1
|
Neugebauer V, Presto P, Yakhnitsa V, Antenucci N, Mendoza B, Ji G. Pain-related cortico-limbic plasticity and opioid signaling. Neuropharmacology 2023; 231:109510. [PMID: 36944393 PMCID: PMC10585936 DOI: 10.1016/j.neuropharm.2023.109510] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
Neuroplasticity in cortico-limbic circuits has been implicated in pain persistence and pain modulation in clinical and preclinical studies. The amygdala has emerged as a key player in the emotional-affective dimension of pain and pain modulation. Reciprocal interactions with medial prefrontal cortical regions undergo changes in pain conditions. Other limbic and paralimbic regions have been implicated in pain modulation as well. The cortico-limbic system is rich in opioids and opioid receptors. Preclinical evidence for their pain modulatory effects in different regions of this highly interactive system, potentially opposing functions of different opioid receptors, and knowledge gaps will be described here. There is little information about cell type- and circuit-specific functions of opioid receptor subtypes related to pain processing and pain-related plasticity in the cortico-limbic system. The important role of anterior cingulate cortex (ACC) and amygdala in MOR-dependent analgesia is most well-established, and MOR actions in the mesolimbic system appear to be similar but remain to be determined in mPFC regions other than ACC. Evidence also suggests that KOR signaling generally serves opposing functions whereas DOR signaling in the ACC has similar, if not synergistic effects, to MOR. A unifying picture of pain-related neuronal mechanisms of opioid signaling in different elements of the cortico-limbic circuitry has yet to emerge. This article is part of the Special Issue on "Opioid-induced changes in addiction and pain circuits".
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.
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Vadim Yakhnitsa
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Nico Antenucci
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Brianna Mendoza
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 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
| |
Collapse
|
2
|
Hou Y, Zou G, Wang X, Guo H, Ma X, Cheng X, Xie Z, Zuo X, Xia J, Mao H, Yuan M, Chen Q, Cao P, Yang Y, Zhang L, Xiong W. Coordinated activity of a central pathway drives associative opioid analgesic tolerance. SCIENCE ADVANCES 2023; 9:eabo5627. [PMID: 36753548 PMCID: PMC9908028 DOI: 10.1126/sciadv.abo5627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 12/22/2022] [Indexed: 06/18/2023]
Abstract
Opioid analgesic tolerance, a root cause of opioid overdose and misuse, can develop through an associative learning. Despite intensive research, the locus and central pathway subserving the associative opioid analgesic tolerance (AOAT) remains unclear. Using a combination of chemo/optogenetic manipulation with calcium imaging and slice physiology, here we identify neuronal ensembles in a hierarchically organized pathway essential for AOAT. The association of morphine-induced analgesia with an environmental condition drives glutamatergic signaling from ventral hippocampus (vHPC) to dorsomedial prefrontal cortex (dmPFC) cholecystokininergic (CCKergic) neurons. Excitation of CCKergic neurons, which project and release CCK to basolateral amygdala (BLA) glutamatergic neurons, relays AOAT signal through inhibition of BLA μ-opioid receptor function, thereby leading to further loss of morphine analgesic efficacy. This work provides evidence for a circuit across different brain regions distinct for opioid analgesic tolerance. The components of this pathway are potential targets to treat opioid overdose and abuse.
Collapse
Affiliation(s)
- Yiwen Hou
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Guichang Zou
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
| | - Xianglian Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Hui Guo
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Xiao Ma
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Xingyu Cheng
- National Institute of Biological Sciences, Beijing 102206, China
| | - Zhiyong Xie
- National Institute of Biological Sciences, Beijing 102206, China
| | - Xin Zuo
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Jing Xia
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Huanhuan Mao
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Man Yuan
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Qi Chen
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Peng Cao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Yupeng Yang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Li Zhang
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Xiong
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei 230026, China
| |
Collapse
|
3
|
McPherson KB, Ingram SL. Cellular and circuit diversity determines the impact of endogenous opioids in the descending pain modulatory pathway. Front Syst Neurosci 2022; 16:963812. [PMID: 36045708 PMCID: PMC9421147 DOI: 10.3389/fnsys.2022.963812] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/15/2022] [Indexed: 01/31/2023] Open
Abstract
The descending pain modulatory pathway exerts important bidirectional control of nociceptive inputs to dampen and/or facilitate the perception of pain. The ventrolateral periaqueductal gray (vlPAG) integrates inputs from many regions associated with the processing of nociceptive, cognitive, and affective components of pain perception, and is a key brain area for opioid action. Opioid receptors are expressed on a subset of vlPAG neurons, as well as on both GABAergic and glutamatergic presynaptic terminals that impinge on vlPAG neurons. Microinjection of opioids into the vlPAG produces analgesia and microinjection of the opioid receptor antagonist naloxone blocks stimulation-mediated analgesia, highlighting the role of endogenous opioid release within this region in the modulation of nociception. Endogenous opioid effects within the vlPAG are complex and likely dependent on specific neuronal circuits activated by acute and chronic pain stimuli. This review is focused on the cellular heterogeneity within vlPAG circuits and highlights gaps in our understanding of endogenous opioid regulation of the descending pain modulatory circuits.
Collapse
Affiliation(s)
- Kylie B. McPherson
- Division of Neuroscience and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy,Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States
| | - Susan L. Ingram
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, United States,*Correspondence: Susan L. Ingram
| |
Collapse
|
4
|
Medina S, O’Daly OG, Howard MA, Feliu-Soler A, Luciano JV. Differential Brain Perfusion Changes Following Two Mind–Body Interventions for Fibromyalgia Patients: an Arterial Spin Labelling fMRI Study. Mindfulness (N Y) 2022; 13:449-461. [PMID: 35222735 PMCID: PMC8831296 DOI: 10.1007/s12671-021-01806-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/04/2021] [Indexed: 12/20/2022]
Abstract
Objectives Further mechanistic insight on mind–body techniques for fibromyalgia (FMS) is needed. Arterial spin labelling (ASL) imaging can capture changes in regional cerebral blood flow (rCBF) that relate to spontaneous pain. Methods We recruited FMS patients undergoing either mindfulness-based stress reduction training (MBSR, n = 14) or a psychoeducational programme (FibroQoL, n = 18), and a control FMS group with no add-on treatment (n = 14). We acquired whole-brain rCBF maps and self-report measures at baseline and following treatment and explored interaction effects in brain perfusion between the treatment group and session with a focus on the amygdala, the insula and the anterior cingulate cortex (ACC). Results We identified a significant interaction effect in the amygdala, which corresponded with rCBF decreases following FibroQoL specifically. At baseline, rCBF in the amygdala for the FibroQoL group correlated with pain catastrophizing and anxiety scores, but not after treatment, suggesting a decoupling between activity in the amygdala and negative emotional symptoms of FMS as a consequence of treatment. Baseline rCBF correlated positively with pain symptoms in the ACC and the anterior insula across all patients; moreover, the correlation between rCBF changes post intervention in the insula and pain improvement was negative for both treatments and significantly different from the control group. We suggest that there is disruption of the typical relationship between clinical pain and activity as a product of these two nonpharmacological therapies. Conclusions We have demonstrated that different mind-to-body treatments correspond to differential changes in clinical symptoms and brain activity patterns, which encourages future research investigating predictors of treatment response. Trial Registration NCT02561416. Supplementary Information The online version contains supplementary material available at 10.1007/s12671-021-01806-2.
Collapse
Affiliation(s)
- Sonia Medina
- Department of Neuroimaging, King’s College London, London, UK
| | - Owen G. O’Daly
- Department of Neuroimaging, King’s College London, London, UK
| | | | - Albert Feliu-Soler
- Department of Clinical & Health Psychology, Autonomous University of Barcelona, Bellaterra, Spain
| | - Juan V. Luciano
- Department of Clinical & Health Psychology, Autonomous University of Barcelona, Bellaterra, Spain
- Research & Innovation Unit, Parc Sanitari Sant Joan de Déu, Sant Boi de Llobregat, Spain
| |
Collapse
|
5
|
Chartier M, Desgagné M, Sousbie M, Rumsby C, Chevillard L, Théroux L, Haroune L, Côté J, Longpré JM, Boudreault PL, Marsault É, Sarret P. Pharmacodynamic and pharmacokinetic profiles of a neurotensin receptor type 2 (NTS2) analgesic macrocyclic analog. Biomed Pharmacother 2021; 141:111861. [PMID: 34229249 DOI: 10.1016/j.biopha.2021.111861] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
The current opioid crisis highlights the urgent need to develop safe and effective pain medications. Thus, neurotensin (NT) compounds represent a promising approach, as the antinociceptive effects of NT are mediated by activation of the two G protein-coupled receptor subtypes (i.e., NTS1 and NTS2) and produce potent opioid-independent analgesia. Here, we describe the synthesis and pharmacodynamic and pharmacokinetic properties of the first constrained NTS2 macrocyclic NT(8-13) analog. The Tyr11 residue of NT(8-13) was replaced with a Trp residue to achieve NTS2 selectivity, and a rationally designed side-chain to side-chain macrocyclization reaction was applied between Lys8 and Trp11 to constrain the peptide in an active binding conformation and limit its recognition by proteolytic enzymes. The resulting macrocyclic peptide, CR-01-64, exhibited high-affinity for NTS2 (Ki 7.0 nM), with a more than 125-fold selectivity over NTS1, as well as an improved plasma stability profile (t1/2 > 24 h) compared with NT (t1/2 ~ 2 min). Following intrathecal administration, CR-01-64 exerted dose-dependent and long-lasting analgesic effects in acute (ED50 = 4.6 µg/kg) and tonic (ED50 = 7.1 µg/kg) pain models as well as strong mechanical anti-allodynic effects in the CFA-induced chronic inflammatory pain model. Of particular importance, this constrained NTS2 analog exerted potent nonopioid antinociceptive effects and potentiated opioid-induced analgesia when combined with morphine. At high doses, CR-01-64 did not cause hypothermia or ileum relaxation, although it did induce mild and short-term hypotension, all of which are physiological effects associated with NTS1 activation. Overall, these results demonstrate the strong therapeutic potential of NTS2-selective analogs for the management of pain.
Collapse
Affiliation(s)
- Magali Chartier
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Michael Desgagné
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Marc Sousbie
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Charles Rumsby
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | | | - Léa Théroux
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Lounès Haroune
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Jérôme Côté
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Jean-Michel Longpré
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Pierre-Luc Boudreault
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Éric Marsault
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Philippe Sarret
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
6
|
A Neural Circuit from Thalamic Paraventricular Nucleus to Central Amygdala for the Facilitation of Neuropathic Pain. J Neurosci 2020; 40:7837-7854. [PMID: 32958568 DOI: 10.1523/jneurosci.2487-19.2020] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 09/04/2020] [Accepted: 09/09/2020] [Indexed: 12/23/2022] Open
Abstract
As one of the thalamic midline nuclei, the thalamic paraventricular nucleus (PVT) is considered to be an important signal integration site for many descending and ascending pathways that modulate a variety of behaviors, including feeding, emotions, and drug-seeking. A recent study has demonstrated that the PVT is implicated in the acute visceral pain response, but it is unclear whether the PVT plays a critical role in the central processing of chronic pain. Here, we report that the neurons in the posterior portion of the PVT (pPVT) and their downstream pathway are involved in descending nociceptive facilitation regarding the development of neuropathic pain conditions in male rats. Lesions or inhibition of pPVT neurons alleviated mechanical allodynia induced by spared nerve injury (SNI). The excitability of pPVT-central amygdala (CeA) projection neurons was significantly increased in SNI rats. Importantly, selective optogenetic activation of the pPVT-CeA pathway induced obvious mechanical hypersensitivity in naive rats. In addition, we used rabies virus (RV)-based and cell-type-specific retrograde transsynaptic tracing techniques to define a novel neuronal circuit in which glutamatergic neurons in the vlPAG were the target of the pPVT-CeA descending facilitation pathway. Our data suggest that this pPVTGlu+-CeA-vlPAGGlu+ circuit mediates central mechanisms of descending pain facilitation underlying persistent pain conditions.SIGNIFICANCE STATEMENT Studies have shown that the interactions between the posterior portion of the thalamic paraventricular nucleus (pPVT) and central amygdala (CeA) play a critical role in pain-related emotional regulation. However, most reports have associated this circuit with fear and anxiety behaviors. Here, an integrative approach of behavioral tests, electrophysiology, and immunohistochemistry was used to advance the novel concept that the pPVT-CeA pathway activation facilitates neuropathic pain processing. Using rabies virus (RV)-based and cell-type-specific retrograde transsynaptic tracing techniques, we found that glutamatergic neurons in the vlPAG were the target of the pPVT-CeA pathway. Thus, this study indicates the involvement of a pPVTGlu+-CeA-vlPAGGlu+ pathway in a descending facilitatory mechanism underlying neuropathic pain.
Collapse
|
7
|
Affiliation(s)
- Marlene A Wilson
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
- Columbia VA Health Care System, Columbia, SC, United States
| | - Alexander J McDonald
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
8
|
Qiao H, Gao Y, Huang Q, Jia R. The central nucleus of the amygdala lesion attenuates orthodontic pain during experimental tooth movement in rats. Brain Behav 2020; 10:e01506. [PMID: 31863574 PMCID: PMC6955828 DOI: 10.1002/brb3.1506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/29/2019] [Accepted: 11/29/2019] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Orthodontic pain is the most common adverse side effect reported in the context of tooth movement. Given its central role in processing pain and negative emotion, the central nucleus of the amygdala (CeA) is thought to be a key site involved in orthodontic pain sensation. METHODS In the present study, we therefore explored whether the CeA is involved in contributing to orthodontic pain in a rat model of tooth movement. For this study, we utilized adult male rats with bilateral sham or electrolytic CeA lesions (400 μA; 25 s), and then we analyzed face grooming behavior as a measure of pain sensation. RESULTS Through this approach, we found that there were time- and force-dependent factors influencing pain levels in these rats. We further found that bilateral CeA lesions markedly reduced tooth movement-induced orofacial pain and that unilateral CeA lesions did so to a lesser extent. CONCLUSIONS As such, these results suggest the CeA is a key area of orthodontic pain, with the results of this study highlighting potential avenues for achieving pain relief in those suffering from orthodontic pain.
Collapse
Affiliation(s)
- Hu Qiao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Xi'an Jiaotong University College of Stomatology, Xi'an, Shaanxi, China.,Department of Orthodontics, Xi'an Jiaotong University College of Stomatology, Xi'an, Shaanxi, China
| | - Yunan Gao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Xi'an Jiaotong University College of Stomatology, Xi'an, Shaanxi, China.,Department of Orthodontics, Xi'an Jiaotong University College of Stomatology, Xi'an, Shaanxi, China
| | - Qianqian Huang
- Department of Orthodontics, Xi'an Jiaotong University College of Stomatology, Xi'an, Shaanxi, China
| | - Ru Jia
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Xi'an Jiaotong University College of Stomatology, Xi'an, Shaanxi, China
| |
Collapse
|
9
|
Pedrón VT, Varani AP, Bettler B, Balerio GN. GABA B receptors modulate morphine antinociception: Pharmacological and genetic approaches. Pharmacol Biochem Behav 2019; 180:11-21. [PMID: 30851293 DOI: 10.1016/j.pbb.2019.02.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 02/28/2019] [Accepted: 02/28/2019] [Indexed: 10/27/2022]
Abstract
Previous studies in our laboratory showed an interaction between the GABAergic and opioid systems involved in the analgesic effect of baclofen (BAC). Furthermore, it is known that sex differences exist regarding various pharmacological responses of morphine (MOR) and they are related to an increased sensitivity to MOR effects in males. The aims of the present study were to evaluate the possible involvement of the GABAB receptors in the antinociceptive responses induced by MOR (1, 3 and 9 mg/kg, s.c.) administration using both pharmacological (BAC 2 mg/kg, i.p.; and 2-OH-saclofen, SAC 0.3 mg/kg, intra cisterna magna) and genetic approaches (GABAB1 knockout mice; GABAB1 KO) in mice of both sexes. In addition, we explored the alterations in c-Fos expression of different brain areas involved in the antinociceptive effect of MOR using both approaches. The pharmacological approach showed a higher dose-dependent antinociceptive effect of MOR in male mice compared to female mice. BAC and SAC pretreatment potentiated and attenuated the antinociceptive effect of MOR, respectively, in both sexes. The genetic approach revealed a dose-dependent antinociceptive effect of MOR in the wild type mice, but not in the GABAB1 KO mice and no sex differences were observed. Additionally, BAC and SAC pretreatment and the lack of GABAB1 subunit of the GABAB receptor prevented the changes observed in c-Fos expression in the cingulate cortex and nucleus accumbens of male mice. Our results suggest that the GABAB receptors are involved in the MOR antinociceptive effect of both male and female mice.
Collapse
Affiliation(s)
- Valeria T Pedrón
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Farmacológicas (ININFA), Buenos Aires, Argentina
| | - Andrés P Varani
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Farmacológicas (ININFA), Buenos Aires, Argentina
| | - Bernhard Bettler
- Department of Biomedicine, Institute of Physiology, Pharmazentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland
| | - Graciela N Balerio
- CONICET - Universidad de Buenos Aires, Instituto de Investigaciones Farmacológicas (ININFA), Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Central Amygdala Circuits Mediate Hyperalgesia in Alcohol-Dependent Rats. J Neurosci 2018; 38:7761-7773. [PMID: 30054393 DOI: 10.1523/jneurosci.0483-18.2018] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/06/2018] [Accepted: 07/21/2018] [Indexed: 01/13/2023] Open
Abstract
Alcohol withdrawal symptoms contribute to excessive alcohol drinking and relapse in alcohol-dependent individuals. Among these symptoms, alcohol withdrawal promotes hyperalgesia, but the neurological underpinnings of this phenomenon are not known. Chronic alcohol exposure alters cell signaling in the central nucleus of the amygdala (CeA), and the CeA is implicated in mediating alcohol dependence-related behaviors. The CeA projects to the periaqueductal gray (PAG), a region critical for descending pain modulation, and may have a role in alcohol withdrawal hyperalgesia. Here, we tested the roles of (1) CeA projections to PAG, (2) CeA melanocortin signaling, and (3) PAG μ-opioid receptor signaling in mediating thermal nociception and alcohol withdrawal hyperalgesia in male Wistar rats. Our results demonstrate that alcohol dependence reduces GABAergic signaling from CeA terminals onto PAG neurons and alters the CeA melanocortin system, that CeA-PAG projections and CeA melanocortin signaling mediate alcohol withdrawal hyperalgesia, and that μ-opioid receptors in PAG filter CeA effects on thermal nociception.SIGNIFICANCE STATEMENT Hyperalgesia is commonly seen in individuals with alcohol use disorder during periods of withdrawal, but the neurological underpinnings behind this phenomenon are not completely understood. Here, we tested whether alcohol dependence exerts its influence on pain modulation via effects on the limbic system. Using behavioral, optogenetic, electrophysiological, and molecular biological approaches, we demonstrate that central nucleus of the amygdala (CeA) projections to periaqueductal gray mediate thermal hyperalgesia in alcohol-dependent and alcohol-naive rats. Using pharmacological approaches, we show that melanocortin receptor-4 signaling in CeA alters alcohol withdrawal hyperalgesia, but this effect is not mediated directly at synaptic inputs onto periaqueductal gray-projecting CeA neurons. Overall, our findings support a role for limbic influence over the descending pain pathway and identify a potential therapeutic target for treating hyperalgesia in individuals with alcohol use disorder .
Collapse
|
11
|
Analgesic Neural Circuits Are Activated by Electroacupuncture at Two Sets of Acupoints. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:3840202. [PMID: 27429635 PMCID: PMC4939346 DOI: 10.1155/2016/3840202] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/03/2016] [Accepted: 06/05/2016] [Indexed: 11/17/2022]
Abstract
To investigate analgesic neural circuits activated by electroacupuncture (EA) at different sets of acupoints in the brain, goats were stimulated by EA at set of Baihui-Santai acupoints or set of Housanli acupoints for 30 min. The pain threshold was measured using the potassium iontophoresis method. The levels of c-Fos were determined with Streptavidin-Biotin Complex immunohistochemistry. The results showed pain threshold induced by EA at set of Baihui-Santai acupoints was 44.74% ± 4.56% higher than that by EA at set of Housanli acupoints (32.64% ± 5.04%). Compared with blank control, EA at two sets of acupoints increased c-Fos expression in the medial septal nucleus (MSN), the arcuate nucleus (ARC), the nucleus amygdala basalis (AB), the lateral habenula nucleus (HL), the ventrolateral periaqueductal grey (vlPAG), the locus coeruleus (LC), the nucleus raphe magnus (NRM), the pituitary gland, and spinal cord dorsal horn (SDH). Compared with EA at set of Housanli points, EA at set of Baihui-Santai points induced increased c-Fos expression in AB but decrease in MSN, the paraventricular nucleus of the hypothalamus, HL, and SDH. It suggests that ARC-PAG-NRM/LC-SDH and the hypothalamus-pituitary may be the common activated neural pathways taking part in EA-induced analgesia at the two sets of acupoints.
Collapse
|
12
|
μ-Opioid Receptor-Mediated Inhibition of Intercalated Neurons and Effect on Synaptic Transmission to the Central Amygdala. J Neurosci 2015; 35:7317-25. [PMID: 25972162 DOI: 10.1523/jneurosci.0204-15.2015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The amygdala is a key region for the processing of information underlying fear, anxiety, and fear extinction. Within the local neuronal networks of the amygdala, a population of inhibitory, intercalated neurons (ITCs) modulates the flow of information among various nuclei of amygdala, including the basal nucleus (BA) and the centromedial nucleus (CeM) of the amygdala. These ITCs have been shown to be important during fear extinction and are target of a variety of neurotransmitters and neuropeptides. Here we provide evidence that the activation of μ-opioid receptors (MORs) by the specific agonist DAMGO ([D-Ala2,N-Me-Phe4,Gly5-ol]-Enkephalin) hyperpolarizes medially located ITCs (mITCs) in acute brain slices of mice. Moreover, we use whole-cell patch-clamp recordings in combination with local electrical stimulation or glutamate uncaging to analyze the effect of MOR activation on local microcircuits. We show that the GABAergic transmission between mITCs and CeM neurons is attenuated by DAMGO, whereas the glutamatergic transmission on CeM neurons and mITCs is unaffected. Furthermore, MOR activation induced by theta burst stimulation in BA suppresses plastic changes of feedforward inhibitory transmission onto CeM neurons as revealed by the MOR antagonist CTAP d-Phe-Cys-Tyr-d-Trp-Arg-Thr-Pen-Thr-NH2. In summary, the mITCs constitute a target for the opioid system, and therefore, the activation of MOR in ITCs might play a central role in the modulation of the information processing between the basolateral complex of the amygdala and central nuclei of the amygdala.
Collapse
|
13
|
Spuz CA, Tomaszycki ML, Borszcz GS. N-methyl-D-aspartate receptor agonism and antagonism within the amygdaloid central nucleus suppresses pain affect: differential contribution of the ventrolateral periaqueductal gray. THE JOURNAL OF PAIN 2014; 15:1305-18. [PMID: 25261341 DOI: 10.1016/j.jpain.2014.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/07/2014] [Accepted: 09/16/2014] [Indexed: 01/12/2023]
Abstract
UNLABELLED The amygdala contributes to the generation of pain affect, and the amygdaloid central nucleus (CeA) receives nociceptive input that is mediated by glutamatergic neurotransmission. The present study compared the contribution of N-methyl-d-aspartate (NMDA) receptor agonism and antagonism in the CeA to generation of the affective response of rats to an acute noxious stimulus. Vocalizations that occur following a brief tail shock (vocalization afterdischarges) are a validated rodent model of pain affect and were preferentially suppressed, in a dose-dependent manner, by bilateral injection into the CeA of NMDA (.1, .25, .5, or 1 μg/side) or the NMDA receptor antagonist d-(-)-2-amino-5-phosphopentanoic acid (AP5; 1, 2, or 4 μg/side). Vocalizations that occur during tail shock were suppressed to a lesser degree, whereas spinal motor reflexes (tail flick and hind limb movements) were unaffected by injection of NMDA or AP5 into the CeA. Injection of NMDA, but not AP5, into the CeA increased c-Fos immunoreactivity in the ventrolateral periaqueductal gray, and unilateral injection of the μ-opiate receptor antagonist H-d-Phe-Cys-Tyr-d-Trp-Arg-Thr-Pen-Thr-NH2 (CTAP; .25 μg) into ventrolateral periaqueductal gray prevented the antinociception generated by injection of NMDA into the CeA. These findings demonstrate that although NMDA receptor agonism and antagonism in the CeA produce similar suppression of pain behaviors, they do so via different neurobiologic mechanisms. PERSPECTIVE The amygdala contributes to production of the emotional dimension of pain. NMDA receptor agonism and antagonism within the CeA suppressed rats' emotional response to acute painful stimulation. Understanding the neurobiology underlying emotional responses to pain will provide insights into new treatments for pain and its associated affective disorders.
Collapse
Affiliation(s)
- Catherine A Spuz
- Department of Psychology, Behavioral & Cognitive Neuroscience Program, Wayne State University, Detroit, Michigan
| | - Michelle L Tomaszycki
- Department of Psychology, Behavioral & Cognitive Neuroscience Program, Wayne State University, Detroit, Michigan
| | - George S Borszcz
- Department of Psychology, Behavioral & Cognitive Neuroscience Program, Wayne State University, Detroit, Michigan.
| |
Collapse
|
14
|
Do patients with functional chest pain have neuroplastic reorganization of the pain matrix? A diffusion tensor imaging study. Scand J Pain 2014; 5:85-90. [DOI: 10.1016/j.sjpain.2013.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 11/25/2013] [Indexed: 02/08/2023]
Abstract
Abstract
Background and aims
In functional chest pain (FCP) of presumed esophageal origin central nervous system hyperexcitability is generally believed to play an important role in pain pathogenesis. However, this theory has recently been challenged. Using magnetic resonance diffusion tensor imaging, the aim was to characterize any microstructural reorganization of the pain neuromatrix in FCP patients.
Methods
13 FCP patients and 20 matched healthy controls were studied in a 3T MR scanner. Inclusion criteria were relevant chest pain, normal coronary angiogram and normal upper gastrointestinal evaluation. Apparent diffusion coefficient (ADC) (i.e. mean diffusivity of water) and fractional anisotropy (FA) (i.e. directionality of water diffusion as a measure of fiber organization) values were assessed in the secondary sensory cortex, cingulate cortex, insula, prefrontal cortex, and amygdala.
Results
Overall, including all regions, no difference in ADC and FA values was found between the patients and controls (P = 0.79 and P = 0.23, respectively). Post-hoc tests revealed no difference in ADC and FA values of the individual regions. However, a trend of patients having increased ADC in the mid insula grey matter and increased FA in the mid insula white matter was observed (both P = 0.065).
Conclusions
This explorative study suggests that microstructural reorganization of the central pain neuromatrix may not be present in well-characterized FCP patients.
Implications
This finding, together with recent neurophysiologal evidence, challenges the theory of visceral hypersensitivity due to changes in the central nervous system in FCP patients.
Collapse
|
15
|
Boules M, Li Z, Smith K, Fredrickson P, Richelson E. Diverse roles of neurotensin agonists in the central nervous system. Front Endocrinol (Lausanne) 2013; 4:36. [PMID: 23526754 PMCID: PMC3605594 DOI: 10.3389/fendo.2013.00036] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 03/06/2013] [Indexed: 01/10/2023] Open
Abstract
Neurotensin (NT) is a tridecapeptide that is found in the central nervous system (CNS) and the gastrointestinal tract. NT behaves as a neurotransmitter in the brain and as a hormone in the gut. Additionally, NT acts as a neuromodulator to several neurotransmitter systems including dopaminergic, sertonergic, GABAergic, glutamatergic, and cholinergic systems. Due to its association with such a wide variety of neurotransmitters, NT has been implicated in the pathophysiology of several CNS disorders such as schizophrenia, drug abuse, Parkinson's disease (PD), pain, central control of blood pressure, eating disorders, as well as, cancer and inflammation. The present review will focus on the role that NT and its analogs play in schizophrenia, endocrine function, pain, psychostimulant abuse, and PD.
Collapse
Affiliation(s)
- Mona Boules
- Neuropsychopharmacology Laboratory, Department of Neuroscience, Mayo Clinic FloridaJacksonville, FL, USA
- *Correspondence: Mona Boules, Neuropsychopharmacology Laboratory, Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA. e-mail:
| | - Zhimin Li
- Neuropsychopharmacology Laboratory, Department of Neuroscience, Mayo Clinic FloridaJacksonville, FL, USA
| | - Kristin Smith
- Neuropsychopharmacology Laboratory, Department of Neuroscience, Mayo Clinic FloridaJacksonville, FL, USA
| | - Paul Fredrickson
- Neuropsychopharmacology Laboratory, Department of Neuroscience, Mayo Clinic FloridaJacksonville, FL, USA
| | - Elliott Richelson
- Neuropsychopharmacology Laboratory, Department of Neuroscience, Mayo Clinic FloridaJacksonville, FL, USA
| |
Collapse
|
16
|
Reduction of Pain Behavior by Stimulation of the Basolateral Amygdalar Nuclei in Freely Moving Rats. NEUROPHYSIOLOGY+ 2012. [DOI: 10.1007/s11062-012-9310-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
17
|
Abstract
The behavioral response to pain is driven by sensory and affective components, each of which is mediated by the CNS. Subjective pain ratings are used as readouts when appraising potential analgesics; however, pain ratings alone cannot enable a characterization of CNS pain circuitry during pain processing or how this circuitry is modulated pharmacologically. Having a more objective readout of potential analgesic effects may allow improved understanding and detection of pharmacological efficacy for pain. The pharmacological/functional magnetic resonance imaging (phMRI/fMRI) methodology can be used to objectively evaluate drug action on the CNS. In this context, we aimed to evaluate two drugs that had been developed as analgesics: one that is efficacious for pain (buprenorphine (BUP)) and one that failed as an analgesic in clinical trials aprepitant (APREP). Using phMRI, we observed that activation induced solely by BUP was present in regions with μ-opioid receptors, whereas APREP-induced activation was seen in regions expressing NK(1) receptors. However, significant pharmacological modulation of functional connectivity in pain-processing pathways was only observed following BUP administration. By implementing an evoked pain fMRI paradigm, these drugs could also be differentiated by comparing the respective fMRI signals in CNS circuits mediating sensory and affective components of pain. We report a correlation of functional connectivity and evoked pain fMRI measures with pain ratings as well as peak drug concentration. This investigation demonstrates how CNS-acting drugs can be compared, and how the phMRI/fMRI methodology may be used with conventional measures to better evaluate candidate analgesics in small subject cohorts.
Collapse
|
18
|
Creech SK, Smith J, Grimes JS, Meagher MW. Written Emotional Disclosure of Trauma and Trauma History Alter Pain Sensitivity. THE JOURNAL OF PAIN 2011; 12:801-10. [DOI: 10.1016/j.jpain.2011.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 01/13/2011] [Accepted: 01/19/2011] [Indexed: 10/18/2022]
|
19
|
McNally GP, Johansen JP, Blair HT. Placing prediction into the fear circuit. Trends Neurosci 2011; 34:283-92. [PMID: 21549434 DOI: 10.1016/j.tins.2011.03.005] [Citation(s) in RCA: 205] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 03/28/2011] [Accepted: 03/29/2011] [Indexed: 10/18/2022]
Abstract
Pavlovian fear conditioning depends on synaptic plasticity at amygdala neurons. Here, we review recent electrophysiological, molecular and behavioral evidence suggesting the existence of a distributed neural circuitry regulating amygdala synaptic plasticity during fear learning. This circuitry, which involves projections from the midbrain periaqueductal gray region, can be linked to prediction error and expectation modulation of fear learning, as described by associative and computational learning models. It controls whether, and how much, fear learning occurs by signaling aversive events when they are unexpected. Functional neuroimaging and clinical studies indicate that this prediction circuit is recruited in humans during fear learning and contributes to exposure-based treatments for clinical anxiety. This aversive prediction error circuit might represent a conserved mechanism for regulating fear learning in mammals.
Collapse
Affiliation(s)
- Gavan P McNally
- School of Psychology, The University of New South Wales, Sydney, NSW, Australia.
| | | | | |
Collapse
|
20
|
Roussy G, Beaudry H, Lafrance M, Belleville K, Beaudet N, Wada K, Gendron L, Sarret P. Altered morphine-induced analgesia in neurotensin type 1 receptor null mice. Neuroscience 2010; 170:1286-94. [PMID: 20727387 DOI: 10.1016/j.neuroscience.2010.08.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 08/02/2010] [Accepted: 08/10/2010] [Indexed: 01/09/2023]
Abstract
Both neurotensin (NT) and opioid agonists have been shown to induce antinociception in rodents after central administration. Besides, previous studies have revealed the existence of functional interactions between NT and opioid systems in the regulation of pain processing. We recently demonstrated that NTS1 receptors play a key role in the mediation of the analgesic effects of NT in long-lasting pain. In the present study, we therefore investigated whether NTS1 gene deletion affected the antinociceptive action of mu opioid drugs. To this end, pain behavioral responses to formalin were determined following systemic administration of morphine in both male and female NTS1 knockout mice. Acute injection of morphine (2 or 5 mg/kg) produced strong antinociceptive effects in both male and female wild-type littermates, with no significant sex differences. On the other hand, morphine analgesia was considerably reduced in NTS1-deficient mice of both sexes compared to their respective controls, indicating that the NTS1 receptor actively participates in mu opioid alleviating pain. By examining specifically the flinching, licking and biting nociceptive behaviors, we also showed that the functional crosstalk between NTS1 and mu opioid receptors influences the supraspinally-mediated behaviors. Interestingly, sexual dimorphic action of morphine-induced pain inhibition was found in NTS1 null mice in the formalin test, suggesting that the endogenous NT system interacts differently with the opioid network in male and female mice. Altogether, these results demonstrated that NTS1 receptor activation operates downstream to the opioidergic transmission and that NTS1-selective agonists combined with morphine may act synergistically to reduce persistent pain.
Collapse
Affiliation(s)
- G Roussy
- Department of Physiology and Biophysics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Jiménez-Velázquez G, López-Muñoz FJ, Fernández-Guasti A. Parallel anxiolytic-like and antinociceptive actions of diazepam in the anterior basolateral amygdala and dorsal periaqueductal gray. Brain Res 2010; 1349:11-20. [DOI: 10.1016/j.brainres.2010.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 06/04/2010] [Accepted: 06/05/2010] [Indexed: 01/10/2023]
|
22
|
Pozza DH, Potes CS, Barroso PA, Azevedo L, Castro-Lopes JM, Neto FL. Nociceptive behaviour upon modulation of mu-opioid receptors in the ventrobasal complex of the thalamus of rats. Pain 2010; 148:492-502. [PMID: 20106595 DOI: 10.1016/j.pain.2009.12.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Revised: 11/18/2009] [Accepted: 12/18/2009] [Indexed: 12/22/2022]
Abstract
The role of mu-opioid receptors (MORs) in the inflammatory pain processing mechanisms within the ventrobasal complex of the thalamus (VB) is not well understood. This study investigated the effect of modulating MOR activity upon nociception, by stereotaxically injecting specific ligands in the VB. Nociceptive behaviour was evaluated in two established animal models of inflammatory pain, by using the formalin (acute and tonic pain) and the ankle-bend (chronic monoarthritic pain) tests. Control (saline intra-VB injection) formalin-injected rats showed acute and tonic pain-related behaviours. In contrast, intrathalamic administration of [D-Ala(2), N-Me-Phe(4), Gly(5)-ol]-enkephalin acetate (DAMGO), a MOR-specific agonist, induced a statistically significant decrease of all tonic phase pain-related behaviours assessed until 30-35min after formalin hind paw injection. In the acute phase only the number of paw-jerks was affected. In monoarthritic rats, there was a noticeable antinociceptive effect with approximately 40min of duration, as denoted by the reduced ankle-bend scores observed after DAMGO injection. Intra-VB injection of D-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH(2) (CTOP), a specific MOR antagonist, or of CTOP followed, 10min after, by DAMGO had no effects in either formalin or ankle-bend tests. Data show that DAMGO-induced MOR activation in the VB has an antinociceptive effect in the formalin test as well as in chronic pain observed in MA rats, suggesting an important and specific role for MORs in the VB processing of inflammatory pain.
Collapse
Affiliation(s)
- Daniel Humberto Pozza
- Instituto de Histologia e Embriologia, Faculdade de Medicina, Universidade do Porto, Portugal IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Portugal Serviço de Bioestatística e Informática Médica, Faculdade de Medicina, Universidade do Porto, Portugal Centro de Investigação em Tecnologias e Sistemas de Informação em Saúde - CINTESIS, Universidade do Porto, Portugal
| | | | | | | | | | | |
Collapse
|
23
|
Gustin SM, Wrigley PJ, Siddall PJ, Henderson LA. Brain anatomy changes associated with persistent neuropathic pain following spinal cord injury. Cereb Cortex 2009; 20:1409-19. [PMID: 19815621 DOI: 10.1093/cercor/bhp205] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Persistent neuropathic pain commonly occurs following spinal cord injury (SCI). It remains one of the most challenging management problems in this condition. In order to develop more effective treatments, a better understanding of the neural changes associated with neuropathic SCI pain is required. The aim of this investigation was to use diffusion tensor imaging (DTI) to determine if persistent neuropathic pain following SCI is associated with changes in regional brain anatomy and connectivity. In 23 subjects with complete thoracic SCI, 12 with below-level neuropathic pain and 11 without pain, and 45 healthy control subjects, a series of whole-brain DTI scans were performed. The mean diffusivity (MD) of each voxel was calculated and values compared between groups. This analysis revealed that neuropathic pain following SCI is associated with significant differences in regional brain anatomy. These anatomical changes were located in pain-related regions as well as regions of the classic reward circuitry, that is, the nucleus accumbens and orbitofrontal, dorsolateral prefrontal, and posterior parietal cortices. The right posterior parietal cortex projected to most regions that displayed an anatomical change. Analysis of the fiber tracts connecting areas of MD differences revealed no significance differences in MD values between the SCI pain, SCI no pain, and control groups.
Collapse
Affiliation(s)
- S M Gustin
- Pain Management Research Institute, Kolling Institute, University of Sydney, Royal North Shore Hospital, St. Leonards, Sydney, NSW 2065, Australia
| | | | | | | |
Collapse
|
24
|
Boules M, Shaw A, Liang Y, Barbut D, Richelson E. NT69L, a novel analgesic, shows synergy with morphine. Brain Res 2009; 1294:22-8. [DOI: 10.1016/j.brainres.2009.07.086] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 07/20/2009] [Accepted: 07/25/2009] [Indexed: 10/20/2022]
|
25
|
DeSantana JM, Da Silva LFS, De Resende MA, Sluka KA. Transcutaneous electrical nerve stimulation at both high and low frequencies activates ventrolateral periaqueductal grey to decrease mechanical hyperalgesia in arthritic rats. Neuroscience 2009; 163:1233-41. [PMID: 19576962 DOI: 10.1016/j.neuroscience.2009.06.056] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Revised: 06/24/2009] [Accepted: 06/27/2009] [Indexed: 10/20/2022]
Abstract
Transcutaneous electric nerve stimulation (TENS) is widely used for the treatment of pain. TENS produces an opioid-mediated antinociception that utilizes the rostroventromedial medulla (RVM). Similarly, antinociception evoked from the periaqueductal grey (PAG) is opioid-mediated and includes a relay in the RVM. Therefore, we investigated whether the ventrolateral or dorsolateral PAG mediates antinociception produced by TENS in rats. Paw and knee joint mechanical withdrawal thresholds were assessed before and after knee joint inflammation (3% kaolin/carrageenan), and after TENS stimulation (active or sham). Cobalt chloride (CoCl(2); 5 mM) or vehicle was microinjected into the ventrolateral periaqueductal grey (vlPAG) or dorsolateral periaqueductal grey (dlPAG) prior to treatment with TENS. Either high (100 Hz) or low (4 Hz) frequency TENS was then applied to the inflamed knee for 20 min. Active TENS significantly increased withdrawal thresholds of the paw and knee joint in the group microinjected with vehicle when compared to thresholds prior to TENS (P<0.001) or to sham TENS (P<0.001). The increases in withdrawal thresholds normally observed after TENS were prevented by microinjection of CoCl(2) into the vlPAG, but not the dlPAG prior to TENS and were significantly lower than controls treated with TENS (P<0.001). In a separate group of animals, microinjection of CoCl(2) into the vlPAG temporarily reversed the decreased mechanical withdrawal threshold suggesting a role for the vlPAG in the facilitation of joint pain. No significant difference was observed for dlPAG. We hypothesize that the effects of TENS are mediated through the vlPAG that sends projections through the RVM to the spinal cord to produce an opioid-mediated analgesia.
Collapse
Affiliation(s)
- J M DeSantana
- Department of Physical Therapy, Federal University of Sergipe, Cidade Universitária Professor José Aloísio de Campos. Av. Marechal Rondon s/n, Jardim Rosa Else, São Cristóvão/Sergipe, Brazil.
| | | | | | | |
Collapse
|
26
|
Decreased gray matter volumes in the cingulo-frontal cortex and the amygdala in patients with fibromyalgia. Psychosom Med 2009; 71:566-73. [PMID: 19414621 DOI: 10.1097/psy.0b013e3181a32da0] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Studies in fibromyalgia syndrome with functional neuroimaging support the hypothesis of central pain augmentation. To determine whether structural changes in areas of the pain system are additional preconditions for the central sensitization in fibromyalgia we performed voxel based morphometry in patients with fibromyalgia and healthy controls. METHODS We performed 3 Tesla magnetic resonance imaging of the brain in 14 patients with fibromyalgia and 14 healthy controls. Regional differences of the segmented and normalized gray matter volumes in brain areas of the pain system between both groups were determined. In those areas in which patients structurally differed from healthy controls, the correlation of disease-related factors with gray matter volumes was analyzed. RESULTS Patients presented a decrease in gray matter volume in the prefrontal cortex, the amygdala, and the anterior cingulate cortex (ACC). The duration of pain or functional pain disability did not correlate with gray matter volumes. A trend of inverse correlation of gray matter volume reduction in the ACC with the duration of pain medication intake has been detected. CONCLUSIONS Our results suggest that structural changes in the pain system are associated with fibromyalgia. As disease factors do not correlate with reduced gray matter volume in areas of the cingulo-frontal cortex and the amygdala in patients, one possible interpretation is that volume reductions might be a precondition for central sensitization in fibromyalgia.
Collapse
|
27
|
Mitchell VA, Kawahara H, Vaughan CW. Neurotensin inhibition of GABAergic transmission via mGluR-induced endocannabinoid signalling in rat periaqueductal grey. J Physiol 2009; 587:2511-20. [PMID: 19359367 DOI: 10.1113/jphysiol.2008.167429] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Neurotensin modulates pain via its actions within descending analgesic pathways which include brain regions such as the midbrain periaqueductal grey (PAG). The aim of this study was to examine the cellular actions of neurotensin on PAG neurons. Whole cell patch clamp recordings were made from rat midbrain PAG slices in vitro to examine the postsynaptic effects of neurotensin and its effects on GABA(A) mediated inhibitory postsynaptic currents (IPSCs). Neurotensin (100-300 nM) produced an inward current in subpopulations of opioid sensitive and insensitive PAG neurons which did not reverse over membrane potentials between -50 and -130 mV. The neurotensin induced current was abolished by the NTS1 and NTS1/2 antagonists SR48692 (300 nM) and SR142948A (300 nM). Neurotensin also produced a reduction in the amplitude of evoked IPSCs, but had no effect on the rate and amplitude of TTX-resistant miniature IPSCs. The neurotensin induced inhibition of evoked IPSCs was reduced by the mGluR5 antagonist MPEP (5microM) and abolished by the cannabinoid CB(1) receptor antagonist AM251 (3 microM). These results suggest that neurotensin produces direct neuronal depolarisation via NTS1 receptors and inhibits GABAergic synaptic transmission within the PAG. The inhibition of synaptic transmission is mediated by neuronal excitation and action potential dependent release of glutamate, leading to mGluR5 mediated production of endocannabinoids which activate presynaptic CB(1) receptors. Thus, neurotensin has cellular actions within the PAG which are consistent with both algesic and analgesic activity, some of which are mediated via the endocannabinoid system.
Collapse
Affiliation(s)
- V A Mitchell
- Pain Management Research Institute, Northern Clinical School, University of Sydney at Royal North Shore Hospital, St Leonards, NSW, Australia
| | | | | |
Collapse
|
28
|
Wilson MA, Junor L. The role of amygdalar mu-opioid receptors in anxiety-related responses in two rat models. Neuropsychopharmacology 2008; 33:2957-68. [PMID: 18216773 PMCID: PMC2705758 DOI: 10.1038/sj.npp.1301675] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Amygdala opioids such as enkephalin appear to play some role in the control of anxiety and the anxiolytic effects of benzodiazepines, although the opioid receptor subtypes mediating such effects are unclear. This study compared the influences of mu-opioid receptor (MOR) activation in the central nucleus of the amygdala (CEA) on unconditioned fear or anxiety-like responses in two models, the elevated plus maze, and the defensive burying test. The role of MORs in the anxiolytic actions of the benzodiazepine agonist diazepam was also examined using both models. Either the MOR agonist [D-Ala(2), NMe-Phe(4), Gly-ol(5)]-enkephalin (DAMGO), or the MOR antagonists Cys-Tyr-D-Trp-Arg-Thr-Pen-Thr-NH(2) (CTAP) or beta-funaltrexamine (FNA) were bilaterally infused into the CEA of rats before testing. The results show that microinjection of DAMGO in the CEA decreased open-arm time in the plus maze, whereas CTAP increased open-arm behaviors. In contrast, DAMGO injections in the CEA reduced burying behaviors and increased rearing following exposure to a predator odor, suggesting a shift in the behavioral response in this context. Amygdala injections of the MOR agonist DAMGO or the MOR antagonist CTAP failed to change the anxiolytic effects of diazepam in either test. Our results demonstrate that MOR activation in the central amygdala exerts distinctive effects in two different models of unconditioned fear or anxiety-like responses, and suggest that opioids may exert context-specific regulation of amygdalar output circuits and behavioral responses during exposure to potential threats (open arms of the maze) vs discrete threats (predator odor).
Collapse
|
29
|
Martin TJ, Ewan E. Chronic pain alters drug self-administration: implications for addiction and pain mechanisms. Exp Clin Psychopharmacol 2008; 16:357-66. [PMID: 18837632 PMCID: PMC3788584 DOI: 10.1037/a0013597] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
This review article focuses on the impact that the presence of pain has on drug self-administration in rodents, and the potential for using self-administration to study both addiction and pain, as well as their interaction. The literature on the effects of noxious input to the brain on both spinal and supraspinal neuronal activity is reviewed as well as the evidence that human and rodent neurobiology is affected similarly by noxious stimulation. The convergence of peripheral input to somatosensory systems with limbic forebrain structures is briefly discussed in the context of how the activity of one system may influence activity within the other system. Finally, the literature on how pain influences drug-seeking behaviors in rodents is reviewed, with a final discussion of how these techniques might be able to contribute to the development of novel analgesic treatments that minimize addiction and tolerance.
Collapse
Affiliation(s)
- Thomas J Martin
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | |
Collapse
|
30
|
Gonçalves L, Silva R, Pinto-Ribeiro F, Pêgo JM, Bessa JM, Pertovaara A, Sousa N, Almeida A. Neuropathic pain is associated with depressive behaviour and induces neuroplasticity in the amygdala of the rat. Exp Neurol 2008; 213:48-56. [DOI: 10.1016/j.expneurol.2008.04.043] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 04/24/2008] [Accepted: 04/28/2008] [Indexed: 01/22/2023]
|
31
|
Zhao ZQ. Neural mechanism underlying acupuncture analgesia. Prog Neurobiol 2008; 85:355-75. [PMID: 18582529 DOI: 10.1016/j.pneurobio.2008.05.004] [Citation(s) in RCA: 706] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 03/19/2008] [Accepted: 05/30/2008] [Indexed: 12/16/2022]
Abstract
Acupuncture has been accepted to effectively treat chronic pain by inserting needles into the specific "acupuncture points" (acupoints) on the patient's body. During the last decades, our understanding of how the brain processes acupuncture analgesia has undergone considerable development. Acupuncture analgesia is manifested only when the intricate feeling (soreness, numbness, heaviness and distension) of acupuncture in patients occurs following acupuncture manipulation. Manual acupuncture (MA) is the insertion of an acupuncture needle into acupoint followed by the twisting of the needle up and down by hand. In MA, all types of afferent fibers (Abeta, Adelta and C) are activated. In electrical acupuncture (EA), a stimulating current via the inserted needle is delivered to acupoints. Electrical current intense enough to excite Abeta- and part of Adelta-fibers can induce an analgesic effect. Acupuncture signals ascend mainly through the spinal ventrolateral funiculus to the brain. Many brain nuclei composing a complicated network are involved in processing acupuncture analgesia, including the nucleus raphe magnus (NRM), periaqueductal grey (PAG), locus coeruleus, arcuate nucleus (Arc), preoptic area, nucleus submedius, habenular nucleus, accumbens nucleus, caudate nucleus, septal area, amygdale, etc. Acupuncture analgesia is essentially a manifestation of integrative processes at different levels in the CNS between afferent impulses from pain regions and impulses from acupoints. In the last decade, profound studies on neural mechanisms underlying acupuncture analgesia predominately focus on cellular and molecular substrate and functional brain imaging and have developed rapidly. Diverse signal molecules contribute to mediating acupuncture analgesia, such as opioid peptides (mu-, delta- and kappa-receptors), glutamate (NMDA and AMPA/KA receptors), 5-hydroxytryptamine, and cholecystokinin octapeptide. Among these, the opioid peptides and their receptors in Arc-PAG-NRM-spinal dorsal horn pathway play a pivotal role in mediating acupuncture analgesia. The release of opioid peptides evoked by electroacupuncture is frequency-dependent. EA at 2 and 100Hz produces release of enkephalin and dynorphin in the spinal cord, respectively. CCK-8 antagonizes acupuncture analgesia. The individual differences of acupuncture analgesia are associated with inherited genetic factors and the density of CCK receptors. The brain regions associated with acupuncture analgesia identified in animal experiments were confirmed and further explored in the human brain by means of functional imaging. EA analgesia is likely associated with its counter-regulation to spinal glial activation. PTX-sesntive Gi/o protein- and MAP kinase-mediated signal pathways as well as the downstream events NF-kappaB, c-fos and c-jun play important roles in EA analgesia.
Collapse
Affiliation(s)
- Zhi-Qi Zhao
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| |
Collapse
|
32
|
Huge V, Rammes G, Beyer A, Zieglgänsberger W, Azad SC. Activation of kappa opioid receptors decreases synaptic transmission and inhibits long-term potentiation in the basolateral amygdala of the mouse. Eur J Pain 2008; 13:124-9. [PMID: 18439862 DOI: 10.1016/j.ejpain.2008.03.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Revised: 03/05/2008] [Accepted: 03/13/2008] [Indexed: 11/25/2022]
Abstract
BACKGROUND The amygdala plays an important role in the processing of chronic pain and pain memory formation. Particularly, it is involved in the emotional and affective components of the pain circuitry. The role of kappa opioid receptors in these pain conditions is only partly known. The present study investigates the effect of kappa receptor activation on synaptic transmission and synaptic plasticity in the amygdala. METHODS Electrophysiological in vitro experiments were carried out in brain slices of male C57BL/6JOlaHsd mice. The effect of the kappa opioid receptor agonist U50,488H (5 microM) and the selective kappa opioid receptor antagonist nor-BNI (3 microM) on field potential (FP) amplitude and the induction of long-term potentiation (LTP) in the basolateral amygdala (BLA) was examined. RESULTS High frequency stimulation (HFS) of afferents in the lateral amygdala with two trains of 100 pulses at 50 Hz increased the FP amplitudes to 119+/-2% (mean+/-SEM; n=6) in the BLA. U50,488H decreased synaptic transmission (baseline: 100+/-0.5%; U50,488H: 86.3+/-2.4%; n=6) and blocked the induction of LTP (U50,488H: 100+/-4.1%; HFS: 102.6+/-7%; n=6). The effect on synaptic transmission and on LTP was completely reversed or prevented by application of nor-BNI, which itself had no effect on synaptic transmission or the induction of LTP. CONCLUSION Kappa opioid receptor activation decreases synaptic transmission and inhibits the induction of LTP in the BLA of the mouse. These findings may be associated with the effects of kappa opioid agonists in chronic pain and pain memory formation.
Collapse
Affiliation(s)
- Volker Huge
- Clinical Neuropharmacology Group, Max-Planck-Institut für Psychiatrie, Kraepelinstrasse 2-10, 80804 Munich, Germany.
| | | | | | | | | |
Collapse
|
33
|
Seo DO, Pang MH, Shin MS, Kim HT, Choi JS. Hippocampal NMDA receptors are necessary for auditory trace fear conditioning measured with conditioned hypoalgesia in rats. Behav Brain Res 2008; 192:264-8. [PMID: 18514922 DOI: 10.1016/j.bbr.2008.04.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2007] [Revised: 03/31/2008] [Accepted: 04/09/2008] [Indexed: 11/18/2022]
Abstract
We tested whether N-methyl-D-aspartate (NMDA) receptors in the dorsal hippocampus (DH) are critical for the acquisition of trace fear conditioning using conditioned hypoalgesia (CHA), decrease in pain reactivity, as the conditioned response (CR) instead of commonly used freezing. Infusions of the NMDA receptor antagonist, DL-2-amino-5-phosphonovaleric acid (APV) into DH prior to conditioning resulted in impaired CHA, measured with the radiant heat tail flick test, only in the trace-conditioning group when they were tested during the trace interval. The same infusion had no effect on CHA in the delay-conditioned animals. The results support that NMDA receptors in DH are critically involved in associating the CS with the US across a temporal gap. In addition, temporal specificity of the CR was revealed as CHA was induced only in the temporal vicinity of the US used for the training.
Collapse
Affiliation(s)
- Dong-Oh Seo
- Department of Psychology, Korea University, 5-1 Anam-Dong, Seongbuk-Gu, Seoul, 136-701, Republic of Korea
| | | | | | | | | |
Collapse
|
34
|
D'Hanis W, Linke R, Yilmazer-Hanke DM. Topography of thalamic and parabrachial calcitonin gene-related peptide (CGRP) immunoreactive neurons projecting to subnuclei of the amygdala and extended amygdala. J Comp Neurol 2007; 505:268-91. [PMID: 17879271 DOI: 10.1002/cne.21495] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Injections of calcitonin gene-related peptide (CGRP) into the amygdala evoke fear-related behaviors and antinociceptive effects. In the present study we therefore characterized CGRP-containing amygdaloid afferents by injecting the retrograde tracer FluoroGold (FG) into subnuclei of the amygdala and adjacent divisions of the extended amygdala, namely, the lateral (LA) and central (CE) amygdaloid nuclei, interstitial nucleus of the posterior limb of the anterior commissure (IPAC), and the amygdalostriatal area (AStr). The distribution of retrogradely FG-labeled neurons and colocalization of CGRP-immunoreactivity with FG-labeling were mapped in the posterior paralaminar thalamic complex and parabrachial nuclei. The analysis of the posterior thalamus revealed that about 50% of CGRP-containing neurons projected to the AStr, the projections originating in the medial part of the medial geniculate body, posterior intralaminar nucleus, parvicellular subparafascicular nucleus, and peripeduncular nucleus. However, the percentage of CGRP-containing thalamic neurons projecting to the adjacent LA, medial part of the CE, and ventrocaudal part of the caudatoputamen rapidly dropped to 3-9%. There were no double-labeled cells after injections into the lateral and capsular parts of the CE and the IPAC. Thus, the AStr received the heaviest CGRP-containing projection from the posterior thalamus. CGRP-containing parabrachial neurons projected to the AStr and lateral, capsular, and medial parts of the CE, the projections originating in the external, crescent, and central parts of the lateral parabrachial nucleus and external part of the medial parabrachial nucleus. The results demonstrate a distinct projection pattern of CGRP-containing thalamic and parabrachial neurons to subnuclei of the amygdala and extended amygdala.
Collapse
Affiliation(s)
- W D'Hanis
- Institute of Anatomy, Medical Faculty, Otto-von-Guericke University, D-39120 Magdeburg, Germany
| | | | | |
Collapse
|
35
|
Hasanein P, Parviz M, Keshavarz M, Javanmardi K. CB1 RECEPTOR ACTIVATION IN THE BASOLATERAL AMYGDALA PRODUCES ANTINOCICEPTION IN ANIMAL MODELS OF ACUTE AND TONIC NOCICEPTION. Clin Exp Pharmacol Physiol 2007; 34:439-49. [PMID: 17439413 DOI: 10.1111/j.1440-1681.2007.04592.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
1. Recent studies have suggested that the basolateral nucleus of the amygdala (BLA) participates in the processing of pain information, especially noxious somatic information. Cannabinoid receptors or CB1 mRNA are expressed more in the BLA than in other nuclei of the amygdala. Thus, the aim of the present study was to examine whether CB1 receptors in the BLA may be involved in modulating acute and/or tonic nociceptive processing. 2. Adult rats were exposed to intra-BLA microinjection of the cannabinoid receptor agonist (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl) pyrrolo [1,2,3,-de]-1,4-benzoxazin-6-yl]-1-naphthalenylmethanone mesylate [WIN 55,212-2 (1, 2.5, 5 or 10 microg/side)] and subjected to the tail flick and formalin tests. 3. The rats demonstrated a dose-dependent increase in latency to withdraw from a thermal noxious stimulus in the tail flick test and a decrease in formalin-induced pain behaviours. The antinociceptive effects of the CB1 receptor agonist WIN 55,212-2 (10 microg/side) in both tests were attenuated in the presence of the selective CB1 receptor antagonist, N-(piperidin-1-yl)-5-(4-iodophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3- carboxamide (AM251; 0.55 ng/side). Administration of the CB1 receptor antagonist AM251 (0.55, 5.5, or 55.5 ng/side) alone did not alter the nociceptive thresholds in either test. Bilateral microinjection of the selective CB2 receptor antagonist N-[(1S)-endo-1,3,3-trimethyl bicyclo [2.2.1] heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide (SR144528; 1 microg/side) had no effect on the antinociception produced by WIN 55,212-2, suggesting that the antinociceptive actions of WIN 55,212-2 are mediated by CB1 receptors. 4. The findings suggest the existence of a CB1-mediated inhibitory system in the BLA that, when activated, can diminish responsivity to acute and tonic noxious stimuli, but that normally has no tonic effect on the response threshold of these stimuli.
Collapse
Affiliation(s)
- Parisa Hasanein
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran
| | | | | | | |
Collapse
|
36
|
Jiménez-Velázquez G, Fernández-Guasti A, López-Muñoz FJ. Influence of pharmacologically-induced experimental anxiety on nociception and antinociception in rats. Eur J Pharmacol 2006; 547:83-91. [PMID: 16952350 DOI: 10.1016/j.ejphar.2006.06.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2006] [Accepted: 06/20/2006] [Indexed: 02/06/2023]
Abstract
Animal studies reveal that diverse environmental stimuli that generate anxiety-like behaviors also induce antinociception; conversely, clinical data show that pain perception is reduced under anxiolysis. This study was conducted to investigate the influence of pharmacologically induced-anxiety on nociception and antinociception. Experimental anxiety levels were measured using the rat burying behavior test. Diazepam (0, 0.5, 1.0 and 2.0 mg/kg, i.p.) or yohimbine (0, 0.5 and 1.0 mg/kg, i.p.) were used as anxiolytic or anxiogenic drugs, respectively. To evaluate the influence of different experimental anxiety levels on nociception, the pain-induced functional impairment in the rat (PIFIR model) was used. Nociception was induced by an intra-articular injection of 15% uric acid into the knee joint of the right hind limb. Diazepam or yohimbine were administered 15 min before uric acid and the ability of the rat to use the injured hind limb was recorded. To analyze the influence of different levels of anxiety on the antinociceptive effects produced by acetylsalicylic acid (0, 31, 100 and 310 mg/kg, p.o.); this analgesic was administered 3.5 h after uric acid. Fifteen min before diazepam (2.0 mg/kg) or yohimbine (1.0 mg/kg) were administered. We found that, in the burying behavior test, diazepam and yohimbine produced a dose-dependent decrease or augment in the cumulative time of burying, effects denoting reduced or increased experimental anxiety, respectively. Diazepam or yohimbine, administered alone, was unable to produce nociception. The results showed an influence of anxiety on nociception since a decreased (by diazepam) or increased (by yohimbine) experimental anxiety prevented nociception. Control experiments showed that acetylsalicylic acid did not modify experimental anxiety in the burying behavior test, but effectively reversed the nociception induced by uric acid (15%) in the PIFIR model. Such antinociceptive effect was unmodified by the anxiolytic or anxiogenic actions of diazepam or yohimbine. Data are discussed on the bases of clinical- and animal-studies revealing interactions between anxiety and nociception.
Collapse
Affiliation(s)
- Guadalupe Jiménez-Velázquez
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados - Sede Sur, Calz. de los Tenorios 235, Col. Granjas Coapa, C.P. 14330, México D.F., Mexico
| | | | | |
Collapse
|
37
|
Padlubnaya DB, Parekh NH, Brown TH. Neurophysiological theory of Kamin blocking in fear conditioning. Behav Neurosci 2006; 120:337-52. [PMID: 16719698 DOI: 10.1037/0735-7044.120.2.337] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Kamin blocking in fear conditioning is thought to reflect diminished processing of the unconditional stimulus (US) in the presence of a conditional stimulus (CS-super(+)) that was previously paired with this US. According to Fanselow's (1998) hypothesis, the CS-super(+) drives output from the amygdala that ultimately produces analgesia by causing opiate release onto afferent pain circuits. This hypothesis was explored quantitatively through neurophysiological simulations. The results suggest that opiate-mediated, negative-feedback control of US processing is too slow for efficient blocking of cue conditioning. The reason is that conditioning-produced synaptic modifications can be induced before the opiate-mediated inhibition has any substantial effect on US processing. The results suggest the existence of an additional, faster-acting, inhibitory neurotransmitter in the blocking circuit.
Collapse
|
38
|
Neugebauer V. Chapter 11 Subcortical processing of nociceptive information: basal ganglia and amygdala. HANDBOOK OF CLINICAL NEUROLOGY 2006; 81:141-58. [PMID: 18808833 DOI: 10.1016/s0072-9752(06)80015-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Finnegan TF, Chen SR, Pan HL. Mu opioid receptor activation inhibits GABAergic inputs to basolateral amygdala neurons through Kv1.1/1.2 channels. J Neurophysiol 2005; 95:2032-41. [PMID: 16306173 DOI: 10.1152/jn.01004.2005] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The basolateral amygdala (BLA) is the major amygdaloid nucleus distributed with mu opioid receptors. The afferent input from the BLA to the central nucleus of the amygdala (CeA) is considered important for opioid analgesia. However, little is known about the effect of mu opioids on synaptic transmission in the BLA. In this study, we examined the effect of mu opioid receptor stimulation on the inhibitory and excitatory synaptic inputs to CeA-projecting BLA neurons. BLA neurons were retrogradely labeled with a fluorescent tracer injected into the CeA of rats. Whole cell voltage-clamp recordings were performed on labeled BLA neurons in brain slices. The specific mu opioid receptor agonist, (D-Ala2,N-Me-Phe4,Gly5-ol)-enkephalin (DAMGO, 1 microM), significantly reduced the frequency of miniature inhibitory postsynaptic currents (mIPSCs) in 77% of cells tested. DAMGO also significantly decreased the peak amplitude of evoked IPSCs in 75% of cells examined. However, DAMGO did not significantly alter the frequency of mEPSCs or the peak amplitude of evoked EPSCs in 90% and 75% of labeled cells, respectively. Bath application of the Kv channel blockers, 4-AP (Kv1.1, 1.2, 1.3, 1.5, 1.6, 3.1, 3.2), alpha-dendrotoxin (Kv1.1, 1.2, 1.6), dendrotoxin-K (Kv1.1), or tityustoxin-Kalpha (Kv1.2) each blocked the inhibitory effect of DAMGO on mIPSCs. Double immunofluorescence labeling showed that some of the immunoreactivities of Kv1.1 and Kv1.2 were colocalized with synaptophysin in the BLA. This study provides new information that activation of presynaptic mu opioid receptors primarily attenuates GABAergic synaptic inputs to CeA-projecting neurons in the BLA through a signaling mechanism involving Kv1.1 and Kv1.2 channels.
Collapse
MESH Headings
- Amygdala/chemistry
- Amygdala/physiology
- Analgesics, Opioid/pharmacology
- Animals
- Bicuculline/pharmacology
- Elapid Venoms/pharmacology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Excitatory Postsynaptic Potentials/physiology
- Kv1.1 Potassium Channel/analysis
- Kv1.1 Potassium Channel/drug effects
- Kv1.1 Potassium Channel/physiology
- Kv1.2 Potassium Channel/analysis
- Kv1.2 Potassium Channel/drug effects
- Kv1.2 Potassium Channel/physiology
- Limbic System/physiology
- Male
- Neurons, Afferent/drug effects
- Neurons, Afferent/physiology
- Patch-Clamp Techniques
- Peptides/pharmacology
- Potassium Channel Blockers/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid/physiology
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/physiology
- Scorpion Venoms/pharmacology
- Signal Transduction/physiology
- Synaptic Transmission/drug effects
- Synaptic Transmission/physiology
- Synaptophysin/analysis
- gamma-Aminobutyric Acid/physiology
- Nociceptin Receptor
Collapse
Affiliation(s)
- Thomas F Finnegan
- Department of Anesthesiology , Pennsylvania State University College of Medicine, The Milton S. Hershey Medical Center, Hershey Pennsylvania, USA
| | | | | |
Collapse
|
40
|
Wang R, Boules M, Gollatz E, Williams K, Tiner W, Richelson E. Effects of 5 daily injections of the neurotensin-mimetic NT69L on the expression of neurotensin receptors in rat brain. ACTA ACUST UNITED AC 2005; 138:24-34. [PMID: 15878217 DOI: 10.1016/j.molbrainres.2005.03.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2004] [Revised: 03/11/2005] [Accepted: 03/27/2005] [Indexed: 11/25/2022]
Abstract
The effects of one or five daily intraperitoneal injections of a neurotensin (NT) receptor agonist NT69L (2 mg/kg, i.p.) on the expression of NT (NTS), dopamine 1 and 2 receptors, tyrosine hydroxylase, and DOPA decarboxylase using immunohistochemical and real-time PCR were investigated in rats. Except for the striatum, acute injection of NT69L did not affect neurotensin receptors as compared to saline control. However, 5 daily injections of NT69L resulted in down-regulation of both NTS-1 protein and mRNA levels in several brain regions with the striatum showing a dramatic decrease in NTS-1 expression (P<0.05). The down-regulation of NTS-1 in the striatum, hypothalamus, and substania nigra (SN) after 5 daily injections was confirmed by autoradiography. Acute injection of NT69L increased NTS-2 mRNA and protein level in prefrontal cortex (PFC). NTS-3 mRNA expression and protein levels were slightly down-regulated in hypothalamus, periaqueductal gray (PAG), and SN, though the difference was not significant. The results indicated a difference in the profile of NT receptors expression in response to NT69L. Tyrosine hydroxylase (TH) and DOPA decarboxylase (DDC) mRNA was significantly down-regulated in striatum but not in SN. Interestingly, Nurr 1, a transcriptional activator of TH, was dramatically up-regulated in striatum, but down-regulated in PFC, suggesting that different modulating mechanisms may participate in NT69L tolerance in different regions. The present results suggest that distinct NT receptors involved in the effects exerted by NT69L may contribute to the interactions of NT69L with both neural networks and cellular proteins.
Collapse
Affiliation(s)
- Rui Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200031, PR China
| | | | | | | | | | | |
Collapse
|
41
|
Shin MS, Helmstetter FJ. Antinociception following application of DAMGO to the basolateral amygdala results from a direct interaction of DAMGO with Mu opioid receptors in the amygdala. Brain Res 2005; 1064:56-65. [PMID: 16289487 DOI: 10.1016/j.brainres.2005.09.065] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2005] [Revised: 09/29/2005] [Accepted: 09/30/2005] [Indexed: 10/25/2022]
Abstract
Previous studies from our laboratory have shown that application of the mu opioid agonist DAMGO into the basolateral region of the amygdala (BLA) suppresses the radiant heat tail flick (TF) reflex in anesthetized rats. This antinociceptive effect can be blocked by lesions of brainstem regions such as the periaqueductal gray (PAG) or the rostral ventromedial medulla (RVM) or by functional inactivation of neurons in these regions, suggesting the activation of brainstem-descending antinociceptive systems from the amygdala. However, little is known about the direct interaction of DAMGO with mu receptors in the amygdala. In the present series of experiments, the BLA was pretreated with opioid receptor antagonists and a G protein inhibitor prior to TF testing with application of DAMGO into the same site. Rats pretreated with the non-selective opioid antagonist naltrexone (1.25-3.75 microg/0.25 microl per side) or the G protein inhibitor pertussis toxin (0.25 microg) failed to show inhibition of TF reflexes following infusion of DAMGO (0.168-0.50 microg), indicating that DAMGO works through G-protein-coupled opioid receptors in the BLA. Furthermore, pretreatment with the mu antagonist beta-FNA (1.00-2.00 microg) attenuated antinociception induced by DAMGO injection, suggesting DAMGO's action on mu receptors in the BLA. Accordingly, we confirm a direct interaction of DAMGO with G-protein-coupled mu receptors in the BLA contributing to induction of opioid antinociception in the amygdala.
Collapse
MESH Headings
- Amygdala/cytology
- Amygdala/drug effects
- Amygdala/metabolism
- Analgesics, Opioid/administration & dosage
- Analgesics, Opioid/metabolism
- Anesthetics, Local/administration & dosage
- Anesthetics, Local/metabolism
- Animals
- Dose-Response Relationship, Drug
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/administration & dosage
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism
- Male
- Microinjections
- Neurons/drug effects
- Neurons/metabolism
- Pain/metabolism
- Pain Threshold/drug effects
- Pain Threshold/physiology
- Rats
- Rats, Long-Evans
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Maeng-Sik Shin
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53201, USA.
| | | |
Collapse
|
42
|
Shin MS. Vasoactive intestinal peptide in the amygdala inhibits tail flick reflexes in rats. Brain Res 2005; 1040:197-201. [PMID: 15804442 DOI: 10.1016/j.brainres.2005.01.071] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2004] [Revised: 01/24/2005] [Accepted: 01/24/2005] [Indexed: 11/25/2022]
Abstract
The present study was conducted to test the capability of a representative type of non-opioid peptides vasoactive intestinal peptide (VIP) in the amygdala to modulate nociception. Bilateral application of VIP into the basolateral region of the amygdala persistently suppressed radiant heat-evoked tail flick reflexes of anesthetized rats. The present result suggests that VIP synapses in the amygdala may play important roles in controlling pain, as with opioid synapses in the amygdala. This result also implies that local VIP in the amygdala is likely to subserve activating the descending antinociceptive systems of the brainstem from the amygdala.
Collapse
Affiliation(s)
- Maeng-Sik Shin
- Department of Psychology, University of Wisconsin-Milwaukee, P.O. Box 413, Milwaukee, WI 53201, USA.
| |
Collapse
|
43
|
Gao YJ, Ren WH, Zhang YQ, Zhao ZQ. Contributions of the anterior cingulate cortex and amygdala to pain- and fear-conditioned place avoidance in rats. Pain 2004; 110:343-53. [PMID: 15275785 DOI: 10.1016/j.pain.2004.04.030] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2003] [Revised: 03/09/2004] [Accepted: 04/12/2004] [Indexed: 12/11/2022]
Abstract
The pain experience includes a sensory-discriminative and an affective-emotional component. The sensory component of pain has been extensively studied, while data about the negative affective component of pain are quite limited. The anterior cingulate cortex (ACC), and amygdala are thought to be key neural substrates underlying emotional responses. Using formalin-induced conditioned place avoidance (F-CPA) and electric foot-shock conditioned place avoidance (S-CPA) models, the present study observed the effects of bilateral excitotoxic (quinolinic acid 200 nmol/microl) lesions of the ACC and amygdala on pain and fear induced negative emotion, as well as on sensory component of pain. In the place-conditioning paradigm, both intraplantar (i.pl.) injection of formalin and electric foot-shock produced conditioned place avoidance. Excitotoxin-induced lesion of either the ACC or amygdala significantly reduced the magnitude of F-CPA. However, the decrease in the magnitude of S-CPA occurred only in the amygdala, but not ACC lesioned animals. Neither ACC nor amygdala lesion significantly changed formalin-induced acute nociceptive behaviors. These results suggest that the amygdala is involved in both pain- and fear-related negative emotion, and the ACC might play a critical role in the expression of pain-related negative emotion.
Collapse
Affiliation(s)
- Yong-Jing Gao
- Institute of Neurobiology, Fudan University, 220 Han Dan Road, Shanghai, 200433, China
| | | | | | | |
Collapse
|
44
|
Finnegan TF, Chen SR, Pan HL. Effect of the μ Opioid on Excitatory and Inhibitory Synaptic Inputs to Periaqueductal Gray-Projecting Neurons in the Amygdala. J Pharmacol Exp Ther 2004; 312:441-8. [PMID: 15388784 DOI: 10.1124/jpet.104.074633] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Opioids are potent analgesics, but the sites of their action and cellular mechanisms are not fully understood. The central nucleus of the amygdala (CeA) is important for opioid analgesia through the projection to the periaquaductal gray (PAG). In this study, we examined the effects of mu opioid receptor stimulation on inhibitory and excitatory synaptic inputs to PAG-projecting CeA neurons retrogradely labeled with a fluorescent tracer injected into the ventrolateral PAG of rats. Whole-cell voltage-clamp recordings were performed on labeled CeA neurons in brain slices. The specific mu opioid receptor agonist, [d-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO, 1 microM), significantly reduced the frequency of miniature inhibitory postsynaptic currents (mIPSCs) without altering the amplitude and decay constant of mIPSCs in 47.6% (10 of 21) of cells tested. DAMGO also significantly decreased the peak amplitude of evoked IPSCs in 69% (9 of 13) of cells examined. However, DAMGO did not significantly alter the frequency of miniature excitatory postsynaptic currents (EPSCs) and the amplitude of evoked EPSCs in 69% (9 of 13) and 83% (10 of 12) of labeled cells, respectively. The IPSCs were blocked by the GABA(A) receptor antagonist bicuculline, whereas the EPSCs were largely abolished by the non-N-methyl-d-aspartate antagonist 6-cyano-7-nitroquinoxaline-2,3-dione. The immunoreactivity of mu opioid receptors was colocalized with synaptophysin, a presynaptic marker, in close appositions to labeled CeA neurons. These results suggest that activation of mu opioid receptors on presynaptic terminals primarily attenuates GABAergic synaptic inputs to PAG-projecting neurons in the CeA.
Collapse
Affiliation(s)
- Thomas F Finnegan
- Department of Anesthesiology, H187, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | |
Collapse
|
45
|
Affiliation(s)
- Howard Fields
- University of California, 513 Parnassus Avenue, San Francisco, California 94143, USA.
| |
Collapse
|
46
|
Abstract
A reciprocal relationship exists between persistent pain and negative affective states such as fear, anxiety, and depression. Accumulating evidence points to the amygdala as an important site of such interaction. Whereas a key role of the amygdala in the neuronal mechanisms of emotionality and affective disorders has been well established, the concept of the amygdala as an important contributor to pain and its emotional component is still emerging. This article will review and discuss evidence from anatomical, neuroimaging, behavioral, electrophysiological, pharmacological, and biochemical data that implicate the amygdala in pain modulation and emotional responses to pain. The latero-capsular division of the central nucleus of the amygdala is now defined as the "nociceptive amygdala" and integrates nociceptive information with poly-modal information about the internal and external bodily environment. Dependent on environmental conditions and affective states, the amygdala appears to play a dual facilitatory and inhibitory role in the modulation of pain behavior and nociceptive processing at different levels of the pain neuraxis. Only recently, electrophysiological, pharmacological, and biochemical neuroplastic changes were shown in the nociceptive amygdala in persistent pain. It is conceivable, however, that amygdala plasticity plays an important role in emotional pain behavior and its modulation by affective state.
Collapse
Affiliation(s)
- Volker Neugebauer
- Department of Anatomy & Neurosciences, University of Texas Medical Branch, Galveston, 77555-1069, USA.
| | | | | | | |
Collapse
|
47
|
Faber ESL, Sah P. Opioids inhibit lateral amygdala pyramidal neurons by enhancing a dendritic potassium current. J Neurosci 2004; 24:3031-9. [PMID: 15044542 PMCID: PMC6729839 DOI: 10.1523/jneurosci.4496-03.2004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pyramidal neurons in the lateral amygdala discharge trains of action potentials that show marked spike frequency adaptation, which is primarily mediated by activation of a slow calcium-activated potassium current. We show here that these neurons also express an alpha-dendrotoxin- and tityustoxin-Kalpha-sensitive voltage-dependent potassium current that plays a key role in the control of spike discharge frequency. This current is selectively targeted to the primary apical dendrite of these neurons. Activation of micro-opioid receptors by application of morphine or d-Ala(2)-N-Me-Phe(4)-Glycol(5)-enkephalin (DAMGO) potentiates spike frequency adaptation by enhancing the alpha-dendrotoxin-sensitive potassium current. The effects of micro-opioid agonists on spike frequency adaptation were blocked by inhibiting G-proteins with N-ethylmaleimide (NEM) and by blocking phospholipase A(2). Application of arachidonic acid mimicked the actions of DAMGO or morphine. These results show that micro-opioid receptor activation enhances spike frequency adaptation in lateral amygdala neurons by modulating a voltage-dependent potassium channel containing Kv1.2 subunits, through activation of the phospholipase A(2)-arachidonic acid-lipoxygenases cascade.
Collapse
Affiliation(s)
- E S Louise Faber
- Division of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra ACT 2601, Australia
| | | |
Collapse
|
48
|
McGaraughty S, Farr DA, Heinricher MM. Lesions of the periaqueductal gray disrupt input to the rostral ventromedial medulla following microinjections of morphine into the medial or basolateral nuclei of the amygdala. Brain Res 2004; 1009:223-7. [PMID: 15120601 DOI: 10.1016/j.brainres.2004.02.048] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2004] [Indexed: 10/26/2022]
Abstract
Microinjections of morphine into the basolateral (BLa) and medial (MEa) nuclei of the amygdala differentially affect rostral ventromedial medulla (RVM) neuronal activity and nocifensive behaviors. PAG lesions attenuated or blocked the effects of both BLa and MEa morphine on RVM cell activity, and interfered with the behavioral antinociception produced by BLa infusions. These results demonstrate that the influences from both the BLa and MEa to the RVM are relayed via the PAG.
Collapse
Affiliation(s)
- Steve McGaraughty
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR 97201, USA.
| | | | | |
Collapse
|
49
|
Gui X, Carraway RE, Dobner PR. Endogenous neurotensin facilitates visceral nociception and is required for stress-induced antinociception in mice and rats. Neuroscience 2004; 126:1023-32. [PMID: 15207335 DOI: 10.1016/j.neuroscience.2004.04.034] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2004] [Indexed: 10/26/2022]
Abstract
Central neurotensin (NT) administration can both facilitate and inhibit somatic and visceral nociception, depending on the dose and administration site. NT microinjection in the rostroventral medulla facilitates nociception at low doses, while NT antagonist microinjection can markedly attenuate nociception, supporting the hypothesis that endogenous NT facilitates nociception. However, higher doses of NT produce a mu-opioid receptor-independent analgesia, similar to that resulting from various intense stressors. Furthermore, intense stress results in increased NT expression in several hypothalamic nuclei that have been implicated in stress-induced antinociception (SIAN); however, there is little direct evidence that endogenous NT is required for SIAN. We have investigated the role of endogenous NT in both basal visceral nociception and SIAN using both NT knockout mice and pharmacological approaches in rats. Visceral nociception was monitored by measuring visceromotor responses during colorectal distension both prior to and following water avoidance stress. Visceral nociception was significantly attenuated in both NT knockout mice and rats pre-treated with the NT antagonist SR 48692. Disruption of NT signaling also blocked SIAN, revealing a novel stress-induced hyperalgesic response that was significantly greater in female than in male rats. NT was also required for acetic acid-induced hyperalgesia. These results indicate that endogenous NT normally facilitates visceral pain responses, is required for irritant-induced hyperalgesia, and plays a critical role in SIAN.
Collapse
Affiliation(s)
- X Gui
- Department of Physiology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | | | | |
Collapse
|
50
|
Manning BH, Martin WJ, Meng ID. The rodent amygdala contributes to the production of cannabinoid-induced antinociception. Neuroscience 2003; 120:1157-70. [PMID: 12927220 DOI: 10.1016/s0306-4522(03)00356-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The amygdala is a temporal lobe region that is implicated in emotional information processing. The amygdala also is associated with the processing and modulation of pain sensation. Recently, we demonstrated that in nonhuman primates, the amygdala is necessary for the full expression of cannabinoid-induced antinociception [J Neurosci 21 (2001) 8238]. The antinociceptive effect of the cannabinoid receptor agonist (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo(1,2,3-de)-1,4-benzoxazin-6-yl]-1-naphthalenylmethanone (WIN55,212-2) was significantly reduced in rhesus monkeys with large bilateral lesions of the amygdaloid complex. In the present study, we investigated the contribution of the amygdala to cannabinoid-induced antinociception in the rat. Using bilateral local microinjections of the GABA(A) receptor agonist muscimol, we inactivated neurons originating from the central nucleus of the amygdala (CeA) or basolateral nucleus of the amygdala (BLA). In rats injected with intra-CeA saline, the cannabinoid receptor agonist WIN55,212-2 produced dose-dependent antinociception on the noxious heat-evoked tail flick assay. In rats treated with intra-CeA muscimol, however, the antinociceptive effect of WIN55,212-2 was significantly reduced. Rats treated with intra-BLA muscimol showed no deficit in WIN55,212-2-induced antinociception. The effect of CeA inactivation on WIN55,212-2-induced suppression of prolonged pain in the formalin test also was tested. In rats treated with intra-CeA saline, WIN55,212-2 reduced the incidence of formalin-induced nociceptive behaviors and also reduced formalin-evoked c-fos expression in both superficial and deep laminae of the spinal cord dorsal horn. In rats treated with intra-CeA muscimol, however, these effects of WIN55,212-2 were significantly reduced. The results constitute the first causal data demonstrating the necessity of descending pain-modulatory circuitry (of which the CeA is a component) for the full expression of cannabinoid-induced antinociception in the rat. Furthermore, the results complement previous findings suggesting an overlap in neural circuitry activated by opioids and cannabinoids.
Collapse
Affiliation(s)
- B H Manning
- Department of Neuroscience, Merck Research Laboratories, 770 Sumneytown Pike WP46-300, West Point, PA 19486-0004, USA.
| | | | | |
Collapse
|