1
|
Lopes EF, West AM, Locke JL, Holleran K, Adrian LA, Dawes MH, Curry AM, McKelvey HA, Martin T, Jones SR. Morphine-Induced Antinociception Is Potentiated and Dopamine Elevations Are Inhibited by the Biased Kappa Opioid Receptor Agonist Triazole 1.1. ACS Chem Neurosci 2025; 16:1377-1387. [PMID: 40129263 DOI: 10.1021/acschemneuro.5c00075] [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] [Indexed: 03/26/2025] Open
Abstract
Traditional analgesic opioid compounds, which act through μ opioid receptors (MORs), engender a high risk for misuse and dependence. κ opioid receptor (KOR) activation, a potential target for pain treatment, produces antinociception without euphoric side effects but results in dysphoria and aversion. Triazole 1.1 is a KOR agonist biased toward G-protein coupled signaling, potentially promoting antinociception without dysphoria. We tested whether triazole 1.1 could provide antinociception and its effects in combination with morphine. We employed a lactic acid abdominal pain model, which induced acute pain behaviors, decreased basal dopamine levels in the nucleus accumbens (NAc), and increased KOR function. We administered several interventions including triazole 1.1 (30 mg/kg) and morphine (12 or 24 mg/kg), individually and in combination. Triazole 1.1 alone reduced the pain behavioral response and changes to KOR function but did not prevent the reduction in basal dopamine levels. Morphine not only dose-dependently prevented behavioral pain responses but also elevated NAc dopamine and did not prevent the pain-induced increase in KOR function. However, combining low-dose morphine with triazole 1.1 prevents behavioral pain responses, changes to NAc dopamine levels, and changes to KOR function. Therefore, we present triazole 1.1 as a dose-sparing pain treatment to be used in combination with a lower dose of morphine, thus reducing the potential for opioid misuse.
Collapse
Affiliation(s)
- Emanuel F Lopes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
| | - Alyssa M West
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
| | - Jason L Locke
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
| | - Katherine Holleran
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
| | - Leighelle A Adrian
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
| | - Monica H Dawes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
| | - Alyson M Curry
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
| | - Harlie A McKelvey
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
| | - Thomas Martin
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Sara R Jones
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
| |
Collapse
|
2
|
Marchette RCN, Vendruscolo LF, Koob GF. The Dynorphin/-Opioid Receptor System at the Interface of Hyperalgesia/Hyperkatifeia and Addiction. CURRENT ADDICTION REPORTS 2025; 12:11. [PMID: 40124896 PMCID: PMC11925990 DOI: 10.1007/s40429-025-00618-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 03/25/2025]
Abstract
Purpose of Review Drug addiction is characterized by compulsive drug seeking and use, accompanied by negative emotional states (hyperkatifeia) and heightened pain sensitivity (hyperalgesia) during withdrawal. Both hyperalgesia and hyperkatifeia are integral components of substance use disorders, negatively impacting treatment and recovery. The underlying neurobiological mechanisms of hyperalgesia and hyperkatifeia involve alterations of brain reward and stress circuits, including the dynorphin/κ-opioid receptor (KOR) system. The dynorphin/KOR system modulates pain perception, negative affect, and addictive behaviors. Here, we review the preclinical evidence of dynorphin/KOR signaling in opioid withdrawal-induced hyperalgesia and hyperkatifeia. Recent Findings In opioid dependence models, pharmacological and genetic interventions of the dynorphin/KOR system attenuate somatic and motivational signs of withdrawal and addictive-like behaviors, highlighting its therapeutic potential. Understanding the intricate interplay between dynorphin/KOR signaling, hyperalgesia, hyperkatifeia, and addiction offers novel insights into treatment strategies for opioid use disorder and other substance use disorders. Summary Further research is needed to elucidate precise mechanisms of the sexual dimorphism of dynorphin/KOR signaling and identify targeted interventions to mitigate hyperalgesia and hyperkatifeia and facilitate recovery from addiction.
Collapse
Affiliation(s)
- Renata C. N. Marchette
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, BRC Room 08A505.19, 251 Bayview Blvd, Baltimore, MD 21224 USA
| | - Leandro F. Vendruscolo
- Stress and Addiction Neuroscience Unit, Integrative Neuroscience Research Branch, Division of Intramural Clinical and Biological Research, National Institute on Drug Abuse, Intramural Research Program, and National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore, MD 21224 USA
| | - George F. Koob
- Neurobiology of Addiction Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, BRC Room 08A505.19, 251 Bayview Blvd, Baltimore, MD 21224 USA
| |
Collapse
|
3
|
Diester CM, Balint H, Gillespie JC, Lichtman AH, Sim-Selley LJ, Selley DE, Negus SS. Effects of Repeated Treatment with the Monoacylglycerol Lipase Inhibitor MJN110 on Pain-Related Depression of Nesting and Cannabinoid 1 Receptor Function in Male and Female Mice. J Pharmacol Exp Ther 2024; 390:291-301. [PMID: 38262742 PMCID: PMC11338278 DOI: 10.1124/jpet.123.001940] [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: 09/24/2023] [Revised: 11/17/2023] [Accepted: 12/08/2023] [Indexed: 01/25/2024] Open
Abstract
MJN110 inhibits the enzyme monoacylglycerol lipase (MAGL) to increase levels of the endocannabinoid 2-arachidonoylglycerol , an endogenous high-efficacy agonist of cannabinoid 1 and 2 receptors (CB1/2R). MAGL inhibitors are under consideration as candidate analgesics, and we reported previously that acute MJN110 produced partial antinociception in an assay of pain-related behavioral depression in mice. Given the need for repeated analgesic administration in many pain patients and the potential for analgesic tolerance during repeated treatment, this study examined antinociceptive effects of repeated MJN110 on pain-related behavioral depression and CB1R-mediated G-protein function. Male and female ICR mice were treated daily for 7 days in a 2 × 2 design with (a) 1.0 mg/kg/d MJN110 or its vehicle followed by (b) intraperitoneal injection of dilute lactic acid (IP acid) or its vehicle as a visceral noxious stimulus to depress nesting behavior. After behavioral testing, G-protein activity was assessed in lumbar spinal cord (LSC) and five brain regions using an assay of CP55,940-stimulated [35S]GTPɣS activation. As reported previously, acute MJN110 produced partial but significant relief of IP acid-induced nesting depression on day 1. After 7 days, MJN110 continued to produce significant but partial antinociception in males, while antinociceptive tolerance developed in females. Repeated MJN110 also produced modest decreases in maximum levels of CP55,940-induced [35S]GTPɣS binding in spinal cord and most brain regions. These results indicate that repeated treatment with a relatively low antinociceptive MJN110 dose produces only partial and sex-dependent transient antinociception associated with the emergence of CB1R desensitization in this model of IP acid-induced nesting depression. SIGNIFICANCE STATEMENT: The drug MJN110 inhibits monoacylglycerol lipase (MAGL) to increase levels of the endogenous cannabinoid 2-arachidonoylglycerol and produce potentially useful therapeutic effects including analgesia. This study used an assay of pain-related behavioral depression in mice to show that repeated MJN110 treatment produced (1) weak but sustained antinociception in male mice, (2) antinociceptive tolerance in females, and (3) modest cannabinoid-receptor desensitization that varied by region and sex. Antinociceptive tolerance may limit the utility of MJN110 for treatment of pain.
Collapse
Affiliation(s)
- Clare M Diester
- Department of Pharmacology and Toxicology (C.M.D., H.B., J.C.G., A.H.L., L.J.S.-S., D.E.S., S.S.N.) and School of Pharmacy (A.H.L.), Virginia Commonwealth University, Richmond, Virginia
| | - Hallie Balint
- Department of Pharmacology and Toxicology (C.M.D., H.B., J.C.G., A.H.L., L.J.S.-S., D.E.S., S.S.N.) and School of Pharmacy (A.H.L.), Virginia Commonwealth University, Richmond, Virginia
| | - James C Gillespie
- Department of Pharmacology and Toxicology (C.M.D., H.B., J.C.G., A.H.L., L.J.S.-S., D.E.S., S.S.N.) and School of Pharmacy (A.H.L.), Virginia Commonwealth University, Richmond, Virginia
| | - Aron H Lichtman
- Department of Pharmacology and Toxicology (C.M.D., H.B., J.C.G., A.H.L., L.J.S.-S., D.E.S., S.S.N.) and School of Pharmacy (A.H.L.), Virginia Commonwealth University, Richmond, Virginia
| | - Laura J Sim-Selley
- Department of Pharmacology and Toxicology (C.M.D., H.B., J.C.G., A.H.L., L.J.S.-S., D.E.S., S.S.N.) and School of Pharmacy (A.H.L.), Virginia Commonwealth University, Richmond, Virginia
| | - Dana E Selley
- Department of Pharmacology and Toxicology (C.M.D., H.B., J.C.G., A.H.L., L.J.S.-S., D.E.S., S.S.N.) and School of Pharmacy (A.H.L.), Virginia Commonwealth University, Richmond, Virginia
| | - S Stevens Negus
- Department of Pharmacology and Toxicology (C.M.D., H.B., J.C.G., A.H.L., L.J.S.-S., D.E.S., S.S.N.) and School of Pharmacy (A.H.L.), Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
4
|
Wilkerson JL. Seeing through the Haze: Monoacylglycerol Lipase Inhibitors As Analgesics. J Pharmacol Exp Ther 2024; 390:288-290. [PMID: 39159976 DOI: 10.1124/jpet.124.002132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/08/2024] [Indexed: 08/21/2024] Open
Affiliation(s)
- Jenny L Wilkerson
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Amarillo, Texas
| |
Collapse
|
5
|
Zhang Y, Ma H, Bai Y, Hou X, Yang Y, Wang G, Li Y. Chronic Neuropathic Pain and Comorbid Depression Syndrome: From Neural Circuit Mechanisms to Treatment. ACS Chem Neurosci 2024; 15:2432-2444. [PMID: 38916052 DOI: 10.1021/acschemneuro.4c00125] [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: 06/26/2024] Open
Abstract
Chronic neuropathic pain and comorbid depression syndrome (CDS) is a major worldwide health problem that affects the quality of life of patients and imposes a tremendous socioeconomic burden. More than half of patients with chronic neuropathic pain also suffer from moderate or severe depression. Due to the complex pathogenesis of CDS, there are no effective therapeutic drugs available. The lack of research on the neural circuit mechanisms of CDS limits the development of treatments. The purpose of this article is to provide an overview of the various circuits involved in CDS. Notably, activating some neural circuits can alleviate pain and/or depression, while activating other circuits can exacerbate these conditions. Moreover, we discuss current and emerging pharmacotherapies for CDS, such as ketamine. Understanding the circuit mechanisms of CDS may provide clues for the development of novel drug treatments for improved CDS management.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Hui Ma
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yafan Bai
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xiaojuan Hou
- Hebei North University, Zhangjiakou, 075000, China
| | - Yixin Yang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Guyan Wang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yunfeng Li
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, 100850, China
| |
Collapse
|
6
|
Fiatcoski F, Jesus CHA, de Melo Turnes J, Chichorro JG, Kopruszinski CM. Sex differences in descending control of nociception (DCN) responses after chronic orofacial pain induction in rats and the contribution of kappa opioid receptors. Behav Brain Res 2024; 459:114789. [PMID: 38036264 DOI: 10.1016/j.bbr.2023.114789] [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: 08/18/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/02/2023]
Abstract
Descending control of nociception (DCN), a measure of efficiency of descending pain inhibition, can be assessed in animals by the combined application of test and conditioning noxious stimuli. Evidence from pre-clinical and clinical studies indicates that this mechanism of pain control may differ between sexes and might be impaired in many chronic pain states. However, little is known about sex differences in DCN efficiency in models of acute and chronic orofacial pain. Herein, we first evaluated DCN responses in male and female rats by the applying formalin into the upper lip or capsaicin into the forepaw as the conditioning stimulus, followed by mechanical stimulation (Randall-Selitto) of the hind paw as the test stimulus. The same protocol (i.e., capsaicin in the forepaw followed by mechanical stimulation of the hind paw) was evaluated in male and female rats on day 3 after intraoral incision and on day 15 and 30 after chronic constriction injury of the infraorbital nerve (CCI-ION). Additionally, we assessed the effect of the kappa opioid receptor (KOR) antagonist Norbinaltorphimine (nor-BNI) on DCN responses of female nerve-injured rats. This study shows that naïve female rats exhibit less efficient DCN compared to males. Postoperative pain did not alter DCN responses in female and male rats, but CCI-ION induced loss of DCN responses in females but not in males. Systemic pretreatment with nor-BNI prevented the loss of DCN induced by CCI-ION in female rats. The results reveal sex differences in DCN responses and female-specific impairment of DCN following chronic orofacial pain. Moreover, the findings suggest that, at least for females, blocking KOR could be a promising therapeutic approach to prevent maladaptive changes in chronic orofacial pain.
Collapse
Affiliation(s)
- Fernanda Fiatcoski
- Department of Pharmacology, Biological Sciences Sector, Federal University of Parana, Curitiba, PR, Brazil
| | | | - Joelle de Melo Turnes
- Department of Pharmacology, Biological Sciences Sector, Federal University of Parana, Curitiba, PR, Brazil
| | - Juliana Geremias Chichorro
- Department of Pharmacology, Biological Sciences Sector, Federal University of Parana, Curitiba, PR, Brazil
| | | |
Collapse
|
7
|
Mazuryk J, Klepacka K, Kutner W, Sharma PS. Glyphosate: Impact on the microbiota-gut-brain axis and the immune-nervous system, and clinical cases of multiorgan toxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 271:115965. [PMID: 38244513 DOI: 10.1016/j.ecoenv.2024.115965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/25/2023] [Accepted: 01/06/2024] [Indexed: 01/22/2024]
Abstract
Glyphosate (GLP) and GLP-based herbicides (GBHs), such as polyethoxylated tallow amine-based GLP surfactants (GLP-SH), developed in the late 70', have become the most popular and controversial agrochemicals ever produced. Nowadays, GBHs have reached 350 million hectares of crops in over 140 countries, with an annual turnover of 5 billion and 11 billion USD in the U.S.A. and worldwide, respectively. Because of the highly efficient inhibitory activity of GLP targeted to the 5-enolpyruvylshikimate-3-phosphate synthase pathway, present in plants and several bacterial strains, the GLP-resistant crop-based genetic agricultural revolution has decreased famine and improved the costs and quality of living in developing countries. However, this progress has come at the cost of the 50-year GBH overuse, leading to environmental pollution, animal intoxication, bacterial resistance, and sustained occupational exposure of the herbicide farm and companies' workers. According to preclinical and clinical studies covered in the present review, poisoning with GLP, GLP-SH, and GBHs devastatingly affects gut microbiota and the microbiota-gut-brain (MGB) axis, leading to dysbiosis and gastrointestinal (GI) ailments, as well as immunosuppression and inappropriate immunostimulation, cholinergic neurotransmission dysregulation, neuroendocrinal system disarray, and neurodevelopmental and neurobehavioral alterations. Herein, we mainly focus on the contribution of gut microbiota (GM) to neurological impairments, e.g., stroke and neurodegenerative and neuropsychiatric disorders. The current review provides a comprehensive introduction to GLP's microbiological and neurochemical activities, including deviation of the intestinal Firmicutes-to-Bacteroidetes ratio, acetylcholinesterase inhibition, excitotoxicity, and mind-altering processes. Besides, it summarizes and critically discusses recent preclinical studies and clinical case reports concerning the harmful impacts of GBHs on the GI tract, MGB axis, and nervous system. Finally, an insightful comparison of toxic effects caused by GLP, GBH-SH, and GBHs is presented. To this end, we propose a first-to-date survey of clinical case reports on intoxications with these herbicides.
Collapse
Affiliation(s)
- Jarosław Mazuryk
- Department of Electrode Processes, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland; Bio & Soft Matter, Institute of Condensed Matter and Nanosciences, Université catholique de Louvain, 1 Place Louis Pasteur, 1348 Louvain-la-Neuve, Belgium.
| | - Katarzyna Klepacka
- Functional Polymers Research Team, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland; ENSEMBLE(3) sp. z o. o., 01-919 Warsaw, Poland
| | - Włodzimierz Kutner
- Department of Electrode Processes, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland; Faculty of Mathematics and Natural Sciences. School of Sciences, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland
| | - Piyush Sindhu Sharma
- Functional Polymers Research Team, Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warsaw, Poland
| |
Collapse
|
8
|
Li L, Chen J, Li YQ. The Downregulation of Opioid Receptors and Neuropathic Pain. Int J Mol Sci 2023; 24:ijms24065981. [PMID: 36983055 PMCID: PMC10053236 DOI: 10.3390/ijms24065981] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Neuropathic pain (NP) refers to pain caused by primary or secondary damage or dysfunction of the peripheral or central nervous system, which seriously affects the physical and mental health of 7-10% of the general population. The etiology and pathogenesis of NP are complex; as such, NP has been a hot topic in clinical medicine and basic research for a long time, with researchers aiming to find a cure by studying it. Opioids are the most commonly used painkillers in clinical practice but are regarded as third-line drugs for NP in various guidelines due to the low efficacy caused by the imbalance of opioid receptor internalization and their possible side effects. Therefore, this literature review aims to evaluate the role of the downregulation of opioid receptors in the development of NP from the perspective of dorsal root ganglion, spinal cord, and supraspinal regions. We also discuss the reasons for the poor efficacy of opioids, given the commonness of opioid tolerance caused by NP and/or repeated opioid treatments, an angle that has received little attention to date; in-depth understanding might provide a new method for the treatment of NP.
Collapse
Affiliation(s)
- Lin Li
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, No. 169, West Changle Road, Xi'an 710032, China
| | - Jing Chen
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, No. 169, West Changle Road, Xi'an 710032, China
| | - Yun-Qing Li
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an 710072, China
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, No. 169, West Changle Road, Xi'an 710032, China
| |
Collapse
|
9
|
Baldwin AN, Banks ML, Marsh SA, Townsend EA, Venniro M, Shaham Y, Negus SS. Acute pain-related depression of operant responding maintained by social interaction or food in male and female rats. Psychopharmacology (Berl) 2022; 239:561-572. [PMID: 35043215 PMCID: PMC10053137 DOI: 10.1007/s00213-021-06048-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/19/2021] [Indexed: 01/26/2023]
Abstract
RATIONALE Clinically relevant pain is often associated with functional impairment and behavioral depression, including depression of social behavior. Moreover, recovery of function is a major goal in pain treatment. We used a recently developed model of operant responding for social interaction in rats to evaluate the vulnerability of social behavior to an experimental pain manipulation and the sensitivity of pain-depressed social behavior to treatment with clinically effective analgesics. METHODS Sprague-Dawley male and female rats were trained to lever press for social access to another rat, and responding was evaluated after treatment with (a) intraperitoneal injection of dilute lactic acid (IP acid; 0.18-5.6%) administered alone as a visceral noxious stimulus, (b) the mu-opioid receptor (MOR) agonist morphine (0.32-10 mg/kg) or nonsteroidal anti-inflammatory drug (NSAID) ketoprofen (10 mg/kg) administered alone, or (c) morphine or ketoprofen administered before IP acid. For comparison, the same treatments were evaluated in separate rats trained to lever press for food delivery. RESULTS Both IP acid alone and morphine alone more potently decreased responding maintained by social interaction than by food, whereas ketoprofen did not affect responding for either reinforcer. In general, analgesics were most effective to rescue operant responding when relatively low IP acid concentrations produced significant but submaximal behavioral depression; however, morphine was not effective to rescue responding for social interaction. CONCLUSIONS Operant responding maintained by social interaction was more sensitive to pain-related disruption and less responsive to opioid analgesic rescue than food-maintained operant responding. Social behavior may be especially vulnerable to depression by pain states.
Collapse
Affiliation(s)
- A N Baldwin
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - M L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - S A Marsh
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - E A Townsend
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - M Venniro
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Y Shaham
- Behavioral Neuroscience Research Branch, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
10
|
Nikolaus S, Wittsack HJ, Beu M, Hautzel H, Antke C, Mamlins E, Cardinale J, Decheva C, Huston JP, Antoch G, Giesel FL, Müller HW. The 5-HT1A receptor antagonist WAY-100635 decreases motor/exploratory behaviors and nigrostriatal and mesolimbocortical dopamine D2/3 receptor binding in adult rats. Pharmacol Biochem Behav 2022; 215:173363. [DOI: 10.1016/j.pbb.2022.173363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 10/19/2022]
|
11
|
Massaly N, Markovic T, Creed M, Al-Hasani R, Cahill CM, Moron JA. Pain, negative affective states and opioid-based analgesics: Safer pain therapies to dampen addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 157:31-68. [PMID: 33648672 DOI: 10.1016/bs.irn.2020.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Across centuries and civilizations opioids have been used to relieve pain. In our modern societies, opioid-based analgesics remain one of the most efficient treatments for acute pain. However, the long-term use of opioids can lead to the development of analgesic tolerance, opioid-induced hyperalgesia, opioid use disorders, and overdose, which can ultimately produce respiratory depressant effects with fatal consequences. In addition to the nociceptive sensory component of pain, negative affective states arising from persistent pain represent a risk factor for developing an opioid use disorder. Several studies have indicated that the increase in prescribed opioid analgesics since the 1990s represents the root of our current opioid epidemic. In this review, we will present our current knowledge on the endogenous opioid system within the pain neuroaxis and the plastic changes occurring in this system that may underlie the occurrence of pain-induced negative affect leading to misuse and abuse of opioid medications. Dissecting the allostatic neuronal changes occurring during pain is the most promising avenue to uncover novel targets for the development of safer pain medications. We will discuss this along with current and potential approaches to treat pain-induced negative affective states that lead to drug misuse. Moreover, this chapter will provide a discussion on potential avenues to reduce the abuse potential of new analgesic drugs and highlight a basis for future research and drug development based on recent advances in this field.
Collapse
Affiliation(s)
- Nicolas Massaly
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States; Washington University in St Louis, Pain Center, St. Louis, MO, United States; Washington University in St Louis, School of Medicine, St. Louis, MO, United States.
| | - Tamara Markovic
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States; Washington University in St Louis, Pain Center, St. Louis, MO, United States; Washington University in St Louis, School of Medicine, St. Louis, MO, United States
| | - Meaghan Creed
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States; Washington University in St Louis, Pain Center, St. Louis, MO, United States; Washington University in St Louis, School of Medicine, 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; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - Ream Al-Hasani
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States; Washington University in St Louis, Pain Center, St. Louis, MO, United States; Washington University in St Louis, School of Medicine, St. Louis, MO, United States; Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, United States; Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Catherine M Cahill
- Department of Psychiatry and Biobehavioural Sciences, University of California, Los Angeles, CA, United States; Shirley and Stefan Hatos Center for Neuropharmacology, University of California Los Angeles, Los Angeles, CA, United States; Jane & Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, United States
| | - Jose A Moron
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States; Washington University in St Louis, Pain Center, St. Louis, MO, United States; Washington University in St Louis, School of Medicine, 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
|
12
|
Serrano NE, Saputra SG, Íbias J, Company M, Nazarian A. Pain-induced impulsivity is sexually dimorphic and mu-opioid receptor sensitive in rats. Psychopharmacology (Berl) 2021; 238:3447-3462. [PMID: 34427720 DOI: 10.1007/s00213-021-05963-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/10/2021] [Indexed: 12/13/2022]
Abstract
RATIONALE AND OBJECTIVES Pain sensation can negatively impact cognitive function, including impulsivity. Pain-induced changes in impulsivity can contribute to development of psychiatric comorbidities found in those with chronic pain conditions. The goal of this study was to determine whether complete Freund's adjuvant (CFA)-induced pain manipulation enhances impulsivity in rats. Whether the pain-induced impulsivity is sexually dimorphic, and if mu-opioid receptors play a role in these processes. METHODS Male and female rats were screened for trait impulsivity and designated as high or low impulsive using a delay discounting task. Rats then received a hind paw injection of CFA, and their impulsivity was assessed for 16 days. The effects of morphine on impulsivity were also examined. In a separate experiment, rats were pretreated with beta-funaltrexamine (β-FNA) to determine the role of mu-opioid receptors on impulsivity. RESULTS CFA treatment increased impulsivity in males and females. The onset of CFA-induced impulsivity was faster in high impulsive females than males. Morphine blocked CFA-induced impulsivity in both sexes in a dose- and time-dependent manner. β-FNA prevented the actions of morphine on CFA-induced impulsivity in high impulsive males, but not high impulsive females. Moreover, β-FNA increased CFA-induced impulsivity in morphine naïve males, but not females. CONCLUSION These findings demonstrate unique sex differences in CFA-induced impulsivity, response to morphine, and the impact of mu-opioid receptors. A better understanding of cognitive deficits and their mechanisms can provide insight into the development of substance abuse and psychiatric comorbidities that occur in people with chronic pain.
Collapse
Affiliation(s)
- Nidia Espinoza Serrano
- Department of Pharmaceutical Sciences, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Samuel G Saputra
- Department of Pharmaceutical Sciences, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Javier Íbias
- Departamento de Metodologίa de Las Ciencias del Comportamiento, Facultad de Psicologίa, Universidad Nacional de Educacίon a Distancia (UNED), 28040, Madrid, Spain
| | - Matthew Company
- Department of Pharmaceutical Sciences, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Arbi Nazarian
- Department of Pharmaceutical Sciences, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
13
|
Banks ML. Environmental influence on the preclinical evaluation of substance use disorder therapeutics. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 93:219-242. [PMID: 35341567 DOI: 10.1016/bs.apha.2021.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Substance use disorders (SUD) develop as a result of complex interactions between the environment, the subject, and the drug of abuse. Preclinical basic research investigating each of these tripartite components of SUD individually has resulted in advancements in our fundamental knowledge regarding the progression from drug abuse to SUD and severe drug addiction and the underlying behavioral and neurobiological mechanisms. How these complex interactions between the environment, the subject, and the drug of abuse impact the effectiveness of candidate or clinically used medications for SUD has not been as extensively investigated. The focus of this chapter will address the current state of our knowledge how these environmental, subject, and pharmacological variables have been shown to impact candidate or clinical SUD medication evaluation in preclinical research using drug self-administration procedures as the primary dependent measure. The results discussed in this chapter highlight the importance of considering environmental variables such as the schedule of reinforcement, concurrent availability of alternative nondrug reinforcers, and experimental housing conditions in the context of SUD therapeutic evaluation. The thesis of this chapter is that improved understanding of environmental variables in the context of SUD research will facilitate the utility of preclinical drug self-administration studies in the evaluation and development of candidate SUD therapeutics.
Collapse
Affiliation(s)
- Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.
| |
Collapse
|
14
|
Garner JB, Marshall LS, Boyer NM, Alapatt V, Miller LL. Effects of Ketoprofen and Morphine on Pain-Related Depression of Nestlet Shredding in Male and Female Mice. FRONTIERS IN PAIN RESEARCH 2021; 2:673940. [PMID: 34485976 PMCID: PMC8415797 DOI: 10.3389/fpain.2021.673940] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/10/2021] [Indexed: 12/30/2022] Open
Abstract
A primary goal in pain treatment is restoration of behaviors that are disrupted by pain. Measures of pain interference indicate the degree to which pain interferes with activities in pain patients, and these measures are used to evaluate the effects of analgesic drugs. As a result of the emphasis on the expression and treatment of functional impairment in clinical settings, preclinical pain researchers have attempted to develop procedures for evaluation of pain-related functional impairment in laboratory animals. The goal of the present study was to develop and validate a low cost procedure for the objective evaluation of pain-related depression of home cage behavior in mice. On test days, a 5 × 5 cm Nestlet was weighed prior to being suspended from the wire lid of the home cage of individually housed male and female ICR mice. Over the course of experimental sessions, mice removed pieces of the suspended Nestlet, and began to build a nest with the material they removed. Thus, the weight of the pieces of Nestlet that remained suspended at various time points in the session provided an indicator of the rate of this behavior. The results indicate that Nestlet shredding was stable with repeated testing, and shredding was depressed by intra-peritoneal injection of 0.32% lactic acid. The non-steroidal anti-inflammatory drug ketoprofen blocked 0.32% lactic acid-induced depression of shredding, but did not block depression of shredding by a pharmacological stimulus, the kappa opioid receptor agonist U69,593. The μ-opioid receptor agonist morphine did not block 0.32% lactic acid-induced depression of shredding when tested up to doses that depressed shredding in the absence of lactic acid. When noxious stimulus intensity was reduced by decreasing the lactic acid concentration to 0.18%, morphine was effective at blocking pain-related depression of behavior. In summary, the data from the present study support consideration of the Nestlet shredding procedure for use in studies examining mechanisms, expression, and treatment of pain-related functional impairment.
Collapse
Affiliation(s)
| | | | | | | | - Laurence L. Miller
- Department of Psychological Sciences, Augusta University, Augusta, GA, United States
| |
Collapse
|
15
|
Diester CM, Santos EJ, Moerke MJ, Negus SS. Behavioral Battery for Testing Candidate Analgesics in Mice. I. Validation with Positive and Negative Controls. J Pharmacol Exp Ther 2021; 377:232-241. [PMID: 33622770 PMCID: PMC8058504 DOI: 10.1124/jpet.120.000464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
This study evaluated a battery of pain-stimulated, pain-depressed, and pain-independent behaviors for preclinical pharmacological assessment of candidate analgesics in mice. Intraperitoneal injection of dilute lactic acid (IP acid) served as an acute visceral noxious stimulus to produce four pain-related behaviors in male and female ICR mice: stimulation of 1) stretching, 2) facial grimace, 3) depression of rearing, and 4) depression of nesting. Additionally, nesting and locomotion in the absence of the noxious stimulus were used to assess pain-independent drug effects. These six behaviors were used to compare effects of two mechanistically distinct but clinically effective positive controls (ketoprofen and oxycodone) and two negative controls that are not clinically approved as analgesics but produce either general motor depression (diazepam) or motor stimulation (amphetamine). We predicted that analgesics would alleviate all IP acid effects at doses that did not alter pain-independent behaviors, whereas negative controls would not. Consistent with this prediction, ketoprofen (0.1-32 mg/kg) produced the expected analgesic profile, whereas oxycodone (0.32-3.2 mg/kg) alleviated all IP acid effects except depression of rearing at doses lower than those that altered pain-independent behaviors. For the negative controls, diazepam (1-10 mg/kg) failed to block IP acid-induced depression of either rearing or nesting and only decreased IP acid-stimulated behaviors at doses that also decreased pain-independent behaviors. Amphetamine (0.32-3.2 mg/kg) alleviated all IP acid effects but only at doses that also stimulated locomotion. These results support utility of this model as a framework to evaluate candidate-analgesic effects in a battery of complementary pain-stimulated, pain-depressed, and pain-independent behavioral endpoints. SIGNIFICANCE STATEMENT: Preclinical assays of pain and analgesia often yield false-positive effects with candidate analgesics. This study used two positive-control analgesics (ketoprofen, oxycodone) and two active negative controls (diazepam, amphetamine) to validate a strategy for distinguishing analgesics from nonanalgesics by profiling drug effects in a battery of complementary pain-stimulated, pain-depressed, and pain-independent behaviors in male and female mice.
Collapse
Affiliation(s)
- C M Diester
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - E J Santos
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - M J Moerke
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - S S Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
16
|
A pain-induced tonic hypodopaminergic state augments phasic dopamine release in the nucleus accumbens. Pain 2021; 161:2376-2384. [PMID: 32453137 DOI: 10.1097/j.pain.0000000000001925] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Diseases and disorders such as Parkinson disease, schizophrenia, and chronic pain are characterized by altered mesolimbic dopaminergic neurotransmission. Dopamine release in the nucleus accumbens influences behavior through both tonic and phasic signaling. Tonic dopamine levels are hypothesized to inversely regulate phasic signals through dopamine D2 receptor feedback inhibition. We tested this hypothesis directly in the context of ongoing pain. Tonic and phasic dopamine signals were measured using fast-scan controlled-adsorption voltammetry and fast-scan cyclic voltammetry, respectively, in the nucleus accumbens shell of male rats with standardized levels of anesthesia. Application of capsaicin to the cornea produced a transient decrease in tonic dopamine levels. During the pain-induced hypodopaminergic state, electrically evoked phasic dopamine release was significantly increased when compared to baseline, evoked phasic release. A second application of capsaicin to the same eye had a lessened effect on tonic dopamine suggesting desensitization of TRPV1 channels in that eye. Capsaicin treatment in the alternate cornea, however, again produced coincident decreased dopaminergic tone and increased phasic dopamine release. These findings occurred independently of stimulus lateralization relative to the hemisphere of dopamine measurement. Our data show that (1) the mesolimbic dopamine circuit reliably encodes acute noxious stimuli; (2) ongoing pain produces decreases in dopaminergic tone; and (3) pain-induced decreases in tonic dopamine correspond to augmented evoked phasic dopamine release. Enhanced phasic dopamine neurotransmission resulting from salient stimuli may contribute to increased impulsivity and cognitive deficits often observed in conditions associated with decreased dopaminergic tone, including Parkinson disease and chronic pain.
Collapse
|
17
|
Negus SS, Marsh SA, Townsend EA. Resistance of Food-Maintained Operant Responding to Mechanical Punishment in Rats: Further Evidence for Weak "Affective/Motivational Pain" in Rat Models of Inflammatory and Neuropathic Pain. Front Pharmacol 2021; 11:615782. [PMID: 33584295 PMCID: PMC7878562 DOI: 10.3389/fphar.2020.615782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/29/2020] [Indexed: 01/25/2023] Open
Abstract
Clinically relevant chronic pain is often associated with functional impairment and behavioral depression as an “affective/motivational” sign of pain; however preclinical animal models of inflammatory and neuropathic pain often produce weak evidence of impaired function. We hypothesized that hindpaw mechanical stimulation produced by a requirement to rear on a textured “NOX” plate would punish operant responding in rats treated with intraplantar complete Freund’s adjuvant (CFA, a model of inflammatory pain) or the chemotherapeutic paclitaxel (PTX, a model of neuropathic pain) and produce sustained pain-related depression of operant behavior. Male Sprague–Dawley rats were trained under a progressive-ratio (PR) schedule of food-maintained operant responding, then treated with CFA (100 µL in left hindpaw), PTX (2.0 mg/kg IP on alternate days for four total injections; 6.6 mg/kg IV on alternate days for three total injections), or saline vehicle. PR break points and mechanical thresholds for paw withdrawal from von Frey filaments were then tracked for 28 days. Subsequently, rats were tested with the opioid receptor antagonist naltrexone to assess latent sensitization and with the kappa opioid receptor (KOR) agonist U69593 to assess KOR function. CFA produced significant mechanical hypersensitivity for 3 weeks but decreased PR breakpoints for only 1 day. Both IP and IV PTX produced mechanical hypersensitivity for at least three weeks; however, only IV PTX decreased PR breakpoints, and this decrease was not alleviated by morphine. After recovery, naltrexone reinstated mechanical hypersensitivity in CFA- but not PTX-treated rats, and it did not reinstate depression of breakpoints in any group. U69593 dose-dependently decreased PR breakpoints in all groups with no difference between control vs. CFA/PTX groups. These results suggest that rearing on a textured NOX plate was not sufficient to punish operant responding in CFA- and PTX-treated rats despite the presence of sustained mechanical hypersensitivity. The rapid recovery of operant responding could not be attributed to latent sensitization, KOR downregulation, or behavioral tolerance. These results extend the range of conditions under which putative chronic pain manipulations produce weak evidence for depression of operant responding as a sign of the “affective/motivational” component of pain in rats.
Collapse
Affiliation(s)
- S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - S A Marsh
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| | - E A Townsend
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
18
|
Han QQ, Yin M, Wang ZY, Liu H, Ao JP, Wang YX. Cynandione A Alleviates Neuropathic Pain Through α7-nAChR-Dependent IL-10/β-Endorphin Signaling Complexes. Front Pharmacol 2021; 11:614450. [PMID: 33584292 PMCID: PMC7873367 DOI: 10.3389/fphar.2020.614450] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022] Open
Abstract
Cynandione A, an acetophenone isolated from Cynanchum Wilfordii Radix, exhibits antineuropathic pain effect. This study further explored the target molecule and signaling mechanisms underlying cynandione-A-induced antineuropathic pain. Intrathecal injection of cynandione A significantly attenuated mechanical allodynia in neuropathic rats and substantially increased spinal expression of IL-10 and β-endorphin but not dynorphin A. Cynandione A treatment also enhanced expression of IL-10 and β-endorphin but not α7 nicotinic acetylcholine receptors (nAChRs) in cultured microglia. The IL-10 antibody attenuated cynandione-A-induced spinal or microglial gene expression of β-endorphin and mechanical allodynia, whereas the β-endorphin antiserum blocked cynandione-A-induced mechanical antiallodynia but not spinal or microglial IL-10 gene expression. The α7 nAChR antagonist methyllycaconitine significantly reduced cynandione-A-induced mechanical antiallodynia and spinal or microglial expression of IL-10 and β-endorphin. Furthermore, cynandione A stimulated microglial phosphorylation of PKA, p38, and CREB in an α7-nAChR-dependent manner, and treatment with their inhibitors attenuated cynandione-A-induced mechanical antiallodynia and spinal or microglial expression of IL-10 and β-endorphin. In addition, cynandione A stimulated spinal phosphorylation of the transcription factor STAT3, which was inhibited by methyllycaconitine, the PKA activation inhibitor or IL-10 antibody. The STAT3 inhibitor NSC74859 also abolished cynandione-A-induced mechanical antiallodynia and spinal expression of β-endorphin. These findings suggest that cynandione A suppresses neuropathic pain through α7-nAChR-dependent IL-10/β-endorphin signaling pathway in spinal microglia.
Collapse
Affiliation(s)
- Qiao-Qiao Han
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai, China
| | - Min Yin
- Jiangsu Key Laboratory for the Research and Utilization of Plants Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing, China
| | - Zi-Ying Wang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai, China
| | - Hao Liu
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai, China
| | - Jun-Ping Ao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yong-Xiang Wang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, Shanghai, China
| |
Collapse
|
19
|
Abstract
Pain is complex and is a unique experience for individuals in that no two people will have exactly the same physiological and emotional response to the same noxious stimulus or injury. Pain is composed of two essential processes: a sensory component that allows for discrimination of the intensity and location of a painful stimulus and an emotional component that underlies the affective, motivational, unpleasant, and aversive response to a painful stimulus. Kappa opioid receptor (KOR) activation in the periphery and throughout the neuroaxis modulates both of these components of the pain experience. In this chapter we focus on recent findings that KORs contribute to the emotional, aversive nature of chronic pain, including how expression in the limbic circuitry contributes to anhedonic states and components of opioid misuse disorder. While the primary focus is on preclinical pain models, we also highlight clinical or human research where there is strong evidence for KOR involvement in negative affective states associated with chronic pain and opioid misuse.
Collapse
|
20
|
Koob GF. Drug Addiction: Hyperkatifeia/Negative Reinforcement as a Framework for Medications Development. Pharmacol Rev 2021; 73:163-201. [PMID: 33318153 PMCID: PMC7770492 DOI: 10.1124/pharmrev.120.000083] [Citation(s) in RCA: 193] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Compulsive drug seeking that is associated with addiction is hypothesized to follow a heuristic framework that involves three stages (binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation) and three domains of dysfunction (incentive salience/pathologic habits, negative emotional states, and executive function, respectively) via changes in the basal ganglia, extended amygdala/habenula, and frontal cortex, respectively. This review focuses on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the addiction cycle. Hyperkatifeia provides an additional source of motivation for compulsive drug seeking via negative reinforcement. Negative reinforcement reflects an increase in the probability of a response to remove an aversive stimulus or drug seeking to remove hyperkatifeia that is augmented by genetic/epigenetic vulnerability, environmental trauma, and psychiatric comorbidity. Neurobiological targets for hyperkatifeia in addiction involve neurocircuitry of the extended amygdala and its connections via within-system neuroadaptations in dopamine, enkephalin/endorphin opioid peptide, and γ-aminobutyric acid/glutamate systems and between-system neuroadaptations in prostress corticotropin-releasing factor, norepinephrine, glucocorticoid, dynorphin, hypocretin, and neuroimmune systems and antistress neuropeptide Y, nociceptin, endocannabinoid, and oxytocin systems. Such neurochemical/neurocircuitry dysregulations are hypothesized to mediate a negative hedonic set point that gradually gains allostatic load and shifts from a homeostatic hedonic state to an allostatic hedonic state. Based on preclinical studies and translational studies to date, medications and behavioral therapies that reset brain stress, antistress, and emotional pain systems and return them to homeostasis would be promising new targets for medication development. SIGNIFICANCE STATEMENT: The focus of this review is on neurochemical/neurocircuitry dysregulations that contribute to hyperkatifeia, defined as a greater intensity of negative emotional/motivational signs and symptoms during withdrawal from drugs of abuse in the withdrawal/negative affect stage of the drug addiction cycle and a driving force for negative reinforcement in addiction. Medications and behavioral therapies that reverse hyperkatifeia by resetting brain stress, antistress, and emotional pain systems and returning them to homeostasis would be promising new targets for medication development.
Collapse
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
21
|
Abstract
Preclinical models that assess "pain" in rodents typically measure increases in behaviors produced by a "pain stimulus." A large literature exists showing that kappa opioid receptor (KOR) agonists can decrease these "pain-stimulated behaviors" following many different pain stimuli. Despite showing apparent antinociceptive properties in these preclinical models, KOR agonists failed as analgesics in clinical trials. Recent studies that assessed decreases in behavior due to a pain stimulus show that KOR agonists are not effective in restoring these "pain-depressed behaviors" to normal levels, which agrees with the lack of effectiveness for KOR agonists in clinical trials. One current explanation for the failure of previous KOR agonists in clinical trials is that those agonists activated beta-arrestin signaling and that KOR agonists with a greater bias for G protein signaling will be more successful. However, neither G protein-biased agonists nor beta-arrestin-biased agonists are very effective in assays of pain-depressed behavior, which suggests that novel biased agonists may still not be effective analgesics. This review provides a concise account of the effectiveness of KOR agonists in preclinical models of pain-stimulated and pain-depressed behaviors following the administration of different pain stimuli. Based on the previous results, it may be appropriate to include both behaviors when testing the analgesic potential of KOR agonists.
Collapse
Affiliation(s)
- Matthew F Lazenka
- Department of Biology and Chemistry, Liberty University, Lynchburg, VA, USA.
| |
Collapse
|
22
|
Parker KE, Sugiarto E, Taylor AMW, Pradhan AA, Al-Hasani R. Pain, Motivation, Migraine, and the Microbiome: New Frontiers for Opioid Systems and Disease. Mol Pharmacol 2020; 98:433-444. [PMID: 32958571 PMCID: PMC7562975 DOI: 10.1124/mol.120.119438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022] Open
Abstract
For decades the broad role of opioids in addiction, neuropsychiatric disorders, and pain states has been somewhat well established. However, in recent years, with the rise of technological advances, not only is the existing dogma being challenged, but we are identifying new disease areas in which opioids play a critical role. This review highlights four new areas of exploration in the opioid field. The most recent addition to the opioid family, the nociceptin receptor system, shows promise as the missing link in understanding the neurocircuitry of motivation. It is well known that activation of the kappa opioid receptor system modulates negative affect and dysphoria, but recent studies now implicate the kappa opioid system in the modulation of negative affect associated with pain. Opioids are critical in pain management; however, the often-forgotten delta opioid receptor system has been identified as a novel therapeutic target for headache disorders and migraine. Lastly, changes to the gut microbiome have been shown to directly contribute to many of the symptoms of chronic opioid use and opioid related behaviors. This review summarizes the findings from each of these areas with an emphasis on identifying new therapeutic targets. SIGNIFICANCE STATEMENT: The focus of this minireview is to highlight new disease areas or new aspects of disease in which opioids have been implicated; this includes pain, motivation, migraine, and the microbiome. In some cases, this has resulted in the pursuit of a novel therapeutic target and resultant clinical trial. We believe this is very timely and will be a refreshing take on reading about opioids and disease.
Collapse
Affiliation(s)
- Kyle E Parker
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Elizabeth Sugiarto
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Anna M W Taylor
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Amynah A Pradhan
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| | - Ream Al-Hasani
- Department of Anesthesiology and Washington University Pain Center, Washington University in St. Louis, Missouri (K.E.P, R.A.-H.); Center for Clinical Pharmacology, Washington University School of Medicine, St. Louis, Missouri (K.E.P., R.A.-H.); Department of Pharmacology, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada (E.S., A.M.W.T.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (A.A.P.); and St. Louis College of Pharmacy, St. Louis, Missouri (R.A.-H.)
| |
Collapse
|
23
|
Legakis LP, Karim-Nejad L, Negus SS. Effects of repeated treatment with monoamine-transporter-inhibitor antidepressants on pain-related depression of intracranial self-stimulation in rats. Psychopharmacology (Berl) 2020; 237:2201-2212. [PMID: 32382785 PMCID: PMC7308219 DOI: 10.1007/s00213-020-05530-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 04/17/2020] [Indexed: 12/20/2022]
Abstract
RATIONALE Synaptic neurotransmission with dopamine (DA), norepinephrine (NE), and serotonin (5-HT) is terminated primarily by reuptake into presynaptic terminals via the DA, NE, and 5-HT transporters (DAT/NET/SERT, respectively). Monoamine transporter inhibitors constitute one class of drugs used to treat both depression and pain, and therapeutic effects by these compounds often require repeated treatment for days or weeks. OBJECTIVES The present study compared antinociceptive effects produced by repeated treatment with monoamine transporter inhibitors in a preclinical assay of pain-related depression of positively reinforced operant responding. METHODS Adult Sprague-Dawley rats equipped with microelectrodes targeting a brain-reward area responded for pulses of electrical brain stimulation in an intracranial self-stimulation (ICSS) procedure. Intraperitoneal injection of dilute lactic acid served as a noxious stimulus that repeatedly depressed ICSS and also produced weight loss during 7 days of repeated acid administration. RESULTS Acid-induced depression of both ICSS and body weight were completely blocked by repeated pretreatment with the nonsteroidal anti-inflammatory drug ketorolac. The DAT-selective inhibitor bupropion also fully blocked acid-induced ICSS depression and weight loss throughout all 7 days of treatment. The NET-selective inhibitor nortriptyline and the SERT-selective inhibitor citalopram were generally less effective, but both drugs blocked acid-induced ICSS depression by the end of the 7-day treatment. Acid-induced depression of ICSS and body weight were not blocked by the kappa opioid receptor (KOR) agonist U69593 or the KOR antagonist norbinaltorphimine. CONCLUSIONS These results support effectiveness of bupropion to alleviate signs of pain-related behavioral depression in rats and further suggest that nortriptyline and citalopram produce significant but less reliable effects.
Collapse
Affiliation(s)
- L P Legakis
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - L Karim-Nejad
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - S S Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
24
|
Mokhtari T, Tu Y, Hu L. Involvement of the hippocampus in chronic pain and depression. BRAIN SCIENCE ADVANCES 2020. [DOI: 10.26599/bsa.2019.9050025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Increases in depressive behaviors have been reported in patients experiencing chronic pain. In these patients, the symptoms of pain and depression commonly coexist, impairing their lives and challenging effective treatment. The hippocampus may play a role in both chronic pain and depression. A reduction in the volume of the hippocampus is related to reduced neurogenesis and neuroplasticity in cases of chronic pain and depression. Moreover, an increase of proinflammatory factors and a reduction of neurotrophic factors have been reported to modulate the hippocampal neurogenesis and neuroplasticity in chronic pain and depression. This review discusses the mechanisms underlying the depressive-like behavior accompanying chronic pain, emphasizing the structural and functional changes in the hippocampus. We also discuss the hypothesis that pro-inflammatory factors and neurotrophic factors expressed in the hippocampus may serve as a therapeutic target for comorbid chronic pain and depression.
Collapse
Affiliation(s)
- Tahmineh Mokhtari
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yiheng Tu
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Li Hu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
25
|
Attenuated dopamine receptor signaling in nucleus accumbens core in a rat model of chemically-induced neuropathy. Neuropharmacology 2020; 166:107935. [PMID: 31917153 DOI: 10.1016/j.neuropharm.2020.107935] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/10/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023]
Abstract
Neuropathy is major source of chronic pain that can be caused by mechanically or chemically induced nerve injury. Intraplantar formalin injection produces local necrosis over a two-week period and has been used to model neuropathy in rats. To determine whether neuropathy alters dopamine (DA) receptor responsiveness in mesolimbic brain regions, we examined dopamine D1-like and D2-like receptor (D1/2R) signaling and expression in male rats 14 days after bilateral intraplantar formalin injections into both rear paws. D2R-mediated G-protein activation and expression of the D2R long, but not short, isoform were reduced in nucleus accumbens (NAc) core, but not in NAc shell, caudate-putamen or ventral tegmental area of formalin- compared to saline-treated rats. In addition, D1R-stimulated adenylyl cyclase activity was also reduced in NAc core, but not in NAc shell or prefrontal cortex, of formalin-treated rats, whereas D1R expression was unaffected. Other proteins involved in dopamine neurotransmission, including dopamine uptake transporter and tyrosine hydroxylase, were unaffected by formalin treatment. In behavioral tests, the potency of a D2R agonist to suppress intracranial self-stimulation (ICSS) was decreased in formalin-treated rats, whereas D1R agonist effects were not altered. The combination of reduced D2R expression and signaling in NAc core with reduced suppression of ICSS responding by a D2R agonist suggest a reduction in D2 autoreceptor function. Altogether, these results indicate that intraplantar formalin produces attenuation of highly specific DA receptor signaling processes in NAc core of male rats and suggest the development of a neuropathy-induced allostatic state in both pre- and post-synaptic DA receptor function.
Collapse
|
26
|
Jacobson ML, Browne CA, Lucki I. Kappa Opioid Receptor Antagonists as Potential Therapeutics for Stress-Related Disorders. Annu Rev Pharmacol Toxicol 2020; 60:615-636. [DOI: 10.1146/annurev-pharmtox-010919-023317] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Exposure to stressful stimuli activates kappa opioid receptor (KOR) signaling, a process known to produce aversion and dysphoria in humans and other species. This endogenous opioid system is dysregulated in stress-related disorders, specifically in major depressive disorder (MDD). These findings serve as the foundation for a growing interest in the therapeutic potential of KOR antagonists as novel antidepressants. In this review, data supporting the hypothesis of dysregulated KOR function in MDD are considered. The clinical data demonstrating the therapeutic efficacy and safety of selective and mixed opioid antagonists are then presented. Finally, the preclinical evidence illustrating the induction of behaviors relevant to the endophenotypes of MDD and KOR antagonist activity in stress-naïve and stress-exposed animals is evaluated. Overall, this review highlights the emergent literature supporting the pursuit of KOR antagonists as novel therapeutics for MDD and other stress-related disorders.
Collapse
Affiliation(s)
- Moriah L. Jacobson
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Caroline A. Browne
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Irwin Lucki
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| |
Collapse
|
27
|
Abstract
Substance use disorders represent a global public health issue. This mental health disorder is hypothesized to result from neurobiological changes as a result of chronic drug exposure and clinically manifests as inappropriate behavioral allocation toward the procurement and use of the abused substance and away from other behaviors maintained by more adaptive nondrug reinforcers (e.g., social relationships, work). The dynorphin/kappa-opioid receptor (KOR) is one receptor system that has been altered following chronic exposure to drugs of abuse (e.g., cocaine, opioids, alcohol) in both laboratory animals and humans, implicating the dynorphin/KOR system in the expression, mechanisms, and treatment of substance use disorders. KOR antagonists have reduced drug self-administration in laboratory animals under certain experimental conditions, but not others. Recently, several human laboratory and clinical trials have evaluated the effectiveness of KOR antagonists as candidate pharmacotherapies for cocaine or tobacco use disorder to test hypotheses generated from preclinical studies. KOR antagonists failed to significantly alter drug use metrics in humans suggesting translational discordance between some preclinical drug self-administration studies and consistent with other preclinical drug self-administration studies that provide concurrent access to an alternative nondrug reinforcer (e.g., food). The implications of this translational discordance and future directions for examining the therapeutic potential of KOR agonists or antagonists as candidate substance use disorder pharmacotherapies are discussed.
Collapse
Affiliation(s)
- Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
28
|
Missig G, Robbins JO, Mokler EL, McCullough KM, Bilbo SD, McDougle CJ, Carlezon WA. Sex-dependent neurobiological features of prenatal immune activation via TLR7. Mol Psychiatry 2020; 25:2330-2341. [PMID: 30610201 PMCID: PMC7515834 DOI: 10.1038/s41380-018-0346-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/16/2018] [Accepted: 12/10/2018] [Indexed: 12/30/2022]
Abstract
Immune activation during pregnancy via infection or autoimmune disease is a risk factor for neuropsychiatric illness. Mouse models of prenatal immune activation often involve maternal administration of agents that activate toll-like receptors (TLRs), a class of pattern recognition receptors that initiate innate immune responses. Such studies have focused primarily on activating the TLR3 or TLR4 subtypes, to mimic immune responses to viral or bacterial infections, respectively. Here, we characterize the effects of prenatal activation of TLR7, which is implicated in the pathogenesis of autoimmune disease. Prenatal TLR7 activation via administration of the selective agonist imiquimod (5.0 mg/kg) induces a phenotype in offspring characterized by reduced anxiety-like behavior, fragmented social behavior, and altered ultrasonic vocalization patterns at 6-12 weeks of age. The characteristics of this phenotype are readily distinguishable from-and in some ways opposite to-those seen following prenatal activation of TLR3 and/or TLR4. Prenatal TLR7-activated mice have normal baseline locomotor activity, but are hyperresponsive to stimuli including social partners, circadian cues, and gonadal hormone fluctuations. These alterations are accompanied by decreases in microglia density but increases in ramifications. RNA-sequencing of dorsal striatum, a region showing profound changes in microglial markers, indicates that prenatal TLR7 activation induces differential expression of hundreds of genes at 13 weeks of age, with virtually no overlap in differentially expressed genes between males and females. Our findings demonstrate that prenatal immune activation can promote a wide range of developmental trajectories, depending on the type and/or pattern of TLR activation and the sex of the offspring.
Collapse
Affiliation(s)
- Galen Missig
- grid.240206.20000 0000 8795 072XBasic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA USA
| | - James O. Robbins
- grid.240206.20000 0000 8795 072XBasic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA USA
| | - Emery L. Mokler
- grid.240206.20000 0000 8795 072XBasic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA USA
| | - Kenneth M. McCullough
- grid.240206.20000 0000 8795 072XBasic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA USA
| | - Staci D. Bilbo
- grid.32224.350000 0004 0386 9924Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA USA ,grid.38142.3c000000041936754XDepartment of Pediatrics, Harvard Medical School, Lexington, MA USA
| | - Christopher J. McDougle
- grid.32224.350000 0004 0386 9924Lurie Center for Autism, Massachusetts General Hospital, Lexington, MA USA ,grid.38142.3c000000041936754XDepartment of Psychiatry, Harvard Medical School, Lexington, MA USA
| | - William A. Carlezon
- grid.240206.20000 0000 8795 072XBasic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA USA
| |
Collapse
|
29
|
Bechara A, Berridge KC, Bickel WK, Morón JA, Williams SB, Stein JS. A Neurobehavioral Approach to Addiction: Implications for the Opioid Epidemic and the Psychology of Addiction. Psychol Sci Public Interest 2019; 20:96-127. [PMID: 31591935 PMCID: PMC7001788 DOI: 10.1177/1529100619860513] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Two major questions about addictive behaviors need to be explained by any worthwhile neurobiological theory. First, why do people seek drugs in the first place? Second, why do some people who use drugs seem to eventually become unable to resist drug temptation and so become "addicted"? We will review the theories of addiction that address negative-reinforcement views of drug use (i.e., taking opioids to alleviate distress or withdrawal), positive-reinforcement views (i.e., taking drugs for euphoria), habit views (i.e., growth of automatic drug-use routines), incentive-sensitization views (i.e., growth of excessive "wanting" to take drugs as a result of dopamine-related sensitization), and cognitive-dysfunction views (i.e., impaired prefrontal top-down control), including those involving competing neurobehavioral decision systems (CNDS), and the role of the insula in modulating addictive drug craving. In the special case of opioids, particular attention is paid to whether their analgesic effects overlap with their reinforcing effects and whether the perceived low risk of taking legal medicinal opioids, which are often prescribed by a health professional, could play a role in the decision to use. Specifically, we will address the issue of predisposition or vulnerability to becoming addicted to drugs (i.e., the question of why some people who experiment with drugs develop an addiction, while others do not). Finally, we review attempts to develop novel therapeutic strategies and policy ideas that could help prevent opioid and other substance abuse.
Collapse
Affiliation(s)
- Antoine Bechara
- Department of Psychology, University of Southern California
- Brain and Creativity Institute, University of Southern California
| | | | - Warren K. Bickel
- Addiction Recovery Research Center & Center for Transformational Research on Health Behaviors, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia
| | - Jose A. Morón
- Department of Anesthesiology, Washington University School of Medicine
- Washington University Pain Center, Washington University School of Medicine
| | - Sidney B. Williams
- Department of Anesthesiology, Washington University School of Medicine
- Washington University Pain Center, Washington University School of Medicine
| | - Jeffrey S. Stein
- Addiction Recovery Research Center & Center for Transformational Research on Health Behaviors, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia
| |
Collapse
|
30
|
Margolis EB, Karkhanis AN. Dopaminergic cellular and circuit contributions to kappa opioid receptor mediated aversion. Neurochem Int 2019; 129:104504. [PMID: 31301327 DOI: 10.1016/j.neuint.2019.104504] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 07/07/2019] [Accepted: 07/10/2019] [Indexed: 01/05/2023]
Abstract
Neural circuits that enable an organism to protect itself by promoting escape from immediate threat and avoidance of future injury are conceptualized to carry an "aversive" signal. One of the key molecular elements of these circuits is the kappa opioid receptor (KOR) and its endogenous peptide agonist, dynorphin. In many cases, the aversive response to an experimental manipulation can be eliminated by selective blockade of KOR function, indicating its necessity in transmitting this signal. The dopamine system, through its contributions to reinforcement learning, is also involved in processing of aversive stimuli, and KOR control of dopamine in the context of aversive behavioral states has been intensely studied. In this review, we have discussed the multiple ways in which the KORs regulate dopamine dynamics with a central focus on dopamine neurons and projections from the ventral tegmental area. At the neuronal level, KOR agonists inhibit dopamine neurons both in the somatodendritic region as well as at terminal release sites, through various signaling pathways and ion channels, and these effects are specific to different synaptic sites. While the dominant hypotheses are that aversive states are driven by decreases in dopamine and increases in dynorphin, reported exceptions to these patterns indicate these ideas require refinement. This is critical given that KOR is being considered as a target for development of new therapeutics for anxiety, depression, pain, and other psychiatric disorders.
Collapse
Affiliation(s)
- Elyssa B Margolis
- Department of Neurology, Alcohol and Addiction Research Group, University of California, San Francisco, 675 Nelson Rising Lane, Box 0444, San Francisco, CA, 94143, USA.
| | - Anushree N Karkhanis
- Department of Psychology, Developmental Exposure Alcohol Research Center, Center for Developmental and Behavioral Neuroscience, Binghamton University - SUNY, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA.
| |
Collapse
|
31
|
Butelman ER, McElroy BD, Prisinzano TE, Kreek MJ. Impact of Pharmacological Manipulation of the κ-Opioid Receptor System on Self-grooming and Anhedonic-like Behaviors in Male Mice. J Pharmacol Exp Ther 2019; 370:1-8. [PMID: 30975792 PMCID: PMC6538891 DOI: 10.1124/jpet.119.256354] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/09/2019] [Indexed: 12/11/2022] Open
Abstract
The kappa (κ) opioid receptor/dynorphin system modulates depression-like states and anhedonia, as well adaptations to stress and exposure to drugs of abuse. Several relatively short-acting small molecule κ-receptor antagonists have been synthesized, and their behavioral profile has been examined under some conditions. The hypothesis of this study is that pharmacological manipulations of the κ-receptor system will result in changes in ethologically relevant anhedonic-like behaviors in mice. Adult male C57BL/6j mice (n = 6-8) were examined for self-grooming behavior in the splash test (in which robust self-grooming is elicited by spraying the dorsum of the mouse with a sucrose solution). The κ-agonist salvinorin A (0.56-1.8 mg/kg) produced dose-dependent decreases in self-grooming, a marker of anhedonia. The selectivity, potency, and duration of action of two relatively short-acting κ-antagonists, LY2444296 [(S)-3-fluoro-4-(4-((2-(3-fluorophenyl) pyrrolidin-1-yl)methyl)phenoxy)benzamide] and LY2795050 [3-chloro-4-(4-(((2S)-2-pyridin-3-ylpyrrolidin-1-yl)methyl) phenoxy)benzamide], were studied for their effectiveness in preventing grooming deficits caused by salvinorin A (1.8 mg/kg). κ-selective doses of both LY2444296 (0.032-1 mg/kg) and LY2795050 (0.032-0.32 mg/kg) dose- and time-dependently prevented the grooming deficits caused by salvinorin A (1.8 m/kg). We also found that a κ-selective dose of each of these antagonists decreased immobility in the forced swim test, a common test of anti-anhedonia effects. This study shows that the κ-receptor system is involved in an ethologically relevant measure of anhedonia, and that κ-selective doses of these antagonists can produce effects consistent with rapid anti-anhedonia. SIGNIFICANCE STATEMENT: Activation of the κ-opioid receptor system results in grooming deficits in mice, an ethologically relevant marker of anhedonia. Shorter acting κ-antagonists are able to cause effects consistent with rapid antianhedonia.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Bryan D McElroy
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Thomas E Prisinzano
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| |
Collapse
|
32
|
Moerke MJ, Negus SS. Interactions between pain states and opioid reward assessed with intracranial self-stimulation in rats. Neuropharmacology 2019; 160:107689. [PMID: 31271771 DOI: 10.1016/j.neuropharm.2019.107689] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 06/19/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
Opioids are an essential component of current clinical treatments for pain, but they also produce side effects that include abuse liability. Recent media attention surrounding the use of opioids in the United States has elevated the discussion of their benefits and drawbacks to one of national concern, leading to increased scrutiny of prescribing practices. Regulatory agencies have responded by recommending stricter limits on the amount and duration of opioid prescriptions for pain treatment; however, the relationship between pain states and the abuse-related effects of opioids is still not completely understood. Intracranial self-stimulation (ICSS) is one preclinical procedure that can be used to study the abuse-related effects of opioids in naïve subjects over the course of initial opioid exposure and in the context of inferred pain states. The goal of this review is to provide a summary of evidence from our laboratory using ICSS to study the modulation of opioid reward by pain states and examine these results in the context of related studies from other groups. This article is part of the Special Issue entitled 'New Vistas in Opioid Pharmacology'.
Collapse
Affiliation(s)
- Megan J Moerke
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N 12th Street, Richmond, VA, 23298, USA
| | - S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 N 12th Street, Richmond, VA, 23298, USA.
| |
Collapse
|
33
|
Abstract
All preclinical procedures for analgesic drug discovery involve two components: 1) a "pain stimulus" (the principal independent variable), which is delivered to an experimental subject with the intention of producing a pain state; and 2) a "pain behavior" (the principal dependent variable), which is measured as evidence of that pain state. Candidate analgesics are then evaluated for their effectiveness to reduce the pain behavior, and results are used to prioritize drugs for advancement to clinical testing. This review describes a taxonomy of preclinical procedures organized into an "antinociception matrix" by reference to their types of pain stimulus (noxious, inflammatory, neuropathic, disease related) and pain behavior (unconditioned, classically conditioned, operant conditioned). Particular emphasis is devoted to pain behaviors and the behavioral principals that govern their expression, pharmacological modulation, and preclinical-to-clinical translation. Strengths and weaknesses are compared and contrasted for procedures using each type of behavioral outcome measure, and the following four recommendations are offered to promote strategic use of these procedures for preclinical-to-clinical analgesic drug testing. First, attend to the degree of homology between preclinical and clinical outcome measures, and use preclinical procedures with behavioral outcome measures homologous to clinically relevant outcomes in humans. Second, use combinations of preclinical procedures with complementary strengths and weaknesses to optimize both sensitivity and selectivity of preclinical testing. Third, take advantage of failed clinical translation to identify drugs that can be back-translated preclinically as active negative controls. Finally, increase precision of procedure labels by indicating both the pain stimulus and the pain behavior in naming preclinical procedures.
Collapse
Affiliation(s)
- S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
34
|
Abstract
Pain has a useful protective role; through avoidance learning, it helps to decrease the probability of engaging in tissue-damaging, or otherwise dangerous experiences. In our modern society, the experience of acute post-surgical pain and the development of chronic pain states represent an unnecessary negative outcome. This has become an important health issue as more than 30% of the US population reports experiencing "unnecessary" pain at any given time. Opioid therapies are often efficacious treatments for severe and acute pain; however, in addition to their powerful analgesic properties, opioids produce potent reinforcing properties and their inappropriate use has led to the current opioid overdose epidemic in North America. Dissecting the allostatic changes occurring in nociceptors and neuronal pathways in response to pain are the first and most important steps in understanding the physiologic changes underlying the opioid epidemic. Full characterization of these adaptations will provide novel targets for the development of safer pharmacotherapies. In this review, we highlight the current efforts toward safer opioid treatments and describe our current knowledge of the interaction between pain and opioid systems.
Collapse
Affiliation(s)
- Nicolas Massaly
- Department of Anesthesiology; Washington University in St. Louis; St. Louis, MO, 63110 ; USA
- Washington University Pain Center; St. Louis, MO, 63110 ; USA
- Washington University in St Louis; School of Medicine; St. Louis, MO, 63110 ; USA
| | - Jose A Morón
- Department of Anesthesiology; Washington University in St. Louis; St. Louis, MO, 63110 ; USA
- Washington University Pain Center; St. Louis, MO, 63110 ; USA
- Washington University in St Louis; School of Medicine; St. Louis, MO, 63110 ; USA
- Department of Neuroscience; Washington University in St. Louis; St. Louis, MO, 63110 ; USA
| |
Collapse
|
35
|
Massaly N, Copits BA, Wilson-Poe AR, Hipólito L, Markovic T, Yoon HJ, Liu S, Walicki MC, Bhatti DL, Sirohi S, Klaas A, Walker BM, Neve R, Cahill CM, Shoghi KI, Gereau RW, McCall JG, Al-Hasani R, Bruchas MR, Morón JA. Pain-Induced Negative Affect Is Mediated via Recruitment of The Nucleus Accumbens Kappa Opioid System. Neuron 2019; 102:564-573.e6. [PMID: 30878290 DOI: 10.1016/j.neuron.2019.02.029] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 09/20/2018] [Accepted: 02/14/2019] [Indexed: 12/26/2022]
Abstract
Negative affective states affect quality of life for patients suffering from pain. These maladaptive emotional states can lead to involuntary opioid overdose and many neuropsychiatric comorbidities. Uncovering the mechanisms responsible for pain-induced negative affect is critical in addressing these comorbid outcomes. The nucleus accumbens (NAc) shell, which integrates the aversive and rewarding valence of stimuli, exhibits plastic adaptations in the presence of pain. In discrete regions of the NAc, activation of the kappa opioid receptor (KOR) decreases the reinforcing properties of rewards and induces aversive behaviors. Using complementary techniques, we report that in vivo recruitment of NAc shell dynorphin neurons, acting through KOR, is necessary and sufficient to drive pain-induced negative affect. Taken together, our results provide evidence that pain-induced adaptations in the kappa opioid system within the NAc shell represent a functional target for therapeutic intervention that could circumvent pain-induced affective disorders. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Nicolas Massaly
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Bryan A Copits
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Adrianne R Wilson-Poe
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Lucia Hipólito
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Valencia 46100, Spain
| | - Tamara Markovic
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Hye Jean Yoon
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Shiwei Liu
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marie C Walicki
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO 63110, USA; Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Dionnet L Bhatti
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Sunil Sirohi
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA
| | - Amanda Klaas
- Department of Radiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Brendan M Walker
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA
| | - Rachael Neve
- Department of Brain and Cognitive Science, Viral Gene Transfer Core, MIT, Cambridge, MA 02139-4307, USA
| | - Catherine M Cahill
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kooresh I Shoghi
- Department of Radiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Robert W Gereau
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jordan G McCall
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO 63110, USA; Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Ream Al-Hasani
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO 63110, USA; Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA.
| | - Michael R Bruchas
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110, USA.
| | - Jose A Morón
- Department of Anesthesiology, Washington University Pain Center, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
36
|
Kappa Opioid Receptors Drive a Tonic Aversive Component of Chronic Pain. J Neurosci 2019; 39:4162-4178. [PMID: 30862664 DOI: 10.1523/jneurosci.0274-19.2019] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 02/05/2019] [Indexed: 12/22/2022] Open
Abstract
Pain is a multidimensional experience and negative affect, or how much the pain is "bothersome", significantly impacts the sufferers' quality of life. It is well established that the κ opioid system contributes to depressive and dysphoric states, but whether this system contributes to the negative affect precipitated by the occurrence of chronic pain remains tenuous. Using a model of persistent pain, we show by quantitative real-time-PCR, florescence in situ hybridization, Western blotting and GTPgS autoradiography an upregulation of expression and the function of κ opioid receptors (KORs) and its endogenous ligand dynorphin in the mesolimbic circuitry in animals with chronic pain compared with surgical controls. Using in vivo microdialysis and microinjection of drugs into the mesolimbic dopamine system, we demonstrate that inhibiting KORs reinstates evoked dopamine release and reward-related behaviors in chronic pain animals. Chronic pain enhanced KOR agonist-induced place aversion in a sex-dependent manner. Using various place preference paradigms, we show that activation of KORs drives pain aversive states in male but not female mice. However, KOR antagonist treatment was effective in alleviating anxiogenic and depressive affective-like behaviors in both sexes. Finally, ablation of KORs from dopamine neurons using AAV-TH-cre in KORloxP mice prevented pain-induced aversive states as measured by place aversion assays. Our results strongly support the use of KOR antagonists as therapeutic adjuvants to alleviate the emotional, tonic-aversive component of chronic pain, which is argued to be the most significant component of the pain experience that impacts patients' quality of life.SIGNIFICANCE STATEMENT We show that KORs are sufficient to drive the tonic-aversive component of chronic pain; the emotional component of pain that is argued to significantly impact a patient's quality of life. The impact of our study is broadly relevant to affective disorders associated with disruption of reward circuitry and thus likely contributes to many of the devastating sequelae of chronic pain, including the poor response to treatment of many patients, debilitating affective disorders (other disorders including anxiety and depression that demonstrate high comorbidity with chronic pain) and substance abuse. Indeed, coexisting psychopathology increases pain intensity, pain-related disability and effectiveness of treatments (Jamison and Edwards, 2013).
Collapse
|
37
|
Abstract
Pain is a significant public health problem, and assessment of pain-related impairment of behavior is a key clinical indicator and treatment target. Similar to opioids and NSAIDs, dopamine (DA) transporter inhibitors block pain-related depression of intracranial self-stimulation (ICSS) in rats. The primary goal of the present study was to determine if the effects of monoamine uptake inhibitors on pain-related depression of ICSS in rats extend to an assay of pain-related depression of nesting in mice. We hypothesized that the DA transporter-selective uptake inhibitor bupropion would block depression of nesting behavior produced by intraperitoneal injection of lactic acid, whereas selective serotonin transporter-selective citalopram, norepinephrine transporter-selective nisoxetine, and the mixed action selective serotonin transporter/norepinephrine transporter inhibitor milnacipran would be ineffective. Effects of the NSAID ketoprofen were also obtained to facilitate interpretation of the effects of the monoamine uptake inhibitors. Consistent with previous findings, ketoprofen blocked pain-related depression of nesting. In contrast, none of the monoamine uptake inhibitors blocked pain-related depression of nesting, although they all blocked pain-related stimulation of stretching. Unlike findings from studies of pain-related depression of ICSS, these results do not support consideration of DA uptake inhibitors for treatment of pain-related depression of behavior.
Collapse
|
38
|
Negus SS, Moerke MJ. Determinants of opioid abuse potential: Insights using intracranial self-stimulation. Peptides 2019; 112:23-31. [PMID: 30391425 PMCID: PMC6342636 DOI: 10.1016/j.peptides.2018.10.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/24/2018] [Accepted: 10/25/2018] [Indexed: 12/14/2022]
Abstract
Intracranial self-stimulation (ICSS) is one procedure that can be used for preclinical abuse potential assessment. In ICSS procedures, subjects with microelectrodes implanted into a brain-reward region are trained to press an operant response lever for pulses of electrical brain stimulation, and drugs are evaluated for their effectiveness to increase or "facilitate" ICSS responding (an abuse-related effect) or to depress ICSS responding (an abuse-limiting effect). ICSS has been used for decades to evaluate determinants of opioid abuse potential, and this article reviews pharmacological and biological determinants of opioid abuse potential as revealed by ICSS studies in rodents. One of the most important observations from ICSS studies is that abused mu opioid receptor (MOR) agonists like morphine often fail to produce abuse-related ICSS facilitation in opioid-naïve subjects, but several days of repeated opioid exposure is sufficient for opioid-induced facilitation to emerge. Future studies with ICSS could help (a) to clarify mechanisms that increase MOR agonist abuse potential during early opioid exposure or during chronic exposure leading to dependence, (b) to evaluate novel opioids either developed as candidate analgesics with reduced abuse potential or identified as designer opioids being synthesized and distributed for illicit use, and (c) to test candidate pharmacotherapies for treatment of opioid abuse in non-dependent and dependent subjects.
Collapse
Affiliation(s)
- S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond VA 23298, United States.
| | - Megan J Moerke
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond VA 23298, United States
| |
Collapse
|
39
|
Sun ML, Ao JP, Wang YR, Huang Q, Li TF, Li XY, Wang YX. Lappaconitine, a C18-diterpenoid alkaloid, exhibits antihypersensitivity in chronic pain through stimulation of spinal dynorphin A expression. Psychopharmacology (Berl) 2018; 235:2559-2571. [PMID: 29926144 DOI: 10.1007/s00213-018-4948-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/08/2018] [Indexed: 12/19/2022]
Abstract
Lappaconitine is a representative C18-diterpenoid alkaloid extracted from Aconitum sinomontanum Nakai and has been prescribed as a pain relief medicine in China for more than 30 years. This study evaluated its antihypersensitivity activity in the rat models of neuropathic and cancer pains and explored its underlying mechanisms. Subcutaneous injection of cumulative doses of lappaconitine produced dose-dependent mechanical antiallodynia and thermal antihyperalgesia in spinal nerve ligation-induced neuropathic rats. The cumulative dose-response analysis exhibited their Emax values of 53.3 and 58.3% MPE, and ED50 values of 1.1 and 1.6 mg/kg. Single intrathecal lappaconitine dose in neuropathy also dose- and time-dependently blocked mechanical allodynia, with an Emax of 66.1% MPE and an ED50 of 0.8 μg. Its multiple twice-daily intrathecal administration over 7 days did not induce mechanical antiallodynic tolerance. Subcutaneous cumulative doses of lappaconitine also produced dose-dependent blockade of mechanical allodynia in the rat bone cancer pain model induced by tibia implantation of cancer cells, with the Emax of 57.9% MPE and ED50 of 2.0 mg/kg. Furthermore, lappaconitine treatment stimulated spinal dynorphin A expression in neuropathic rats, and in primary cultures of microglia but not neurons or astrocytes. Intrathecal pretreatment with the specific microglia depletor liposome-encapsulated clodronate, dynorphin A antibody, and κ-opioid receptor antagonist GNTI totally suppressed intrathecal and subcutaneous lappaconitine-induced mechanical antiallodynia. This study suggests that lappaconitine exhibits antinociception through directly stimulating spinal microglial dynorphin A expression. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Ming-Li Sun
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jun-Ping Ao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China
| | - Yi-Rui Wang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China
| | - Qian Huang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China
| | - Teng-Fei Li
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China
| | - Xin-Yan Li
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China
| | - Yong-Xiang Wang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai, 200240, China. .,State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China.
| |
Collapse
|
40
|
Ong WY, Stohler CS, Herr DR. Role of the Prefrontal Cortex in Pain Processing. Mol Neurobiol 2018; 56:1137-1166. [PMID: 29876878 PMCID: PMC6400876 DOI: 10.1007/s12035-018-1130-9] [Citation(s) in RCA: 413] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/14/2018] [Indexed: 12/16/2022]
Abstract
The prefrontal cortex (PFC) is not only important in executive functions, but also pain processing. The latter is dependent on its connections to other areas of the cerebral neocortex, hippocampus, periaqueductal gray (PAG), thalamus, amygdala, and basal nuclei. Changes in neurotransmitters, gene expression, glial cells, and neuroinflammation occur in the PFC during acute and chronic pain, that result in alterations to its structure, activity, and connectivity. The medial PFC (mPFC) could serve dual, opposing roles in pain: (1) it mediates antinociceptive effects, due to its connections with other cortical areas, and as the main source of cortical afferents to the PAG for modulation of pain. This is a ‘loop’ where, on one side, a sensory stimulus is transformed into a perceptual signal through high brain processing activity, and perceptual activity is then utilized to control the flow of afferent sensory stimuli at their entrance (dorsal horn) to the CNS. (2) It could induce pain chronification via its corticostriatal projection, possibly depending on the level of dopamine receptor activation (or lack of) in the ventral tegmental area-nucleus accumbens reward pathway. The PFC is involved in biopsychosocial pain management. This includes repetitive transcranial magnetic stimulation, transcranial direct current stimulation, antidepressants, acupuncture, cognitive behavioral therapy, mindfulness, music, exercise, partner support, empathy, meditation, and prayer. Studies demonstrate the role of the PFC during placebo analgesia, and in establishing links between pain and depression, anxiety, and loss of cognition. In particular, losses in PFC grey matter are often reversible after successful treatment of chronic pain.
Collapse
Affiliation(s)
- Wei-Yi Ong
- Department of Anatomy, National University of Singapore, Singapore, 119260, Singapore.
- Neurobiology and Ageing Research Programme, National University of Singapore, Singapore, 119260, Singapore.
| | | | - Deron R Herr
- Department of Pharmacology, National University of Singapore, Singapore, 119260, Singapore.
| |
Collapse
|
41
|
Shafiei I, Vatankhah M, Zarepour L, Ezzatpanah S, Haghparast A. Role of D1- and D2-like dopaminergic receptors in the nucleus accumbens in modulation of formalin-induced orofacial pain: Involvement of lateral hypothalamus. Physiol Behav 2018; 188:25-31. [DOI: 10.1016/j.physbeh.2018.01.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 01/24/2018] [Accepted: 01/24/2018] [Indexed: 11/16/2022]
|
42
|
Legakis LP, Bigbee JW, Negus SS. Lack of paclitaxel effects on intracranial self-stimulation in male and female rats: comparison to mechanical sensitivity. Behav Pharmacol 2018; 29:290-298. [PMID: 29369054 PMCID: PMC5854530 DOI: 10.1097/fbp.0000000000000378] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Paclitaxel is a cancer chemotherapy with adverse effects that include peripheral neuropathy, neuropathic pain, and depression of behavior and mood. In rodents, hypersensitive paw-withdrawal reflexes from mechanical stimuli serve as one common measure of paclitaxel-induced pain-related behavior. This study tested the hypothesis that paclitaxel would also depress rates of positively reinforced operant responding as a measure of pain-related behavioral depression. Male and female Sprague-Dawley rats were equipped with electrodes targeting the medial forebrain bundle, trained to lever press for electrical brain stimulation in an assay of intracranial self-stimulation (ICSS), and treated with four injections of varying paclitaxel doses (0.67, 2.0, or 6.0 mg/kg/injection×4 injections on alternate days). Mechanical sensitivity, body weight, and ICSS were evaluated before, during, and for 3 weeks after paclitaxel treatment. Paclitaxel doses sufficient to produce mechanical hypersensitivity did not reliably depress ICSS in male or female rats. Moreover, the degree of behavioral suppression in individual rats did not correlate with mechanical sensitivity. Paclitaxel treatment regimens commonly used to model chemotherapy-induced neuropathic pain in rats are not sufficient to depress ICSS.
Collapse
Affiliation(s)
| | - John W Bigbee
- Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | | |
Collapse
|
43
|
Lazenka ML, Moerke MJ, Townsend EA, Freeman KB, Carroll FI, Negus SS. Dissociable effects of the kappa opioid receptor agonist nalfurafine on pain/itch-stimulated and pain/itch-depressed behaviors in male rats. Psychopharmacology (Berl) 2018; 235:203-213. [PMID: 29063139 PMCID: PMC5750069 DOI: 10.1007/s00213-017-4758-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/03/2017] [Indexed: 12/18/2022]
Abstract
RATIONALE Nalfurafine is a G protein signaling-biased kappa opioid receptor (KOR) agonist approved in Japan for second-line treatment of uremic pruritus. Neither nalfurafine nor any other KOR agonist is currently approved anywhere for treatment of pain, but recent evidence suggests that G protein signaling-biased KOR agonists may have promise as candidate analgesics/antipruritics with reduced side effects compared to nonbiased or ß-arrestin-signaling-biased KOR agonists. OBJECTIVES This study compared nalfurafine effects in rats using assays of pain-stimulated and pain-depressed behavior used previously to evaluate other candidate analgesics. Nalfurafine effects were also examined in complementary assays of itch-stimulated and itch-depressed behavior. METHODS Intraperitoneal lactic acid (IP acid) and intradermal serotonin (ID 5HT) served as noxious and pruritic stimuli, respectively, in male Sprague Dawley rats to stimulate stretching (IP acid) or scratching (ID 5HT) or to depress positively reinforced operant responding in an assay of intracranial self-stimulation (ICSS; both stimuli). RESULTS Nalfurafine was equipotent to decrease IP acid-stimulated stretching and ID 5HT-stimulated scratching; however, doses of nalfurafine that decreased these pain/itch-stimulated behaviors also decreased control ICSS performance. Moreover, nalfurafine failed to alleviate either IP acid- or ID 5HT-induced depression of ICSS. CONCLUSIONS These results suggest that nalfurafine-induced decreases in pain/itch-stimulated behaviors may reflect nonselective decreases in motivated behavior rather than analgesia or antipruritus against the noxious and pruritic stimuli used here. This conclusion agrees with the absence of clinical data for nalfurafine analgesia and the weak clinical data for nalfurafine antipruritus. Nalfurafine bias for G protein signaling may not be sufficient for clinically safe and reliable analgesia or antipruritus.
Collapse
Affiliation(s)
- Matthew L Lazenka
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Megan J Moerke
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - E Andrew Townsend
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kevin B Freeman
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - F Ivy Carroll
- Center for Organic and Medicinal Chemistry, Research Triangle Institute, Research Triangle Park, NC, USA
| | - S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
44
|
Pharmacological modulation of neuropathic pain-related depression of behavior: effects of morphine, ketoprofen, bupropion and [INCREMENT]9-tetrahydrocannabinol on formalin-induced depression of intracranial self-stimulation in rats. Behav Pharmacol 2017; 27:364-76. [PMID: 26588213 DOI: 10.1097/fbp.0000000000000207] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuropathic pain is often associated with behavioral depression. Intraplantar formalin produces sustained, neuropathy-associated depression of intracranial self-stimulation (ICSS) in rats. This study evaluated pharmacological modulation of formalin-induced ICSS depression. Rats with intracranial electrodes targeting the medial forebrain bundle responded for electrical brain stimulation in an ICSS procedure. Bilateral intraplantar formalin administration depressed ICSS for 14 days. Morphine (0.32-3.2 mg/kg), ketoprofen (0.1-10 mg/kg), bupropion (3.2-32 mg/kg), and [INCREMENT]9-tetrahydrocannabinol (THC; 0.32-3.2 mg/kg) were evaluated for their effectiveness to reverse formalin-induced depression of ICSS. Drug effects on formalin-induced mechanical allodynia were evaluated for comparison. Morphine and bupropion reversed both formalin-induced ICSS depression and mechanical allodynia, and effects on ICSS were sustained during repeated treatment. Ketoprofen failed to reverse either formalin effect. THC blocked mechanical allodynia, but decreased ICSS in control rats and exacerbated formalin-induced depression of ICSS. The failure of ketoprofen to alter formalin effects suggests that formalin effects result from neuropathy rather than inflammation. The effectiveness of morphine and bupropion to reverse formalin effects agrees with other evidence that these drugs block pain-depressed behavior in rats and relieve neuropathic pain in humans. The effects of THC suggest general behavioral suppression and do not support the use of THC to treat neuropathic pain.
Collapse
|
45
|
Lazenka MF, Freitas KC, Henck S, Negus SS. Relief of Pain-Depressed Behavior in Rats by Activation of D1-Like Dopamine Receptors. J Pharmacol Exp Ther 2017; 362:14-23. [PMID: 28411257 PMCID: PMC5454591 DOI: 10.1124/jpet.117.240796] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/13/2017] [Indexed: 12/11/2022] Open
Abstract
Clinically significant pain often includes a decrease in both behavior and mesolimbic dopamine signaling. Indirect and/or direct dopamine receptor agonists may alleviate pain-related behavioral depression. To test this hypothesis, the present study compared effects of indirect and direct dopamine agonists in a preclinical assay of pain-depressed operant responding. Male Sprague-Dawley rats with chronic indwelling microelectrodes in the medial forebrain bundle were trained in an intracranial self-stimulation (ICSS) procedure to press a lever for pulses of electrical brain stimulation. Intraperitoneal injection of dilute lactic acid served as an acute noxious stimulus to depress ICSS. Intraperitoneal lactic acid-induced depression of ICSS was dose-dependently blocked by the dopamine transporter inhibitor methylphenidate and the D1-selective agonist SKF82958, but not by the D2/3-selective agonists quinpirole, pramipexole, or sumanirole. The antinociceptive effects of methylphenidate and SKF82958 were blocked by the D1-selective antagonist SCH39166. Acid-induced stimulation of a stretching response was evaluated in separate groups of rats, but all agonists decreased acid-stimulated stretching, and antagonism experiments were inconclusive due to direct effects of the antagonists when administered alone. Taken together, these results suggest that D1-receptor stimulation is both sufficient to block acid-induced depression of ICSS and necessary for methylphenidate antinociception in this procedure. Conversely, D2/3-receptor stimulation is not sufficient to relieve pain-depressed behavior. These results support the hypothesis that pain-related depression of dopamine D1 receptor signaling contributes to pain-related depression of behavior in rats. Additionally, these results support further consideration of indirect dopamine agonists and direct D1 receptor agonists as candidate treatments for pain-related behavioral depression.
Collapse
Affiliation(s)
- Matthew F Lazenka
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Kelen C Freitas
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Sydney Henck
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - S Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
46
|
Li TF, Wu HY, Wang YR, Li XY, Wang YX. Molecular signaling underlying bulleyaconitine A (BAA)-induced microglial expression of prodynorphin. Sci Rep 2017; 7:45056. [PMID: 28327597 PMCID: PMC5361206 DOI: 10.1038/srep45056] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 02/20/2017] [Indexed: 12/30/2022] Open
Abstract
Bulleyaconitine (BAA) has been shown to possess antinociceptive activities by stimulation of dynorphin A release from spinal microglia. This study investigated its underlying signal transduction mechanisms. The data showed that (1) BAA treatment induced phosphorylation of CREB (rather than NF-κB) and prodynorphin expression in cultured primary microglia, and antiallodynia in neuropathy, which were totally inhibited by the CREB inhibitor KG-501; (2) BAA upregulated phosphorylation of p38 (but not ERK or JNK), and the p38 inhibitor SB203580 (but not ERK or JNK inhibitor) and p38β gene silencer siRNA/p38β (but not siRNA/p38α) completely blocked BAA-induced p38 phosphorylation and/or prodynorphin expression, and antiallodynia; (3) BAA stimulated cAMP production and PKA phosphorylation, and the adenylate cyclase inhibitor DDA and PKA inhibitor H-89 entirely antagonized BAA-induced prodynorphin expression and antiallodynia; (4) The Gs-protein inhibitor NF449 completely inhibited BAA-increased cAMP level, prodynorphin expression and antiallodynia, whereas the antagonists of noradrenergic, corticotrophin-releasing factor, A1 adenosine, formyl peptide, D1/D2 dopamine, and glucagon like-peptide-1 receptors failed to block BAA-induced antiallodynia. The data indicate that BAA-induced microglial expression of prodynorphin is mediated by activation of the cAMP-PKA-p38β-CREB signaling pathway, suggesting that its possible target is a Gs-protein-coupled receptor - "aconitine receptor", although the chemical identity is not illustrated.
Collapse
Affiliation(s)
- Teng-Fei Li
- King’s Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai 200240, China
| | - Hai-Yun Wu
- King’s Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai 200240, China
| | - Yi-Rui Wang
- King’s Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai 200240, China
| | - Xin-Yan Li
- King’s Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai 200240, China
| | - Yong-Xiang Wang
- King’s Lab, Shanghai Jiao Tong University School of Pharmacy, 800 Dongchuan Road, Shanghai 200240, China
| |
Collapse
|
47
|
Mesolimbic dopamine signaling in acute and chronic pain: implications for motivation, analgesia, and addiction. Pain 2017; 157:1194-1198. [PMID: 26797678 PMCID: PMC4866581 DOI: 10.1097/j.pain.0000000000000494] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
48
|
Bryant C, Cockburn R, Plante AF, Chia A. The psychological profile of women presenting to a multidisciplinary clinic for chronic pelvic pain: high levels of psychological dysfunction and implications for practice. J Pain Res 2016; 9:1049-1056. [PMID: 27895510 PMCID: PMC5118022 DOI: 10.2147/jpr.s115065] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective Chronic pelvic pain (CPP) is widely acknowledged as a common problem with significant consequences for those diagnosed with this condition. There is a lack of studies with good sample size that provide a comprehensive psychological profile of women presenting to specialist chronic pain clinics. Therefore, the objective of this study was to describe the psychological profile of a representative sample of women presenting with CPP at a tertiary referral center. Design This was a cross-sectional study. Women were asked to complete a questionnaire assessing symptoms of anxiety and depression, pain severity and interference, pain self-efficacy and catastrophizing beliefs, and sexual functioning. Methods One-hundred and seventy-five women with CPP were recruited when they attended their initial assessment at a specialist CPP clinic of the Royal Women’s Hospital, a public hospital in Melbourne, Australia. Results Over 75% of the participants had experienced pain for longer than 2 years. Fifty-three percent of women experienced either moderate or severe anxiety, and 26.7% experienced moderate-to-severe depression. There were strong correlations between depressive symptoms and pain interference, pain catastrophizing and self-efficacy beliefs. Conclusion Our findings confirm previous evidence for high levels of psychological distress and functional impairment associated with this condition, and extend these findings by including measures that are highly relevant to treatment planning, such as thinking styles and pain self-efficacy. Therefore, treatment of this complex condition needs to be holistic, and a multidisciplinary approach is likely to be the best way to achieve this.
Collapse
Affiliation(s)
- Christina Bryant
- Centre for Women's Mental Health, Royal Women's Hospital, Parkville; Melbourne School of Psychological Sciences, University of Melbourne
| | - Rebecca Cockburn
- Centre for Women's Mental Health, Royal Women's Hospital, Parkville
| | | | - Angela Chia
- Department of Anaesthesia, Royal Women's Hospital, Parkville, VIC, Australia
| |
Collapse
|
49
|
Massaly N, Morón JA, Al-Hasani R. A Trigger for Opioid Misuse: Chronic Pain and Stress Dysregulate the Mesolimbic Pathway and Kappa Opioid System. Front Neurosci 2016; 10:480. [PMID: 27872581 PMCID: PMC5097922 DOI: 10.3389/fnins.2016.00480] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 10/06/2016] [Indexed: 12/17/2022] Open
Abstract
Pain and stress are protective mechanisms essential in avoiding harmful or threatening stimuli and ensuring survival. Despite these beneficial roles, chronic exposure to either pain or stress can lead to maladaptive hormonal and neuronal modulations that can result in chronic pain and a wide spectrum of stress-related disorders including anxiety and depression. By inducing allostatic changes in the mesolimbic dopaminergic pathway, both chronic pain and stress disorders affect the rewarding values of both natural reinforcers, such as food or social interaction, and drugs of abuse. Despite opioids representing the best therapeutic strategy in pain conditions, they are often misused as a result of these allostatic changes induced by chronic pain and stress. The kappa opioid receptor (KOR) system is critically involved in these neuronal adaptations in part through its control of dopamine release in the nucleus accumbens. Therefore, it is likely that changes in the kappa opioid system following chronic exposure to pain and stress play a key role in increasing the misuse liability observed in pain patients treated with opioids. In this review, we will discuss how chronic pain and stress-induced pathologies can affect mesolimbic dopaminergic transmission, leading to increased abuse liability. We will also assess how the kappa opioid system may underlie these pathological changes.
Collapse
Affiliation(s)
- Nicolas Massaly
- Basic Research Division, Department of Anesthesiology, Washington University School of MedicineSt. Louis, MO, USA; Washington University Pain Center, Department of Anesthesiology, Washington University School of MedicineSt. Louis, MO, USA
| | - Jose A Morón
- Basic Research Division, Department of Anesthesiology, Washington University School of MedicineSt. Louis, MO, USA; Washington University Pain Center, Department of Anesthesiology, Washington University School of MedicineSt. Louis, MO, USA
| | - Ream Al-Hasani
- Basic Research Division, Department of Anesthesiology, Washington University School of Medicine St. Louis, MO, USA
| |
Collapse
|
50
|
Li TF, Gong N, Wang YX. Ester Hydrolysis Differentially Reduces Aconitine-Induced Anti-hypersensitivity and Acute Neurotoxicity: Involvement of Spinal Microglial Dynorphin Expression and Implications for Aconitum Processing. Front Pharmacol 2016; 7:367. [PMID: 27761113 PMCID: PMC5051147 DOI: 10.3389/fphar.2016.00367] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 09/26/2016] [Indexed: 01/06/2023] Open
Abstract
Aconitines, including bulleyaconitine A, probably the most bioactive and abundant alkaloids in Aconitum plant, are a group of diester C19-diterpenoid alkaloids with one acetylester group attached to C8 of the diterpenoid skeleton and one benzoylester group to C14. Hydrolysis of both groups is involved in the processing of Aconitum, a traditional Chinese medicinal approach. We recently demonstrated that bulleyaconitine A produced anti-hypersensitivity, which was mediated by stimulation of spinal microglial dynorphin A expression. This study aimed to elucidate whether the acetylester and benzoylester groups are involved in aconitine-induced dynorphin A expression, anti-hypersensitivity, neurotoxicity in neuropathic rats. Intrathecal administration of aconitine and benzoylaconine (but not aconine) attenuated mechanical allodynia and heat hyperalgesia, with normalized ED50 values of 35 pmol and 3.6 nmol, respectively. Aconitine and benzoylaconine anti-allodynia was completely blocked by the microglial inhibitor, dynorphin A antiserum, and κ-opioid receptor antagonist. Aconitine and benzoylaconine, but not aconine, stimulated dynorphin A expression in cultured primary spinal microglia, with EC50 values of 32 nM and 3 μM, respectively. Intrathecal aconitine, benzoylaconine and aconine induced flaccid paralysis and death, with normalized TD50 values of 0.5 nmol, 0.2 μmol, and 1.6 μmol, respectively. The TD50/ED50 ratios of aconitine and benzolyaconine were 14:1 and 56:1. Our results suggest that both the C8-acetyl and C14-benzoyl groups are essential for aconitine to stimulate spinal microglial dynorphin A expression and subsequent anti-hypersensitivity, which can be separated from neurotoxicity, because both benzoylaconine and aconine differentially produced anti-hypersensitivity and neurotoxicity due to their different stimulatory ability on dynorphin A expression. Our results support the scientific rationale for Aconitum processing, but caution should be taken to avoid overprocessing and excess hydrolysis of benzolyaconine to aconine.
Collapse
Affiliation(s)
- Teng-Fei Li
- King's Lab, Shanghai Jiao Tong University School of Pharmacy Shanghai, China
| | - Nian Gong
- King's Lab, Shanghai Jiao Tong University School of Pharmacy Shanghai, China
| | - Yong-Xiang Wang
- King's Lab, Shanghai Jiao Tong University School of Pharmacy Shanghai, China
| |
Collapse
|