1
|
Sun Q, Weng RX, Li YC, Jia SM, Ma CT, Zhang HH, Tang Y, Li R, Xu GY. Potentiation of visualized exosomal miR-1306-3p from primary sensory neurons contributes to chronic visceral pain via spinal P2X3 receptors. Pain 2025:00006396-990000000-00814. [PMID: 39907482 DOI: 10.1097/j.pain.0000000000003537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/31/2024] [Indexed: 02/06/2025]
Abstract
ABSTRACT Exosomes served as "communicators" to exchange information among different cells in the nervous system. Our previous study demonstrated that the enhanced spinal synaptic transmission contributed to chronic visceral pain in irritable bowel syndrome. However, the underlying mechanism of primary sensory neuron (PSN)-derived exosomes on spinal transmission remains unclear. In this study, an exosome visualization method was established to specifically track exosomes derived from PSNs in CD63-GFPf/+ (green fluorescent protein) mice. Neonatal maternal deprivation (NMD) was adopted to induce chronic visceral pain in CD63-GFPf/+ male mice. The exosome visualization technology demonstrated that NMD increased visible PSN-derived exosomes in the spinal dorsal horn, enhanced spinal synaptic transmission, and led to visceral pain in CD63-GFPf/+ male mice. The PSN-derived exosomal miR-1306-3p sorted from spinal dorsal horn activated P2X3R, enhanced spinal synaptic transmission, and led to visceral pain in NMD mice. Moreover, upregulation of Rab27a in dorsal root ganglia mediated the increased release of PSN-derived exosomes, and intrathecal injection of siR-Rab27a reduced visible PSN-derived exosomes in spinal cord, suppressed spinal synaptic transmission, and alleviated visceral pain in NMD mice. This and future studies would reveal the detailed mechanisms of PSN-derived exosomes and provide a potential target for clinical treatment of chronic visceral pain in patients with irritable bowel syndrome.
Collapse
Affiliation(s)
- Qian Sun
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, P. R. China
| | - Rui-Xia Weng
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China
| | - Yong-Chang Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, P. R. China
| | - Shu-Man Jia
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China
| | - Chun-Tao Ma
- Department of Gastroenterology, Suzhou Xiangcheng People's Hospital, Suzhou, P. R. China
| | - Hong-Hong Zhang
- Department of Endocrinology, the Second Affiliated Hospital, Soochow University, Suzhou, P. R. China
| | - Yong Tang
- International Collaborative Centre on Big Science Plan for Purine Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China
| | - Guang-Yin Xu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, P. R. China
| |
Collapse
|
2
|
Koponen ME, Naray E, Hales TG, Forget P. Pharmacological interventions for remifentanil-induced hyperalgesia: A systematic review and network meta-analysis of preclinical trials. PLoS One 2024; 19:e0313749. [PMID: 39636808 PMCID: PMC11620364 DOI: 10.1371/journal.pone.0313749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND To improve perioperative pain management, several interventions have been suggested for the prevention of increased pain sensitivity caused by opioids (called opioid-induced hyperalgesia). It is currently unclear which intervention is the most effective or appropriate in preventing opioid-induced hyperalgesia. Remifentanil is the most investigated opioid causing opioid-induced hyperalgesia. Thus, to guide future research, we conducted a systematic review and a network meta-analysis of preclinical trials investigating pharmacological interventions for remifentanil-induced hyperalgesia. METHODS To identify relevant articles, electronic database searches were conducted in Embase, PubMed, Web of Science, and Google Scholar. Study characteristics were extracted, and the risk of bias was evaluated. Studies were included in the network meta-analysis if they shared similar characteristics with at least one other study. The interventions were ranked based on P-scores. RESULTS Overall, the 62 eligible trials tested 86 individual interventions and 6 combination interventions. Thirty-five studies eligible in the network meta-analysis formed five groups which were further divided into subgroups based on the quantitative sensory tests used. The best-ranked interventions within the subgroups were Anxa12-26, MRS2179, salicylaldehyde isonicotinoyl hydrazone (SIH), ANA-12, TDZD-8, ketamine, dexmedetomidine, JWH015, and the combination of KN93 and ketamine. DISCUSSION The current literature is too heterogeneous to produce a clear answer on which intervention is the most effective in preventing remifentanil-induced hyperalgesia. Future research in this field should prioritise finding the most effective intervention over testing the efficacy of new options. The results of our work can be used in planning which comparisons should be included in new trials.
Collapse
Affiliation(s)
- Mia E. Koponen
- MSc Clinical Pharmacology, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Emily Naray
- MSci Biomedical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Tim G. Hales
- Division of Systems Medicine, School of Medicine, Institute of Academic Anaesthesia, Ninewells Hospital, University of Dundee, Dundee, United Kingdom
| | - Patrice Forget
- Institute of Applied Health Sciences, Epidemiology Group, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
- Department of Anaesthesia, National Health Service (NHS) Grampian, Aberdeen, United Kingdom
- Emergency and Pain Medicine Division, IMAGINE UR UM 103, Montpellier University, Anesthesia Critical Care, Nîmes University Hospital, Nîmes, France
- Pain and Opioids after Surgery (PANDOS) European Society of Anaesthesia (ID ESAIC_RG_PAND) Research Group, Brussels, Belgium
| |
Collapse
|
3
|
Wen T, Wen J, Yao C. Remimazolam inhibits postoperative cognitive impairment after cardiopulmonary bypass by alleviating neuroinflammation and promoting microglia M2 polarization. Brain Res 2024; 1838:148975. [PMID: 38702024 DOI: 10.1016/j.brainres.2024.148975] [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: 03/20/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Postoperative cognitive impairment (POCD) is a complication of cardiopulmonary bypass (CPB). Remimazolam is an ultra-short acting benzodiazepine that can be used for anesthesia or sedation during surgery. This study investigated the role of remimazolam in inflammasome activation and microglia polarization using CPB rat model and lipopolysaccharide (LPS)-induced microglia model. The cognitive function of rats was evaluated by Morris water maze. TUNEL assay was performed to detect apoptosis. Inflammatory cytokines concentration were analyzed by enzyme-linked immunosorbent assay. Reverse transcription-polymerase chain reaction was used to assess the expression of inflammasome and M1/M2-related microglia markers. Flow cytometry was performed to evaluate the expression of CD16/32 and CD206 in microglia. The results showed that remimazolam improved the memory and learning abilities in CPB rats. CPB rats and LPS-treated microglia showed increased apoptosis, pro-inflammatory cytokines level, and inflammasome expression as well as decreased microglia activation, while the results were reversed after remimazolam treatment. Besides, remimazolam treatment promoted the expression of M2-related markers in LPS-treated microglia. Nigericin treatment reversed the increased M2-related mRNA levels and the decreased apoptosis and inflammatory responses induced by remimazolam treatment. In conclusion, remimazolam attenuated POCD after CPB through regulating neuroinflammation and microglia M2 polarization, suggesting a new insight into the clinical treatment of POCD after CPB.
Collapse
Affiliation(s)
- Tao Wen
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, China
| | - Jing Wen
- Laboratory Department of Peking University Shenzhen Hospital, Shenzhen, China
| | - Cuicui Yao
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, China.
| |
Collapse
|
4
|
Liu X, Cai H, Peng L, Ma H, Yan Y, Li W, Zhao J. Microglial Nrf2/HO-1 signaling gates remifentanil-induced hyperalgesia via suppressing TRPV4-mediated M1 polarization. Free Radic Biol Med 2024; 214:87-100. [PMID: 38295888 DOI: 10.1016/j.freeradbiomed.2024.01.047] [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: 11/07/2023] [Revised: 12/29/2023] [Accepted: 01/27/2024] [Indexed: 02/18/2024]
Abstract
Remifentanil-induced hyperalgesia (RIH) represents a significant clinical challenge due to the widespread use of opioids in pain management. However, the molecular and cellular mechanisms underlying RIH remain elusive. This study aimed to unravel the role of spinal cord microglia, focusing on the Nrf2/HO-1 signaling pathway and TRPV4 channels in the development of RIH. We used both in vivo and in vitro models to investigate the activation state of spinal cord microglia, the expression of TRPV4 channels, and the modulation of the Nrf2/HO-1 pathway under remifentanil exposure. In addition, we evaluated the potential therapeutic effects of dexmedetomidine, a perioperative α2-adrenergic agonist, on RIH and its related molecular pathways. Our results revealed a prominent role of spinal cord microglia in RIH, demonstrating an apparent microglial M1 polarization and increased TRPV4 channel expression. A notable observation was the downregulation of the Nrf2/HO-1 pathway, which was associated with increased neuroinflammation and mechanical allodynia. By upregulating or overexpressing Nrf2, we confirmed its ability to inhibit TRPV4 and thereby attenuate RIH-associated mechanical allodynia, M1 polarization, and neuroinflammation. Encouragingly, dexmedetomidine demonstrated therapeutic potential by positively modulating the Nrf2-TRPV4 nexus, attenuating mechanical allodynia, and reducing microglial inflammation. Our research highlights the critical role of spinal cord microglia in RIH mediated by the Nrf2-TRPV4 axis. The ability of dexmedetomidine to modulate this axis suggests its potential as an adjunctive therapy to remifentanil in mitigating RIH. Further studies are imperative to explore the broader implications and practical applicability of our findings.
Collapse
Affiliation(s)
- Xiaowen Liu
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Huamei Cai
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Liang Peng
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Hongli Ma
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Yun Yan
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China; Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Weixia Li
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jing Zhao
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
5
|
Lu J, Yang B, Zhang W, Cheng H, Zeng J, Wang Y, Wei W, Liu Z. Transplantation of olfactory ensheathing cells can alleviate neuroinflammatory responses in rats with trigeminal neuralgia. Brain Res 2024; 1825:148732. [PMID: 38104922 DOI: 10.1016/j.brainres.2023.148732] [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: 05/12/2023] [Revised: 09/07/2023] [Accepted: 12/14/2023] [Indexed: 12/19/2023]
Abstract
Trigeminal neuralgia (TN) is a common form of facial pain, which primarily manifests as severe pain similar to facial acupuncture and electric shock. Olfactory ensheathing cells (OECs) are glial cells with high bioactivity; these cells are essential for the periodic regeneration of the olfactory nerve and have been utilized for the repair of nerve injuries. A member of the P2X receptor family, P2X7R, is an ion channel type receptor that has been confirmed to participate in various pain response processes. In this study, we transplanted OECs into trigeminal nerve-model rats with distal infraorbital nerve ligation to observe the therapeutic effect of transplanted OECs in rats. Additionally, we utilized the P2X7R-specific inhibitor brilliant blue G (BBG) to study the therapeutic mechanisms of cell transplantation. The facial mechanical pain threshold of these rats significantly increased following cell transplantation. The immunohistochemistry, immunoblotting, and RT-qPCR results demonstrated that the levels of P2X7R, (NOD)-like receptor protein-3 (NLRP3), nuclear factor-κB (NF-κB), interleukin (IL)-1β, and IL-18 in the trigeminal ganglion of rats treated with OEC transplantation or BBG treatment were significantly lower than those in the injured group without treatment. Overall, our results demonstrate that OEC transplantation can alleviate TN in rats, and it can reduce the expression of P2X7R related inflammatory factors in TN rats, reducing neuroinflammatory response in TG.
Collapse
Affiliation(s)
- Jiafeng Lu
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Baolin Yang
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Wenjun Zhang
- Rehabilitation Medicine Department, Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Hui Cheng
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Jingnan Zeng
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Yuanli Wang
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Wei Wei
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China
| | - Zengxu Liu
- Department of Anatomy, Basic Medical School, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
6
|
Zare N, Sharafeddin F, Montazerolghaem A, Moradiannezhad N, Araghizadeh M. NLRs and inflammasome signaling in opioid-induced hyperalgesia and tolerance. Inflammopharmacology 2024; 32:127-148. [PMID: 38153538 DOI: 10.1007/s10787-023-01402-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/18/2023] [Indexed: 12/29/2023]
Abstract
We investigated the role that innate immunological signaling pathways, principally nod-like receptors (NLRs) and inflammasomes, in the manifestation of the contradictory outcomes associated with opioids, namely hyperalgesia, and tolerance. The utilization of opioids for pain management is prevalent; nonetheless, it frequently leads to an increased sensitivity to pain (hyperalgesia) and reduced efficacy of the medication (tolerance) over an extended period. This, therefore, represents a major challenge in the area of chronic pain treatment. Recent studies indicate that the aforementioned negative consequences are partially influenced by the stimulation of NLRs, specifically the NLRP3 inflammasome, and the subsequent assembly of the inflammasome. This process ultimately results in the generation of inflammatory cytokines and the occurrence of neuroinflammation and the pathogenesis of hyperalgesia. We also explored the putative downstream signaling cascades activated by NOD-like receptors (NLRs) and inflammasomes in response to opioid stimuli. Furthermore, we probed potential therapeutic targets for modifying opioid-induced hyperalgesia, with explicit emphasis on the activation of the NLRP3 inflammasome. Ultimately, our findings underscore the significance of conducting additional research in this area that includes an examination of the involvement of various NLRs, immune cells, and genetic variables in the development of opioid-induced hyperalgesia and tolerance. The present review provides substantial insight into the possible pathways contributing to the occurrence of hyperalgesia and tolerance in individuals taking opioids.
Collapse
Affiliation(s)
- Nasrin Zare
- Clinical Research Development Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran.
- School of Medicine, Najafabad Branch, Islamic Azad University, Najafabad, Iran.
| | - Fateme Sharafeddin
- Clinical Research Development Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran
- School of Medicine, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - AmirMahdi Montazerolghaem
- Clinical Research Development Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran
- School of Medicine, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Nastaran Moradiannezhad
- Clinical Research Development Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran
- School of Medicine, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Mohammaderfan Araghizadeh
- Clinical Research Development Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran
- School of Medicine, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| |
Collapse
|
7
|
Liu W, Li Y, Zhao T, Gong M, Wang X, Zhang Y, Xu L, Li W, Li Y, Jia J. The role of N-methyl-D-aspartate glutamate receptors in Alzheimer's disease: From pathophysiology to therapeutic approaches. Prog Neurobiol 2023; 231:102534. [PMID: 37783430 DOI: 10.1016/j.pneurobio.2023.102534] [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: 02/27/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
N-Methyl-D-aspartate glutamate receptors (NMDARs) are involved in multiple physiopathological processes, including synaptic plasticity, neuronal network activities, excitotoxic events, and cognitive impairment. Abnormalities in NMDARs can initiate a cascade of pathological events, notably in Alzheimer's disease (AD) and even other neuropsychiatric disorders. The subunit composition of NMDARs is plastic, giving rise to a diverse array of receptor subtypes. While they are primarily found in neurons, NMDAR complexes, comprising both traditional and atypical subunits, are also present in non-neuronal cells, influencing the functions of various peripheral tissues. Furthermore, protein-protein interactions within NMDAR complexes has been linked with Aβ accumulation, tau phosphorylation, neuroinflammation, and mitochondrial dysfunction, all of which potentially served as an obligatory relay of cognitive impairment. Nonetheless, the precise mechanistic link remains to be fully elucidated. In this review, we provided an in-depth analysis of the structure and function of NMDAR, investigated their interactions with various pathogenic proteins, discussed the current landscape of NMDAR-based therapeutics, and highlighted the remaining challenges during drug development.
Collapse
Affiliation(s)
- Wenying Liu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Tan Zhao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Min Gong
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Xuechu Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Yue Zhang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Wenwen Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China.
| |
Collapse
|
8
|
Abstract
This paper is the forty-fifth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2022 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, USA.
| |
Collapse
|
9
|
Smith PA. Neuropathic pain; what we know and what we should do about it. FRONTIERS IN PAIN RESEARCH 2023; 4:1220034. [PMID: 37810432 PMCID: PMC10559888 DOI: 10.3389/fpain.2023.1220034] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Neuropathic pain can result from injury to, or disease of the nervous system. It is notoriously difficult to treat. Peripheral nerve injury promotes Schwann cell activation and invasion of immunocompetent cells into the site of injury, spinal cord and higher sensory structures such as thalamus and cingulate and sensory cortices. Various cytokines, chemokines, growth factors, monoamines and neuropeptides effect two-way signalling between neurons, glia and immune cells. This promotes sustained hyperexcitability and spontaneous activity in primary afferents that is crucial for onset and persistence of pain as well as misprocessing of sensory information in the spinal cord and supraspinal structures. Much of the current understanding of pain aetiology and identification of drug targets derives from studies of the consequences of peripheral nerve injury in rodent models. Although a vast amount of information has been forthcoming, the translation of this information into the clinical arena has been minimal. Few, if any, major therapeutic approaches have appeared since the mid 1990's. This may reflect failure to recognise differences in pain processing in males vs. females, differences in cellular responses to different types of injury and differences in pain processing in humans vs. animals. Basic science and clinical approaches which seek to bridge this knowledge gap include better assessment of pain in animal models, use of pain models which better emulate human disease, and stratification of human pain phenotypes according to quantitative assessment of signs and symptoms of disease. This can lead to more personalized and effective treatments for individual patients. Significance statement: There is an urgent need to find new treatments for neuropathic pain. Although classical animal models have revealed essential features of pain aetiology such as peripheral and central sensitization and some of the molecular and cellular mechanisms involved, they do not adequately model the multiplicity of disease states or injuries that may bring forth neuropathic pain in the clinic. This review seeks to integrate information from the multiplicity of disciplines that seek to understand neuropathic pain; including immunology, cell biology, electrophysiology and biophysics, anatomy, cell biology, neurology, molecular biology, pharmacology and behavioral science. Beyond this, it underlines ongoing refinements in basic science and clinical practice that will engender improved approaches to pain management.
Collapse
Affiliation(s)
- Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
10
|
Dello Russo C, Cappoli N, Tabolacci E, Sollazzi L, Navarra P, Aceto P. Remifentanil does not affect human microglial immune activation in response to pro-inflammatory cytokines. EXCLI JOURNAL 2023; 22:295-309. [PMID: 37220493 PMCID: PMC10201013 DOI: 10.17179/excli2022-5667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/20/2023] [Indexed: 05/25/2023]
Abstract
Remifentanil is a potent ultra-short acting μ-opioid analgesic drug, frequently used in anaesthesia due to its favorable pharmacodynamic and pharmacokinetic profile. It may be associated with the occurrence of hyperalgesia. Preclinical studies suggest a potential role of microglia, although the molecular mechanisms have not been fully elucidated. Considering the role of microglia in brain inflammation and the relevant differences among species, the effects of remifentanil were studied on the human microglial C20 cells. The drug was tested at clinically relevant concentrations under basal and inflammatory conditions. In the C20 cells, the expression and secretion of interleukin 6, interleukin 8 and the monocyte chemotactic protein 1 were rapidly induced by a mixture of pro-inflammatory cytokines. This stimulatory effect was sustained up to 24 h. Remifentanil did not exert any toxic effect nor modify the production of these inflammatory mediators, thus suggesting the lack of direct immune modulatory actions on human microglia.
Collapse
Affiliation(s)
- Cinzia Dello Russo
- Dipartimento di Sicurezza e Bioetica, Sezione di Farmacologia, Università Cattolica Del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Pharmacology & Therapeutics, Institute of Systems Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool, United Kingdom
| | - Natalia Cappoli
- Dipartimento di Sicurezza e Bioetica, Sezione di Farmacologia, Università Cattolica Del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elisabetta Tabolacci
- Dipartimento di Scienze Della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Liliana Sollazzi
- Dipartimento di Scienze dell'Emergenza, Anestesiologiche e della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Pierluigi Navarra
- Dipartimento di Sicurezza e Bioetica, Sezione di Farmacologia, Università Cattolica Del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Paola Aceto
- Dipartimento di Scienze dell'Emergenza, Anestesiologiche e della Rianimazione, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
11
|
Temmermand R, Barrett JE, Fontana ACK. Glutamatergic systems in neuropathic pain and emerging non-opioid therapies. Pharmacol Res 2022; 185:106492. [PMID: 36228868 PMCID: PMC10413816 DOI: 10.1016/j.phrs.2022.106492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 01/14/2023]
Abstract
Neuropathic pain, a disease of the somatosensory nervous system, afflicts many individuals and adequate management with current pharmacotherapies remains elusive. The glutamatergic system of neurons, receptors and transporters are intimately involved in pain but, to date, there have been few drugs developed that therapeutically modulate this system. Glutamate transporters, or excitatory amino acid transporters (EAATs), remove excess glutamate around pain transmitting neurons to decrease nociception suggesting that the modulation of glutamate transporters may represent a novel approach to the treatment of pain. This review highlights and summarizes (1) the physiology of the glutamatergic system in neuropathic pain, (2) the preclinical evidence for dysregulation of glutamate transport in animal pain models, and (3) emerging novel therapies that modulate glutamate transporters. Successful drug discovery requires continuous focus on basic and translational methods to fully elucidate the etiologies of this disease to enable the development of targeted therapies. Increasing the efficacy of astrocytic EAATs may serve as a new way to successfully treat those suffering from this devastating disease.
Collapse
Affiliation(s)
- Rhea Temmermand
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - James E Barrett
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Andréia C K Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA.
| |
Collapse
|