pain sensitivity caused by rapid eye movement sleep deprivation. Pain 2017; 159:5-6. [DOI: 10.1097/j.pain.0000000000001073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|