101
|
Zamyad M, Abbasnejad M, Esmaeili-Mahani S, Sheibani V, Raoof M. Pain influences food preference and food-related memory by activating the basolateral amygdala in rats. Exp Brain Res 2021; 239:79-93. [PMID: 33104830 DOI: 10.1007/s00221-020-05961-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 10/14/2020] [Indexed: 11/24/2022]
Abstract
The amygdala has been demonstrated to contribute to pain-related behavior and food preference. Here, the effect of pain on food preference and food-matched visual-cue memory, in the presence or absence of a basolateral amygdala (BLA) lesion, has been evaluated using a novel innovative apparatus and protocol. Forty adult male Wistar rats were randomly divided into five groups (n = 8) as follows: control, pain, ibuprofen + pain, BLA lesion, BLA lesion + pain groups. Bilateral lesions of the BLA were produced by passing a current of 1.5 mA for 7 s. Pain was induced on the right hind paw of the rats by sub-plantar injection of 50 μl of 2.5% formalin. The animals were encountered with four different meals including wholemeal, wholemeal + sugar, white flour, and biscuit. Each test session consisted of six trials with inter-trial intervals of 15 min. The number of visits to each meal zone and port, the amount of time spent in each food zone and port, traveled distance in each food zone, food consumption per each visit and the total food consumption were recorded. The control group showed a high biscuit preference and low white flour preference. Rats suffering BLA lesion and rats in the BLA lesion + pain group exhibited a shifted preference curve. They had a bias toward eating wholemeal + sugar rather than white flour and biscuit. This group also showed an impaired spatial memory. In conclusion, our findings suggest that the BLA may be involved in pain-induced food preference and food-matched visual-cue memory.
Collapse
Affiliation(s)
- Mahnaz Zamyad
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, P.O. Box: 76135-133, Kerman, Iran
| | - Mehdi Abbasnejad
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, P.O. Box: 76135-133, Kerman, Iran.
| | - Saeed Esmaeili-Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, P.O. Box: 76135-133, Kerman, Iran
- Laboratory of Molecular Neuroscience, Kerman Neuroscience Research Center (KNRC), Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Laboratory of Molecular Neuroscience, Kerman Neuroscience Research Center (KNRC), Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Raoof
- Endodontology Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Orofacial Pain and Dysfunction, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
102
|
Heo K, Kim KM, Han SM, Cho KH, Chu MK. Nasal pain as an aura: Amygdala origin? Seizure 2020; 83:13-16. [PMID: 33075671 DOI: 10.1016/j.seizure.2020.09.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/26/2020] [Accepted: 09/30/2020] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Nasal pain, as an epileptic aura, has been poorly recognized. This study aims to demonstrate clinical features of patients with epilepsy who have nasal pain as an aura. METHODS We retrospectively investigated consecutive patients who visited the epilepsy clinic of tertiary hospital from April 2000 to September 2019. All included patients underwent epilepsy-dedicated, high-resolution magnetic resonance imaging (MRI) examinations. All MRI studies were analyzed by visual inspection. RESULTS Seven patients who presented nasal pain as an aura, were identified. Four patients reported nasal pain as the first aura. Four patients had right amygdala enlargement (isolated amygdala enlargement in three patients; amygdala enlargement in addition to hippocampal sclerosis in one patient), and one patient with compression of an internal carotid-posterior communicating artery aneurysm to right amygdala on brain MRI. Interictal epileptiform or ictal discharges on EEG were found in the right temporal region in five patients. In all four patients with amygdala enlargement, amygdala enlargement was ipsilateral to EEG anomalies. In all patients, nasal pain was accompanied by ictal semiological features, such as autonomic, olfactory, abdominal, or psychic auras, and focal impaired awareness seizures, which are typically associated with mesial temporal lobe epilepsy. CONCLUSIONS Our findings suggest that nasal pain can occur as an epileptic aura in patients with temporal lobe epilepsy with probable involvement of the amygdala.
Collapse
Affiliation(s)
- Kyoung Heo
- Department of Neurology, Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Kyung Min Kim
- Department of Neurology, Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Min Han
- Department of Neurology, Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyoo Ho Cho
- Department of Neurology, Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Kyung Chu
- Department of Neurology, Epilepsy Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
103
|
Lin YW, Chou AIW, Su H, Su KP. Transient receptor potential V1 (TRPV1) modulates the therapeutic effects for comorbidity of pain and depression: The common molecular implication for electroacupuncture and omega-3 polyunsaturated fatty acids. Brain Behav Immun 2020; 89:604-614. [PMID: 32640285 DOI: 10.1016/j.bbi.2020.06.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 01/02/2023] Open
Abstract
Chronic pain and depression are conditions that are highly comorbid and present with overlapping clinical presentations and common pathological biological pathways in neuroinflammation, both of which can be reversed by the use of electroacupuncture (EA) and omega-3 polyunsaturated fatty acids (PUFAs). Transient receptor potential V1 (TRPV1), a Ca2+ permeable ion channel that can be activated by inflammation, is reported to be involved in the development of chronic pain and depression. Here, we investigated the role of TRPV1 and its related pathways in the murine models of cold stress-induced nociception and depression. Female C57BL/6 wild type and TRPV1 knockout mice were subjected to intermittent cold-stress (ICS) to initiate depressive-like and chronic pain behaviors, respectively. The Bio-Plex ELISA technique was utilized to analyze inflammatory mediators in mice plasma. The western blot and immunostaining techniques were used to analyze the presence of TRPV1 and related molecules in the medial prefrontal cortex (mPFC), hippocampus, periaqueductal gray (PAG), and amygdala. The ICS model significantly induced chronic pain (mechanical: 2.55 ± 0.31 g; thermal: 8.12 ± 0.87 s) and depressive-like behaviors (10.95 ± 0.95% in the center zone; 53.14 ± 4.01% in immobility). The treatment efficacy of EA, docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA) were observed in both nociceptive and depression test results. Inflammatory mediators were increased after ICS induction and further reversed by the use of EA, EPA and DHA. A majority of TRPV1 proteins and related molecules were significantly decreased in the mPFC, hippocampus and PAG of mice. This decrease can be reversed by the use of EA, EPA and DHA. In contrast, these molecules were increased in the mice's amygdala, and were attenuated by the use of EA, EPA and DHA. Our findings indicate that these inflammatory mediators can regulate the TRPV1 signaling pathway and initiate new potential therapeutic targets for chronic pain and depression treatment.
Collapse
Affiliation(s)
- Yi-Wen Lin
- College of Chinese Medicine, Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
| | - Ana Isabel Wu Chou
- Department of Psychiatry, Taipei Medical University-Wan Fang Medical Center, Taipei, Taiwan
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Kuan-Pin Su
- College of Medicine, China Medical University, Taichung, Taiwan; Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan; An-Nan Hospital, China Medical University, Tainan, Taiwan.
| |
Collapse
|
104
|
Ji G, Neugebauer V. Kappa opioid receptors in the central amygdala modulate spinal nociceptive processing through an action on amygdala CRF neurons. Mol Brain 2020; 13:128. [PMID: 32948219 PMCID: PMC7501648 DOI: 10.1186/s13041-020-00669-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
The amygdala plays an important role in the emotional-affective aspects of behaviors and pain, but can also modulate sensory aspect of pain ("nociception"), likely through coupling to descending modulatory systems. Here we explored the functional coupling of the amygdala to spinal nociception. We found that pharmacological activation of neurons in the central nucleus of the amygdala (CeA) increased the activity of spinal dorsal horn neurons; and this effect was blocked by optogenetic silencing of corticotropin releasing factor (CRF) positive CeA neurons. A kappa opioid receptor (KOR) agonist (U-69,593) was administered into the CeA by microdialysis. KOR was targeted because of their role in averse-affective behaviors through actions in limbic brain regions. Extracellular single-unit recordings were made of CeA neurons or spinal dorsal horn neurons in anesthetized transgenic Crh-Cre rats. Neurons responded more strongly to noxious than innocuous stimuli. U-69,593 increased the responses of CeA and spinal neurons to innocuous and noxious mechanical stimulation of peripheral tissues. The facilitatory effect of the agonist was blocked by optical silencing of CRF-CeA neurons though light activation of halorhodopsin expressed in these neurons by viral-vector. The CRF system in the amygdala has been implicated in aversiveness and pain modulation. The results suggest that the amygdala can modulate spinal nociceptive processing in a positive direction through CRF-CeA neurons and that KOR activation in the amygdala (CeA) has pro-nociceptive effects.
Collapse
Affiliation(s)
- Guangchen Ji
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, 3601 4th St, Lubbock, TX, 79430-6592, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, 3601 4th St, Lubbock, TX, 79430-6592, USA.
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
105
|
Wilson TD, Valdivia S, Khan A, Ahn HS, Adke AP, Martinez Gonzalez S, Sugimura YK, Carrasquillo Y. Dual and Opposing Functions of the Central Amygdala in the Modulation of Pain. Cell Rep 2020; 29:332-346.e5. [PMID: 31597095 PMCID: PMC6816228 DOI: 10.1016/j.celrep.2019.09.011] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 07/27/2019] [Accepted: 09/05/2019] [Indexed: 12/22/2022] Open
Abstract
Pain perception is essential for survival and can be amplified or suppressed by expectations, experiences, and context. The neural mechanisms underlying bidirectional modulation of pain remain largely unknown. Here, we demonstrate that the central nucleus of the amygdala (CeA) functions as a pain rheostat, decreasing or increasing pain-related behaviors in mice. This dual and opposing function of the CeA is encoded by opposing changes in the excitability of two distinct subpopulations of GABAergic neurons that receive excitatory inputs from the parabrachial nucleus (PB). Thus, cells expressing protein kinase C-delta (CeA-PKCδ) are sensitized by nerve injury and increase pain-related responses. In contrast, cells expressing somatostatin (CeA-Som) are inhibited by nerve injury and their activity drives antinociception. Together, these results demonstrate that the CeA can amplify or suppress pain in a cell-type-specific manner, uncovering a previously unknown mechanism underlying bidirectional control of pain in the brain. The brain can bidirectionally influence behavioral responses to painful stimuli. Wilson et al identify a cellular mechanism underlying a pain rheostat system within the forebrain, with activation of CeA-Som neurons attenuating pain-related responses and increases in the activity of CeA-PKCδ neurons promoting amplification of pain-related behaviors following injury.
Collapse
Affiliation(s)
- Torri D Wilson
- National Center of Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States
| | - Spring Valdivia
- National Center of Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States
| | - Aleisha Khan
- National Center of Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States
| | - Hye-Sook Ahn
- National Center of Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States
| | - Anisha P Adke
- National Center of Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States
| | - Santiago Martinez Gonzalez
- National Center of Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States
| | - Yae K Sugimura
- National Center of Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States
| | - Yarimar Carrasquillo
- National Center of Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
106
|
Hu X, Liu Y, Wu J, Liu Y, Liu W, Chen J, Yang F. Inhibition of P2X7R in the amygdala ameliorates symptoms of neuropathic pain after spared nerve injury in rats. Brain Behav Immun 2020; 88:507-514. [PMID: 32311494 DOI: 10.1016/j.bbi.2020.04.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/23/2020] [Accepted: 04/11/2020] [Indexed: 12/26/2022] Open
Abstract
The amygdala circuitry and P2X7 receptor (P2X7R) have both been shown to play important roles in the modulation of neuropathic pain (NP). However, little is known about the functional role of P2X7R in the amygdala for the regulation of NP. This study aims to evaluate the alleviative effect of intra-amygdala microinfusion of a pharmacological antagonist of P2X7R (A-438079) on NP and explore its possible mechanism of action. Male Sprague-Dawley rats were used to construct the animal model of NP through spared nerve injury (SNI). The SNI rats randomly received chronic bilateral microinjection of A-438079 (100 pmol/side) or saline into the amygdalae via cannulas. Mechanical paw withdrawal threshold (MWT) and thermal withdrawal duration (TWD) were measured by von Frey monofilaments. Besides, tail suspension test (TST), forced swimming test (FST), open field test (OFT) and sucrose preference test (SPT) were performed to assess depression- and anxiety-like behaviors. Immunofluorescence assay was employed to determine the levels of glial fibrillary acidic protein (GFAP), ionized calcium binding adaptor molecule 1 (IBA-1) and connexin 43 (Cx43) in the spinal cord. In addition, the change of growth associated protein 43 (GAP43) level in the spinal cord was assessed by Western blot. Our data showed that chronic treatment with A-438079 increased MWT and decreased TWD on days 11-21 post-SNI while decreased depression-like and anxiety-like behaviors. A-438079 administration significantly attenuated the elevated immunoreactivities of IBA-1 and GFAP in microglia and astrocytes after SNI. Furthermore, the decreased expression of GAP-43 in the spinal cord due to SNI was significantly attenuated by A-438079. However, when A-438079 and a pharmacological agonist (BzATP) of P2X7R were given simultaneously, all the effects caused by A-438079 alone were reversed. In brief, our study revealed the protective role of inhibiting P2X7R in the amygdala against symptoms associated with NP, possibly attributing to its inhibitory effects on spinal microglia and astrocytes.
Collapse
Affiliation(s)
- Xiaoling Hu
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China
| | - Yiming Liu
- Department of Anesthesiology, Affiliated Nanhua Hospital, University of South China, Hunan Province 421001, China
| | - Junting Wu
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China
| | - Yu Liu
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China
| | - Wenjie Liu
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China
| | - Ji Chen
- Department of Endocrinology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China
| | - Fengrui Yang
- Department of Anesthesiology, The First Affiliated Hospital of University of South China, Hunan Province 421001, China; Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
107
|
Fitzgerald JM, Belleau EL, Ehret LE, Trevino C, Brasel KJ, Larson C, deRoon-Cassini T. DACC Resting State Functional Connectivity as a Predictor of Pain Symptoms Following Motor Vehicle Crash: A Preliminary Investigation. THE JOURNAL OF PAIN 2020; 22:171-179. [PMID: 32736035 DOI: 10.1016/j.jpain.2020.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/26/2020] [Accepted: 07/25/2020] [Indexed: 12/25/2022]
Abstract
There is significant heterogeneity in pain outcomes following motor vehicle crashes (MVCs), such that a sizeable portion of individuals develop symptoms of chronic pain months after injury while others recover. Despite variable outcomes, the pathogenesis of chronic pain is currently unclear. Previous neuroimaging work implicates the dorsal anterior cingulate cortex (dACC) in adaptive control of pain, while prior resting state functional magnetic resonance imaging studies find increased functional connectivity (FC) between the dACC and regions involved in pain processing in those with chronic pain. Hyper-connectivity of the dACC to regions that mediate pain response may therefore relate to pain severity. The present study completed rsfMRI scans on N = 22 survivors of MVCs collected within 2 weeks of the incident to test whole-brain dACC-FC as a predictor of pain severity 6 months later. At 2 weeks, pain symptoms were predicted by positive connectivity between the dACC and the premotor cortex. Controlling for pain symptoms at 2 weeks, pain symptoms at 6 months were predicted by negative connectivity between the dACC and the precuneus. Previous research implicates the precuneus in the individual subjective awareness of pain. Given a relatively small sample size, approximately half of which did not experience chronic pain at 6 months, findings warrant replication. Nevertheless, this study provides preliminary evidence of enhanced dACC connectivity with motor regions and decreased connectivity with pain processing regions as immediate and prospective predictors of pain following MVC. PERSPECTIVE: This article presents evidence of distinct neural vulnerabilities that predict chronic pain in MVC survivors based on whole-brain connectivity with the dorsal anterior cingulate cortex.
Collapse
Affiliation(s)
| | - Emily L Belleau
- Department of Psychiatry, McLean Hospital, Belmont, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | | | - Colleen Trevino
- Division of Trauma & Critical Care, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Christine Larson
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin
| | - Terri deRoon-Cassini
- Division of Trauma & Critical Care, Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
108
|
Increased immediate early gene activation in the basolateral amygdala following persistent peripheral inflammation. Neuroreport 2020; 31:724-729. [DOI: 10.1097/wnr.0000000000001480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
109
|
Delery EC, Edwards S. Neuropeptide and cytokine regulation of pain in the context of substance use disorders. Neuropharmacology 2020; 174:108153. [PMID: 32470337 DOI: 10.1016/j.neuropharm.2020.108153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/23/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022]
Abstract
Substance use disorders (SUDs) are frequently accompanied by affective symptoms that promote negative reinforcement mechanisms contributing to SUD maintenance or progression. Despite their widespread use as analgesics, chronic or excessive exposure to alcohol, opioids, and nicotine produces heightened nociceptive sensitivity, termed hyperalgesia. This review focuses on the contributions of neuropeptide (CRF, melanocortin, opioid peptide) and cytokine (IL-1β, TNF-α, chemokine) systems in the development and maintenance of substance-induced hyperalgesia. Few effective therapies exist for either chronic pain or SUD, and the common interaction of these disease states likely complicates their effective treatment. Here we highlight promising new discoveries as well as identify gaps in research that could lead to more effective and even simultaneous treatment of SUDs and co-morbid hyperalgesia symptoms.
Collapse
Affiliation(s)
- Elizabeth C Delery
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health Sciences Center, New Orleans, LA, 70112, USA
| | - Scott Edwards
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health Sciences Center, New Orleans, LA, 70112, USA.
| |
Collapse
|
110
|
Bravo L, Llorca-Torralba M, Suárez-Pereira I, Berrocoso E. Pain in neuropsychiatry: Insights from animal models. Neurosci Biobehav Rev 2020; 115:96-115. [PMID: 32437745 DOI: 10.1016/j.neubiorev.2020.04.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 04/11/2020] [Accepted: 04/23/2020] [Indexed: 02/08/2023]
Abstract
Pain is the most common symptom reported in clinical practice, meaning that it is associated with many pathologies as either the origin or a consequence of other illnesses. Furthermore, pain is a complex emotional and sensorial experience, as the correspondence between pain and body damage varies considerably. While these issues are widely acknowledged in clinical pain research, until recently they have not been extensively considered when exploring animal models, important tools for understanding pain pathophysiology. Interestingly, chronic pain is currently considered a risk factor to suffer psychiatric disorders, mainly stress-related disorders like anxiety and depression. Conversely, pain appears to be altered in many psychiatric disorders, such as depression, anxiety and schizophrenia. Thus, pain and psychiatric disorders have been linked in epidemiological and clinical terms, although the neurobiological mechanisms involved in this pathological bidirectional relationship remain unclear. Here we review the evidence obtained from animal models about the co-morbidity of pain and psychiatric disorders, placing special emphasis on the different dimensions of pain.
Collapse
Affiliation(s)
- Lidia Bravo
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Meritxell Llorca-Torralba
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Suárez-Pereira
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, 11003 Cádiz, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Berrocoso
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Avda. Ana de Viya 21, 11009 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Neuropsychopharmacology and Psychobiology Research Group, Department of Psychology, University of Cádiz, 11510 Puerto Real, Cádiz, Spain.
| |
Collapse
|
111
|
Nelson S, Burns M, McEwen B, Borsook D. Stressful experiences in youth: "Set-up" for diminished resilience to chronic pain. Brain Behav Immun Health 2020; 5:100095. [PMID: 34589863 PMCID: PMC8474662 DOI: 10.1016/j.bbih.2020.100095] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/07/2020] [Indexed: 01/01/2023] Open
Abstract
Chronic pain in youth is common, with prevalence rates in some reports exceeding 50%. Given the plasticity of brain systems in youth and their general level of activity, the underlying processes relating to the evolution of chronic pain may be different from that observed in adults. One aspect that affects brain development is childhood stress. Preliminary research indicates that maladaptive responses to stressful events that induce biological and psychological inability to adapt may be related to pain chronicity in youth. This relationship is particularly notable given the high rates of exposure to stressful events in pediatric pain populations. A review of the literature was performed in the areas of biological, cognitive, psychological and social processes associated with chronic pain and psychological stress and trauma in youth and adult populations. The current review presents a theoretical framework, adapted from McEwen's model (1998) on stress and allostatic load, which aims to outline the potential connection between exposure to stressful events and pediatric chronic pain. Avenues for future investigation are also identified.
Collapse
Affiliation(s)
- Sarah Nelson
- Center for Pain and the Brain, Departments of Anesthesiology, Critical Care, and Pain Medicine and Radiology, Boston Children’s Hospital, USA
- Department of Psychiatry, Harvard Medical School, USA
| | - Maureen Burns
- Center for Pain and the Brain, Departments of Anesthesiology, Critical Care, and Pain Medicine and Radiology, Boston Children’s Hospital, USA
| | | | - David Borsook
- Center for Pain and the Brain, Departments of Anesthesiology, Critical Care, and Pain Medicine and Radiology, Boston Children’s Hospital, USA
- Department of Anesthesia, Harvard Medical School, USA
| |
Collapse
|
112
|
Qian Y, Wang Z, Zhou S, Zhao W, Yin C, Cao J, Wang Z, Li Y. MKP1 in the medial prefrontal cortex modulates chronic neuropathic pain via regulation of p38 and JNK1/2. Int J Neurosci 2020; 130:643-652. [PMID: 31518515 DOI: 10.1080/00207454.2019.1667785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aim: The medial prefrontal context (mPFC) plays pivotal roles in initiation, development, and maintenance of chronic pain, whereas the underlying molecular mechanisms remain elusive, which invited investigation of potential involvement of MKP1 in mPFC in mice in neuropathic pain, and its cellular and molecular mechanisms.Materials and methods: Neuropathic pain model was established in adult male Kunming mice via chronic constrictive injury (CCI) of the sciatic nerve. Paw withdrawal latency (PWL) was measured at the plantar area by radiant heat test. Stereotaxic microinjection was applied in mice as per the atlas of Mouse Brain in Stereotaxic Coordinates. mRNA levels of MKP1 in mPFC in CCI mice were assessed by RT-PCR; protein expressions of MKP1, p-p38, p-JNK and p-ERK in mPFC in CCI mice were analyzed by Western blotting, and expressions of the c-Fos in mPFC in CCI mice evaluated by immunohistochemistry. Moreover, Lenti-MKP1 particles or BCI treatment was employed to inhibit MKP1 in mPFC contralateral to the injury.Results: MKP1 was activated and persistently upregulated in mPFC neurons in CCI mice. Inhibition of MKP1 in the mPFC contralateral to the injury could reverse CCI-induced pain behavior and neuronal activity either via employment of Lenti-MKP1 particles or BCI treatment. MKP1 in the mPFC modulated neuropathic pain via dephosphorization of p38 and JNK1/2.Conclusion: The findings demonstrated that MKP1 in mPFC could play a paramount role in the modulation of neuropathic pain, which might be associated to the increased neuronal excitability in the mPFC and downregulated p-p38 and p-JNK expression.
Collapse
Affiliation(s)
- Yiling Qian
- Department of Anesthesiology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Zhiyong Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Siqi Zhou
- Department of Gastroenterology, The Drum Tower Clinical College of Nanjing Medical University, Nanjing, China
| | - Weinan Zhao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Cui Yin
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Junli Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China.,Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhiping Wang
- Department of Anesthesiology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China.,Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yanqiang Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
113
|
Nahman‐Averbuch H, Schneider VJ, Chamberlin LA, Kroon Van Diest AM, Peugh JL, Lee GR, Radhakrishnan R, Hershey AD, King CD, Coghill RC, Powers SW. Alterations in Brain Function After Cognitive Behavioral Therapy for Migraine in Children and Adolescents. Headache 2020; 60:1165-1182. [DOI: 10.1111/head.13814] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Hadas Nahman‐Averbuch
- Division of Behavioral Medicine and Clinical Psychology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Center for Understanding Pediatric Pain Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Victor J. Schneider
- Division of Behavioral Medicine and Clinical Psychology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Leigh Ann Chamberlin
- Division of Behavioral Medicine and Clinical Psychology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | | | - James L. Peugh
- Division of Behavioral Medicine and Clinical Psychology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati OH USA
| | - Gregory R. Lee
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati OH USA
- Department of Radiology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Pediatric NeuroImaging Research Consortium Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Rupa Radhakrishnan
- Department of Radiology and Imaging Sciences Indiana University School of Medicine Riley Hospital for Children at Indiana University Health Indianapolis IN USA
| | - Andrew D. Hershey
- Center for Understanding Pediatric Pain Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati OH USA
- Division of Neurology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Christopher D. King
- Division of Behavioral Medicine and Clinical Psychology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Center for Understanding Pediatric Pain Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati OH USA
| | - Robert C. Coghill
- Division of Behavioral Medicine and Clinical Psychology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Center for Understanding Pediatric Pain Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati OH USA
- Pediatric NeuroImaging Research Consortium Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Scott W. Powers
- Division of Behavioral Medicine and Clinical Psychology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Center for Understanding Pediatric Pain Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati OH USA
| |
Collapse
|
114
|
Zhang-Molina C, Schmit MB, Cai H. Neural Circuit Mechanism Underlying the Feeding Controlled by Insula-Central Amygdala Pathway. iScience 2020; 23:101033. [PMID: 32311583 PMCID: PMC7168768 DOI: 10.1016/j.isci.2020.101033] [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: 08/28/2019] [Revised: 01/02/2020] [Accepted: 03/31/2020] [Indexed: 12/16/2022] Open
Abstract
The Central nucleus of amygdala (CeA) contains distinct populations of neurons that play opposing roles in feeding. The circuit mechanism of how CeA neurons process information sent from their upstream inputs to regulate feeding is still unclear. Here we show that activation of the neural pathway projecting from insular cortex neurons to the CeA suppresses food intake. Surprisingly, we find that the inputs from insular cortex form excitatory connections with similar strength to all types of CeA neurons. To reconcile this puzzling result, and previous findings, we developed a conductance-based dynamical systems model for the CeA neuronal network. Computer simulations showed that both the intrinsic electrophysiological properties of individual CeA neurons and the overall synaptic organization of the CeA circuit play a functionally significant role in shaping CeA neural dynamics. We successfully identified a specific CeA circuit structure that reproduces the desired circuit output consistent with existing experimentally observed feeding behaviors. Activation of the insular cortex→central amygdala (CeA) pathway suppresses feeding Insular cortex neurons send similar excitatory inputs to different types of CeA neurons Model suggests a required circuit with both late firing and regular spiking cells The circuit model can explain current and previous CeA-mediated feeding behaviors
Collapse
Affiliation(s)
| | - Matthew B Schmit
- Department of Neuroscience, University of Arizona, Tucson, AZ 85721, USA; Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ 85721, USA
| | - Haijiang Cai
- Department of Neuroscience, University of Arizona, Tucson, AZ 85721, USA; Bio5 Institute and Department of Neurology, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
115
|
Abstract
The amygdala has emerged as an important brain area for the emotional-affective dimension of pain and pain modulation. The amygdala receives nociceptive information through direct and indirect routes. These excitatory inputs converge on the amygdala output region (central nucleus) and can be modulated by inhibitory elements that are the target of (prefrontal) cortical modulation. For example, inhibitory neurons in the intercalated cell mass in the amygdala project to the central nucleus to serve gating functions, and so do inhibitory (PKCdelta) interneurons within the central nucleus. In pain conditions, synaptic plasticity develops in output neurons because of an excitation-inhibition imbalance and drives pain-like behaviors and pain persistence. Mechanisms of pain related neuroplasticity in the amygdala include classical transmitters, neuropeptides, biogenic amines, and various signaling pathways. An emerging concept is that differences in amygdala activity are associated with phenotypic differences in pain vulnerability and resilience and may be predetermining factors of the complexity and persistence of pain.
Collapse
Affiliation(s)
- Volker Neugebauer
- Professor and Chair, Department of Pharmacology and Neuroscience, Giles McCrary Endowed Chair in Addiction Medicine, Director, Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center
- School of Medicine, 3601 4th Street
- Mail Stop 6592, Lubbock, Texas 79430-6592
| |
Collapse
|
116
|
Neugebauer V, Mazzitelli M, Cragg B, Ji G, Navratilova E, Porreca F. Amygdala, neuropeptides, and chronic pain-related affective behaviors. Neuropharmacology 2020; 170:108052. [PMID: 32188569 DOI: 10.1016/j.neuropharm.2020.108052] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/04/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022]
Abstract
Neuropeptides play important modulatory roles throughout the nervous system, functioning as direct effectors or as interacting partners with other neuropeptide and neurotransmitter systems. Limbic brain areas involved in learning, memory and emotions are particularly rich in neuropeptides. This review will focus on the amygdala, a limbic region that plays a key role in emotional-affective behaviors and pain modulation. The amygdala is comprised of different nuclei; the basolateral (BLA) and central (CeA) nuclei and in between, the intercalated cells (ITC), have been linked to pain-related functions. A wide range of neuropeptides are found in the amygdala, particularly in the CeA, but this review will discuss those neuropeptides that have been explored for their role in pain modulation. Calcitonin gene-related peptide (CGRP) is a key peptide in the afferent nociceptive pathway from the parabrachial area and mediates excitatory drive of CeA neurons. CeA neurons containing corticotropin releasing factor (CRF) and/or somatostatin (SOM) are a source of long-range projections and serve major output functions, but CRF also acts locally to excite neurons in the CeA and BLA. Neuropeptide S (NPS) is associated with inhibitory ITC neurons that gate amygdala output. Oxytocin and vasopressin exert opposite (inhibitory and excitatory, respectively) effects on amygdala output. The opioid system of mu, delta and kappa receptors (MOR, DOR, KOR) and their peptide ligands (β-endorphin, enkephalin, dynorphin) have complex and partially opposing effects on amygdala function. Neuropeptides therefore serve as valuable targets to regulate amygdala function in pain conditions. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Bryce Cragg
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Edita Navratilova
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Frank Porreca
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
117
|
Zhou C, Wu Y, Ding X, Shi N, Cai Y, Pan ZZ. SIRT1 Decreases Emotional Pain Vulnerability with Associated CaMKIIα Deacetylation in Central Amygdala. J Neurosci 2020; 40:2332-2342. [PMID: 32005763 PMCID: PMC7083291 DOI: 10.1523/jneurosci.1259-19.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 01/09/2020] [Accepted: 01/24/2020] [Indexed: 11/21/2022] Open
Abstract
Emotional disorders are common comorbid conditions that further exacerbate the severity and chronicity of chronic pain. However, individuals show considerable vulnerability to the development of chronic pain under similar pain conditions. In this study on male rat and mouse models of chronic neuropathic pain, we identify the histone deacetylase Sirtuin 1 (SIRT1) in central amygdala as a key epigenetic regulator that controls the development of comorbid emotional disorders underlying the individual vulnerability to chronic pain. We found that animals that were vulnerable to developing behaviors of anxiety and depression under the pain condition displayed reduced SIRT1 protein levels in central amygdala, but not those animals resistant to the emotional disorders. Viral overexpression of local SIRT1 reversed this vulnerability, but viral knockdown of local SIRT1 mimicked the pain effect, eliciting the pain vulnerability in pain-free animals. The SIRT1 action was associated with CaMKIIα downregulation and deacetylation of histone H3 lysine 9 at the CaMKIIα promoter. These results suggest that, by transcriptional repression of CaMKIIα in central amygdala, SIRT1 functions to guard against the emotional pain vulnerability under chronic pain conditions. This study indicates that SIRT1 may serve as a potential therapeutic molecule for individualized treatment of chronic pain with vulnerable emotional disorders.SIGNIFICANCE STATEMENT Chronic pain is a prevalent neurological disease with no effective treatment at present. Pain patients display considerably variable vulnerability to developing chronic pain, indicating individual-based molecular mechanisms underlying the pain vulnerability, which is hardly addressed in current preclinical research. In this study, we have identified the histone deacetylase Sirtuin 1 (SIRT1) as a key regulator that controls this pain vulnerability. This study reveals that the SIRT1-CaMKIIaα pathway in central amygdala acts as an epigenetic mechanism that guards against the development of comorbid emotional disorders under chronic pain, and that its dysfunction causes increased vulnerability to the development of chronic pain. These findings suggest that SIRT1 activators may be used in a novel therapeutic approach for individual-based treatment of chronic pain.
Collapse
Affiliation(s)
- Chenghua Zhou
- Department of Anesthesiology and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, and
| | - Yuqing Wu
- Jiangsu Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 266061, People's Republic of China
| | - Xiaobao Ding
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, and
| | - Naihao Shi
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, and
| | - Youqin Cai
- Department of Anesthesiology and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Zhizhong Z Pan
- Department of Anesthesiology and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030,
| |
Collapse
|
118
|
Lai G, Langevin JP, Koek RJ, Krahl SE, Bari AA, Chen JWY. Acute Effects and the Dreamy State Evoked by Deep Brain Electrical Stimulation of the Amygdala: Associations of the Amygdala in Human Dreaming, Consciousness, Emotions, and Creativity. Front Hum Neurosci 2020; 14:61. [PMID: 32158384 PMCID: PMC7052301 DOI: 10.3389/fnhum.2020.00061] [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: 04/04/2019] [Accepted: 02/10/2020] [Indexed: 01/06/2023] Open
Abstract
Accurate localization of complex human experiences such as emotions, dreaming, creativity, and consciousness to specific cerebral structures or neural networks has remained elusive despite technological advances. We report the use of acute deep brain stimulation (DBS) to evoke behavioral and emotional effects by applying electrical stimulation (ES) at various voltage strengths to the basolateral and central subnuclei of the amygdala in addition to the head of hippocampus (HC) for two subjects with medically refractory post-traumatic stress disorder (PTSD). Our results suggest that the amygdala could be a node in a neural network responsible for the generation of complex vivid mental imagery and integrated sensory experiences similar to John Hughlings Jackson's "dreamy state" and "double consciousness," which have been classically associated with temporal lobe epilepsy during uncinate seizures. That we were able to elicit similar vivid, dynamic, complex, bizarre, and original mental imagery with ES in non-epileptic subjects suggests that Jackson's seizure related "dreamy state" and "double consciousness" may arise from heightened innate brain mechanisms with the amygdala acting as a node in the neural network responsible for physiologic dreaming and creative functions. Furthermore, our subjects experienced different emotions with different stimulation strengths at various electrode contacts. Our results suggest that higher voltage stimulation of the amygdala and HC at 4-5 V leads to predominantly negative responses and 2-4 V stimulation showed inversely coupled positive and negative responses of the amygdala in either hemisphere which may imply hemispheric dominance of emotional valences without relation to handedness. Due to the unique and complex responses dependent on location and strength of stimulation, we advise that all patients receiving DBS of the amygdala undergo acute stimulation mapping in a monitored setting before selecting therapeutic parameters for chronic stimulation.
Collapse
Affiliation(s)
- George Lai
- Neurology Service, VA Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Department of Neurology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jean-Philippe Langevin
- Neurosurgery Service, VA Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - Ralph J Koek
- Psychiatry and Mental Health Service, VA Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Department of Psychiatry and Behavior Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| | - Scott E Krahl
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States.,Research and Development, VA Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Ausaf A Bari
- Neurosurgery Service, VA Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - James W Y Chen
- Neurology Service, VA Greater Los Angeles Healthcare System, Los Angeles, CA, United States.,Department of Neurology, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
119
|
Mamelak M. Nightmares and the Cannabinoids. Curr Neuropharmacol 2020; 18:754-768. [PMID: 31934840 PMCID: PMC7536831 DOI: 10.2174/1570159x18666200114142321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 12/21/2019] [Accepted: 01/11/2020] [Indexed: 11/22/2022] Open
Abstract
The cannabinoids, Δ9 tetrahydrocannabinol and its analogue, nabilone, have been found to reliably attenuate the intensity and frequency of post-traumatic nightmares. This essay examines how a traumatic event is captured in the mind, after just a single exposure, and repeatedly replicated during the nights that follow. The adaptive neurophysiological, endocrine and inflammatory changes that are triggered by the trauma and that alter personality and behavior are surveyed. These adaptive changes, once established, can be difficult to reverse. But cannabinoids, uniquely, have been shown to interfere with all of these post-traumatic somatic adaptations. While cannabinoids can suppress nightmares and other symptoms of post-traumatic stress disorder, they are not a cure. There may be no cure. The cannabinoids may best be employed, alone, but more likely in conjunction with other agents, in the immediate aftermath of a trauma to mitigate or even abort the metabolic changes which are set in motion by the trauma and which may permanently alter the reactivity of the nervous system. Steps in this direction have already been taken.
Collapse
Affiliation(s)
- Mortimer Mamelak
- Department of Psychiatry, University of Toronto, Baycrest Hospital, Permanent Address: 19 Tumbleweed Road, Toronto, OntarioM2J 2N2, Canada
| |
Collapse
|
120
|
Brain activity changes in a monkey model of central post-stroke pain. Exp Neurol 2020; 323:113096. [DOI: 10.1016/j.expneurol.2019.113096] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/25/2019] [Accepted: 10/31/2019] [Indexed: 01/27/2023]
|
121
|
Suwito J, Putra ST, Sulistyono A. The Affirmation – Tapping on Pain Perception and Serotonin Serum Level of Post – Caesarian Section patients. JURNAL NERS 2019. [DOI: 10.20473/jn.v14i2.16421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Introduction: Affirmation - tapping interventions have been shown to reduce pain complaints in post-operative patients completing conventional treatment. This is thought to be due to serotonin performance but clinical studies have not been conducted. The aim was to compare the mean perception of the pain reported by post-operative patients given affirmation- tapping treatment with another treatment as a complementary nursing intervention. This was to see if the performance of the serotonin serum level is different from in other treatments.Methods: We used a randomized post-test only control group design carried out in parallel in post-caesarean section patients. The sample totaled 40 patients divided into four groups (10 in affirmation, 10 in tapping, 10 in affirmation-tapping and 10 in the control). They were obtained through simple random sampling. The instruments included affirmation-tapping guidelines, Elisa kits and the McGill - Melzack Pain Questionnaire short-form (MPQsf). The independent variable was the intervention of affirmation-tapping and the dependent variables were pain perception and serotonin level. The data was analyzed using simple linear regression.Results: The average variant of the serotonin levels in the affirmation-tapping treatment group was higher and thus differed significantly from the other groups.Conclusion: Affirmation-tapping as a complementary nursing intervention can increase the serotonin serum levels of the post-caesarean section patients by complementing conventional treatments. Participant pain complaints were lowest in the affirmation-tapping group with the highest serotonin levels present and these were significantly different to the other groups. Affirmation – tapping was recommended as a complementary intervention in nursing post-operative patients that complements conventional treatment.
Collapse
|
122
|
Ji G, Neugebauer V. Contribution of Corticotropin-Releasing Factor Receptor 1 (CRF1) to Serotonin Receptor 5-HT 2CR Function in Amygdala Neurons in a Neuropathic Pain Model. Int J Mol Sci 2019; 20:E4380. [PMID: 31489921 PMCID: PMC6770811 DOI: 10.3390/ijms20184380] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 08/30/2019] [Accepted: 09/01/2019] [Indexed: 12/30/2022] Open
Abstract
The amygdala plays a key role in emotional-affective aspects of pain and in pain modulation. The central nucleus (CeA) serves major amygdala output functions related to emotional-affective behaviors and pain modulation. Our previous studies implicated the corticotropin-releasing factor (CRF) system in amygdala plasticity and pain behaviors in an arthritis model. We also showed that serotonin (5-HT) receptor subtype 5-HT2CR in the basolateral amygdala (BLA) contributes to increased CeA output and neuropathic pain-like behaviors. Here, we tested the novel hypothesis that 5-HT2CR in the BLA drives CRF1 receptor activation to increase CeA neuronal activity in neuropathic pain. Extracellular single-unit recordings of CeA neurons in anesthetized adult male rats detected increased activity in neuropathic rats (spinal nerve ligation model) compared to sham controls. Increased CeA activity was blocked by local knockdown or pharmacological blockade of 5-HT2CR in the BLA, using stereotaxic administration of 5-HT2CR short hairpin RNA (shRNA) viral vector or a 5-HT2CR antagonist (SB242084), respectively. Stereotaxic administration of a CRF1 receptor antagonist (NBI27914) into the BLA also decreased CeA activity in neuropathic rats and blocked the facilitatory effects of a 5-HT2CR agonist (WAY161503) administered stereotaxically into the BLA. Conversely, local (BLA) knockdown of 5-HT2CR eliminated the inhibitory effect of NBI27914 and the facilitatory effect of WAY161503 in neuropathic rats. The data suggest that 5-HT2CR activation in the BLA contributes to neuropathic pain-related amygdala (CeA) activity by engaging CRF1 receptor signaling.
Collapse
Affiliation(s)
- Guangchen Ji
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA.
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA.
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79424, USA.
| |
Collapse
|
123
|
Modulation of the Negative Affective Dimension of Pain: Focus on Selected Neuropeptidergic System Contributions. Int J Mol Sci 2019; 20:ijms20164010. [PMID: 31426473 PMCID: PMC6720937 DOI: 10.3390/ijms20164010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/11/2022] Open
Abstract
It is well known that emotions can interfere with the perception of physical pain, as well as with the development and maintenance of painful conditions. On the other hand, somatic pain can have significant consequences on an individual’s affective behavior. Indeed, pain is defined as a complex and multidimensional experience, which includes both sensory and emotional components, thus exhibiting the features of a highly subjective experience. Over the years, neural pathways involved in the modulation of the different components of pain have been identified, indicating the existence of medial and lateral pain systems, which, respectively, project from medial or lateral thalamic nuclei to reach distinct cortex regions relating to specific functions. However, owing to the limited information concerning how mood state and painful input affect each other, pain treatment is frequently unsatisfactory. Different neuromodulators, including endogenous neuropeptides, appear to be involved in pain-related emotion and in its affective influence on pain perception, thus playing key roles in vulnerability and clinical outcome. Hence, this review article focuses on evidence concerning the modulation of the sensory and affective dimensions of pain, with particular attention given to some selected neuropeptidergic system contributions.
Collapse
|
124
|
Tolou-Dabbaghian B, Delphi L, Rezayof A. Blockade of NMDA Receptors and Nitric Oxide Synthesis Potentiated Morphine-Induced Anti-Allodynia via Attenuating Pain-Related Amygdala pCREB/CREB Signaling Pathway. THE JOURNAL OF PAIN 2019; 20:885-897. [DOI: 10.1016/j.jpain.2019.01.329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 11/30/2018] [Accepted: 01/11/2019] [Indexed: 01/26/2023]
|
125
|
Mazzitelli M, Neugebauer V. Amygdala group II mGluRs mediate the inhibitory effects of systemic group II mGluR activation on behavior and spinal neurons in a rat model of arthritis pain. Neuropharmacology 2019; 158:107706. [PMID: 31306647 DOI: 10.1016/j.neuropharm.2019.107706] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/03/2019] [Accepted: 07/10/2019] [Indexed: 12/16/2022]
Abstract
The amygdala plays a critical role in emotional-affective aspects of behaviors and pain modulation. The central nucleus of amygdala (CeA) serves major output functions, and neuroplasticity in the CeA is linked to pain-related behaviors in different models. Activation of Gi/o-coupled group II metabotropic glutamate receptors (mGluRs), which consist of mGluR2 and mGluR3, can decrease neurotransmitter release and regulate synaptic plasticity. Group II mGluRs have emerged as targets for neuropsychiatric disorders and can inhibit pain-related processing and behaviors. Surprisingly, site and mechanism of antinociceptive actions of systemically applied group II mGluR agonists are not clear. Our previous work showed that group II mGluR activation in the amygdala inhibits pain-related CeA activity, but behavioral and spinal consequences remain to be determined. Here we studied the contribution of group II mGluRs in the amygdala to the antinociceptive effects of a systemically applied group II mGluR agonist (LY379268) on behavior and spinal dorsal horn neuronal activity, using the kaolin/carrageenan-induced knee joint arthritis pain model. Audible and ultrasonic vocalizations (emotional responses) and mechanical reflex thresholds were measured in adult rats with and without arthritis (5-6 h postinduction). Extracellular single-unit recordings were made from spinal dorsal horn wide dynamic range neurons of anesthetized (isoflurane) rats with and without arthritis (5-6 h postinduction). Systemic (intraperitoneal) application of a group II mGluR agonist (LY379268) decreased behaviors and activity of spinal neurons in the arthritis pain model but not under normal conditions. Stereotaxic administration of LY379268 into the CeA mimicked the effects of systemic application. Conversely, stereotaxic administration of a group II mGluR antagonist (LY341495) into the CeA reversed the effects of systemic application of LY379268 on behaviors and dorsal horn neuronal activity in arthritic rats. The data show for the first time that the amygdala is the critical site of action for the antinociceptive behavioral and spinal neuronal effects of systemically applied group II mGluR agonists.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA; Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, 79430-6592, USA.
| |
Collapse
|
126
|
Chronic Pain: Structural and Functional Changes in Brain Structures and Associated Negative Affective States. Int J Mol Sci 2019; 20:ijms20133130. [PMID: 31248061 PMCID: PMC6650904 DOI: 10.3390/ijms20133130] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 12/22/2022] Open
Abstract
Chronic pain is a condition in which pain progresses from an acute to chronic state and persists beyond the healing process. Chronic pain impairs function and decreases patients’ quality of life. In recent years, efforts have been made to deepen our understanding of chronic pain and to develop better treatments to alleviate chronic pain. In this review, we summarize the results of previous studies, focusing on the mechanisms underlying chronic pain development and the identification of neural areas related to chronic pain. We review the association between chronic pain and negative affective states. Further, we describe the structural and functional changes in brain structures that accompany the chronification of pain and discuss various neurotransmitter families involved. Our review aims to provide guidance for the development of future therapeutic approaches that could be used in the management of chronic pain.
Collapse
|
127
|
Cardenas A, Blanca M, Dimitrov E. Persistent pain intensifies recall of consolidated fear memories. Neurobiol Stress 2019; 10:100163. [PMID: 31193505 PMCID: PMC6535623 DOI: 10.1016/j.ynstr.2019.100163] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 12/04/2022] Open
Abstract
Ensembles of principal neurons in the basolateral amygdala (BLA) generate the initial engrams for fear memories, while projections from the BLA to the medial prefrontal cortex (mPFC) are essential for the encoding, transfer and storage of remote fear memories. We tested the effects of chronic pain on remote fear memories in mice. Male mice underwent classic fear conditioning by pairing a single tone (conditional stimulus, CS) with a single electric foot shock (unconditional stimulus, US). Sciatic nerve constriction was used to induce neuropathic pain at various time points before or after the fear conditioning. The mice with sciatic nerve cuffs implanted 48 h after the fear conditioning showed an increased freezing response to CS when compared to mice without cuffs or when compared to mice in which the nerve cuffing was performed 48 h before the fear conditioning. The enhancing effect of pain on consolidated fear memory was further tested and mice in which the nerve cuffing was performed 14 days after the fear conditioning also showed an increased fear response when tested 56 days later. We used immunostaining to detect morphological changes in the BLA that could suggest a mechanism for the observed increase in fear response. We found an increased number of calbindin/parvalbumin positive neurons in the BLA and increased perisomatic density of GAD65 on projection neurons that connect BLA to mPFC in mice with nerve cuffs. Despite the strong increase of c-Fos expression in BLA and mPFC that was induced by fear recall, neither the BLA to mPFC nor the mPFC to BLA projection neurons were activated in mice with nerve cuffs. Furthermore, non-injured mice had an increased fear response when BLA to mPFC projections were inhibited by a chemogenetic method. In conclusion, this study provides evidence that persistent pain has a significant impact on consolidated fear memories. Very likely the underlying mechanism for this phenomenon is increased inhibitory input onto the BLA to mPFC projection neurons, possibly from neurons with induced parvalbumin expression. Conceivably, the increased fear response to consolidated fear memory is a harbinger for the later development of anxiety and depression symptoms associated with chronic pain.
Collapse
Affiliation(s)
- Andrea Cardenas
- Department of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Michelle Blanca
- Department of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Eugene Dimitrov
- Department of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| |
Collapse
|
128
|
Metabotropic Glutamate Receptor 5 and 8 Modulate the Ameliorative Effect of Ultramicronized Palmitoylethanolamide on Cognitive Decline Associated with Neuropathic Pain. Int J Mol Sci 2019; 20:ijms20071757. [PMID: 30970677 PMCID: PMC6480075 DOI: 10.3390/ijms20071757] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/23/2022] Open
Abstract
This study investigated whether metabotropic glutamate receptor (mGluR) 5 and 8 are involved in the effect of ultramicronizedpalmitoylethanolamide (um-PEA) on the cognitive behavior and long term potentiation (LTP) at entorhinal cortex (LEC)-dentate gyrus (DG) pathway in mice rendered neuropathic by the spare nerve injury (SNI). SNI reduced discriminative memory and LTP. Um-PEA treatment started after the development of neuropathic pain had no effects in sham mice, whereas it restored cognitive behavior and LTP in SNI mice. 2-Methyl-6-(phenylethynyl) pyridine (MPEP), a selective mGluR5 antagonist, improved cognition in SNI mice and produced a chemical long term depression of the field excitatory postsynaptic potentials (fEPSPs) in sham and SNI mice. After theta burst stimulation (TBS) MPEP restored LTP in SNI mice. In combination with PEA, MPEP antagonized the PEA effect on discriminative memory and decreased LTP in SNI mice. The (RS)-4-(1-amino-1-carboxyethyl)phthalic acid (MDCPG), a selective mGluR8 antagonist, did not affect discriminative memory, but it induced a chemical LTP and prevented the enhancement of fEPSPs after TBS in SNI mice which were treated or not treated with PEA. The effect of PEA on LTP and cognitive behavior was modulated by mGluR5 and mGluR8. In particular in the SNI conditions, the mGluR5 blockade facilitated memory and LTP, but prevented the beneficial effects of PEA on discriminative memory while the mGluR8 blockade, which was ineffective in itself, prevented the favorable action of the PEA on LTP. Thus, although their opposite roles (excitatory/inhibitory of the two receptor subtypes on the glutamatergic system), they appeared to be required for the neuroprotective effect of PEA in conditions of neuropathic pain.
Collapse
|
129
|
Chen Q, Heinricher MM. Plasticity in the Link between Pain-Transmitting and Pain-Modulating Systems in Acute and Persistent Inflammation. J Neurosci 2019; 39:2065-2079. [PMID: 30651329 PMCID: PMC6507088 DOI: 10.1523/jneurosci.2552-18.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/26/2018] [Accepted: 01/04/2019] [Indexed: 01/08/2023] Open
Abstract
There is strong evidence that spinoparabrachial neurons in the superficial dorsal horn contribute to persistent pain states, and that the lateral parabrachial complex (PB) conveys relevant nociceptive information to higher structures. The role of PB itself in hyperalgesia and how it recruits descending facilitation has nevertheless received significantly less attention. The current study is a first step toward delineating the functional dynamics of PB and its link to descending control in acute and persistent inflammatory pain. In lightly anesthetized rats, we recorded behavioral withdrawal evoked by mechanical stimulation of the hindpaw and, simultaneously, the activity of identified pain-modulating neurons, "ON-cells" and "OFF-cells," in the rostral ventromedial medulla (RVM). This was done before and after the inactivation of PB, contralateral or ipsilateral to an inflamed paw [1 h, 1 d, or 5-6 d after intraplantar injection of Complete Freund's Adjuvant (CFA)]. The inactivation of contralateral, but not ipsilateral, PB interfered with nociceptive input to RVM under basal conditions, as well as in acute inflammation. By contrast, blocking ipsilateral, but not contralateral, PB in established inflammation interfered with behavioral hyperalgesia and ON-cell and OFF-cell responses. The lesioning of contralateral PB before CFA injection prevented this recruitment of ipsilateral PB in persistent inflammation. These experiments show that contralateral PB is required to initiate hyperalgesia, which is then maintained by ipsilateral PB, most likely in both cases via the engagement of pain-modulating neurons of the RVM.SIGNIFICANCE STATEMENT The lateral parabrachial complex (PB) relays nociceptive information to brain circuits that are important for the transmission and modulation of pain, but its specific role in persistent pain and engagement of descending control mechanisms has received relatively little attention. We show here that PB contralateral and ipsilateral to an inflammatory insult demonstrate different functions as inflammation persists, likely by engaging pain-facilitating neurons of the rostral ventromedial medulla. While the contralateral PB, the target of the major spinoparabrachial pathway, relays acute nociceptive information, the ipsilateral PB is recruited or unmasked in persistent inflammation to maintain hyperalgesia. These data point to plasticity in the PB itself or its direct and indirect connections with pain-modulating systems as central to the development and maintenance of persistent pain.
Collapse
Affiliation(s)
| | - Mary M Heinricher
- Departments of Neurological Surgery and
- Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
| |
Collapse
|
130
|
Abstract
PURPOSE OF REVIEW The goal of the review was to highlight recent advances in our understanding of descending pain-modulating systems and how these contribute to persistent pain states, with an emphasis on the current state of knowledge around "bottom-up" (sensory) and "top-down" (higher structures mediating cognitive and emotional processing) influences on pain-modulating circuits. RECENT FINDINGS The connectivity, physiology, and function of these systems have been characterized extensively over the last 30 years. The field is now beginning to ask how and when these systems are engaged to modulate pain. A recent focus is on the parabrachial complex, now recognized as the major relay of nociceptive information to pain-modulating circuits, and plasticity in this circuit and its connections to the RVM is marked in persistent inflammatory pain. Top-down influences from higher structures, including hypothalamus, amygdala, and medial prefrontal areas, are also considered. The challenge will be to tease out mechanisms through which a particular behavioral context engages distinct circuits to enhance or suppress pain, and to understand how these mechanisms contribute to chronic pain.
Collapse
|
131
|
Zhang Q, Yu S, Wang Y, Wang M, Yang Y, Wei W, Guo X, Zeng F, Liang F, Yang J. Abnormal reward system network in primary dysmenorrhea. Mol Pain 2019; 15:1744806919862096. [PMID: 31286840 PMCID: PMC6616063 DOI: 10.1177/1744806919862096] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 04/23/2019] [Accepted: 06/11/2019] [Indexed: 12/27/2022] Open
Abstract
Neuroimaging studies have demonstrated that reward system is associated with chronic pain diseases. In addition, previous studies have also demonstrated abnormal functional and structural brain regions in primary dysmenorrhea. However, the relation of reward system and primary dysmenorrhea is still unknown. Using the resting state functional magnetic resonance imaging, we aimed to investigate the functional connectivity changes of reward system during periovulatory phase in primary dysmenorrhea. Forty-one primary dysmenorrhea patients and 39 matched female healthy controls participated in this study. Compared to healthy controls, primary dysmenorrhea patients showed decreased connectivity of left nucleus accumbens with the bilateral anterior insula and the left amygdala and decreased connectivity of right nucleus accumbens with ventral tegmental area, the left hippocampus, the right orbital frontal cortex, and the right anterior insula. In addition, the decreased functional connectivity between the right nucleus accumbens-ventral tegmental area negatively correlated with the level of prostaglandin F2 alpha. Our findings provide neuroimaging evidence in support of the abnormal reward system connectivity in primary dysmenorrhea patients, which might contribute to a better understanding of the cerebral pathophysiology of primary dysmenorrhea.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siyi Yu
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanan Wang
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Minyu Wang
- Damian Honghe Community Health Service Center of Longquanyi, Chengdu, China
| | - Ya Yang
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Wei
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoli Guo
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fang Zeng
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fanrong Liang
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Yang
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
132
|
Lin WY, Chu WH, Chao THH, Sun WZ, Yen CT. Longitudinal FDG-PET scan study of brain changes in mice with cancer-induced bone pain and after morphine analgesia. Mol Pain 2019; 15:1744806919841194. [PMID: 30868934 PMCID: PMC6492350 DOI: 10.1177/1744806919841194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 02/14/2019] [Accepted: 02/20/2019] [Indexed: 01/21/2023] Open
Abstract
Morphine is the most commonly used drug for treating physical and psychological suffering caused by advanced cancer. Although morphine is known to elicit multiple supraspinal analgesic effects, its behavioral correlates with respect to the whole-brain metabolic activity during cancer-induced bone pain have not been elucidated. We injected 4T1 mouse breast cancer cells into the left femur bone marrow cavity of BALB/c mice. All mice developed limb use deficits, mechanical allodynia, and hypersensitivity to cold, which were effectively suppressed with morphine. Serial 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) was performed for each mouse before cancer induction (0 day), after cancer-induced bone pain was established (14 days), and during effective morphine treatment (16 days). The longitudinal FDG-PET imaging analysis demonstrated that cancer-induced bone pain increased glucose uptake in the insular cortex and hypothalamus and decreased the activity of the retrosplenial cortex. Morphine reversed the activation of the insular cortex and hypothalamus. Furthermore, morphine activated the amygdala and rostral ventromedial medulla and suppressed the activity of anterior cingulate cortex. Our findings of hypothalamic and insular cortical activation support the hypothesis that cancer-induced bone pain has strong inflammatory and affective components in freely moving animals. Morphine may provide descending inhibitory and facilitatory actions in the treatment of cancer-induced bone pain in a clinical setting.
Collapse
Affiliation(s)
- Wen-Ying Lin
- Department of Life Science, National Taiwan University,
Taipei
- Department of Anesthesiology, National Taiwan University
Hospital, Taipei
- National Taiwan University Cancer Center, National Taiwan
University College of Medicine, Taipei
| | - Wen-Hua Chu
- Department of Life Science, National Taiwan University,
Taipei
| | | | - Wen-Zen Sun
- Department of Anesthesiology, National Taiwan University
Hospital, Taipei
| | - Chen-Tung Yen
- Department of Life Science, National Taiwan University,
Taipei
| |
Collapse
|
133
|
Paretkar T, Dimitrov E. Activation of enkephalinergic (Enk) interneurons in the central amygdala (CeA) buffers the behavioral effects of persistent pain. Neurobiol Dis 2018; 124:364-372. [PMID: 30572023 DOI: 10.1016/j.nbd.2018.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/23/2018] [Accepted: 12/10/2018] [Indexed: 12/25/2022] Open
Abstract
Enk neurons in CeA modulate the activity of the amygdala projection neurons and it is very likely that changes of Enk signaling cause the heightened anxiety that accompanies chronic pain. We use chemogenetics and transgenic mice to investigate the effects of acute and continuous activation of the amygdala Enk neurons on persistent pain and anxiodepressive-like behavior in mice. Enk-cre mice were injected bilaterally into the CeA with cre-activated AAV-DREADD/Gq/mCherry, while neuropathic pain was induced by sciatic nerve constriction. A single injection of DREADD's ligand CNO decreased the anxiety-like behavior in both, uninjured mice and in mice with neuropathic pain and produced robust analgesia that lasted for 24 h. Furthermore, the activation of Enk neurons by the DREADD ligand led to increased c-Fos expression in PKC-δ interneurons of the CeA and in non-serotonergic neurons in the ventrolateral periaqueductal gray (vlPAG), a brain structure that is an essential part of the descending pain inhibitory system. Next, we added CNO to the drinking water of the experimental mice for 14 days in order to assess the effects of continuous activation of CeA Enk interneurons on anxiodepressive-like behavior, which is affected by chronic pain. The prolonged activation of the CeA Enk interneurons reduced neohypophagia in the novelty suppressed feeding test and increased ΔFosB (a marker for sustained neuronal activation) in the vlPAG of mice with chronic pain. All together, the results of our experiments point to an important role of the CeA Enk neurons in the control of both nociception and emotion. Activation of Enk neurons resulted in sustained analgesia accompanied by anxiolysis and antidepressant effects. Very likely, these effects of CeA Enk neurons are result of the activation of vlPAG, a brain region that is essential not only for descending inhibition of pain but it is also a core element in the resilience to stress.
Collapse
Affiliation(s)
- Tanvi Paretkar
- Department of Physiology and Biophysics, Chicago Medical School Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd., North Chicago, IL 60064, United States.
| | - Eugene Dimitrov
- Department of Physiology and Biophysics, Chicago Medical School Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd., North Chicago, IL 60064, United States.
| |
Collapse
|
134
|
Abstract
Pain has a strong emotional component and is defined by its unpleasantness. Chronic pain represents a complex disorder with anxio-depressive symptoms and cognitive deficits. Underlying mechanisms are still not well understood but an important role for interactions between prefrontal cortical areas and subcortical limbic structures has emerged. Evidence from preclinical studies in the rodent brain suggests that neuroplastic changes in prefrontal (anterior cingulate, prelimbic and infralimbic) cortical and subcortical (amygdala and nucleus accumbens) brain areas and their interactions (corticolimbic circuitry) contribute to the complexity and persistence of pain and may be predetermining factors as has been proposed in recent human neuroimaging studies.
Collapse
Affiliation(s)
- Jeremy M Thompson
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, United States
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, United States; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States.
| |
Collapse
|
135
|
Filippini HF, Scalzilli PA, Costa KM, Freitas RDS, Campos MM. Activation of trigeminal ganglion satellite glial cells in CFA-induced tooth pulp pain in rats. PLoS One 2018; 13:e0207411. [PMID: 30419075 PMCID: PMC6231674 DOI: 10.1371/journal.pone.0207411] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023] Open
Abstract
This study further investigated the mechanisms underlying the rat model of tooth pulp inflammatory pain elicited by complete Freund's adjuvant (CFA), in comparison to other pulpitis models. Pulps of the left maxillary first molars were accessed. In the CFA group, the pulps were exposed, and CFA application was followed by dental sealing. In the open group, the pulps were left exposed to the oral cavity. For the closed group, the pulps were exposed, and the teeth were immediately sealed. Naïve rats were used as negative controls. Several parameters were evaluated at 1, 2, 3 and 8 days. There was no statistical significant difference among the groups when body weight variation, food or water consumption were compared. Analysis of serum cytokines (IL-1β, TNF or IL-6) or differential blood cell counts did not reveal any evidence of systemic inflammation. The CFA group displayed a significant reduction in the locomotor activity (at 1 and 3 days), associated with an increased activation of satellite glial cells in the ipsilateral trigeminal ganglion (TG; for up to 8 days). Amygdala astrocyte activation was unaffected in any experimental groups. We provide novel evidence indicating that CFA-induced pulp inflammation impaired the locomotor activity, with persistent activation of ipsilateral TG satellite cells surrounding sensory neurons, without any evidence of systemic inflammation or amygdala astrogliosis.
Collapse
Affiliation(s)
- Helena F. Filippini
- Programa de Pós-graduação em Odontologia, Escola de Ciências da Saúde, PUCRS, Porto Alegre, RS, Brasil
| | - Paulo A. Scalzilli
- Laboratório de Patologia, Escola de Ciência da Saúde, PUCRS, Porto Alegre, RS, Brasil
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde, PUCRS, Porto Alegre, RS, Brasil
| | - Kesiane M. Costa
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, PUCRS, Porto Alegre, RS, Brasil
| | - Raquel D. S. Freitas
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde, PUCRS, Porto Alegre, RS, Brasil
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, PUCRS, Porto Alegre, RS, Brasil
| | - Maria M. Campos
- Programa de Pós-graduação em Odontologia, Escola de Ciências da Saúde, PUCRS, Porto Alegre, RS, Brasil
- Laboratório de Patologia, Escola de Ciência da Saúde, PUCRS, Porto Alegre, RS, Brasil
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde, PUCRS, Porto Alegre, RS, Brasil
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, PUCRS, Porto Alegre, RS, Brasil
| |
Collapse
|
136
|
Mazzitelli M, Palazzo E, Maione S, Neugebauer V. Group II Metabotropic Glutamate Receptors: Role in Pain Mechanisms and Pain Modulation. Front Mol Neurosci 2018; 11:383. [PMID: 30356691 PMCID: PMC6189308 DOI: 10.3389/fnmol.2018.00383] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022] Open
Abstract
Glutamate is the main excitatory neurotransmitter in the nervous system and plays a critical role in nociceptive processing and pain modulation. G-protein coupled metabotropic glutamate receptors (mGluRs) are widely expressed in the central and peripheral nervous system, and they mediate neuronal excitability and synaptic transmission. Eight different mGluR subtypes have been identified so far, and are classified into Groups I-III. Group II mGluR2 and mGluR3 couple negatively to adenylyl cyclase through Gi/Go proteins, are mainly expressed presynaptically, and typically inhibit the release of neurotransmitters, including glutamate and GABA. Group II mGluRs have consistently been linked to pain modulation; they are expressed in peripheral, spinal and supraspinal elements of pain-related neural processing. Pharmacological studies have shown anti-nociceptive/analgesic effects of group II mGluR agonists in preclinical models of acute and chronic pain, although much less is known about mechanisms and sites of action for mGluR2 and mGluR3 compared to other mGluRs. The availability of orthosteric and new selective allosteric modulators acting on mGluR2 and mGluR3 has provided valuable tools for elucidating (subtype) specific contributions of these receptors to the pathophysiological mechanisms of pain and other disorders and their potential as therapeutic targets. This review focuses on the important role of group II mGluRs in the neurobiology of pain mechanisms and behavioral modulation, and discusses evidence for their therapeutic potential in pain.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Enza Palazzo
- Section of Pharmacology L. Donatelli, Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Sabatino Maione
- Section of Pharmacology L. Donatelli, Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
137
|
Boccella S, Cristiano C, Romano R, Iannotta M, Belardo C, Farina A, Guida F, Piscitelli F, Palazzo E, Mazzitelli M, Imperatore R, Tunisi L, de Novellis V, Cristino L, Di Marzo V, Calignano A, Maione S, Luongo L. Ultra-micronized palmitoylethanolamide rescues the cognitive decline-associated loss of neural plasticity in the neuropathic mouse entorhinal cortex-dentate gyrus pathway. Neurobiol Dis 2018; 121:106-119. [PMID: 30266286 DOI: 10.1016/j.nbd.2018.09.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/10/2018] [Accepted: 09/24/2018] [Indexed: 02/08/2023] Open
Abstract
Chronic pain is associated with cognitive deficits. Palmitoylethanolamide (PEA) has been shown to ameliorate pain and pain-related cognitive impairments by restoring glutamatergic synapses functioning in the spared nerve injury (SNI) of the sciatic nerve in mice. SNI reduced mechanical and thermal threshold, spatial memory and LTP at the lateral entorhinal cortex (LEC)-dentate gyrus (DG) pathway. It decreased also postsynaptic density, volume and dendrite arborization of DG and increased the expression of metabotropic glutamate receptor 1 and 7 (mGluR1 and mGluR7), of the GluR1, GluR1s845 and GluR1s831 subunits of AMPA receptor and the levels of glutamate in the DG. The level of the endocannabinoid 2-arachidonoylglycerol (2-AG) was instead increased in the LEC. Chronic treatment with PEA, starting from when neuropathic pain was fully developed, was able to reverse mechanical allodynia and thermal hyperalgesia, memory deficit and LTP in SNI wild type, but not in PPARα null, mice. PEA also restored the level of glutamate and the expression of phosphorylated GluR1 subunits, postsynaptic density and neurogenesis. Altogether, these results suggest that neuropathic pain negatively affects cognitive behavior and related LTP, glutamatergic synapse and synaptogenesis in the DG. In these conditions PEA treatment alleviates pain and cognitive impairment by restoring LTP and synaptic maladaptative changes in the LEC-DG pathway. These outcomes open new perspectives for the use of the N-acylethanolamines, such as PEA, for the treatment of neuropathic pain and its central behavioural sequelae.
Collapse
Affiliation(s)
- Serena Boccella
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Claudia Cristiano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Rosaria Romano
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Carmela Belardo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Antonio Farina
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
| | - Enza Palazzo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX
| | - Roberta Imperatore
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Lea Tunisi
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
| | - Vito de Novellis
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Luigia Cristino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
| | - Antonio Calignano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Pharmacology Division, University of Campania "L. Vanvitelli", 80138 Naples, Italy.
| |
Collapse
|
138
|
Kiritoshi T, Neugebauer V. Pathway-Specific Alterations of Cortico-Amygdala Transmission in an Arthritis Pain Model. ACS Chem Neurosci 2018; 9:2252-2261. [PMID: 29630339 PMCID: PMC6146017 DOI: 10.1021/acschemneuro.8b00022] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Medial prefrontal cortex (mPFC) and amygdala are closely interconnected brain areas that play a key role in cognitive-affective aspects of pain through their reciprocal interactions. Clinical and preclinical evidence suggests that dysfunctions in the mPFC-amygdala circuitry underlie pain-related cognitive-affective deficits. However, synaptic mechanisms of pain-related changes in these long-range pathways are largely unknown. Here we used optogenetics and brain slice physiology to analyze synaptic transmission in different types of amygdala neurons driven by inputs from infralimbic (IL) and prelimbic (PL) subdivisions of the mPFC. We found that IL inputs evoked stronger synaptic inhibition of neurons in the latero-capsular division of the central nucleus (CeLC) of the amygdala than PL inputs, and this inhibition was impaired in an arthritis pain model. Furthermore, inhibition-excitation ratio in basolateral amygdala neurons was increased in the pain model in the IL pathway but not in the PL pathway. These results suggest that IL rather than PL controls CeLC activity, and that changes in this acute pain model occur predominantly in the IL-amygdala pathway.
Collapse
Affiliation(s)
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience
- Center of Excellence for Translational Neuroscience and Therapeutics Texas Tech University Health Sciences Center (TTUHSC), School of Medicine 3601 4th Street, Lubbock, TX 79430-6592
| |
Collapse
|
139
|
Seno MDJ, Assis DV, Gouveia F, Antunes GF, Kuroki M, Oliveira CC, Santos LCT, Pagano RL, Martinez RCR. The critical role of amygdala subnuclei in nociceptive and depressive-like behaviors in peripheral neuropathy. Sci Rep 2018; 8:13608. [PMID: 30206257 PMCID: PMC6134132 DOI: 10.1038/s41598-018-31962-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/30/2018] [Indexed: 01/05/2023] Open
Abstract
The amygdala is an important component of the limbic system that participates in the control of the pain response and modulates the affective-motivational aspect of pain. Neuropathic pain is a serious public health problem and has a strong affective-motivational component that makes it difficult to treat. The central (CeA), basolateral (BLA) and lateral (LA) nuclei of the amygdala are involved in the processing and regulation of chronic pain. However, the roles of these nuclei in the maintenance of neuropathic pain, anxiety and depression remain unclear. Thus, the main objective of this study was to investigate the role of amygdala subnuclei in the modulation of neuropathic pain, including the affective-motivational axis, in an experimental model of peripheral neuropathy. The specific goals were as follows: (1) To evaluate the nociceptive responses and the patterns of activation of the CeA, BLA and LA in neuropathic rats; and (2) To evaluate the effect of inactivating the amygdala nuclei on the nociceptive response, anxiety and depressive behaviors, motor activity, and plasma stress hormones in animals with neuropathic pain. Thus, mechanical hyperalgesia and allodynia, and the pattern of c-Fos staining in the amygdala subnuclei were evaluated in rats with chronic constriction of the sciatic nerve, as well as sham-operated and naïve rats. Once the amygdala subnuclei involved in neuropathic pain response were defined, those subnuclei were pharmacological inactivated. The effect of muscimol inactivation on the nociceptive response (hyperalgesia and allodynia), anxiety (elevated plus-maze), depressive-like behavior (forced swim test), motor activity (open field), and plasma stress hormone levels (corticosterone and adrenocorticotropic hormone) were evaluated in sham-operated and neuropathic animals. The results showed that the anterior and posterior portions of the BLA and the central portion of the CeA are involved in controlling neuropathic pain. The inactivation of these nuclei reversed hyperalgesia, allodynia and depressive-like behavior in animals with peripheral neuropathy. Taken together, our findings improve our understanding of the neurocircuitry involved in persistent pain and the roles of specific amygdala subnuclei in the modulation of neuropathic pain, including the neurocircuitry that processes the affective-motivational component of pain.
Collapse
Affiliation(s)
- Midiã D J Seno
- Laboratory of Neuroscience, Hospital Sirio-Libanes, Sao Paulo, SP, Brazil
| | - Danielle V Assis
- Laboratory of Neuroscience, Hospital Sirio-Libanes, Sao Paulo, SP, Brazil
| | - Flávia Gouveia
- Laboratory of Neuroscience, Hospital Sirio-Libanes, Sao Paulo, SP, Brazil
| | - Geiza F Antunes
- Laboratory of Neuroscience, Hospital Sirio-Libanes, Sao Paulo, SP, Brazil
| | - Mayra Kuroki
- Laboratory of Neuroscience, Hospital Sirio-Libanes, Sao Paulo, SP, Brazil
| | | | - Lennon C T Santos
- Laboratory of Neuroscience, Hospital Sirio-Libanes, Sao Paulo, SP, Brazil
| | - Rosana L Pagano
- Laboratory of Neuroscience, Hospital Sirio-Libanes, Sao Paulo, SP, Brazil
| | | |
Collapse
|
140
|
Jensen JR, Pitcher MH, Yuan ZX, Ramsden CE, Domenichiello AF. Concentrations of oxidized linoleic acid derived lipid mediators in the amygdala and periaqueductal grey are reduced in a mouse model of chronic inflammatory pain. Prostaglandins Leukot Essent Fatty Acids 2018; 135:128-136. [PMID: 30103924 PMCID: PMC6269101 DOI: 10.1016/j.plefa.2018.07.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 07/17/2018] [Accepted: 07/17/2018] [Indexed: 12/26/2022]
Abstract
Chronic pain is both a global public health concern and a serious source of personal suffering for which current treatments have limited efficacy. Recently, oxylipins derived from linoleic acid (LA), the most abundantly consumed polyunsaturated fatty acid in the modern diet, have been implicated as mediators of pain in the periphery and spinal cord. However, oxidized linoleic acid derived mediators (OXLAMs) remain understudied in the brain, particularly during pain states. In this study, we employed a mouse model of chronic inflammatory pain followed by a targeted lipidomic analysis of the animals' amygdala and periaqueductal grey (PAG) using LC-MS/MS to investigate the effect of chronic inflammatory pain on oxylipin concentrations in these two brain nuclei known to participate in pain sensation and perception. From punch biopsies of these brain nuclei, we detected twelve OXLAMs in both the PAG and amygdala and one arachidonic acid derived mediator, 15-HETE, in the amygdala only. In the amygdala, we observed an overall decrease in the concentration of the majority of OXLAMs detected, while in the PAG the concentrations of only the epoxide LA derived mediators, 9,10-EpOME and 12,13-EpOME, and one trihydroxy LA derived mediator, 9,10,11-TriHOME, were reduced. This data provides the first evidence that OXLAM concentrations in the brain are affected by chronic pain, suggesting that OXLAMs may be relevant to pain signaling and adaptation to chronic pain in pain circuits in the brain and that the current view of OXLAMs in nociception derived from studies in the periphery is incomplete.
Collapse
Affiliation(s)
- J R Jensen
- Lipid Mediators, Inflammation and Pain Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, United States
| | - M H Pitcher
- National Center for Complementary and Integrative Health, NIH, Bethesda, MD, United States
| | - Z X Yuan
- Lipid Mediators, Inflammation and Pain Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, United States
| | - C E Ramsden
- Lipid Mediators, Inflammation and Pain Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, United States; Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, United States
| | - A F Domenichiello
- Lipid Mediators, Inflammation and Pain Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, United States.
| |
Collapse
|
141
|
Reduced resting-state functional connectivity of the basolateral amygdala to the medial prefrontal cortex in preweaning rats exposed to chronic early-life stress. Brain Struct Funct 2018; 223:3711-3729. [DOI: 10.1007/s00429-018-1720-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/15/2018] [Indexed: 10/28/2022]
|
142
|
Thompson JM, Yakhnitsa V, Ji G, Neugebauer V. Small conductance calcium activated potassium (SK) channel dependent and independent effects of riluzole on neuropathic pain-related amygdala activity and behaviors in rats. Neuropharmacology 2018; 138:219-231. [PMID: 29908238 DOI: 10.1016/j.neuropharm.2018.06.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/04/2018] [Accepted: 06/11/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND PURPOSE Chronic neuropathic pain is an important healthcare issue with significant emotional components. The amygdala is a brain region involved in pain and emotional-affective states and disorders. The central amygdala output nucleus (CeA) contains small-conductance calcium-activated potassium (SK) channels that can control neuronal activity. A clinically available therapeutic, riluzole can activate SK channels and may have antinociceptive effects through a supraspinal action. We tested the hypothesis that riluzole inhibits neuropathic pain behaviors by inhibiting pain-related changes in CeA neurons, in part at least through SK channel activation. EXPERIMENTAL APPROACH Brain slice physiology and behavioral assays were done in adult Sprague Dawley rats. Audible and ultrasonic vocalizations and von Frey thresholds were measured in sham and neuropathic rats 4 weeks after left L5 spinal nerve ligation (SNL model). Whole cell patch-clamp recordings of regular firing CeA neurons in brain slices were used to measure synaptic transmission and neuronal excitability. KEY RESULTS In brain slices, riluzole increased the SK channel-mediated afterhyperpolarization and synaptic inhibition, but inhibited neuronal excitability through an SK channel independent action. SNL rats had increased vocalizations and decreased withdrawal thresholds compared to sham rats, and intra-CeA administration of riluzole inhibited vocalizations and depression-like behaviors but did not affect withdrawal thresholds. Systemic riluzole administration also inhibited these changes, demonstrating the clinical utility of this strategy. SK channel blockade in the CeA attenuated the inhibitory effects of systemic riluzole on vocalizations, confirming SK channel involvement in these effects. CONCLUSIONS AND IMPLICATIONS The results suggest that riluzole has beneficial effects on neuropathic pain behaviors through SK channel dependent and independent mechanisms in the amygdala.
Collapse
Affiliation(s)
- Jeremy M Thompson
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, USA
| | - Vadim Yakhnitsa
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, USA
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|