1
|
Reilimo MI, Sainio M, Liira J, Laurola M. The effect of peer group management intervention on chronic pain intensity, number of areas of pain, and pain self-efficacy. Scand J Pain 2025; 25:sjpain-2024-0018. [PMID: 40014757 DOI: 10.1515/sjpain-2024-0018] [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: 02/24/2024] [Accepted: 01/07/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVES Chronic pain causes loss of workability, and pharmacological treatment is often not sufficient, whereas psychosocial treatments may relieve continual pain. This study aimed to investigate the effect of peer group management intervention among patients with chronic pain. METHODS The participants were 18-65-year-old employees of the Municipality of Helsinki (women 83%) who visited an occupational health care physician, nurse, psychologist, or physiotherapist for chronic pain lasting at least 3 months. An additional inclusion criterion was an elevated risk of work disability. Our study was a stepped wedge cluster, randomized controlled trial, and group interventions used mindfulness, relaxation, cognitive behavioral therapy, and acceptance and commitment therapy. We randomized sixty participants to either a pain management group intervention or to a waiting list with the same intervention 5 months later. After dropouts, 48 employees participated in 6 weekly group meetings. We followed up participants from groups A, B, and C for 12 months and groups D, E, and F for 6 months. As outcome measures, we used the pain Self-Efficacy Questionnaire, the number of areas of pain, the visual analog scale of pain, and the pain self-efficacy. We adjusted the results before and after the intervention for panel data, clustering effect, and time interval. RESULTS The peer group intervention decreased the number of areas of pain by 40%, from 5.96 (1-10) to 3.58 (p < 0.001), and increased the pain self-efficacy by 15%, from 30.4 to 37.5 (p < 0.001). Pain intensity decreased slightly, but not statistically significantly, from 7.1 to 6.8. CONCLUSIONS Peer group intervention for 6 weeks among municipal employees with chronic pain is partially effective. The number of areas of pain and pain self-efficacy were more sensitive indicators of change than the pain intensity. Any primary care unit with sufficient resources may implement the intervention.
Collapse
Affiliation(s)
| | | | - Juha Liira
- Occupational Health, University of Turku, Helsinki, Finland
| | | |
Collapse
|
2
|
Abbasloo E, Esmaeili-Mahani S, Kobeissy F, Currier Thomas T. Evidence for the Induction of Analgesic Cross-Tolerance Between Opioid and Apelin/APJ Systems in Male Rats. J Stud Alcohol Drugs 2024; 85:704-712. [PMID: 38517751 PMCID: PMC11533929 DOI: 10.15288/jsad.23-00377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 02/28/2024] [Indexed: 03/24/2024] Open
Abstract
OBJECTIVE Opioids are potent pain relievers for managing severe pain. However, their effectiveness is hindered by tolerance, which causes the need for higher doses and leads to adverse effects. In a previous study, we found that prolonged use of apelin, similar to opioids, results in a tolerance to its analgesic effects. It remains unclear whether there is a cross-tolerance between morphine and apelin, meaning if the analgesic effects of one can reduce the effectiveness of the other. METHOD The tail-flick test was used to assess the nociceptive threshold. All experiments were carried out on 63 male Wistar rats, which received intrathecal apelin (3 μg/rat) or morphine (15 μg/rat) for 7 days. To determine cross-tolerance between the analgesic effect of morphine and apelin, the analgesic property of apelin or morphine was assessed in chronic morphine- or apelin-treated groups, respectively. To determine the role of apelin and opioid receptors signaling on the development of analgesic cross-tolerance, F13-A and naloxone, as apelin and opioid receptor antagonists, were injected simultaneously with morphine or apelin. At the end of the tests, the expression levels of apelin and μ-opioid receptors were evaluated by western blotting. RESULTS The data indicated that chronic apelin or morphine use produced tolerance to the antinociceptive effects of each other. F13-A and naloxone could inhibit the induction of such cross-tolerance. The molecular data showed that there was a significant downregulation of apelin receptors in chronic morphine-treated rats and vice versa. CONCLUSIONS Chronic administration of apelin or morphine induces analgesic cross-tolerance that may, in part, be mediated through receptor interactions and downregulation. The demonstrated efficacy of F13-A in these experiments highlights its potential as a novel target for improving pain management through the inhibition of the apelin/APJ signaling pathway, meriting further investigation.
Collapse
Affiliation(s)
- Elham Abbasloo
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeed Esmaeili-Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix Children's Hospital, Phoenix, Arizona
| |
Collapse
|
3
|
Sarzi-Puttini P, Pellegrino G, Giorgi V, Bongiovanni SF, Varrassi G, Di Lascio S, Fornasari D, Sirotti S, Di Carlo M, Salaffi F. "Inflammatory or non-inflammatory pain in inflammatory arthritis - How to differentiate it?". Best Pract Res Clin Rheumatol 2024; 38:101970. [PMID: 39004557 DOI: 10.1016/j.berh.2024.101970] [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: 04/30/2024] [Accepted: 05/04/2024] [Indexed: 07/16/2024]
Abstract
Pain is a significant issue in rheumatoid arthritis (RA) and psoriatic arthritis (PSA) and can have a negative impact on patients' quality of life. Despite optimal control of inflammatory disease, residual chronic pain remains a major unmet medical need in RA. Pain in RA can be secondary to inflammation but can also generate neuroendocrine responses that initiate neurogenic inflammation and enhance cytokine release, leading to persistent hyperalgesia. In addition to well-known cytokines such as TNFα and IL-6, other cytokines and the JAK-STAT pathway play a role in pain modulation and inflammation. The development of chronic pain in RA involves processes beyond inflammation or structural damage. Residual pain is often observed in patients even after achieving remission or low disease activity, suggesting the involvement of non-inflammatory and central sensitization mechanisms. Moreover, fibromyalgia syndrome (FMS) is prevalent in RA patients and may contribute to persistent pain. Factors such as depression, sleep disturbance, and pro-inflammatory cytokines may contribute to the development of fibromyalgia in RA. It is essential to identify and diagnose concomitant FMS in RA patients to better manage their symptoms. Further research is needed to unravel the complexities of pain in RA. Finally, recent studies have shown that JAK inhibitors effectively reduce residual pain in RA patients, suggesting pain-reducing effects independent of their anti-inflammatory properties.
Collapse
Affiliation(s)
- Piercarlo Sarzi-Puttini
- Rheumatology Unit, IRCCS Ospedale Galeazzi- Sant' Ambrogio, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.
| | - Greta Pellegrino
- Rheumatology Unit, IRCCS Ospedale Galeazzi- Sant' Ambrogio, Milan, Italy
| | - Valeria Giorgi
- Unità di Ricerca Clinica, Gruppo Ospedaliero Moncucco, Lugano, Svizzera
| | | | | | - Simona Di Lascio
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, Milan, Italy
| | - Diego Fornasari
- Department of Medical Biotechnology and Translational Medicine, Università Degli Studi di Milano, Milan, Italy
| | - Silvia Sirotti
- Rheumatology Unit, IRCCS Ospedale Galeazzi- Sant' Ambrogio, Milan, Italy
| | - Marco Di Carlo
- Rheumatology Clinic, Università Politecnica Delle Marche, Jesi, Ancona, Italy
| | - Fausto Salaffi
- Rheumatology Clinic, Università Politecnica Delle Marche, Jesi, Ancona, Italy
| |
Collapse
|
4
|
Yildiz Pehlivan D, Kara AY, Koyu A, Simsek F. Enhancing fentanyl antinociception and preventing tolerance with α-2 adrenoceptor agonists in rats. Behav Brain Res 2024; 457:114726. [PMID: 37865211 DOI: 10.1016/j.bbr.2023.114726] [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: 08/08/2023] [Revised: 10/06/2023] [Accepted: 10/17/2023] [Indexed: 10/23/2023]
Abstract
Fentanyl (FEN) is a potent opioid analgesic used for pain management. Opioid analgesic tolerance poses a significant challenge to the clinical utility of opioid agonists. Preventing the development of tolerance to opioid analgesia is crucial for improving its efficacy and safety. The noradrenergic system is involved in pain regulation. This study examined the effects of α-2 adrenoceptor (AR) agonists, dexmedetomidine (DEX), and xylazine (XYL) on FEN tolerance and antinociception, and their impact on μ-opioid receptor (MOR) expression in the posterior horn of the spinal cord (SC). Male rats were divided into six groups and treated with different drug combinations for three consecutive days. Analgesia tests and motor performance assessments were conducted, followed by SC analysis using immunohistochemistry (IHC). Analgesia tests revealed the development of FEN tolerance on the second day, but the groups receiving combined drugs did not develop tolerance. Instead, FEN antinociception was enhanced, with a prolonged duration of its effects. None of the drugs caused sedation or motor impairment, and SC morphology appeared normal. MOR expression levels did not differ significantly between the groups based on IHC analysis. These findings suggest that changes in the secondary messenger system may play a role in the early development of FEN tolerance. Combining drugs can prevent tolerance, while enhancing FEN's antinociceptive effects. These results have promising implications for chronic pain management; however, further research is needed to explore the molecular effects of α-2 AR agonists on FEN tolerance. Overall, this study sheds light on the mechanism of FEN tolerance and identifies potential avenues for future research.
Collapse
Affiliation(s)
- Deniz Yildiz Pehlivan
- Izmir Katip Celebi University, Faculty of Medicine, Department of Physiology, Izmir, Turkey
| | - Ali Yucel Kara
- Izmir Katip Celebi University, Faculty of Medicine, Department of Physiology, Izmir, Turkey.
| | - Ahmet Koyu
- Izmir Katip Celebi University, Faculty of Medicine, Department of Physiology, Izmir, Turkey
| | - Fatma Simsek
- Izmir Katip Celebi University, Faculty of Medicine, Department of Histology and Embryology, Izmir, Turkey
| |
Collapse
|
5
|
Alami K, Ghasemi E, Semnanian S, Azizi H. Adolescent morphine exposure changes the endogenous vlPAG opioid response to inflammatory pain in rats. Dev Psychobiol 2024; 66:e22447. [PMID: 38131239 DOI: 10.1002/dev.22447] [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: 05/26/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023]
Abstract
Adolescence is one of the most critical periods for brain development, and exposure to morphine during this period can have long-life effects on pain-related behaviors. The opioid system in the periaqueductal gray (PAG) is highly vulnerable to drug exposure. However, the impact of adolescent morphine exposure (AME) on the endogenous opioid system in the PAG is currently unknown. This study aims to investigate the long-lasting effects of AME on the endogenous opioid system and its involvement in altering nociceptive behaviors. Adolescent rats were given escalating doses of morphine (2.5-25 mg/kg, subcutaneous) or an equal volume of saline twice daily for 10 consecutive days (PND 31-40). After a 30-day washout period, adult rats underwent formalin tests following microinjection of morphine, naloxone, or saline into the ventrolateral PAG (vlPAG) region. The results indicated that morphine microinjection into the vlPAG of the adolescent morphine-treated group significantly reduced the nociceptive score. However, the analgesic response to morphine in this group was significantly lower compared to the saline-treated group during adolescence. Additionally, the nociceptive score significantly increased following naloxone but not saline microinjection into the vlPAG of the saline-treated group during adolescence, rather than the morphine-treated one. These findings indicate that AME has long-lasting effects on the endogenous opioid system in the vlPAG, which can consequently alter behaviors related to inflammatory pain in adulthood.
Collapse
Affiliation(s)
- Kawsar Alami
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Elmira Ghasemi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Semnanian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
6
|
Santi MD, Zhang M, Liu N, Viet CT, Xie T, Jensen DD, Amit M, Pan H, Ye Y. Repurposing EGFR Inhibitors for Oral Cancer Pain and Opioid Tolerance. Pharmaceuticals (Basel) 2023; 16:1558. [PMID: 38004424 PMCID: PMC10674507 DOI: 10.3390/ph16111558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Oral cancer pain remains a significant public health concern. Despite the development of improved treatments, pain continues to be a debilitating clinical feature of the disease, leading to reduced oral mobility and diminished quality of life. Opioids are the gold standard treatment for moderate-to-severe oral cancer pain; however, chronic opioid administration leads to hyperalgesia, tolerance, and dependence. The aim of this review is to present accumulating evidence that epidermal growth factor receptor (EGFR) signaling, often dysregulated in cancer, is also an emerging signaling pathway critically involved in pain and opioid tolerance. We presented preclinical and clinical data to demonstrate how repurposing EGFR inhibitors typically used for cancer treatment could be an effective pharmacological strategy to treat oral cancer pain and to prevent or delay the development of opioid tolerance. We also propose that EGFR interaction with the µ-opioid receptor and glutamate N-methyl-D-aspartate receptor could be two novel downstream mechanisms contributing to pain and morphine tolerance. Most data presented here support that repurposing EGFR inhibitors as non-opioid analgesics in oral cancer pain is promising and warrants further research.
Collapse
Affiliation(s)
- Maria Daniela Santi
- Translational Research Center, College of Dentistry, New York University, New York, NY 10010, USA; (M.D.S.); (M.Z.); (N.L.); (D.D.J.)
- Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY 10010, USA
| | - Morgan Zhang
- Translational Research Center, College of Dentistry, New York University, New York, NY 10010, USA; (M.D.S.); (M.Z.); (N.L.); (D.D.J.)
- Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY 10010, USA
| | - Naijiang Liu
- Translational Research Center, College of Dentistry, New York University, New York, NY 10010, USA; (M.D.S.); (M.Z.); (N.L.); (D.D.J.)
- Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY 10010, USA
| | - Chi T. Viet
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Tongxin Xie
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.X.); (M.A.)
| | - Dane D. Jensen
- Translational Research Center, College of Dentistry, New York University, New York, NY 10010, USA; (M.D.S.); (M.Z.); (N.L.); (D.D.J.)
- Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY 10010, USA
| | - Moran Amit
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (T.X.); (M.A.)
| | - Huilin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Yi Ye
- Translational Research Center, College of Dentistry, New York University, New York, NY 10010, USA; (M.D.S.); (M.Z.); (N.L.); (D.D.J.)
- Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY 10010, USA
| |
Collapse
|
7
|
Todorov P, Georgieva S, Tchekalarova J, Peneva P, Mateeva P, Assenov B, Dzhambazova E, Pechlivanova D. Synthesis, characterization and evaluation of anti-hyperalgesia, anticonvulsant and antioxidant activity of novel VV-hemorphin-5 analogs. Arch Pharm (Weinheim) 2023; 356:e2300267. [PMID: 37532631 DOI: 10.1002/ardp.202300267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/04/2023] [Accepted: 07/17/2023] [Indexed: 08/04/2023]
Abstract
Two series of new VV-hemorphin-5 analogs with structures Val-Val-Tyr-Xxx-Trp-Thr-Gln-NH2 and Adam-Val-Val-Tyr-Xxx-Trp-Thr-Gln-NH2 , where Xxx is Ac5c (1-aminocyclopentane-1-carboxylic acid), Ac6c (1-aminocyclohexane-1-carboxylic acid), Ac7c (1-aminocycloheptane-1-carboxylic acid), and Adam is the low-molecular-weight lipophilic adamantyl building block, were synthesized, characterized electrochemically and evaluated for antioxidant, anti-hyperalgesia, and anticonvulsant activity. The design of the compounds followed the strategy to improve the propensity for aqueous solubility and/or to increase their affinity for the target receptor or enzyme. The partition coefficient value shows that the peptide scaffold goes from hydrophilic to lipophilic with the increasing size of the cycloalkane ring and even more with the introduction of the adamantane. The peptides C5-V and C7-V were the only analogs that provoked an immediate antinociceptive effect changing the mechanical pain threshold. The six new peptide analogs produced a significant and long-lasting carrageenan model of inflammatory pain in rats. While the adamantane hemorphin analog Ad7-V was the only compound with the potency to suppress psychomotor seizures in the 6-Hz test, the C6-V and Ad6-V exhibited protective activity against the seizure spread in the maximal electroshock seizure test in mice. The active analogs did not show neurotoxicity or sedative effects. Our results revealed a structure-related specific activity of a newly designed hemorphin analog that could be used as a template for future modification and preparation of compounds with potential analgesic and anticonvulsant activity.
Collapse
Affiliation(s)
- Petar Todorov
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, Sofia, Bulgaria
| | - Stela Georgieva
- Department of Analytical Chemistry, University of Chemical Technology and Metallurgy, Sofia, Bulgaria
| | - Jana Tchekalarova
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, Sofia, Bulgaria
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Petia Peneva
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, Sofia, Bulgaria
| | - Polina Mateeva
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Borislav Assenov
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
- Faculty of Medicine, Sofia University "St. Kliment Ohridski", Sofia, Bulgaria
| | - Elena Dzhambazova
- Faculty of Medicine, Sofia University "St. Kliment Ohridski", Sofia, Bulgaria
| | - Daniela Pechlivanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
- Faculty of Medicine, Sofia University "St. Kliment Ohridski", Sofia, Bulgaria
| |
Collapse
|
8
|
Tucker MH, Tiwari P, Carter BS. The physiology, assessment, and treatment of neonatal pain. Semin Fetal Neonatal Med 2023; 28:101465. [PMID: 37236846 DOI: 10.1016/j.siny.2023.101465] [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: 05/28/2023]
Abstract
Studies have clearly shown that development of pain receptors starts as early as 20-weeks' gestation. Despite contrary belief, the human fetus develops a similar number of receptive pain fibers as seen in adults. These receptors' maturation is based on response to sensory stimuli received after birth which makes the NICU a critical place for developing central nervous system's pain perception. In practice, the assessment of pain relies mostly on bedside staff. In this review we will discuss the various developing features of pain pathways in the neonatal brain and the modification of pain perception secondary to various interactions immediately after birth. We also discuss the various tools utilized in the NICU for pain assessment that rely on physiological and behavioral patterns. Finally, we address the management of pain in the NICU by either pharmacological or non-pharmacological intervention while highlighting potential benefits, disadvantages, and situations where one may be preferred over another.
Collapse
Affiliation(s)
- Megan H Tucker
- Department of Pediatrics, Division of Neonatology, Children's Mercy-Kansas City, Kansas City, MO, USA
| | - Priya Tiwari
- Department of Pediatrics, Division of Neonatology, Children's Mercy-Kansas City, Kansas City, MO, USA
| | - Brian S Carter
- Department of Pediatrics, Division of Neonatology, Children's Mercy-Kansas City, Kansas City, MO, USA; Bioethics Center, Children's Mercy-Kansas City, Kansas City, MO, USA; Department of Medical Humanities & Bioethics, University of Missouri-Kansas City, Kansas City, MO, USA.
| |
Collapse
|
9
|
Nguyen E, Grajales-Reyes JG, Gereau RW, Ross SE. Cell type-specific dissection of sensory pathways involved in descending modulation. Trends Neurosci 2023; 46:539-550. [PMID: 37164868 PMCID: PMC10836406 DOI: 10.1016/j.tins.2023.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/22/2023] [Accepted: 04/07/2023] [Indexed: 05/12/2023]
Abstract
Decades of research have suggested that stimulation of supraspinal structures, such as the periaqueductal gray (PAG) and rostral ventromedial medulla (RVM), inhibits nocifensive responses to noxious stimulation through a process known as descending modulation. Electrical stimulation and pharmacologic manipulations of the PAG and RVM identified transmitters and neuronal firing patterns that represented distinct cell types. Advances in mouse genetics, in vivo imaging, and circuit tracing methods, in addition to chemogenetic and optogenetic approaches, allowed the characterization of the cells and circuits involved in descending modulation in further detail. Recent work has revealed the importance of PAG and RVM neuronal cell types in the descending modulation of pruriceptive as well as nociceptive behaviors, underscoring their roles in coordinating complex behavioral responses to sensory input. This review summarizes how new technical advances that enable cell type-specific manipulation and recording of neuronal activity have supported, as well as expanded, long-standing views on descending modulation.
Collapse
Affiliation(s)
- Eileen Nguyen
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jose G Grajales-Reyes
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Robert W Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine in St Louis, St Louis, MO 63110, USA
| | - Sarah E Ross
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
10
|
Lynch N, Lima JD, Spinieli RL, Kaur S. Opioids, sleep, analgesia and respiratory depression: Their convergence on Mu (μ)-opioid receptors in the parabrachial area. Front Neurosci 2023; 17:1134842. [PMID: 37090798 PMCID: PMC10117663 DOI: 10.3389/fnins.2023.1134842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/21/2023] [Indexed: 04/25/2023] Open
Abstract
Opioids provide analgesia, as well as modulate sleep and respiration, all by possibly acting on the μ-opioid receptors (MOR). MOR's are ubiquitously present throughout the brain, posing a challenge for understanding the precise anatomical substrates that mediate opioid induced respiratory depression (OIRD) that ultimately kills most users. Sleep is a major modulator not only of pain perception, but also for changing the efficacy of opioids as analgesics. Therefore, sleep disturbances are major risk factors for developing opioid overuse, withdrawal, poor treatment response for pain, and addiction relapse. Despite challenges to resolve the neural substrates of respiratory malfunctions during opioid overdose, two main areas, the pre-Bötzinger complex (preBötC) in the medulla and the parabrachial (PB) complex have been implicated in regulating respiratory depression. More recent studies suggest that it is mediation by the PB that causes OIRD. The PB also act as a major node in the upper brain stem that not only receives input from the chemosensory areas in medulla, but also receives nociceptive information from spinal cord. We have previously shown that the PB neurons play an important role in mediating arousal from sleep in response to hypercapnia by its projections to the forebrain arousal centers, and it may also act as a major relay for the pain stimuli. However, due to heterogeneity of cells in the PB, their precise roles in regulating, sleep, analgesia, and respiratory depression, needs addressing. This review sheds light on interactions between sleep and pain, along with dissecting the elements that adversely affects respiration.
Collapse
Affiliation(s)
| | | | | | - Satvinder Kaur
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Vamvakas A, Lawn T, Veronese M, Williams SCR, Tsougos I, Howard MA. Neurotransmitter receptor densities are associated with changes in regional Cerebral blood flow during clinical ongoing pain. Hum Brain Mapp 2022; 43:5235-5249. [PMID: 35796178 PMCID: PMC9812236 DOI: 10.1002/hbm.25999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/07/2022] [Accepted: 06/19/2022] [Indexed: 01/15/2023] Open
Abstract
Arterial spin labelling (ASL) plays an increasingly important role in neuroimaging pain research but does not provide molecular insights regarding how regional cerebral blood flow (rCBF) relates to underlying neurotransmission. Here, we integrate ASL with positron emission tomography (PET) and brain transcriptome data to investigate the molecular substrates of rCBF underlying clinically relevant pain states. Two data sets, representing acute and chronic ongoing pain respectively, were utilised to quantify changes in rCBF; one examining pre-surgical versus post-surgical pain, and the second comparing patients with painful hand Osteoarthritis to a group of matched controls. We implemented a whole-brain spatial correlation analysis to explore associations between change in rCBF (ΔCBF) and neurotransmitter receptor distributions derived from normative PET templates. Additionally, we utilised transcriptomic data from the Allen Brain Atlas to inform distributions of receptor expression. Both datasets presented significant correlations of ΔCBF with the μ-opioid and dopamine-D2 receptor expressions, which play fundamental roles in brain activity associated with pain experiences. ΔCBF also correlated with the gene expression distributions of several receptors involved in pain processing. Overall, this is the first study illustrating the molecular basis of ongoing pain ASL indices and emphasises the potential of rCBF as a biomarker in pain research.
Collapse
Affiliation(s)
- Alexandros Vamvakas
- Medical Physics Department, Medical SchoolUniversity of ThessalyLarisaGreece
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Timothy Lawn
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Steven C. R. Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Ioannis Tsougos
- Medical Physics Department, Medical SchoolUniversity of ThessalyLarisaGreece
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Matthew A. Howard
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| |
Collapse
|
12
|
Toma W, Paris JJ, Warncke UO, Nass SR, Caillaud M, McKiver B, Ondo O, Bagdas D, Bigbee J, Knapp PE, Hauser KF, Damaj MI. Persistent sensory changes and sex differences in transgenic mice conditionally expressing HIV-1 Tat regulatory protein. Exp Neurol 2022; 358:114226. [PMID: 36096180 PMCID: PMC10053560 DOI: 10.1016/j.expneurol.2022.114226] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 08/19/2022] [Accepted: 09/06/2022] [Indexed: 11/04/2022]
Abstract
HIV-associated sensory neuropathies (HIV-SN) are prevalent in >50% of patients aged over 45 years many of which report moderate to severe chronic pain. Previous preclinical studies have investigated the mechanisms by which HIV-1 causes sensory neuropathies and pain-like behaviors. The aim of the present study is to delineate the role of chronic HIV-1 trans-activator of transcription protein (Tat) exposure in the development of neuropathy in mice. The temporal effects of conditional Tat expression on the development of hypersensitivity to mechanical (von Frey filaments) and thermal (heat or cold) stimuli were tested in male and female mice that transgenically expressed HIV-1 Tat in a doxycycline-inducible manner. Inducing Tat expression produced an allodynic response to mechanical or cold (but not heat) stimuli that respectively persisted for at least 23-weeks (mechanical hypersensitivity) or at least 8-weeks (cold hypersensitivity). Both allodynic states were greater in magnitude among females, compared to males, and mechanical increased hypersensitivity progressively in females over time. Acute morphine or gabapentin treatment partly attenuated allodynia in males, but not females. Irrespective of sex, Tat reduced intraepidermal nerve fiber density, the mean amplitude of sensory nerve action potentials (but not conductance), engagement in some pain-related ethological behaviors (cage-hanging and rearing), and down-regulated PPAR-α gene expression in lumbar spinal cord while upregulating TNF-α expression in dorsal root ganglion. Taken together, these data reveal fundamental sex differences in mechanical and cold hypersensitivity in response to Tat and demonstrate the intractable nature in female mice to current therapeutics. Understanding the role of Tat in these pathologies may aid the design of future therapies aimed at mitigating the peripheral sensory neuropathies that accompany neuroHIV.
Collapse
Affiliation(s)
- Wisam Toma
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jason J Paris
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, USA; Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, USA
| | - Urszula O Warncke
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Sara R Nass
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Martial Caillaud
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Bryan McKiver
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Olivia Ondo
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Deniz Bagdas
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - John Bigbee
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - Pamela E Knapp
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA; Institute for Drug and Alcohol Studies, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Department of Anatomy and Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA; Institute for Drug and Alcohol Studies, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; Translational Research Initiative for Pain and Neuropathy, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
13
|
Alencar Neto JFD, Oliveira Júnior RR, Dias AJA, Ferreira Neto ODC, Lira ACSD, Bastos BF, Rocha ML, Marques LFF, Queiroga PHM, Lemos NB, Melo Neto FDP, Lopes AAP, Bem Junior LS, Azevedo Filho HRCD. Neurophysiology and neuroanatomy of spinal cord electrode stimulation for the treatment of chronic pain – State of art. INTERDISCIPLINARY NEUROSURGERY 2022. [DOI: 10.1016/j.inat.2022.101660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
14
|
Lim SY, Cengiz P. Opioid tolerance and opioid-induced hyperalgesia: Is TrkB modulation a potential pharmacological solution? Neuropharmacology 2022; 220:109260. [PMID: 36165856 DOI: 10.1016/j.neuropharm.2022.109260] [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: 03/14/2022] [Revised: 06/23/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022]
Abstract
Opioids are widely prescribed for moderate to severe pain in patients with acute illness, cancer pain, and chronic noncancer pain. However, long-term opioid use can cause opioid tolerance and opioid-induced hyperalgesia (OIH), contributing to the opioid misuse and addiction crisis. Strategies to mitigate opioid tolerance and OIH are needed to reduce opioid use and its sequelae. Currently, there are few effective pharmacological strategies that reduce opioid tolerance and OIH. The intrinsic tyrosine kinase receptor B (TrkB) ligand, brain-derived neurotrophic factor (BDNF), has been shown to modulate pain. The BDNF-TrkB signaling plays a role in initiating and sustaining elevated pain sensitivity; however, increasing evidence has shown that BDNF and 7,8-dihydroxyflavone (7,8-DHF), a potent blood-brain barrier-permeable ligand to TrkB, exert neuroprotective, anti-inflammatory, and antioxidant effects that may protect against opioid tolerance and OIH. As such, TrkB signaling may be an important therapeutic avenue in opioid tolerance and OIH. Here, we review 1) the mechanisms of pain, opioid analgesia, opioid tolerance, and OIH; 2) the role of BDNF-TrkB in pain modulation; and 3) the neuroprotective effects of 7,8-DHF and their implications for opioid tolerance and OIH.
Collapse
Affiliation(s)
- Sin Yin Lim
- Pharmacy Practice and Translational Research Division, University of Wisconsin-Madison School of Pharmacy, Madison, WI, United States.
| | - Pelin Cengiz
- Department of Pediatrics, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States; Waisman Center, University of Wisconsin-Madison, United States.
| |
Collapse
|
15
|
Higginbotham JA, Markovic T, Massaly N, Morón JA. Endogenous opioid systems alterations in pain and opioid use disorder. Front Syst Neurosci 2022; 16:1014768. [PMID: 36341476 PMCID: PMC9628214 DOI: 10.3389/fnsys.2022.1014768] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022] Open
Abstract
Decades of research advances have established a central role for endogenous opioid systems in regulating reward processing, mood, motivation, learning and memory, gastrointestinal function, and pain relief. Endogenous opioid systems are present ubiquitously throughout the central and peripheral nervous system. They are composed of four families, namely the μ (MOPR), κ (KOPR), δ (DOPR), and nociceptin/orphanin FQ (NOPR) opioid receptors systems. These receptors signal through the action of their endogenous opioid peptides β-endorphins, dynorphins, enkephalins, and nociceptins, respectfully, to maintain homeostasis under normal physiological states. Due to their prominent role in pain regulation, exogenous opioids-primarily targeting the MOPR, have been historically used in medicine as analgesics, but their ability to produce euphoric effects also present high risks for abuse. The ability of pain and opioid use to perturb endogenous opioid system function, particularly within the central nervous system, may increase the likelihood of developing opioid use disorder (OUD). Today, the opioid crisis represents a major social, economic, and public health concern. In this review, we summarize the current state of the literature on the function, expression, pharmacology, and regulation of endogenous opioid systems in pain. Additionally, we discuss the adaptations in the endogenous opioid systems upon use of exogenous opioids which contribute to the development of OUD. Finally, we describe the intricate relationship between pain, endogenous opioid systems, and the proclivity for opioid misuse, as well as potential advances in generating safer and more efficient pain therapies.
Collapse
Affiliation(s)
- Jessica A. Higginbotham
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
- Pain Center, Washington University in St. Louis, St. Louis, MO, United States
- School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Tamara Markovic
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nicolas Massaly
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
- Pain Center, Washington University in St. Louis, St. Louis, MO, United States
- School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Jose A. Morón
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States
- Pain Center, Washington University in St. Louis, St. Louis, MO, United States
- School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, United States
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
16
|
Afrose L, McDermott MV, Bhuiyan AI, Pathak SK, Bobeck EN. GPR171 activation regulates morphine tolerance but not withdrawal in a test-dependent manner in mice. Behav Pharmacol 2022; 33:442-451. [PMID: 35942845 PMCID: PMC9477863 DOI: 10.1097/fbp.0000000000000692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/07/2022] [Indexed: 11/26/2022]
Abstract
A newly deorphanized G protein-coupled receptor, GPR171, is found to be highly expressed within the periaqueductal gray, a pain-modulating region in the brain. Our recent research has shown that a GPR171 agonist increases morphine antinociception in male mice and opioid signaling in vitro . The objective of this study was to evaluate the effects of combination treatment in females as well as whether chronic treatment can be used without exacerbating morphine-induced tolerance and withdrawal in female and male mice. Our results demonstrate that activation of GPR171 with an agonist attenuates morphine tolerance in both female and male mice on the tail-flick test, but not the hotplate test. Importantly, the GPR171 agonist in combination with morphine does not exacerbate morphine-induced tolerance and withdrawal during long-term morphine treatment. Taken together, these data suggest that the GPR171 agonist may be combined with morphine to maintain antinociception while reducing the dose of morphine and therefore reducing side effects and abuse liability. The outcome of this study is clearly an important step toward understanding the functional interactions between opioid receptors and GPR171 and developing safer therapeutics for long-term pain management.
Collapse
Affiliation(s)
| | - Max V. McDermott
- Department of Biology
- Interdisciplinary Neuroscience Program, Utah State University, Logan, Utah
| | - Ashif I. Bhuiyan
- Chemistry and Biochemistry Department, Queens College of The City University of New York, Flushing, New York
- Chemistry Doctoral Program
| | - Sanjai K. Pathak
- Chemistry and Biochemistry Department, Queens College of The City University of New York, Flushing, New York
- Chemistry Doctoral Program
- Biochemistry Doctoral Program, The Graduate Center of The City University of New York, New York
| | - Erin N. Bobeck
- Department of Biology
- Interdisciplinary Neuroscience Program, Utah State University, Logan, Utah
| |
Collapse
|
17
|
Carranza-Aguilar CJ, Hernández-Mendoza A, Mejias-Aponte C, Rice KC, Morales M, González-Espinosa C, Cruz SL. Morphine and Fentanyl Repeated Administration Induces Different Levels of NLRP3-Dependent Pyroptosis in the Dorsal Raphe Nucleus of Male Rats via Cell-Specific Activation of TLR4 and Opioid Receptors. Cell Mol Neurobiol 2022; 42:677-694. [PMID: 32926257 PMCID: PMC11441185 DOI: 10.1007/s10571-020-00957-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023]
Abstract
Morphine promotes neuroinflammation after NOD-like receptor protein 3 (NLRP3) oligomerization in glial cells, but the capacity of other opioids to induce neuroinflammation and its relationship to the development of analgesic tolerance is unknown. We studied the effects of morphine and fentanyl on NLRP3 inflammasome activation in glial and neuronal cells in the dorsal raphe nucleus (DRN), a region involved in pain regulation. Male Wistar rats received i.p. injections of morphine (10 mg/kg) or fentanyl (0.1 mg/kg) 3 × daily for 7 days and were tested for nociception. Two hours after the last (19th) administration, we analyzed NLRP3 oligomerization, caspase-1 activation and gasdermin D-N (GSDMD-N) expression in microglia (CD11b positive cells), astrocytes (GFAP-positive cells) and neurons (NeuN-positive cells). Tolerance developed to both opioids, but only fentanyl produced hyperalgesia. Morphine and fentanyl activated NLRP3 inflammasome in astrocytes and serotonergic (TPH-2-positive) neurons, but fentanyl effects were more pronounced. Both opioids increased GFAP and CD11b immunoreactivity, caspase-1 and GSDMD activation, indicating pyroptotic cell death. The opioid receptor antagonist (-)-naloxone, but not the TLR4 receptor antagonist (+)-naloxone, prevented microglia activation and NLRP3 oligomerization. Only (+)-naloxone prevented astrocytes' activation. The anti-inflammatory agent minocycline and the NLRP3 inhibitor MCC950 delayed tolerance to morphine and fentanyl antinociception and prevented fentanyl-induced hyperalgesia. MCC950 also prevented opioid-induced NLRP3 oligomerization. In conclusion, morphine and fentanyl differentially induce cell-specific activation of NLRP3 inflammasome and pyroptosis in the DRN through TLR4 receptors in astrocytes and through opioid receptors in neurons, indicating that neuroinflammation is involved in opioid-induced analgesia and fentanyl-induced hyperalgesia after repeated administrations.
Collapse
Affiliation(s)
- César J Carranza-Aguilar
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav, IPN), Sede Sur, Calzada de los Tenorios No. 235, Col. Granjas Coapa, Del. Tlalpan, C.P. 14330, Ciudad de México, Mexico
| | - Araceli Hernández-Mendoza
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav, IPN), Sede Sur, Calzada de los Tenorios No. 235, Col. Granjas Coapa, Del. Tlalpan, C.P. 14330, Ciudad de México, Mexico
| | - Carlos Mejias-Aponte
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard Suite 200, Baltimore, MD, 21224, USA
| | - Kenner C Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, Intramural Research Program, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Marisela Morales
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard Suite 200, Baltimore, MD, 21224, USA
| | - Claudia González-Espinosa
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav, IPN), Sede Sur, Calzada de los Tenorios No. 235, Col. Granjas Coapa, Del. Tlalpan, C.P. 14330, Ciudad de México, Mexico
| | - Silvia L Cruz
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav, IPN), Sede Sur, Calzada de los Tenorios No. 235, Col. Granjas Coapa, Del. Tlalpan, C.P. 14330, Ciudad de México, Mexico.
| |
Collapse
|
18
|
Mungoven TJ, Marciszewski KK, Macefield VG, Macey PM, Henderson LA, Meylakh N. Alterations in pain processing circuitries in episodic migraine. J Headache Pain 2022; 23:9. [PMID: 35033014 PMCID: PMC8903545 DOI: 10.1186/s10194-021-01381-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/28/2021] [Indexed: 01/13/2023] Open
Abstract
Background The precise underlying mechanisms of migraine remain unknown. Although we have previously shown acute orofacial pain evoked changes within the brainstem of individuals with migraine, we do not know if these brainstem alterations are driven by changes in higher cortical regions. The aim of this investigation is to extend our previous investigation to determine if higher brain centers display altered activation patterns and connectivity in migraineurs during acute orofacial noxious stimuli. Methods Functional magnetic resonance imaging was performed in 29 healthy controls and 25 migraineurs during the interictal and immediately (within 24-h) prior to migraine phases. We assessed activation of higher cortical areas during noxious orofacial heat stimulation using a thermode device and assessed whole scan and pain-related changes in connectivity. Results Despite similar overall pain intensity ratings between all three groups, migraineurs in the group immediately prior to migraine displayed greater activation of the ipsilateral nucleus accumbens, the contralateral ventrolateral prefrontal cortex and two clusters in the dorsolateral prefrontal cortex (dlPFC). Reduced whole scan dlPFC [Z + 44] connectivity with cortical/subcortical and brainstem regions involved in pain modulation such as the putamen and primary motor cortex was demonstrated in migraineurs. Pain-related changes in connectivity of the dlPFC and the hypothalamus immediately prior to migraine was also found to be reduced with brainstem pain modulatory areas such as the rostral ventromedial medulla and dorsolateral pons. Conclusions These data reveal that the modulation of brainstem pain modulatory areas by higher cortical regions may be aberrant during pain and these alterations in this descending pain modulatory pathway manifests exclusively prior to the development of a migraine attack.
Collapse
Affiliation(s)
- Tiffani J Mungoven
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Kasia K Marciszewski
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | | | - Paul M Macey
- UCLA School of Nursing and Brain Research Institute, University of California, Los Angeles, California, 90095, USA
| | - Luke A Henderson
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, Camperdown, NSW, 2050, Australia.
| | - Noemi Meylakh
- School of Medical Sciences (Neuroscience), Brain and Mind Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| |
Collapse
|
19
|
Kulik K, Żyżyńska-Granica B, Kowalczyk A, Kurowski P, Gajewska M, Bujalska-Zadrożny M. Magnesium and Morphine in the Treatment of Chronic Neuropathic Pain-A Biomedical Mechanism of Action. Int J Mol Sci 2021; 22:13599. [PMID: 34948397 PMCID: PMC8707930 DOI: 10.3390/ijms222413599] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023] Open
Abstract
The effectiveness of opioids in the treatment of neuropathic pain is limited. It was demonstrated that magnesium ions (Mg2+), physiological antagonists of N-methyl-D-aspartate receptor (NMDAR), increase opioid analgesia in chronic pain. Our study aimed to determine the molecular mechanism of this action. Early data indicate the cross-regulation of µ opioid receptor (MOR) and NMDAR in pain control. Morphine acting on MOR stimulates protein kinase C (PKC), while induction of NMDAR recruits protein kinase A (PKA), leading to a disruption of the MOR-NMDAR complex and promoting functional changes in receptors. The mechanical Randall-Selitto test was used to assess the effect of chronic Mg2+ and morphine cotreatment on streptozotocin-induced hyperalgesia in Wistar rats. The level of phosphorylated NMDAR NR1 subunit (pNR1) and phosphorylated MOR (pMOR) in the periaqueductal gray matter was determined with the Western blot method. The activity of PKA and PKC was examined by standard enzyme immunoassays. The experiments showed a reduction in hyperalgesia after coadministration of morphine (5 mg/kg intraperitoneally) and Mg2+ (40 mg/kg intraperitoneally). Mg2+ administered alone significantly decreased the level of pNR1, pMOR, and activity of both tested kinases. The results suggest that blocking NMDAR signaling by Mg2+ restores the MOR-NMDAR complex and thus enables morphine analgesia in neuropathic rats.
Collapse
Affiliation(s)
- Kamila Kulik
- Centre for Preclinical Research and Technology, Department of Pharmacodynamics, Medical University of Warsaw, Banacha 1b Str., 02-097 Warsaw, Poland; (B.Ż.-G.); (A.K.); (P.K.); (M.B.-Z.)
| | - Barbara Żyżyńska-Granica
- Centre for Preclinical Research and Technology, Department of Pharmacodynamics, Medical University of Warsaw, Banacha 1b Str., 02-097 Warsaw, Poland; (B.Ż.-G.); (A.K.); (P.K.); (M.B.-Z.)
- Chair and Department of Biochemistry, Medical University of Warsaw, Banacha 1 Str., 02-097 Warsaw, Poland
| | - Agnieszka Kowalczyk
- Centre for Preclinical Research and Technology, Department of Pharmacodynamics, Medical University of Warsaw, Banacha 1b Str., 02-097 Warsaw, Poland; (B.Ż.-G.); (A.K.); (P.K.); (M.B.-Z.)
| | - Przemysław Kurowski
- Centre for Preclinical Research and Technology, Department of Pharmacodynamics, Medical University of Warsaw, Banacha 1b Str., 02-097 Warsaw, Poland; (B.Ż.-G.); (A.K.); (P.K.); (M.B.-Z.)
| | - Małgorzata Gajewska
- Department of Physiological Sciences, Warsaw University of Life Sciences, Nowoursynowska 159 Str., 02-776 Warsaw, Poland;
| | - Magdalena Bujalska-Zadrożny
- Centre for Preclinical Research and Technology, Department of Pharmacodynamics, Medical University of Warsaw, Banacha 1b Str., 02-097 Warsaw, Poland; (B.Ż.-G.); (A.K.); (P.K.); (M.B.-Z.)
| |
Collapse
|
20
|
Ram A, Edwards TM, McCarty A, McDermott MV, Bobeck EN. Morphine-induced kinase activation and localization in the periaqueductal gray of male and female mice. J Neurochem 2021; 159:590-602. [PMID: 34499746 DOI: 10.1111/jnc.15506] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/17/2022]
Abstract
Morphine is a potent opioid analgesic with high propensity for the development of antinociceptive tolerance. Morphine antinociception and tolerance are partially regulated by the midbrain ventrolateral periaqueductal gray (vlPAG). However, the majority of research evaluating mu-opioid receptor signaling has focused on males. Here, we investigate kinase activation and localization patterns in the vlPAG following acute and chronic morphine treatment in both sexes. Male and female mice developed rapid antinociceptive tolerance to morphine (10 mg/kg i.p.) on the hot plate assay, but tolerance did not develop in males on the tail flick assay. Quantitative fluorescence immunohistochemistry was used to map and evaluate the activation of extracellular signal-regulated kinase 1/2 (ERK 1/2), protein kinase-C (PKC), and protein kinase-A (PKA). We observed significantly greater phosphorylated ERK 1/2 in the vlPAG of chronic morphine-treated animals which co-localized with the endosomal marker, Eea1. We note that pPKC is significantly elevated in the vlPAG of both sexes following chronic morphine treatment. We also observed that although PKA activity is elevated following chronic morphine treatment in both sexes, there is a significant reduction in the nuclear translocation of its phosphorylated substrate. Taken together, this study demonstrates increased activation of ERK 1/2, PKC, and PKA in response to repeated morphine treatment. The study opens avenues to explore the impact of chronic morphine treatment on G-protein signaling and kinase nuclear transport.
Collapse
Affiliation(s)
- Akila Ram
- Department of Biology, Utah State University, Logan, Utah, USA
| | | | - Ashley McCarty
- Department of Biology, Utah State University, Logan, Utah, USA
| | - Max V McDermott
- Department of Biology, Utah State University, Logan, Utah, USA
- Interdisciplinary Neuroscience Program, Utah State University, Logan, Utah, USA
| | - Erin N Bobeck
- Department of Biology, Utah State University, Logan, Utah, USA
- Interdisciplinary Neuroscience Program, Utah State University, Logan, Utah, USA
| |
Collapse
|
21
|
Király K, Karádi DÁ, Zádor F, Mohammadzadeh A, Galambos AR, Balogh M, Riba P, Tábi T, Zádori ZS, Szökő É, Fürst S, Al-Khrasani M. Shedding Light on the Pharmacological Interactions between μ-Opioid Analgesics and Angiotensin Receptor Modulators: A New Option for Treating Chronic Pain. Molecules 2021; 26:6168. [PMID: 34684749 PMCID: PMC8537077 DOI: 10.3390/molecules26206168] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/20/2022] Open
Abstract
The current protocols for neuropathic pain management include µ-opioid receptor (MOR) analgesics alongside other drugs; however, there is debate on the effectiveness of opioids. Nevertheless, dose escalation is required to maintain their analgesia, which, in turn, contributes to a further increase in opioid side effects. Finding novel approaches to effectively control chronic pain, particularly neuropathic pain, is a great challenge clinically. Literature data related to pain transmission reveal that angiotensin and its receptors (the AT1R, AT2R, and MAS receptors) could affect the nociception both in the periphery and CNS. The MOR and angiotensin receptors or drugs interacting with these receptors have been independently investigated in relation to analgesia. However, the interaction between the MOR and angiotensin receptors has not been excessively studied in chronic pain, particularly neuropathy. This review aims to shed light on existing literature information in relation to the analgesic action of AT1R and AT2R or MASR ligands in neuropathic pain conditions. Finally, based on literature data, we can hypothesize that combining MOR agonists with AT1R or AT2R antagonists might improve analgesia.
Collapse
MESH Headings
- Analgesics/pharmacology
- Analgesics, Opioid/pharmacology
- Animals
- Chronic Pain/drug therapy
- Humans
- Neuralgia/drug therapy
- Nociception/drug effects
- Pain Management/methods
- Proto-Oncogene Mas
- Receptors, Angiotensin/drug effects
- Receptors, Angiotensin/metabolism
- Receptors, Opioid/agonists
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Kornél Király
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Dávid Á. Karádi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Ferenc Zádor
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (T.T.); (É.S.)
| | - Amir Mohammadzadeh
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Anna Rita Galambos
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Mihály Balogh
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Pál Riba
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Tamás Tábi
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (T.T.); (É.S.)
| | - Zoltán S. Zádori
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Éva Szökő
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary; (T.T.); (É.S.)
| | - Susanna Fürst
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, P.O. Box 370, H-1445 Budapest, Hungary; (D.Á.K.); (F.Z.); (A.M.); (A.R.G.); (M.B.); (P.R.); (Z.S.Z.); (S.F.)
| |
Collapse
|
22
|
Dworsky-Fried Z, Chadwick CI, Kerr BJ, Taylor AMW. Multiple Sclerosis and the Endogenous Opioid System. Front Neurosci 2021; 15:741503. [PMID: 34602975 PMCID: PMC8484329 DOI: 10.3389/fnins.2021.741503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/26/2021] [Indexed: 01/10/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by chronic inflammation, neuronal degeneration and demyelinating lesions within the central nervous system. The mechanisms that underlie the pathogenesis and progression of MS are not fully known and current therapies have limited efficacy. Preclinical investigations using the murine experimental autoimmune encephalomyelitis (EAE) model of MS, as well as clinical observations in patients with MS, provide converging lines of evidence implicating the endogenous opioid system in the pathogenesis of this disease. In recent years, it has become increasingly clear that endogenous opioid peptides, binding μ- (MOR), κ- (KOR) and δ-opioid receptors (DOR), function as immunomodulatory molecules within both the immune and nervous systems. The endogenous opioid system is also well known to play a role in the development of chronic pain and negative affect, both of which are common comorbidities in MS. As such, dysregulation of the opioid system may be a mechanism that contributes to the pathogenesis of MS and associated symptoms. Here, we review the evidence for a connection between the endogenous opioid system and MS. We further explore the mechanisms by which opioidergic signaling might contribute to the pathophysiology and symptomatology of MS.
Collapse
Affiliation(s)
- Zoë Dworsky-Fried
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | - Caylin I. Chadwick
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Bradley J. Kerr
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, AB, Canada
| | - Anna M. W. Taylor
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
23
|
Piras S, Murineddu G, Loriga G, Carta A, Battistello E, Merighi S, Gessi S, Corona P, Asproni B, Ibba R, Temml V, Schuster D, Pinna GA. Biological Effects on μ-Receptors Affinity and Selectivity of Arylpropenyl Chain Structural Modification on Diazatricyclodecane Derivatives. Molecules 2021; 26:5448. [PMID: 34576918 PMCID: PMC8467848 DOI: 10.3390/molecules26185448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/26/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Opioid analgesics are clinically used to relieve severe pain in acute postoperative and cancer pain, and also in the long term in chronic pain. The analgesic action is mediated by μ-, δ-, and κ-receptors, but currently, with few exceptions for k-agonists, μ-agonists are the only ones used in therapy. Previously synthesized compounds with diazotricyclodecane cores (DTDs) have shown their effectiveness in binding opioid receptors. Fourteen novel diazatricyclodecanes belonging to the 9-propionyl-10-substituted-9,10-diazatricyclo[4.2.1.12,5]decane (compounds 20-23, 53, 57 and 59) and 2-propionyl-7-substituted-2,7-diazatricyclo[4.4.0.03,8]decane (compounds 24-27, 54, 58 and 60) series, respectively, have been synthesized and their ability to bind to the opioid μ-, δ- and κ-receptors was evaluated. Five of these derivatives, compounds 20, 21, 24, 26 and 53, showed μ-affinity in the nanomolar range with a negligible affinity towards δ- and κ-receptors and high μ-receptor selectivity. The synthesized compounds showed μ-receptor selectivity higher than those of previously reported methylarylcinnamyl analogs.
Collapse
Affiliation(s)
- Sandra Piras
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Gabriele Murineddu
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Giovanni Loriga
- Institute of Biomolecular Chemistry, National Research Council, Traversa La Crucca 3, 07100 Sassari, Italy;
| | - Antonio Carta
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Enrica Battistello
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.B.); (S.M.); (S.G.)
| | - Stefania Merighi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.B.); (S.M.); (S.G.)
| | - Stefania Gessi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (E.B.); (S.M.); (S.G.)
| | - Paola Corona
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Battistina Asproni
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Roberta Ibba
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| | - Veronika Temml
- Department of Pharmaceutical Chemistry, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; (V.T.); (D.S.)
| | - Daniela Schuster
- Department of Pharmaceutical Chemistry, Paracelsus Medical University Salzburg, Strubergasse 21, 5020 Salzburg, Austria; (V.T.); (D.S.)
| | - Gérard Aimè Pinna
- Department of Chemistry and Pharmacy, University of Sassari, via F. Muroni 23/A, 07100 Sassari, Italy; (G.M.); (A.C.); (P.C.); (B.A.); (R.I.); (G.A.P.)
| |
Collapse
|
24
|
Vázquez-León P, Miranda-Páez A, Chávez-Reyes J, Allende G, Barragán-Iglesias P, Marichal-Cancino BA. The Periaqueductal Gray and Its Extended Participation in Drug Addiction Phenomena. Neurosci Bull 2021; 37:1493-1509. [PMID: 34302618 DOI: 10.1007/s12264-021-00756-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/11/2021] [Indexed: 12/19/2022] Open
Abstract
The periaqueductal gray (PAG) is a complex mesencephalic structure involved in the integration and execution of active and passive self-protective behaviors against imminent threats, such as immobility or flight from a predator. PAG activity is also associated with the integration of responses against physical discomfort (e.g., anxiety, fear, pain, and disgust) which occurs prior an imminent attack, but also during withdrawal from drugs such as morphine and cocaine. The PAG sends and receives projections to and from other well-documented nuclei linked to the phenomenon of drug addiction including: (i) the ventral tegmental area; (ii) extended amygdala; (iii) medial prefrontal cortex; (iv) pontine nucleus; (v) bed nucleus of the stria terminalis; and (vi) hypothalamus. Preclinical models have suggested that the PAG contributes to the modulation of anxiety, fear, and nociception (all of which may produce physical discomfort) linked with chronic exposure to drugs of abuse. Withdrawal produced by the major pharmacological classes of drugs of abuse is mediated through actions that include participation of the PAG. In support of this, there is evidence of functional, pharmacological, molecular. And/or genetic alterations in the PAG during the impulsive/compulsive intake or withdrawal from a drug. Due to its small size, it is difficult to assess the anatomical participation of the PAG when using classical neuroimaging techniques, so its physiopathology in drug addiction has been underestimated and poorly documented. In this theoretical review, we discuss the involvement of the PAG in drug addiction mainly via its role as an integrator of responses to the physical discomfort associated with drug withdrawal.
Collapse
Affiliation(s)
- Priscila Vázquez-León
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, 20131, Aguascalientes, Ags., Mexico
| | - Abraham Miranda-Páez
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Wilfrido Massieu esq. Manuel Stampa s/n Col. Nueva Industrial Vallejo, 07738, Gustavo A. Madero, Mexico City, Mexico
| | - Jesús Chávez-Reyes
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, 20131, Aguascalientes, Ags., Mexico
| | - Gonzalo Allende
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, 20131, Aguascalientes, Ags., Mexico
| | - Paulino Barragán-Iglesias
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, 20131, Aguascalientes, Ags., Mexico.
| | - Bruno A Marichal-Cancino
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, 20131, Aguascalientes, Ags., Mexico.
| |
Collapse
|
25
|
Abstract
This paper is the forty-second consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2019 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, 65-30 Kissena Blvd., Flushing, NY, 11367, United States.
| |
Collapse
|
26
|
Analgesic effect of central relaxin receptor activation on persistent inflammatory pain in mice: behavioral and neurochemical data. Pain Rep 2021; 6:e937. [PMID: 34159282 PMCID: PMC8213244 DOI: 10.1097/pr9.0000000000000937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/26/2021] [Accepted: 04/23/2021] [Indexed: 01/02/2023] Open
Abstract
Supplemental Digital Content is Available in the Text. Relaxin peptide analogues produce strong but transient analgesia in inflammatory pain in mouse. Relaxin and its RXFP1 receptor represent a new peptidergic system that modulates pain processing in the forebrain areas. Introduction: The relaxin peptide signaling system is involved in diverse physiological processes, but its possible roles in the brain, including nociception, are largely unexplored. Objective: In light of abundant expression of relaxin receptor (RXFP1) mRNA/protein in brain regions involved in pain processing, we investigated the effects of central RXFP1 activation on nociceptive behavior in a mouse model of inflammatory pain and examined the neurochemical phenotype and connectivity of relaxin and RXFP1 mRNA-positive neurons. Methods: Mice were injected with Complete Freund Adjuvant (CFA) into a hind paw. After 4 days, the RXFP1 agonist peptides, H2-relaxin or B7-33, ± the RXFP1 antagonist, B-R13/17K-H2, were injected into the lateral cerebral ventricle, and mechanical and thermal sensitivity were assessed at 30 to 120 minutes. Relaxin and RXFP1 mRNA in excitatory and inhibitory neurons were examined using multiplex, fluorescent in situ hybridization. Relaxin-containing neurons were detected using immunohistochemistry and their projections assessed using fluorogold retrograde tract-tracing. Results: Both H2-relaxin and B7-33 produced a strong, but transient, reduction in mechanical and thermal sensitivity of the CFA-injected hind paw alone, at 30 minutes postinjection. Notably, coinjection of B-R13/17K-H2 blocked mechanical, but not thermal, analgesia. In the claustrum, cingulate cortex, and subiculum, RXFP1 mRNA was expressed in excitatory neurons. Relaxin immunoreactivity was detected in neurons in forebrain and midbrain areas involved in pain processing and sending projections to the RXFP1-rich, claustrum and cingulate cortex. No changes were detected in CFA mice. Conclusion: Our study identified a previously unexplored peptidergic system that can control pain processing in the brain and produce analgesia.
Collapse
|
27
|
Khan F, Mehan A. Addressing opioid tolerance and opioid-induced hypersensitivity: Recent developments and future therapeutic strategies. Pharmacol Res Perspect 2021; 9:e00789. [PMID: 34096178 PMCID: PMC8181203 DOI: 10.1002/prp2.789] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/07/2021] [Indexed: 01/07/2023] Open
Abstract
Opioids are a commonly prescribed and efficacious medication for the treatment of chronic pain but major side effects such as addiction, respiratory depression, analgesic tolerance, and paradoxical pain hypersensitivity make them inadequate and unsafe for patients requiring long-term pain management. This review summarizes recent advances in our understanding of the outcomes of chronic opioid administration to lay the foundation for the development of novel pharmacological strategies that attenuate opioid tolerance and hypersensitivity; the two main physiological mechanisms underlying the inadequacies of current therapeutic strategies. We also explore mechanistic similarities between the development of neuropathic pain states, opioid tolerance, and hypersensitivity which may explain opioids' lack of efficacy in certain patients. The findings challenge the current direction of analgesic research in developing non-opioid alternatives and we suggest that improving opioids, rather than replacing them, will be a fruitful avenue for future research.
Collapse
Affiliation(s)
- Faris Khan
- School of Clinical MedicineUniversity of CambridgeCambridgeUK
| | - Aman Mehan
- School of Clinical MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
28
|
Pantazis CB, Gonzalez LA, Tunstall BJ, Carmack SA, Koob GF, Vendruscolo LF. Cues conditioned to withdrawal and negative reinforcement: Neglected but key motivational elements driving opioid addiction. SCIENCE ADVANCES 2021; 7:7/15/eabf0364. [PMID: 33827822 PMCID: PMC8026136 DOI: 10.1126/sciadv.abf0364] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/19/2021] [Indexed: 05/07/2023]
Abstract
Opioid use disorder (OUD) is a debilitating disorder that affects millions of people. Neutral cues can acquire motivational properties when paired with the positive emotional effects of drug intoxication to stimulate relapse. However, much less research has been devoted to cues that become conditioned to the aversive effects of opioid withdrawal. We argue that environmental stimuli promote motivation for opioids when cues are paired with withdrawal (conditioned withdrawal) and generate opioid consumption to terminate conditioned withdrawal (conditioned negative reinforcement). We review evidence that cues associated with pain drive opioid consumption, as patients with chronic pain may misuse opioids to escape physical and emotional pain. We highlight sex differences in withdrawal-induced stress reactivity and withdrawal cue processing and discuss neurocircuitry that may underlie withdrawal cue processing in dependent individuals. These studies highlight the importance of studying cues associated with withdrawal in dependent individuals and point to areas for exploration in OUD research.
Collapse
Affiliation(s)
- Caroline B Pantazis
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
| | - Luis A Gonzalez
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
- Department of Psychological and Brain Sciences, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Brendan J Tunstall
- Department of Pharmacology, Addiction Science, and Toxicology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Stephanie A Carmack
- Center for Adaptive Systems of Brain-Body Interactions, George Mason University, Fairfax, VA, USA
| | - George F Koob
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Leandro F Vendruscolo
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
29
|
WeiWei Y, WenDi F, Mengru C, Tuo Y, Chen G. The cellular mechanism by which the rostral ventromedial medulla acts on the spinal cord during chronic pain. Rev Neurosci 2021; 32:545-558. [PMID: 33565739 DOI: 10.1515/revneuro-2020-0121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/18/2020] [Indexed: 11/15/2022]
Abstract
Clinical therapies for chronic pain are limited. While targeted drugs are promising therapies for chronic pain, they exhibit insufficient efficacy and poor targeting. The occurrence of chronic pain partly results from central changes caused by alterations in neurons in the rostral ventromedial medulla (RVM) in the brainstem regulatory pathway. The RVM, which plays a key role in the descending pain control pathway, greatly contributes to the development and maintenance of pain. However, the exact roles of the RVM in chronic pain remain unclear, making it difficult to develop new drugs targeting the RVM and related pathways. Here, we first discuss the roles of the RVM and related circuits in chronic pain. Then, we analyze synaptic transmission between RVM neurons and spinal cord neurons, specifically focusing on the release of neurotransmitters, to explore the cellular mechanisms by which the RVM regulates chronic pain. Finally, we propose some ideas for the development of drugs targeting the RVM.
Collapse
Affiliation(s)
- Yu WeiWei
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong226001, China
| | - Fei WenDi
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong226001, China
| | - Cui Mengru
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong226001, China
| | - Yang Tuo
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun130033, China
| | - Gang Chen
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong226001, China.,Department of Tissue and Embryology, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong University, Nantong226001, China
| |
Collapse
|
30
|
Faramarzi G, Charmchi E, Salehi S, Zendehdel M, Haghparast A. Intra-accumbal dopaminergic system modulates the restraint stress-induced antinociceptive behaviours in persistent inflammatory pain. Eur J Pain 2021; 25:862-871. [PMID: 33342008 DOI: 10.1002/ejp.1716] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 12/13/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Stress activates several neural pathways that inhibit pain sensation. Nucleus accumbens (NAc), as an important component of the mesolimbic dopaminergic system, has a major role in pain modulation and is differentially affected by stress. Based on the nature of stressors, the direction of this effect is controversial. We previously showed that forced swim stress-induced analgesia through activation of NAc dopamine receptors. In this study, we aimed to examine the role of dopamine receptors within the NAc in restraint stress (RS)-induced analgesia. METHODS Male Wistar rats weighing 230-250 g were unilaterally implanted with a cannula into the NAc. D1-like dopamine receptor antagonist, SCH-23390 (0.25, 1 and 4 µg/0.5 µL saline), or D2-like dopamine receptor antagonist, Sulpiride (0.25, 1 and 4µg/0.5µl DMSO), were microinjected into NAc in two separate super groups 5 min prior to exposure to RS. Their control groups just received intra-accumbal saline or DMSO (0.5 µl) respectively. The formalin test was performed after animals were subjected to RS using Plexiglas tubes. RESULTS The results demonstrated that RS produces analgesia in both phases of the formalin test. Intra-NAc injection of SCH-23390 equally reduced RS-induced analgesia in both early and late phases of the formalin test, while Sulpiride reduced RS-induced analgesia just at the late phase. CONCLUSIONS These findings suggest that the dopaminergic system might act as a potential endogenous pain control system in stress conditions. However, the lack of evaluation of the role of the dopaminergic system in RS-induced antinociception in acute pain conditions is considered as a limitation for this study. In addition, a comprehensive evaluation of this endogenous pain control system in animal and clinical studies will guide future efforts for developing more effective medication. SIGNIFICANCE Restraint stress (RS) induces the antinociceptive behaviors in both phases of formalin test. Blockade of intra-accumbal dopamine receptors impresses the antinociception induced by RS. Blockade of D1-like dopamine receptor equally reduced RS-induced analgesia in both early and late phases of the formalin test. Blockade of D2-like dopamine receptor reduced RS-induced analgesia just at the late phase.
Collapse
Affiliation(s)
- Golnaz Faramarzi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Physiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Elham Charmchi
- Department of Physiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Sakineh Salehi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Medicine, Ardabil Medical Sciences Branch, Islamic Azad University, Ardabil, Iran
| | - Morteza Zendehdel
- Department of Physiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Wang L, Yin C, Xu X, Liu T, Wang B, Abdul M, Zhou Y, Cao J, Lu C. Pellino1 Contributes to Morphine Tolerance by Microglia Activation via MAPK Signaling in the Spinal Cord of Mice. Cell Mol Neurobiol 2020; 40:1117-1131. [PMID: 31989355 PMCID: PMC11448779 DOI: 10.1007/s10571-020-00797-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 01/19/2020] [Indexed: 10/25/2022]
Abstract
Chronic morphine-induced antinociceptive tolerance is a major unresolved issue in clinical practices, which is associated with microglia activation in the spinal cord. E3 ubiquitin ligase Pellino1 (Peli1) is known to be an important microglia-specific regulator. However, it is unclear whether Peli1 is involved in morphine tolerance. Here, we found that Peli1 levels in the spinal cord were significantly elevated in morphine tolerance mouse model. Notably, Peli1 was expressed in a great majority of microglia in the spinal dorsal horn, while downregulation of spinal Peli1 attenuated the development of morphine tolerance and associated hyperalgesia. Our biochemical data revealed that morphine tolerance-induced increase in Peli1 was accompanied by spinal microglia activation, activation of mitogen-activated protein kinase (MAPK) signaling, and production of proinflammatory cytokines. Peli1 additionally was found to promote K63-linked ubiquitination of tumor necrosis factor receptor-associated factor 6 (TRAF6) in the spinal cord after repeated morphine treatment. Furthermore, knocking down Peli1 in cultured BV2 microglial cells significantly attenuated inflammatory reactions in response to morphine challenge. Therefore, we conclude that the upregulation of Peli1 in the spinal cord plays a curial role in the development of morphine tolerance via Peli1-dependent mobilization of spinal microglia, activation of MAPK signaling, and production of proinflammatory cytokines. Modulation of Peli1 may be a potential strategy for the prevention of morphine tolerance.
Collapse
Affiliation(s)
- Lijuan Wang
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Cui Yin
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Xiangying Xu
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Tianya Liu
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
- Department of Pharmacy, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, 221002, China
| | - Bin Wang
- Department of Anesthesiology, The First People's Hospital of Lianyungang City, Lianyungang, 222000, China
| | - Mannan Abdul
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China
| | - Yan Zhou
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, 221002, China
| | - Junli Cao
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China.
| | - Chen Lu
- School of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China.
| |
Collapse
|
32
|
Acupuncture for Cancer-Induced Bone Pain in Animal Models: A Systemic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:5606823. [PMID: 32802130 PMCID: PMC7414378 DOI: 10.1155/2020/5606823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 06/11/2020] [Accepted: 07/02/2020] [Indexed: 12/09/2022]
Abstract
Background Cancer-induced bone pain (CIBP) is a highly prevalent symptom, which afflicts vast majority of patients who suffer from cancer. The current treatment options failed to achieve satisfactory effect and the side effects were prominent. Recent randomized controlled trials (RCTs) of animal demonstrate the benefit of acupuncture for CIBP. We sought to determine if the pooled data from available RCTs supports the use of acupuncture for CIBP. Methods A literature search for randomized controlled trials was conducted in six electronic databases from inception to May 31, 2019. Meta-analysis was performed with Review Manager 5.3 software; the publication bias was assessed by Stata 12.0 software. We used random effects model for pooling data because heterogeneity is absolute among studies to some extent. Results Twenty-four trials were included in the review, of which 12 trials provided detailed data for meta-analyses. Preliminary evidence indicates that compared to wait list/sham group, acupuncture was effective on increasing paw withdrawal threshold (PWT) and paw withdrawal latency (PWL). Compared to medicine, acupuncture was less effective on PWT, but as effective as medicine on PWL. Acupuncture can reinforce medicine's effect on PWT and PWL. Compared to the control group, acupuncture was superior to increase body weight (BW), decrease spinal cord glial fibrillary acidic protein (GFAP), and interleukin-1β (IL-1β). Furthermore, some studies showed acupuncture delay or partially reverse morphine tolerance. Three studies found acupuncture has no effect on PWT, but 2 of them found acupuncture could enhance small dose of Celebrex's effect on CIBP. Conclusions Acupuncture was superior to wait list/sham acupuncture on increasing PWT and has no less effect on increasing PWL compared to medicine; acupuncture improved the efficacy of drugs, increased the CIBP animals' body weight, and decreased their spinal cord GFAP and IL-1β. High-quality studies are necessary to confirm the results.
Collapse
|
33
|
The Cerebral Localization of Pain: Anatomical and Functional Considerations for Targeted Electrical Therapies. J Clin Med 2020; 9:jcm9061945. [PMID: 32580436 PMCID: PMC7355617 DOI: 10.3390/jcm9061945] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/18/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022] Open
Abstract
Millions of people in the United States are affected by chronic pain, and the financial cost of pain treatment is weighing on the healthcare system. In some cases, current pharmacological treatments may do more harm than good, as with the United States opioid crisis. Direct electrical stimulation of the brain is one potential non-pharmacological treatment with a long history of investigation. Yet brain stimulation has been far less successful than peripheral or spinal cord stimulation, perhaps because of our limited understanding of the neural circuits involved in pain perception. In this paper, we review the history of using electrical stimulation of the brain to treat pain, as well as contemporary studies identifying the structures involved in pain networks, such as the thalamus, insula, and anterior cingulate. We propose that the thermal grill illusion, an experimental pain model, can facilitate further investigation of these structures. Pairing this model with intracranial recording will provide insight toward disentangling the neural correlates from the described anatomic areas. Finally, the possibility of altering pain perception with brain stimulation in these regions could be highly informative for the development of novel brain stimulation therapies for chronic pain.
Collapse
|