1
|
Nelson TS, Allen HN, Khanna R. Neuropeptide Y and Pain: Insights from Brain Research. ACS Pharmacol Transl Sci 2024; 7:3718-3728. [PMID: 39698268 PMCID: PMC11651174 DOI: 10.1021/acsptsci.4c00333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 12/20/2024]
Abstract
Neuropeptide Y (NPY) is a highly conserved neuropeptide with widespread distribution in the central nervous system and diverse physiological functions. While extensively studied for its inhibitory effects on pain at the spinal cord level, its role in pain modulation within the brain remains less clear. This review aims to summarize the complex landscape of supraspinal NPY signaling in pain processing. We discuss the expression and function of NPY receptors in key pain-related brain regions, including the parabrachial nucleus, periaqueductal gray, amygdala, and nucleus accumbens. Additionally, we highlight the potent efficacy of NPY in attenuating pain sensitivity and nociceptive processing throughout the central nervous system. NPY-based therapeutic interventions targeting the central nervous system represent a promising avenue for novel analgesic strategies and pain-associated comorbidities.
Collapse
Affiliation(s)
- Tyler S. Nelson
- Department
of Pharmacology and Therapeutics, McKnight Brain Institute, College
of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Heather N. Allen
- Department
of Pharmacology and Therapeutics, McKnight Brain Institute, College
of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Rajesh Khanna
- Department
of Pharmacology and Therapeutics, McKnight Brain Institute, College
of Medicine, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
2
|
Bertagna NB, Holmgren EB, Engi SA, Ha L, Cruz FC, Albrechet-Souza L, Wills TA. BNST CRF receptor type 1 modulates mechanical hypersensitivity induced by adolescent alcohol exposure in adult female mice. Psychopharmacology (Berl) 2024; 241:2513-2523. [PMID: 39348004 DOI: 10.1007/s00213-024-06693-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024]
Abstract
RATIONALE Alcohol exposure during adolescence has been linked to long-lasting behavioral consequences, contributing to the development of alcohol use disorder. Negative affect and chronic pain during alcohol withdrawal are critical factors influencing problematic alcohol use and relapse. Our previous research demonstrated that adolescent intermittent ethanol (AIE) vapor exposure elicits sex-specific negative affect-like behavior in adult mice following stress exposure. Additionally, AIE induces persistent mechanical hypersensitivity, which is accompanied by increased activation of corticotropin-releasing factor receptor type 1 (CRFR1) neurons in the dorsolateral bed nucleus of the stria terminalis (dlBNST). OBJECTIVES This study extends previous work by investigating plasma corticosterone levels and CRFR1 protein expression in the dlBNST following restraint stress exposure in adult mice with an AIE history. We also aim to explore the role of dlBNST CRFR1 signaling in mediating negative affect-like behavior and mechanical hypersensitivity. RESULTS Female mice exhibited elevated plasma corticosterone levels compared to males following restraint stress. Moreover, females with AIE history showed higher expression of CRFR1 protein in the dlBNST compared to air controls. Antagonism of CRFR1 in the dlBNST blocked AIE-induced mechanical hypersensitivity in adult females but did not affect stress-induced negative affect-like behavior. In alcohol-naïve females, intra-dlBNST administration of a CRFR1 agonist induced mechanical hypersensitivity. CONCLUSIONS These findings provide new insights into the neurobiological mechanisms underlying stress-induced negative affect and pain-related behavior, both influenced by a history of adolescent alcohol exposure. The results suggest that CRFR1 antagonists warrant further investigation for their potential in addressing alcohol-related chronic pain.
Collapse
Affiliation(s)
- Natalia B Bertagna
- Department of Cell Biology & Anatomy, School of Medicine, Louisiana State University Health Sciences Center, 533 Bolivar Street, New Orleans, LA, 70112, USA
- Department of Pharmacology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Eleanor B Holmgren
- Department of Cell Biology & Anatomy, School of Medicine, Louisiana State University Health Sciences Center, 533 Bolivar Street, New Orleans, LA, 70112, USA
- Department of Pharmacology, School of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Sheila A Engi
- Department of Cell Biology & Anatomy, School of Medicine, Louisiana State University Health Sciences Center, 533 Bolivar Street, New Orleans, LA, 70112, USA
- Alcohol & Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Linh Ha
- Department of Cell Biology & Anatomy, School of Medicine, Louisiana State University Health Sciences Center, 533 Bolivar Street, New Orleans, LA, 70112, USA
| | - Fabio C Cruz
- Department of Pharmacology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Lucas Albrechet-Souza
- Department of Cell Biology & Anatomy, School of Medicine, Louisiana State University Health Sciences Center, 533 Bolivar Street, New Orleans, LA, 70112, USA.
- Alcohol & Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| | - Tiffany A Wills
- Department of Cell Biology & Anatomy, School of Medicine, Louisiana State University Health Sciences Center, 533 Bolivar Street, New Orleans, LA, 70112, USA
- Alcohol & Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
3
|
Uchida R, Mukai Y, Amano T, Sakimura K, Itoi K, Yamanaka A, Minami M. Chronic pain enhances excitability of corticotropin-releasing factor-expressing neurons in the oval part of the bed nucleus of the stria terminalis. Mol Brain 2024; 17:22. [PMID: 38702738 PMCID: PMC11071157 DOI: 10.1186/s13041-024-01094-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/25/2024] [Indexed: 05/06/2024] Open
Abstract
We previously reported that enhanced corticotropin-releasing factor (CRF) signaling in the bed nucleus of the stria terminalis (BNST) caused the aversive responses during acute pain and suppressed the brain reward system during chronic pain. However, it remains to be examined whether chronic pain alters the excitability of CRF neurons in the BNST. In this study we investigated the chronic pain-induced changes in excitability of CRF-expressing neurons in the oval part of the BNST (ovBNSTCRF neurons) by whole-cell patch-clamp electrophysiology. CRF-Cre; Ai14 mice were used to visualize CRF neurons by tdTomato. Electrophysiological recordings from brain slices prepared from a mouse model of neuropathic pain revealed that rheobase and firing threshold were significantly decreased in the chronic pain group compared with the sham-operated control group. Firing rate of the chronic pain group was higher than that of the control group. These data indicate that chronic pain elevated neuronal excitability of ovBNSTCRF neurons.
Collapse
Affiliation(s)
- Ryoko Uchida
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Yasutaka Mukai
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Aichi, Japan
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
- Postdoctoral Research Fellow, Japan Society for the Promotion of Science, Tokyo, 102-0083, Japan
| | - Taiju Amano
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Keiichi Itoi
- Department of Nursing, Tohoku Fukushi University, Sendai, 981-8522, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601, Aichi, Japan
- Chinese Institute for Brain Research, Beijing, 102206, China
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjuku, Tokyo, 160-8582, Japan
- National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8585, Aichi, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
| |
Collapse
|
4
|
Bertagna NB, Wilson L, Bailey CK, Cruz FC, Albrechet-Souza L, Wills TA. Long-lasting mechanical hypersensitivity and CRF receptor type-1 neuron activation in the BNST following adolescent ethanol exposure. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:48-57. [PMID: 38206283 PMCID: PMC10784637 DOI: 10.1111/acer.15228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/26/2023] [Accepted: 11/08/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Adolescent alcohol use can produce long-lasting alterations in brain function, potentially leading to adverse health outcomes in adulthood. Emerging evidence suggests that chronic alcohol use can increase pain sensitivity or exacerbate existing pain conditions, but the potential neural mechanisms underlying these effects require further investigation. Here, we evaluate the impact of chronic ethanol vapor on mechanical sensitivity over the course of acute and protracted withdrawal in adolescent and adult male and female mice, and its potential association with alterations in corticotropin-releasing factor (CRF) signaling within the bed nucleus of the stria terminalis (BNST). METHODS Adolescent and adult male and female mice underwent intermittent ethanol vapor exposure on 4 days/week for 2 weeks. Mechanical thresholds were evaluated 5 h and 7, 14, 21, and 28 d after cessation of ethanol exposure using the von Frey test. For mice with a history of adolescent ethanol exposure, brains were collected for in situ RNAscope processing after the final test. Messenger RNA expression of c-Fos, Crfr1, and Crf in the BNST subregions was examined. RESULTS Exposure to intermittent ethanol vapor induced persistent mechanical hypersensitivity during withdrawal in both adolescent and adult mice. Notably, the effect was more transient in mice exposed to ethanol during adulthood, resolving by day 28 after ethanol exposure. Furthermore, both male and female mice with a history of adolescent ethanol exposure exhibited increased activation of CRF receptor type 1 (CRFR1) neurons within the dorsolateral BNST. CONCLUSIONS Our results support the conclusion that intermittent ethanol exposure can induce mechanical hypersensitivity, potentially through the activation of BNST CRFR1 neurons. These findings provide a basis for future studies aimed at evaluating specific subpopulations of BNST neurons and their contribution to pain in individuals with a history of alcohol use.
Collapse
Affiliation(s)
- Natalia B. Bertagna
- Department of Cell Biology & Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Department of Pharmacology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Lisa Wilson
- Department of Cell Biology & Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Alcohol & Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Connor K. Bailey
- Department of Cell Biology & Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Fabio C. Cruz
- Department of Pharmacology, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Lucas Albrechet-Souza
- Department of Cell Biology & Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Alcohol & Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Tiffany A. Wills
- Department of Cell Biology & Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Alcohol & Drug Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
5
|
van de Poll Y, Cras Y, Ellender TJ. The neurophysiological basis of stress and anxiety - comparing neuronal diversity in the bed nucleus of the stria terminalis (BNST) across species. Front Cell Neurosci 2023; 17:1225758. [PMID: 37711509 PMCID: PMC10499361 DOI: 10.3389/fncel.2023.1225758] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/03/2023] [Indexed: 09/16/2023] Open
Abstract
The bed nucleus of the stria terminalis (BNST), as part of the extended amygdala, has become a region of increasing interest regarding its role in numerous human stress-related psychiatric diseases, including post-traumatic stress disorder and generalized anxiety disorder amongst others. The BNST is a sexually dimorphic and highly complex structure as already evident by its anatomy consisting of 11 to 18 distinct sub-nuclei in rodents. Located in the ventral forebrain, the BNST is anatomically and functionally connected to many other limbic structures, including the amygdala, hypothalamic nuclei, basal ganglia, and hippocampus. Given this extensive connectivity, the BNST is thought to play a central and critical role in the integration of information on hedonic-valence, mood, arousal states, processing emotional information, and in general shape motivated and stress/anxiety-related behavior. Regarding its role in regulating stress and anxiety behavior the anterolateral group of the BNST (BNSTALG) has been extensively studied and contains a wide variety of neurons that differ in their electrophysiological properties, morphology, spatial organization, neuropeptidergic content and input and output synaptic organization which shape their activity and function. In addition to this great diversity, further species-specific differences are evident on multiple levels. For example, classic studies performed in adult rat brain identified three distinct neuron types (Type I-III) based on their electrophysiological properties and ion channel expression. Whilst similar neurons have been identified in other animal species, such as mice and non-human primates such as macaques, cross-species comparisons have revealed intriguing differences such as their comparative prevalence in the BNSTALG as well as their electrophysiological and morphological properties, amongst other differences. Given this tremendous complexity on multiple levels, the comprehensive elucidation of the BNSTALG circuitry and its role in regulating stress/anxiety-related behavior is a major challenge. In the present Review we bring together and highlight the key differences in BNSTALG structure, functional connectivity, the electrophysiological and morphological properties, and neuropeptidergic profiles of BNSTALG neurons between species with the aim to facilitate future studies of this important nucleus in relation to human disease.
Collapse
Affiliation(s)
- Yana van de Poll
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Yasmin Cras
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tommas J. Ellender
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
Wang X, Ge S, Zhang C. Bed nuclei of the stria terminalis: A key hub in the modulation of anxiety. Eur J Neurosci 2023; 57:900-917. [PMID: 36725691 DOI: 10.1111/ejn.15926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 01/12/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023]
Abstract
The bed nuclei of the stria terminalis (BST) is recognised as a pivotal integrative centre for monitoring emotional valence. It is implicated in the regulation of diverse affective states and motivated behaviours, and decades of research have firmly established its critical role in anxiety-related behavioural processes. Researchers have recently intricately dissected the BST's dynamic activities, its connection patterns and its functions with respect to specific cell types using multiple techniques such as optogenetics, in vivo calcium imaging and transgenic tools to unmask the complex circuitry mechanisms that underlie anxiety. In this review, we principally focus on studies of anxiety-involved neuromodulators within the BST and provide a comprehensive architecture of the anxiety network-highlighting the BST as a key hub in orchestrating anxiety-like behaviour. We posit that these promising efforts will contribute to the identification of an accurate roadmap for future treatment of anxiety disorders.
Collapse
Affiliation(s)
- Xinxin Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shenglin Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chengxin Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
7
|
Zhao N, Shu Y, Jian C, Zhou Z, Bao H, Li X, Cheng X, Zhao Y, Jin S, Shu X. Lactobacillus Ameliorates SD-Induced Stress Responses and Gut Dysbiosis by Increasing the Absorption of Gut-Derived GABA in Rhesus Monkeys. Front Immunol 2022; 13:915393. [PMID: 35874738 PMCID: PMC9302489 DOI: 10.3389/fimmu.2022.915393] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022] Open
Abstract
Sleep deprivation (SD) has become a health problem in the modern society. Although probiotics supplementation has been proven to improve SD-induced gut dysbiosis, the potential neuroendocrine mechanisms remain elusive. In this study, thirty rhesus monkeys (RMs) were recruited. Paradoxical sleep, bright light, and noise were used to build an RM SD model. We examined the plasma γ-aminobutyric acid (GABA), stress hormones, and inflammatory cytokines using ELISAs. 16S ribosomal DNA sequencing and untargeted metabolomics sequencing were employed to detect gut microbial community and metabolites, respectively. The results of our study showed that RMs subjected to SD had elevated plasma stress hormones (such as cortisol and norepinephrine) and proinflammatory cytokines (such as TNF-α, IL-6, and IL-8), and a decreased anti‐inflammatory cytokine IL-10 level. Additionally, SD could give rise to a significant change in gut microbiota and metabolites. The differential gut microbiota and metabolites caused by SD were enriched in the signaling pathways related to GABA metabolism. Pearson correlation analysis revealed that there is a significant correlation between plasma GABA and SD-induced stress responses and gut dysbiosis. The supplementation of GABA-producing probiotics could significantly increase the relative abundance of Lactobacillus and plasma GABA levels, and reverse SD‐induced stress responses and gut dysbiosis. Therefore, we speculated that SD-induced stress response and gut dysbiosis might be an outcome of reduced gut-derived GABA absorption. The supplementation of GABA-producing Lactobacillus might be beneficial for the treatment of SD-induced intestinal dysfunction.
Collapse
Affiliation(s)
- Ning Zhao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Shu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenxing Jian
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zili Zhou
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haijun Bao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianguo Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xukai Cheng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Zhao
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xiaogang Shu, ; Si Jin,
| | - Xiaogang Shu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xiaogang Shu, ; Si Jin,
| |
Collapse
|
8
|
Custers ML, Nestor L, De Bundel D, Van Eeckhaut A, Smolders I. Current Approaches to Monitor Macromolecules Directly from the Cerebral Interstitial Fluid. Pharmaceutics 2022; 14:pharmaceutics14051051. [PMID: 35631637 PMCID: PMC9146401 DOI: 10.3390/pharmaceutics14051051] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 01/27/2023] Open
Abstract
Gaining insights into the pharmacokinetic and pharmacodynamic properties of lead compounds is crucial during drug development processes. When it comes to the treatment of brain diseases, collecting information at the site of action is challenging. There are only a few techniques available that allow for the direct sampling from the cerebral interstitial space. This review concerns the applicability of microdialysis and other approaches, such as cerebral open flow microperfusion and electrochemical biosensors, to monitor macromolecules (neuropeptides, proteins, …) in the brain. Microdialysis and cerebral open flow microperfusion can also be used to locally apply molecules at the same time at the site of sampling. Innovations in the field are discussed, together with the pitfalls. Moreover, the ‘nuts and bolts’ of the techniques and the current research gaps are addressed. The implementation of these techniques could help to improve drug development of brain-targeted drugs.
Collapse
|
9
|
Yamauchi N, Sato K, Sato K, Murakawa S, Hamasaki Y, Nomura H, Amano T, Minami M. Chronic pain-induced neuronal plasticity in the bed nucleus of the stria terminalis causes maladaptive anxiety. SCIENCE ADVANCES 2022; 8:eabj5586. [PMID: 35476439 PMCID: PMC9045713 DOI: 10.1126/sciadv.abj5586] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The comorbidity of chronic pain and mental dysfunctions such as depression and anxiety disorders has long been recognized, but the underlying mechanisms remain poorly understood. Here, using a mouse model of neuropathic pain, we demonstrated neuronal plasticity in the bed nucleus of the stria terminalis (BNST), which plays a critical role in chronic pain-induced maladaptive anxiety. Electrophysiology demonstrated that chronic pain increased inhibitory inputs to lateral hypothalamus (LH)-projecting BNST neurons. Chemogenetic manipulation revealed that sustained suppression of LH-projecting BNST neurons played a crucial role in chronic pain-induced anxiety. Furthermore, using a molecular genetic approach, we demonstrated that chronic pain elevated the excitability of a specific subpopulation of BNST neurons, which express cocaine- and amphetamine-regulated transcript (CART). The elevated excitability of CART-positive neurons caused the increased inhibitory inputs to LH-projecting BNST neurons, thereby inducing anxiety-like behavior. These findings shed light on how chronic pain induces psychiatric disorders, characterized by maladaptive anxiety.
Collapse
|
10
|
Hagiwara H, Sakimura K, Abe M, Itoi K, Kamiya Y, Akema T, Funabashi T. Sex differences in pain-induced modulation of corticotropin-releasing hormone neurons in the dorsolateral part of the stria terminalis in mice. Brain Res 2021; 1773:147688. [PMID: 34644526 DOI: 10.1016/j.brainres.2021.147688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/22/2021] [Accepted: 10/06/2021] [Indexed: 01/06/2023]
Abstract
We earlier reported female-biased, sex-specific involvement of the dorsolateral bed nucleus of the stria terminalis (dl BST) in the formalin-induced pain response in rats. The present study investigated pain effects on mice behaviors. Because the dl BST is densely populated with corticotropin-releasing hormone (CRH) neurons, we examined sex differences in these parameters for the dl BST CRH neurons in male and female mice of a mouse line for which the CRH gene promoter (corticotropin-releasing factor [CRF]-Venus ΔNeo) controls the expression of the modified yellow fluorescent protein (Venus). Approximately 92% of Venus-positive cells in the dl BST were also CRH mRNA-positive, irrespective of sex. Therefore, the cells identified using Venus fluorescence were regarded as CRH neurons. A female-biased sex difference was observed in pain-induced behaviors during the interphase (5-15 min after formalin injection) but not during the later phase (phase 2, 15-60 min) in wild-type mice. In CRF-Venus ΔNeo mice, a female-biased difference was observed in either the earlier phase (phase 1, 0-5 min) or the interphase, but not in phase 2. Patch-clamp recordings taken using an acute BST slice obtained from a CRF-Venus ΔNeo mouse after formalin injection showed miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs). Remarkably, the mEPSCs frequency was higher in the Venus-expressing cells of formalin-injected female mice than in vehicle-treated female mice. Male mice showed no increase in mEPSC frequency by formalin injection. Formalin injection had no effect on mEPSC or mIPSC amplitudes in either sex. Pain-induced changes in mEPSC frequency in putative CRH neurons were phase-dependent. Results show that excitatory synaptic inputs to BST CRH neurons are temporally enhanced along with behavioral sex differences in pain response, suggesting that pain signals alter the BST CRH neurons excitability in a sex-dependent manner.
Collapse
Affiliation(s)
- Hiroko Hagiwara
- Department of Physiology, St. Marianna University School of Medicine, 2-16-1 Sugao Miyamae-ku, Kawasaki 216-8511, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori Chuo-ku, Niigata 951-8585, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori Chuo-ku, Niigata 951-8585, Japan
| | - Keiichi Itoi
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, 6-3-09 Aramaki-aza Aoba-ku, Sendai 980-8579, Japan
| | - Yoshinori Kamiya
- Division of Anesthesiology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata 950-8510, Japan
| | - Tatsuo Akema
- Department of Physiology, St. Marianna University School of Medicine, 2-16-1 Sugao Miyamae-ku, Kawasaki 216-8511, Japan
| | - Toshiya Funabashi
- Department of Physiology, St. Marianna University School of Medicine, 2-16-1 Sugao Miyamae-ku, Kawasaki 216-8511, Japan.
| |
Collapse
|
11
|
Gilpin NW, Yu W, Kash TL. Forebrain-Midbrain Circuits and Peptides Involved in Hyperalgesia After Chronic Alcohol Exposure. Alcohol Res 2021; 41:13. [PMID: 34729286 PMCID: PMC8549866 DOI: 10.35946/arcr.v41.1.13] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
People living with pain report drinking alcohol to relieve pain. Acute alcohol use reduces pain, and chronic alcohol use facilitates the emergence or exaggeration of pain. Recently, funding agencies and neuroscientists involved in basic research have turned their attention to understanding the neurobiological mechanisms that underlie pain-alcohol interactions, with a focus on circuit and molecular mediators of alcohol-induced changes in pain-related behavior. This review briefly discusses some examples of work being done in this area, with a focus on reciprocal projections between the midbrain and extended amygdala, as well as some neurochemical mediators of pain-related phenotypes after alcohol exposure. Finally, as more work accumulates on this topic, the authors highlight the need for the neuroscience field to carefully consider sex and age in the design and analysis of pain-alcohol interaction experiments.
Collapse
Affiliation(s)
- Nicholas W Gilpin
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,Department of Neuroscience, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,Biomedical Laboratory Research and Development and Clinical Science Research and Development Intramural Program, Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana
| | - Waylin Yu
- Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina
| | - Thomas L Kash
- Department of Pharmacology, Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
12
|
Kaneko T, Hara R, Amano T, Minami M. Diverse intracellular signaling pathways mediate the effects of neurotensin on the excitability of type II neurons in the rat dorsolateral bed nucleus of the stria terminalis. J Pharmacol Sci 2021; 147:86-94. [PMID: 34294377 DOI: 10.1016/j.jphs.2021.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 10/21/2022] Open
Abstract
We examined the effects of neurotensin (NTS) on the excitability of type II neurons in the rat dorsolateral bed nucleus of the stria terminalis (dlBNST) using whole-cell patch-clamp electrophysiology. Bath-application of NTS depolarized type II dlBNST neurons. Analyses of the steady-state I-V relationships implied that the depolarizing effect of NTS is due to potassium conductance blocking. The depolarizing effect of NTS was abolished in the presence of a PLC inhibitor, but not affected by a protein kinase C inhibitor. In the presence of a CaMKII inhibitor, NTS showed depolarizing effects via the increase in non-selective cation conductance in addition to the decrease in potassium conductance. Unexpectedly, in the presence of a PKA inhibitor, NTS hyperpolarized type II dlBNST neurons. These results reveal that diverse signaling pathways mediate the effects of NTS on the excitability of type II dlBNST neurons. The elevation of intracellular Ca2+ levels via the inositol phosphate-mediated signaling activates both Ca2+-dependent adenylate cyclase (AC) and CaMKII. Activation of the AC-cAMP-PKA pathway exerts depolarizing effects on type II dlBNST neurons by decreasing potassium conductance and increasing non-selective cation conductance, whereas activation of the CaMKII pathway exerts hyperpolarizing effects on dlBNST neurons by decreasing non-selective cation conductance.
Collapse
Affiliation(s)
- Tomoyuki Kaneko
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Ryuto Hara
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Taiju Amano
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
| |
Collapse
|
13
|
Yu W, Caira CM, Del R Rivera Sanchez N, Moseley GA, Kash TL. Corticotropin-releasing factor neurons in the bed nucleus of the stria terminalis exhibit sex-specific pain encoding in mice. Sci Rep 2021; 11:12500. [PMID: 34127705 PMCID: PMC8203647 DOI: 10.1038/s41598-021-91672-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
The bed nucleus of the stria terminalis (BNST) plays an emerging role in pain regulation. Pharmacological studies have found that inhibiting corticotropin-releasing factor (CRF) signaling in the BNST can selectively mitigate the sensory and affective-motivational components of pain. However, mechanistic insight on the source of CRF that drives BNST responses to these harmful experiences remains unknown. In the present study, we used a series of genetic approaches to show that CRF in the BNST is engaged in the processing and modulation of pain. We conducted cell-type specific in vivo calcium imaging in CRF-Cre mice and found robust and synchronized recruitment of BNSTCRF neurons during acute exposures to noxious heat. Distinct patterns of recruitment were observed by sex, as the magnitude and timing of heat responsive activity in BNSTCRF neurons differed for male and female mice. We then used a viral approach in Floxed-CRF mice to selectively reduce CRF expression in the BNST and found it decreased nociceptive sensitivity for both sexes and increased paw attending for females. Together, these findings reveal that CRF in the BNST influences multiple facets of the pain experience to impact the sex-specific expression of pain-related behaviors.
Collapse
Affiliation(s)
- Waylin Yu
- Department of Pharmacology, School of Medicine, University of North Carolina At Chapel Hill, CB 7178 Thurston Bowles Building, 104 Manning Drive, Chapel Hill, NC, 27599, USA
- Bowles Center for Alcohol Studies, University of North Carolina At Chapel Hill, Chapel Hill, NC, 27599, USA
- Curriculum in Pharmacology, School of Medicine, University of North Carolina At Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Christina M Caira
- Department of Pharmacology, School of Medicine, University of North Carolina At Chapel Hill, CB 7178 Thurston Bowles Building, 104 Manning Drive, Chapel Hill, NC, 27599, USA
- Bowles Center for Alcohol Studies, University of North Carolina At Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Natalia Del R Rivera Sanchez
- Department of Pharmacology, School of Medicine, University of North Carolina At Chapel Hill, CB 7178 Thurston Bowles Building, 104 Manning Drive, Chapel Hill, NC, 27599, USA
- Bowles Center for Alcohol Studies, University of North Carolina At Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Garrett A Moseley
- Department of Pharmacology, School of Medicine, University of North Carolina At Chapel Hill, CB 7178 Thurston Bowles Building, 104 Manning Drive, Chapel Hill, NC, 27599, USA
- Bowles Center for Alcohol Studies, University of North Carolina At Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Thomas L Kash
- Department of Pharmacology, School of Medicine, University of North Carolina At Chapel Hill, CB 7178 Thurston Bowles Building, 104 Manning Drive, Chapel Hill, NC, 27599, USA.
- Bowles Center for Alcohol Studies, University of North Carolina At Chapel Hill, Chapel Hill, NC, 27599, USA.
- Curriculum in Pharmacology, School of Medicine, University of North Carolina At Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
14
|
Yu W, Pati D, Pina MM, Schmidt KT, Boyt KM, Hunker AC, Zweifel LS, McElligott ZA, Kash TL. Periaqueductal gray/dorsal raphe dopamine neurons contribute to sex differences in pain-related behaviors. Neuron 2021; 109:1365-1380.e5. [PMID: 33740416 PMCID: PMC9990825 DOI: 10.1016/j.neuron.2021.03.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/02/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023]
Abstract
Sex differences in pain severity, response, and pathological susceptibility are widely reported, but the neural mechanisms that contribute to these outcomes remain poorly understood. Here we show that dopamine (DA) neurons in the ventrolateral periaqueductal gray/dorsal raphe (vlPAG/DR) differentially regulate pain-related behaviors in male and female mice through projections to the bed nucleus of the stria terminalis (BNST). We find that activation of vlPAG/DRDA+ neurons or vlPAG/DRDA+ terminals in the BNST reduces nociceptive sensitivity during naive and inflammatory pain states in male mice, whereas activation of this pathway in female mice leads to increased locomotion in the presence of salient stimuli. We additionally use slice physiology and genetic editing approaches to demonstrate that vlPAG/DRDA+ projections to the BNST drive sex-specific responses to pain through DA signaling, providing evidence of a novel ascending circuit for pain relief in males and contextual locomotor response in females.
Collapse
Affiliation(s)
- Waylin Yu
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dipanwita Pati
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Melanie M Pina
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Karl T Schmidt
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kristen M Boyt
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Avery C Hunker
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Larry S Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
| | - Zoe A McElligott
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas L Kash
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
15
|
Urien L, Stein N, Ryckman A, Bell L, Bauer EP. Extended amygdala circuits are differentially activated by context fear conditioning in male and female rats. Neurobiol Learn Mem 2021; 180:107401. [PMID: 33581315 PMCID: PMC8076097 DOI: 10.1016/j.nlm.2021.107401] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 02/06/2023]
Abstract
As the incidence of anxiety disorders is more prevalent in females, comparing the neural underpinnings of anxiety in males and females is imperative. The bed nucleus of the stria terminalis (BNST) contributes to long-lasting, anxiety-like states including the expression of context fear conditioning. Currently, there is conflicting evidence as to which nuclei of the BNST contribute to these behaviors. The anterolateral portion of the BNST (BNST-AL) located dorsal to the anterior commissure and lateral to the stria terminalis sends robust projections to the central nucleus of the amygdala (CE). Here we asked whether the BNST-AL is active during the expression of context fear conditioning in both male and female rats. At the cellular level, the expression of context fear produced upregulation of the immediate-early gene ARC in the BNST-AL as well as an upregulation of ARC specifically in neurons projecting to the CE, as labeled by the retrograde tracer Fluorogold infused into the CE. However, this pattern of ARC expression was observed in male rats only. Excitotoxic lesions of the BNST reduced context fear expression in both sexes, suggesting that a different set of BNST subnuclei may be recruited by the expression of fear and anxiety-like behaviors in females. Overall, our data highlight the involvement of the BNST-AL in fear expression in males, and suggest that subnuclei of the BNST may be functionally different in male and female rats.
Collapse
Affiliation(s)
- Louise Urien
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Nicole Stein
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Abigail Ryckman
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Lindsey Bell
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Elizabeth P Bauer
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States.
| |
Collapse
|
16
|
Lin R, Liang J, Luo M. The Raphe Dopamine System: Roles in Salience Encoding, Memory Expression, and Addiction. Trends Neurosci 2021; 44:366-377. [PMID: 33568331 DOI: 10.1016/j.tins.2021.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/21/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023]
Abstract
Dopamine (DA) neurons of the dorsal raphe nucleus (DRN) were traditionally viewed as an extension of the ventral tegmental area (VTA) DA population. While the VTA DA population is known to play important roles in reward processing, emerging evidence now supports the view that DRN DA neurons are a specialized midbrain DA subsystem that performs distinct functions in parallel to the VTA DA population. Recent studies have shed new light on the roles of DRN DA neurons in encoding incentive salience and in regulating memory expression and arousal. Here, we review recent findings using mouse models about the physiology and behavioral functions of DRN DA neurons, highlight the engagement of DRN DA neurons and their upstream circuits in opioid addiction, and discuss emerging lines of investigation that reveal multifaceted heterogeneity among DRN DA neurons.
Collapse
Affiliation(s)
- Rui Lin
- National Institute of Biological Sciences (NIBS), Beijing 102206, China.
| | - Jingwen Liang
- National Institute of Biological Sciences (NIBS), Beijing 102206, China
| | - Minmin Luo
- National Institute of Biological Sciences (NIBS), Beijing 102206, China; Chinese Institute for Brain Research, Beijing 102206, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
17
|
Hara R, Takahashi D, Takehara T, Amano T, Minami M. Inhibitory synaptic transmissions to the bed nucleus of the stria terminalis neurons projecting to the ventral tegmental area are enhanced in rats exposed to chronic mild stress. Mol Brain 2020; 13:139. [PMID: 33059723 PMCID: PMC7560094 DOI: 10.1186/s13041-020-00684-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/09/2020] [Indexed: 11/19/2022] Open
Abstract
The comorbidities of depression and chronic pain have long been recognized in the clinic, and several preclinical studies have demonstrated depression-like behaviors in animal models of chronic pain. These findings suggest a common neuronal basis for depression and chronic pain. Recently, we reported that the mesolimbic dopaminergic system was tonically suppressed during chronic pain by enhanced inhibitory synaptic inputs to neurons projecting from the dorsolateral bed nucleus of the stria terminalis (dlBNST) to the ventral tegmental area (VTA), suggesting that tonic suppression of the mesolimbic dopaminergic system by this neuroplastic change may be involved in chronic pain-induced depression-like behaviors. In this study, we hypothesized that inhibitory synaptic inputs to VTA-projecting dlBNST neurons are also enhanced in animal models of depression, thereby suppressing the mesolimbic dopaminergic system. To test this hypothesis, we performed whole-cell patch-clamp electrophysiology using brain slices prepared from rats exposed to chronic mild stress (CMS), a widely used animal model of depression. The results showed a significant enhancement in the frequency of spontaneous inhibitory postsynaptic currents in VTA-projecting dlBNST neurons in the CMS group compared with the no stress group. The findings revealed enhanced inhibitory synaptic inputs to VTA-projecting dlBNST neurons in this rat model of depression, suggesting that this neuroplastic change is a neuronal mechanism common to depression and chronic pain that causes dysfunction of the mesolimbic dopaminergic system, thereby inducing depression-like behaviors.
Collapse
Affiliation(s)
- Ryuto Hara
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Daiki Takahashi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Tatsuhiro Takehara
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Taiju Amano
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
| |
Collapse
|
18
|
Wang X, Tian Z, Ma J, Feng Z, Ou Y, Zhou M, Peng J, Lv Y, Gao G, Qi S. NPY alterations induced by chronic morphine exposure affect the maintenance and reinstatement of morphine conditioned place preference. Neuropharmacology 2020; 181:108350. [PMID: 33027625 DOI: 10.1016/j.neuropharm.2020.108350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/28/2020] [Accepted: 10/02/2020] [Indexed: 01/23/2023]
Abstract
Opioid addiction is a brain disease that severely harms society and personal health. Although the tremendous numbers of patients worldwide and emerged negative events, effective treatments for opioid addiction are still lacking. Neuropeptide Y (NPY) is one of the main orexigenic peptides that play vital roles in food intake and energy metabolism. However, increasing evidence indicates that NPY may have great potential in mediating reward effects and drug dependence. In the present study, we assessed the expression changes of NPY in the nucleus accumbens at different timepoints following morphine conditioned place preference (CPP) and investigated the functional importance of potential NPY changes. Our results showed that NPY expression significantly decreased in the nucleus accumbens shell (AcbSh) immediately after chronic morphine exposure. Subsequently, it increased rapidly at first and then gradually returned to normal levels. Further data indicated that these NPY changes were involved in morphine reward memory, demonstrated by a reduction in the extinction period after blocking of the Y5 receptor by L-152,804 in the AcbSh and a prolonged duration of the extinction period following the application of NPY. More importantly, the additional results revealed that L-152,804 also remarkably suppressed the reinstatement of morphine CPP. Together, our results indicate that a complicated plasticity of the NPY pathway in AcbSh occurs following morphine CPP, and this plasticity plays an important role in modulating morphine reward memory. These findings may enhance our understanding of the role of the NPY system in opioid addiction and indicate a promising target for opioid addiction treatment.
Collapse
Affiliation(s)
- Xingqin Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhen Tian
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China; Department of Pharmacy, Guangzhou Women and Children's Medical Center, 9 Jinsui Road, Guangzhou, China
| | - Jie Ma
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Zhanpeng Feng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yichao Ou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Mingfeng Zhou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Junjie Peng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yunfei Lv
- Department of Anesthesiology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Guodong Gao
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China.
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
19
|
Rajbhandari AK, Bakshi VP. Repeated norepinephrine receptor stimulation in the BNST induces sensorimotor gating deficits via corticotropin releasing factor. Neuropharmacology 2020; 172:108090. [PMID: 32360378 DOI: 10.1016/j.neuropharm.2020.108090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/15/2020] [Accepted: 04/02/2020] [Indexed: 11/19/2022]
Abstract
Intense stress precipitates symptoms in disorders such as post-traumatic stress (PTSD) and schizophrenia. Patients with these disorders have dysfunctional sensorimotor gating as indexed by disrupted prepulse inhibition of the startle response (PPI), which refers to decreased startle response when a weak pre-stimulus precedes a startling stimulus. Stress promotes release of norepinephrine (NE) and corticotrophin releasing factor (CRF) within the brain, neurotransmitters that also modulate PPI. We have shown that repeated stress causes sensitization of NE receptors within the basolateral amygdala (BLA) via CRF receptors and promotes long-lasting PPI disruptions and startle abnormalities. The bed nucleus of the stria terminalis (BNST) is another crucial brain region that could be involved in stress-induced alterations in NE and CRF functions to promote PPI changes as this anatomical structure is enriched in CRF and NE receptors that have been shown to regulate each other. We hypothesized that repeated infusions of NE into the BNST would cross-sensitize CRF receptors or vice versa to alter PPI. Separate groups of male Sprague Dawley rats received, CRF (200ng/0.5 μl), NE (20μg/0.5 μl), or vehicle into the BNST, once/day for 3 days and PPI was tested after each infusion. Repeated CRF-or vehicle-treated rats were then challenged with a subthreshold dose of NE (0.3μg/0.5 μl) while repeated NE-treated rats were challenged with CRF (200ng/0.5 μl), and PPI was measured. Surprisingly, initial/repeated CRF or vehicle in the BNST had no effects on PPI. In contrast, initial and repeated NE disrupted PPI. Sub-threshold NE challenge in rats that previously received repeated CRF had no effect on PPI. Interestingly though, intra-BNST challenge dose of CRF significantly disrupted PPI in rats that previously had received repeated NE infusions. Taken together, these results indicate that repeated stress-induced NE release could alter the activity of CRF receptors in the BNST to modulate sensorimotor gating as measured through PPI.
Collapse
Affiliation(s)
- Abha Karki Rajbhandari
- Dept. of Psychiatry and Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53719, USA; AKR Is Now at Icahn School of Medicine at Mount Sinai, New York, 10029, USA.
| | - Vaishali P Bakshi
- Dept. of Psychiatry and Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53719, USA; AKR Is Now at Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| |
Collapse
|
20
|
Hara R, Asaoka Y, Takahashi D, Nomura H, Amano T, Minami M. Disappearance of the inhibitory effect of neuropeptide Y within the dorsolateral bed nucleus of the stria terminalis in rats with chronic pain. Neurosci Lett 2020; 728:134958. [PMID: 32278943 DOI: 10.1016/j.neulet.2020.134958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/25/2020] [Accepted: 04/04/2020] [Indexed: 01/24/2023]
Abstract
We recently showed that the mesolimbic dopaminergic system was tonically suppressed during chronic pain by enhanced corticotropin releasing factor (CRF) signaling within the dorsolateral bed nucleus of the stria terminalis (dlBNST), and that inhibition of intra-dlBNST CRF signaling restored the mesolimbic dopaminergic system function. Specifically, bilateral intra-dlBNST injections of the CRF type 1 receptor antagonist NBI27914 increased intra-nucleus accumbens dopamine release and induced reward-related behaviors in rats with chronic pain. Here, we used a conditioned place preference (CPP) test to explore whether intra-dlBNST injections of neuropeptide Y (NPY) restored the mesolimbic reward system function in chronic pain rats, because we previously showed that NPY had an effect opposite to that of CRF in dlBNST neurons. Specifically, CRF depolarized type II dlBNST neurons whereas NPY hyperpolarized them. However, unexpectedly, intra-dlBNST NPY injections had no effect on CPP test outcomes. Then, we compared the effects of NPY on the membrane potentials of type II dlBNST neurons of sham-operated control rats and those of chronic pain animals. Whole-cell patch-clamp electrophysiology revealed that NPY hyperpolarized type II dlBNST neurons in the sham-operated group. By contrast, in the chronic pain group, NPY did not hyperpolarize, but rather depolarized, type II dlBNST neurons. These results indicate that NPY no longer hyperpolarizes type II dlBNST neurons in rats with chronic pain, therefore it does not reverse the excitatory effects of CRF. This may be why intra-dlBNST injections of NPY into chronic pain rats did not exhibit a rewarding effect in the CPP test, whereas intra-dlBNST injections of NBI27914 did. This is the first study to demonstrate a chronic pain-induced neuroplastic change in NPY signaling in the dlBNST. Such a change may be involved in the dysfunction of the mesolimbic reward system under the chronic pain condition.
Collapse
Affiliation(s)
- Ryuto Hara
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Yuta Asaoka
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Daiki Takahashi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Hiroshi Nomura
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Taiju Amano
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan.
| |
Collapse
|
21
|
Beyeler A, Dabrowska J. Neuronal diversity of the amygdala and the bed nucleus of the stria terminalis. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2020; 26:63-100. [PMID: 32792868 DOI: 10.1016/b978-0-12-815134-1.00003-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Anna Beyeler
- Neurocentre Magendie, French National Institutes of Health (INSERM) unit 1215, Neurocampus of Bordeaux University, Bordeaux, France
| | - Joanna Dabrowska
- Center for the Neurobiology of Stress Resilience and Psychiatric Disorders, Discipline of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
22
|
Takahashi D, Asaoka Y, Kimura K, Hara R, Arakaki S, Sakasai K, Suzuki H, Yamauchi N, Nomura H, Amano T, Minami M. Tonic Suppression of the Mesolimbic Dopaminergic System by Enhanced Corticotropin-Releasing Factor Signaling Within the Bed Nucleus of the Stria Terminalis in Chronic Pain Model Rats. J Neurosci 2019; 39:8376-8385. [PMID: 31451580 PMCID: PMC6794933 DOI: 10.1523/jneurosci.3047-18.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 07/01/2019] [Accepted: 08/16/2019] [Indexed: 02/02/2023] Open
Abstract
Although dysfunction of the mesolimbic dopaminergic system has been implicated in chronic pain, the underlying mechanisms remain to be elucidated. We hypothesized that increased inhibitory inputs to the neuronal pathway from the dorsolateral bed nucleus of the stria terminalis (dlBNST) to the ventral tegmental area (VTA) during chronic pain may induce tonic suppression of the mesolimbic dopaminergic system. To test this hypothesis, male Sprague Dawley rats were subjected to spinal nerve ligation to induce neuropathic pain and then spontaneous IPSCs (sIPSCs) were measured in this neuronal pathway. Whole-cell patch-clamp electrophysiology of brain slices containing the dlBNST revealed that the frequency of sIPSCs significantly increased in VTA-projecting dlBNST neurons 4 weeks after surgery. Next, the role of corticotropin-releasing factor (CRF) signaling within the dlBNST in the increased sIPSCs was examined. CRF increased the frequency of sIPSCs in VTA-projecting dlBNST neurons in sham-operated controls, but not in chronic pain rats. By contrast, NBI27914, a CRF type 1 receptor antagonist, decreased the frequency of sIPSCs in VTA-projecting dlBNST neurons in the chronic pain rats, but not in the control animals. In addition, histological analyses revealed the increased expression of CRF mRNA in the dlBNST. Finally, bilateral injections of NBI27914 into the dlBNST of chronic pain rats activated mesolimbic dopaminergic neurons and induced conditioned place preference. Together, these results suggest that the mesolimbic dopaminergic system is tonically suppressed during chronic pain by enhanced CRF signaling within the dlBNST via increased inhibitory inputs to VTA-projecting dlBNST neurons.SIGNIFICANCE STATEMENT The comorbidity of chronic pain and depression has long been recognized. Although dysfunction of the mesolimbic dopaminergic system has been implicated in both chronic pain and depression, the underlying mechanisms remain to be elucidated. Here, we show that the inhibitory inputs to the neuronal pathway from the dorsolateral bed nucleus of the stria terminalis (dlBNST) to the ventral tegmental area increase during chronic pain. This neuroplastic change is mediated by enhanced corticotropin-releasing factor signaling within the dlBNST that leads to tonic suppression of the mesolimbic dopaminergic system, which may be involved in the depressive mood and anhedonia under the chronic pain condition.
Collapse
Affiliation(s)
- Daiki Takahashi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Yuta Asaoka
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Keisuke Kimura
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Ryuto Hara
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Saya Arakaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Keisuke Sakasai
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Hiroe Suzuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Naoki Yamauchi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Hiroshi Nomura
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Taiju Amano
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| |
Collapse
|
23
|
Modulation of the Negative Affective Dimension of Pain: Focus on Selected Neuropeptidergic System Contributions. Int J Mol Sci 2019; 20:ijms20164010. [PMID: 31426473 PMCID: PMC6720937 DOI: 10.3390/ijms20164010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/11/2022] Open
Abstract
It is well known that emotions can interfere with the perception of physical pain, as well as with the development and maintenance of painful conditions. On the other hand, somatic pain can have significant consequences on an individual’s affective behavior. Indeed, pain is defined as a complex and multidimensional experience, which includes both sensory and emotional components, thus exhibiting the features of a highly subjective experience. Over the years, neural pathways involved in the modulation of the different components of pain have been identified, indicating the existence of medial and lateral pain systems, which, respectively, project from medial or lateral thalamic nuclei to reach distinct cortex regions relating to specific functions. However, owing to the limited information concerning how mood state and painful input affect each other, pain treatment is frequently unsatisfactory. Different neuromodulators, including endogenous neuropeptides, appear to be involved in pain-related emotion and in its affective influence on pain perception, thus playing key roles in vulnerability and clinical outcome. Hence, this review article focuses on evidence concerning the modulation of the sensory and affective dimensions of pain, with particular attention given to some selected neuropeptidergic system contributions.
Collapse
|
24
|
de Miguel E, Vekovischeva O, Elsilä LV, Panhelainen A, Kankuri E, Aitta-Aho T, Korpi ER. Conditioned Aversion and Neuroplasticity Induced by a Superagonist of Extrasynaptic GABA A Receptors: Correlation With Activation of the Oval BNST Neurons and CRF Mechanisms. Front Mol Neurosci 2019; 12:130. [PMID: 31178693 PMCID: PMC6543524 DOI: 10.3389/fnmol.2019.00130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/03/2019] [Indexed: 11/13/2022] Open
Abstract
THIP (gaboxadol), a superagonist of the δ subunit-containing extrasynaptic GABAA receptors, produces persistent neuroplasticity in dopamine (DA) neurons of the ventral tegmental area (VTA), similarly to rewarding drugs of abuse. However, unlike them THIP lacks abuse potential and induces conditioned place aversion in mice. The mechanism underlying the aversive effects of THIP remains elusive. Here, we show that mild aversive effects of THIP were detected 2 h after administration likely reflecting an anxiety-like state with increased corticosterone release and with central recruitment of corticotropin-releasing factor corticotropin-releasing factor receptor 1 (CRF1) receptors. A detailed immunohistochemical c-Fos expression mapping for THIP-activated brain areas revealed a correlation between the activation of CRF-expressing neurons in the oval nucleus of the bed nuclei of stria terminalis and THIP-induced aversive effects. In addition, the neuroplasticity of mesolimbic DA system (24 h after administration) and conditioned place aversion by THIP after four daily acute sessions were dependent on extrasynaptic GABAA receptors (abolished in δ-GABAA receptor knockout mice) and activation of the CRF1 receptors (abolished in wildtype mice by a CRF1 receptor antagonist). A selective THIP-induced activation of CRF-expressing neurons in the oval part of the bed nucleus of stria terminalis may constitute a novel mechanism for inducing plasticity in a population of VTA DA neurons and aversive behavioral states.
Collapse
Affiliation(s)
- Elena de Miguel
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olga Vekovischeva
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Lauri V Elsilä
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anne Panhelainen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Teemu Aitta-Aho
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Esa R Korpi
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
25
|
Rossetti I, Zambusi L, Maccioni P, Sau R, Provini L, Castelli MP, Gonciarz K, Colombo G, Morara S. Predisposition to Alcohol Drinking and Alcohol Consumption Alter Expression of Calcitonin Gene-Related Peptide, Neuropeptide Y, and Microglia in Bed Nucleus of Stria Terminalis in a Subnucleus-Specific Manner. Front Cell Neurosci 2019; 13:158. [PMID: 31114482 PMCID: PMC6502997 DOI: 10.3389/fncel.2019.00158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 04/08/2019] [Indexed: 01/01/2023] Open
Abstract
Excessive alcohol consumption is often linked to anxiety states and has a major relay center in the anterior part of bed nucleus of stria terminalis (BNST). We analyzed the impact of (i) genetic predisposition to high alcohol preference and consumption, and (ii) alcohol intake on anterior BNST, namely anterolateral (AL), anteromedial (AM), and anteroventral (lateral + medial subdivisions: AVl, AVm) subnuclei. We used two rat lines selectively bred for low- and high-alcohol preference and consumption, named Sardinian alcohol-non preferring (sNP) and -preferring (sP), respectively, the latter showing also inherent anxiety-related behaviors. We analyzed the modulation of calcitonin gene-related peptide (CGRP; exerting anxiogenic effects in BNST), neuropeptide Y (NPY; exerting mainly anxiolytic effects), and microglia activation (neuroinflammation marker, thought to increase anxiety). Calcitonin gene-related peptide-immunofluorescent fibers/terminals did not differ between alcohol-naive sP and sNP rats. Fiber/terminal NPY-immunofluorescent intensity was lower in BNST-AM and BNST-AVm of alcohol-naive sP rats. Activation of microglia (revealed by morphological analysis) was decreased in BNST-AM and increased in BNST-AVm of alcohol-naive sP rats. Prolonged (30 consecutive days), voluntary alcohol intake under the homecage 2-bottle “alcohol vs. water” regimen strongly increased CGRP intensity in BNST of sP rats in a subnucleus-specific manner: in BNST-AL, BNST-AVm, and BNST-AM. CGRP area sum, however, decreased in BNST-AM, without changes in other subnuclei. Alcohol consumption increased NPY expression, in a subnucleus-specific manner, in BNST-AL, BNST-AVl, and BNST-AVm. Alcohol consumption increased many size/shapes parameters in microglial cells, indicative of microglia de-activation. Finally, microglia density was increased in ventral anterior BNST (BNST-AVl, BNST-AVm) by alcohol consumption. In conclusion, genetic predisposition of sP rats to high alcohol intake could be in part mediated by anterior BNST subnuclei showing lower NPY expression and differential microglia activation. Alcohol intake in sP rats produced complex subnucleus-specific changes in BNST, affecting CGRP/NPY expression and microglia and leading to hypothesize that these changes might contribute to the anxiolytic effects of voluntarily consumed alcohol repeatedly observed in sP rats.
Collapse
Affiliation(s)
- Ilaria Rossetti
- Institute of Neuroscience, National Research Council of Italy, Milan, Italy
| | - Laura Zambusi
- Institute of Neuroscience, National Research Council of Italy, Milan, Italy
| | - Paola Maccioni
- Institute of Neuroscience, National Research Council of Italy, Milan, Italy
| | - Roberta Sau
- Institute of Neuroscience, National Research Council of Italy, Milan, Italy
| | - Luciano Provini
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - M Paola Castelli
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Krzysztof Gonciarz
- Center for Systems Biology Dresden, Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Giancarlo Colombo
- Institute of Neuroscience, National Research Council of Italy, Milan, Italy
| | - Stefano Morara
- Institute of Neuroscience, National Research Council of Italy, Milan, Italy.,Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
26
|
The role of the bed nucleus of the stria terminalis in pain-induced aversive motivation. Curr Opin Behav Sci 2019. [DOI: 10.1016/j.cobeha.2018.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
27
|
Ch'ng S, Fu J, Brown RM, McDougall SJ, Lawrence AJ. The intersection of stress and reward: BNST modulation of aversive and appetitive states. Prog Neuropsychopharmacol Biol Psychiatry 2018; 87:108-125. [PMID: 29330137 DOI: 10.1016/j.pnpbp.2018.01.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 12/27/2017] [Accepted: 01/08/2018] [Indexed: 12/13/2022]
Abstract
The bed nucleus of the stria terminalis (BNST) is widely acknowledged as a brain structure that regulates stress and anxiety states, as well as aversive and appetitive behaviours. The diverse roles of the BNST are afforded by its highly modular organisation, neurochemical heterogeneity, and complex intrinsic and extrinsic circuitry. There has been growing interest in the BNST in relation to psychopathologies such as anxiety and addiction. Although research on the human BNST is still in its infancy, there have been extensive preclinical studies examining the molecular signature and hodology of the BNST and their involvement in stress and reward seeking behaviour. This review examines the neurochemical phenotype and connectivity of the BNST, as well as electrophysiological correlates of plasticity in the BNST mediated by stress and/or drugs of abuse.
Collapse
Affiliation(s)
- Sarah Ch'ng
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Jingjing Fu
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Robyn M Brown
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Stuart J McDougall
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Andrew J Lawrence
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia.
| |
Collapse
|
28
|
Walter AL, Bartsch JC, Datunashvili M, Blaesse P, Lange MD, Pape HC. Physiological Profile of Neuropeptide Y-Expressing Neurons in Bed Nucleus of Stria Terminalis in Mice: State of High Excitability. Front Cell Neurosci 2018; 12:393. [PMID: 30455634 PMCID: PMC6231247 DOI: 10.3389/fncel.2018.00393] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 10/15/2018] [Indexed: 12/17/2022] Open
Abstract
Both, the anterior bed nucleus of the stria terminalis (BNST) and the neuropeptide Y (NPY) system are involved in shaping fear and defensive responses that adapt the organism to potentially life-threatening conditions. NPY is expressed in the BNST but NPY-expressing neurons in this critical hub in the stress response network have not been addressed before. Therefore, we performed whole-cell patch-clamp recordings in acute slices of anterior BNST from Npy-hrGFP transgenic mice to identify and characterize NPY-expressing neurons. We show that NPY-positive and NPY-negative neurons in anterior BNST match the previous classification scheme of type I (Regular Spiking), type II (Low-Threshold Bursting), and type III (fast Inward Rectifying) cells, although the proportion of these physiological phenotypes was similar within both neuronal subpopulations. However, NPY-positive and NPY-negative neurons possessed distinct intrinsic electrophysiological properties. NPY-positive neurons displayed higher input resistance and lower membrane capacitance, corresponding to small cell bodies and shorter less ramified dendrites, as compared to their NPY-negative counterparts. Furthermore, NPY-positive neurons generated higher frequent series of action potentials upon membrane depolarization and displayed significantly lower GABAA receptor-mediated synaptic responsiveness during evoked, spontaneous, and elementary synaptic activity. Taken together, these properties indicate an overall state of high excitability in NPY-positive neurons in anterior BNST. In view of the role of the anterior BNST in anxiety- and stress-related behaviors, these findings suggest a scenario where NPY-positive neurons are preferentially active and responsive to afferent inputs, thereby contributing to adaptation of the organism to stressful environmental encounters.
Collapse
Affiliation(s)
- Achim Leonhard Walter
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | - Maia Datunashvili
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Peter Blaesse
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Maren Denise Lange
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Hans-Christian Pape
- Institute of Physiology I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| |
Collapse
|
29
|
Yamauchi N, Takahashi D, Sugimura YK, Kato F, Amano T, Minami M. Activation of the neural pathway from the dorsolateral bed nucleus of the stria terminalis to the central amygdala induces anxiety-like behaviors. Eur J Neurosci 2018; 48:3052-3061. [PMID: 30240530 DOI: 10.1111/ejn.14165] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 09/02/2018] [Accepted: 09/17/2018] [Indexed: 12/24/2022]
Abstract
The bed nucleus of the stria terminalis (BNST) and the central amygdala (CeA) comprise a forebrain unit that has been described as the "extended amygdala". These two nuclei send dense projections to each other and have been implicated in the regulation of negative emotional states, including anxiety and fear. The present study employed an optogenetic technique to examine whether stimulation of CeA-projecting dorsolateral BNST (dlBNST) neuron terminals would influence anxiety-like behaviors in male Sprague-Dawley rats. Photostimulation of CeA-projecting dlBNST neuron terminals produced anxiogenic effects in an elevated plus maze test. This finding is inconsistent with previous reports showing that optogenetic stimulation of BNST neurons projecting to the lateral hypothalamus (LH) and ventral tegmental area (VTA) produces anxiolytic rather than anxiogenic effects. To address this issue, electrophysiological analyses were conducted to characterize dlBNST neurons projecting to the CeA, LH, and VTA. dlBNST neurons can be electrophysiologically classified into three distinct cell types (types I-III) according to their responses to depolarizing and hyperpolarizing current injections. Whole-cell patch-clamp recordings revealed that more than 60% of the CeA-projecting dlBNST neurons were type II, whereas approximately 80% of the LH- and VTA-projecting dlBNST neurons were type III. These electrophysiological results will help elucidate the mechanisms underlying the heterogeneity of BNST neurons during the regulation of anxiety-like behaviors.
Collapse
Affiliation(s)
- Naoki Yamauchi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Daiki Takahashi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yae K Sugimura
- Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
| | - Fusao Kato
- Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
| | - Taiju Amano
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
30
|
Ide S, Yamamoto R, Suzuki H, Takeda H, Minami M. Roles of noradrenergic transmission within the ventral part of the bed nucleus of the stria terminalis in bidirectional brain-intestine interactions. Neuropsychopharmacol Rep 2018; 38:182-188. [PMID: 30264532 PMCID: PMC7292287 DOI: 10.1002/npr2.12032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 02/06/2023] Open
Abstract
Aims The bed nucleus of the stria terminalis (BNST) is a limbic structure mediating autonomic and neuroendocrine responses and negative affective states such as anxiety and fear. We previously demonstrated that noradrenergic transmission via β‐adrenoceptors within the ventral part of BNST (vBNST) is involved in bidirectional interactions between the brain and the upper gastrointestinal (GI) tract. The present study aimed to examine the roles of intra‐vBNST noradrenergic transmission via β‐adrenoceptors in bidirectional interactions between the brain and lower GI tract. Methods In vivo microdialysis experiments were performed to examine colorectal distention (CRD)‐induced noradrenaline release within the vBNST of freely moving male Sprague‐Dawley rats. Colonic transit and abdominal pain perception were examined following intra‐vBNST injections of isoproterenol, a β‐adrenoceptor agonist, with and without co‐administration of timolol, a β‐adrenoceptor antagonist. Results CRD increased extracellular noradrenaline levels within the vBNST and evoked abdominal contractions in a pressure‐dependent manner (30‐60 mm Hg). Bilateral intra‐vBNST injections of isoproterenol (30 nmol/side) significantly increased CRD (30 mm Hg)‐induced abdominal contractions. Intra‐vBNST injections of isoproterenol (30 nmol/side) significantly increased colonic transit, which was reversed by co‐administration of timolol (30 nmol/side). Conclusion The results of this study suggest (a) the existence of a positive feedback loop between intra‐vBNST noradrenaline release and abdominal pain perception, and (b) the modulation of colonic motility by intra‐vBNST noradrenergic transmission via β‐adrenoceptors. Dysfunction of the lower GI tract may increase noradrenaline release within the vBNST, which, in turn, may exacerbate impairment of its motility and pain perception. In vivo microdialysis experiments demonstrated that colorectal distention (CRD) increased extracellular levels of noradrenaline within the vBNST. Intra‐vBNST injections of isoproterenol, a β‐adrenoceptor agonist, induced visceral hypersensitivity to CRD and increased colonic transit, and the increase in colonic transit was reversed by co‐administration of timolol, a β‐adrenoceptor antagonist. The present findings demonstrated important roles of noradrenergic transmission via β‐adrenoceptors within the vBNST in bidirectional brain‐intestine interactions.
![]()
Collapse
Affiliation(s)
- Soichiro Ide
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.,Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Ryuta Yamamoto
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Hacchi Suzuki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Hiroshi Takeda
- Laboratory of Pathophysiology and Therapeutics, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
31
|
Estradiol Increases Glutamate and GABA Neurotransmission into GnRH Neurons via Retrograde NO-Signaling in Proestrous Mice during the Positive Estradiol Feedback Period. eNeuro 2018; 5:eN-NWR-0057-18. [PMID: 30079374 PMCID: PMC6073979 DOI: 10.1523/eneuro.0057-18.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/26/2018] [Accepted: 07/09/2018] [Indexed: 12/20/2022] Open
Abstract
Surge release of gonadotropin-releasing hormone (GnRH) is essential in the activation of pituitary gonadal unit at proestrus afternoon preceded by the rise of serum 17β-estradiol (E2) level during positive feedback period. Here, we describe a mechanism of positive estradiol feedback regulation acting directly on GnRH-green fluorescent protein (GFP) neurons of mice. Whole-cell clamp and loose patch recordings revealed that a high physiological dose of estradiol (200 pM), significantly increased firing rate at proestrus afternoon. The mPSC frequency at proestrus afternoon also increased, whereas it decreased at metestrus afternoon and had no effect at proestrus morning. Inhibition of the estrogen receptor β (ERβ), intracellular blockade of the Src kinase and phosphatidylinositol 3 kinase (PI3K) and scavenge of nitric oxide (NO) inside GnRH neurons prevented the facilitatory estradiol effect indicating involvement of the ERβ/Src/PI3K/Akt/nNOS pathway in this fast, direct stimulatory effect. Immunohistochemistry localized soluble guanylate cyclase, the main NO receptor, in both glutamatergic and GABAergic terminals innervating GnRH neurons. Accordingly, estradiol facilitated neurotransmissions to GnRH neurons via both GABAA-R and glutamate/AMPA/kainate-R. These results indicate that estradiol acts directly on GnRH neurons via the ERβ/Akt/nNOS pathway at proestrus afternoon generating NO that retrogradely accelerates GABA and glutamate release from the presynaptic terminals contacting GnRH neurons. The newly explored mechanism might contribute to the regulation of the GnRH surge, a fundamental prerequisite of the ovulation.
Collapse
|
32
|
Xiao X, Zhang YQ. A new perspective on the anterior cingulate cortex and affective pain. Neurosci Biobehav Rev 2018; 90:200-211. [DOI: 10.1016/j.neubiorev.2018.03.022] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/22/2018] [Accepted: 03/22/2018] [Indexed: 12/24/2022]
|
33
|
Verma D, Tasan R, Sperk G, Pape HC. Neuropeptide Y2 receptors in anteroventral BNST control remote fear memory depending on extinction training. Neurobiol Learn Mem 2018; 149:144-153. [PMID: 29408468 DOI: 10.1016/j.nlm.2018.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 12/15/2017] [Accepted: 01/23/2018] [Indexed: 12/12/2022]
Abstract
The anterior bed nucleus of stria terminalis (BNST) is involved in reinstatement of extinguished fear, and neuropeptide Y2 receptors influence local synaptic signaling. Therefore, we hypothesized that Y2 receptors in anteroventral BNST (BNSTav) interfere with remote fear memory and that previous fear extinction is an important variable. C57BL/6NCrl mice were fear-conditioned, and a Y2 receptor-specific agonist (NPY3-36) or antagonist (JNJ-5207787) was applied in BNSTav before fear retrieval at the following day. Remote fear memory was tested on day 16 in two groups of mice, which had (experiment 1) or had not (experiment 2) undergone extinction training after conditioning. In the group with extinction training, tests of remote fear memory revealed partial retrieval of extinction, which was prevented after blockade of Y2 receptors in BNSTav. No such effect was observed in the group with no extinction training, but stimulation of Y2 receptors in BNSTav mimicked the influence of extinction during tests of remote fear memory. Pharmacological manipulation of Y2 receptors in BNSTav before fear acquisition (experiment 3) had no effect on fear memory retrieval, extinction or remote fear memory. Furthermore, partial retrieval of extinction during tests of remote fear memory was associated with changes in number of c-Fos expressing neurons in BNSTav, which was prevented or mimicked upon Y2 blockade or stimulation in BNSTav. These results indicate that Y2 receptor manipulation in BNSTav interferes with fear memory and extinction retrieval at remote stages, likely through controlling neuronal activity in BNSTav during extinction training.
Collapse
Affiliation(s)
- Dilip Verma
- Institute of Physiology 1, Westfälische Wilhelms-University, D-48149 Münster, Germany
| | - Ramon Tasan
- Institute of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Guenther Sperk
- Institute of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Hans-Christian Pape
- Institute of Physiology 1, Westfälische Wilhelms-University, D-48149 Münster, Germany.
| |
Collapse
|
34
|
Bud Craig AD. Central neural substrates involved in temperature discrimination, thermal pain, thermal comfort, and thermoregulatory behavior. HANDBOOK OF CLINICAL NEUROLOGY 2018; 156:317-338. [PMID: 30454598 DOI: 10.1016/b978-0-444-63912-7.00019-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A phylogenetically novel pathway that emerged with primate encephalization is described, which conveys high-fidelity cutaneous thermosensory activity in "labeled lines" to a somatotopic map in the dorsal posterior insular cortex. It originates in lamina I of the superficial dorsal horn and ascends by way of the lateral spinothalamic tract and a distinct region in posterolateral thalamus. It evolved from the homeostatic sensory activity that represents the physiologic (interoceptive) condition of the body and drives the central autonomic network, which underlies all affective feelings from the body. Accordingly, human discriminative thermal sensations are accompanied by thermally motivated behaviors and thermal feelings of comfort or discomfort (unless neutral), which evidence suggests are associated with activity in the insular, cingulate, and orbitofrontal cortices, respectively. Yet, the substrates for thermoregulatory behavior have not been established, and several strong candidates (including the hypothalamus and the bed nucleus of the stria terminalis) are discussed. Finally, the neural underpinnings for relationships between thermal affect and social feelings (warm-positive/cold-negative) are addressed, including the association of hyperthermia with clinical depression.
Collapse
Affiliation(s)
- Arthur D Bud Craig
- Atkinson Research Laboratory, Barrow Neurological Institute, Phoenix, AZ, United States.
| |
Collapse
|
35
|
Maruyama C, Deyama S, Nagano Y, Ide S, Kaneda K, Yoshioka M, Minami M. Suppressive effects of morphine injected into the ventral bed nucleus of the stria terminalis on the affective, but not sensory, component of pain in rats. Eur J Neurosci 2017; 47:40-47. [PMID: 29131433 DOI: 10.1111/ejn.13776] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/31/2017] [Accepted: 11/06/2017] [Indexed: 10/18/2022]
Abstract
Pain is a complex experience with both sensory and affective components. Clinical and preclinical studies have shown that the affective component of pain can be reduced by doses of morphine lower than those necessary to reduce the sensory component. Although the neural mechanisms underlying the effects of morphine on the sensory component of pain have been investigated extensively, those influencing the affective component remain to be elucidated. The bed nucleus of the stria terminalis (BNST) has been implicated in the regulation of various negative emotional states, including aversion, anxiety and fear. Thus, this study aimed to clarify the role of the ventral part of the BNST (vBNST) in the actions of morphine on the affective and sensory components of pain. First, the effects of intra-vBNST injections of morphine on intraplantar formalin-induced conditioned place aversion (CPA) and nociceptive behaviors were investigated. Intra-vBNST injections of morphine reduced CPA without affecting nociceptive behaviors, which suggests that intra-vBNST morphine alters the affective, but not sensory, component of pain. Next, to examine the effects of morphine on neuronal excitability in type II vBNST neurons, whole-cell patch-clamp recordings were performed in brain slices. Bath application of morphine hyperpolarized type II vBNST neurons. Thus, the suppressive effects of intra-vBNST morphine on pain-induced aversion may be due to its inhibitory effects on neuronal excitability in type II vBNST neurons. These results suggest that the vBNST is a key brain region involved in the suppressive effects of morphine on the affective component of pain.
Collapse
Affiliation(s)
- Chikashi Maruyama
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo, 060-0812, Japan
| | - Satoshi Deyama
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo, 060-0812, Japan.,Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yusuke Nagano
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo, 060-0812, Japan
| | - Soichiro Ide
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo, 060-0812, Japan.,Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Katsuyuki Kaneda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo, 060-0812, Japan.,Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Mitsuhiro Yoshioka
- Department of Neuropharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita 12, Nishi 6, Kita-ku, Sapporo, 060-0812, Japan
| |
Collapse
|
36
|
Functional Heterogeneity in the Bed Nucleus of the Stria Terminalis. J Neurosci 2017; 36:8038-49. [PMID: 27488624 DOI: 10.1523/jneurosci.0856-16.2016] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/05/2016] [Indexed: 11/21/2022] Open
Abstract
Early work stressed the differing involvement of the central amygdala (CeA) and bed nucleus of the stria terminalis (BNST) in the genesis of fear versus anxiety, respectively. In 2009, Walker, Miles, and Davis proposed a model of amygdala-BNST interactions to explain these functional differences. This model became extremely influential and now guides a new wave of studies on the role of BNST in humans. Here, we consider evidence for and against this model, in the process highlighting central principles of BNST organization. This analysis leads us to conclude that BNST's influence is not limited to the generation of anxiety-like responses to diffuse threats, but that it also shapes the impact of discrete threatening stimuli. It is likely that BNST-CeA interactions are involved in modulating responses to such threats. In addition, whereas current views emphasize the contributions of the anterolateral BNST region in anxiety, accumulating data indicate that the anteromedial and anteroventral regions also play a critical role. The presence of multiple functional subregions within the small volume of BNST raises significant technical obstacles for functional imaging studies in humans.
Collapse
|
37
|
Deyama S, Sugano Y, Mori S, Amano T, Yoshioka M, Kaneda K, Minami M. Activation of the NMDA receptor–neuronal nitric oxide synthase pathway within the ventral bed nucleus of the stria terminalis mediates the negative affective component of pain. Neuropharmacology 2017; 118:59-68. [DOI: 10.1016/j.neuropharm.2017.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/05/2017] [Accepted: 03/06/2017] [Indexed: 12/23/2022]
|
38
|
Reichard RA, Subramanian S, Desta MT, Sura T, Becker ML, Ghobadi CW, Parsley KP, Zahm DS. Abundant collateralization of temporal lobe projections to the accumbens, bed nucleus of stria terminalis, central amygdala and lateral septum. Brain Struct Funct 2017; 222:1971-1988. [PMID: 27704219 PMCID: PMC5378696 DOI: 10.1007/s00429-016-1321-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/28/2016] [Indexed: 10/20/2022]
Abstract
Behavioral flexibility is subserved in part by outputs from the cerebral cortex to telencephalic subcortical structures. In our earlier evaluation of the organization of the cortical-subcortical output system (Reynolds and Zahm, J Neurosci 25:11757-11767, 2005), retrograde double-labeling was evaluated in the prefrontal cortex following tracer injections into pairs of the following subcortical telencephalic structures: caudate-putamen, core and shell of the accumbens (Acb), bed nucleus of stria terminalis (BST) and central nucleus of the amygdala (CeA). The present study was done to assess patterns of retrograde labeling in the temporal lobe after similar paired tracer injections into most of the same telencephalic structures plus the lateral septum (LS). In contrast to the modest double-labeling observed in the prefrontal cortex in the previous study, up to 60-80 % of neurons in the basal and accessory basal amygdaloid nuclei and amygdalopiriform transition area exhibited double-labeling in the present study. The most abundant double-labeling was generated by paired injections into structures affiliated with the extended amygdala, including the CeA, BST and Acb shell. Injections pairing the Acb core with the BST or CeA produced significantly fewer double-labeled neurons. The ventral subiculum exhibited modest amounts of double-labeling associated with paired injections into the Acb, BST, CeA and LS. The results raise the issue of how an extraordinarily collateralized output from the temporal lobe may contribute to behavioral flexibility.
Collapse
Affiliation(s)
- Rhett A Reichard
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Suriya Subramanian
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Mikiyas T Desta
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Tej Sura
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Mary L Becker
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Comeron W Ghobadi
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Kenneth P Parsley
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA
| | - Daniel S Zahm
- Department of Pharmacological and Physiological Science, School of Medicine, Saint Louis University, 1402 S, Grand Blvd., Saint Louis, MO, 63104, USA.
| |
Collapse
|
39
|
Singh AK, Zajdel J, Mirrasekhian E, Almoosawi N, Frisch I, Klawonn AM, Jaarola M, Fritz M, Engblom D. Prostaglandin-mediated inhibition of serotonin signaling controls the affective component of inflammatory pain. J Clin Invest 2017; 127:1370-1374. [PMID: 28287401 DOI: 10.1172/jci90678] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 01/17/2017] [Indexed: 01/03/2023] Open
Abstract
Pain is fundamentally unpleasant and induces a negative affective state. The affective component of pain is mediated by circuits that are distinct from those mediating the sensory-discriminative component. Here, we have investigated the role of prostaglandins in the affective dimension of pain using a rodent pain assay based on conditioned place aversion to formalin injection, an inflammatory noxious stimulus. We found that place aversion induced by inflammatory pain depends on prostaglandin E2 that is synthesized by cyclooxygenase 2 in neural cells. Further, mice lacking the prostaglandin E2 receptor EP3 selectively on serotonergic cells or selectively in the area of the dorsal raphe nucleus failed to form an aversion to formalin-induced pain, as did mice lacking the serotonin transporter. Chemogenetic manipulations revealed that EP3 receptor activation elicited conditioned place aversion to pain via inhibition of serotonergic neurons. In contrast to their role in inflammatory pain aversion, EP3 receptors on serotonergic cells were dispensable for acute nociceptive behaviors and for aversion induced by thermal pain or a κ opioid receptor agonist. Collectively, our findings show that prostaglandin-mediated modulation of serotonergic transmission controls the affective component of inflammatory pain.
Collapse
|
40
|
Kaneko T, Kaneda K, Ohno A, Takahashi D, Hara T, Amano T, Ide S, Yoshioka M, Minami M. Activation of adenylate cyclase-cyclic AMP-protein kinase A signaling by corticotropin-releasing factor within the dorsolateral bed nucleus of the stria terminalis is involved in pain-induced aversion. Eur J Neurosci 2016; 44:2914-2924. [PMID: 27690274 DOI: 10.1111/ejn.13419] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 09/22/2016] [Accepted: 09/26/2016] [Indexed: 01/23/2023]
Abstract
Pain is a complex experience involving sensory and affective components. Although the neuronal mechanisms underlying the sensory component of pain have been extensively studied, those underlying its affective component have yet to be elucidated. Recently, we reported that corticotrophin-releasing factor (CRF)-induced depolarization in type II neurons within the dorsolateral bed nucleus of the stria terminalis (dlBNST) is critical for pain-induced aversive responses in rats. However, the intracellular signaling underlying the excitatory effects of CRF and the contribution of such signaling to the induction of pain-induced aversion remain unclear. In the present study, we addressed these issues by conducting whole-cell patch-clamp recordings in rat brain slices and by undertaking behavioral pharmacological analyses. Intracellular perfusion of protein kinase A (PKA) inhibitor Rp-cyclic adenosine monophosphorothioate (Rp-cAMPS) or KT5720 suppressed the excitatory effects of CRF in type II dlBNST neurons, and bath application of Rp-cAMPS also suppressed it. In addition, bath application of forskolin, an adenylate cyclase (AC) activator, mimicked the effects of CRF, and pretreatment with forskolin diminished the excitatory effects of CRF. Furthermore, a conditioned place aversion (CPA) test showed that co-administration of Rp-cAMPS with CRF into the dlBNST suppressed CRF-induced CPA. Intra-dlBNST injection of Rp-cAMPS also suppressed pain-induced CPA. These results suggest that CRF increases excitability of type II dlBNST neurons through activation of the AC-cAMP-PKA pathway, thereby causing pain-induced aversive responses. The present findings shed light on the neuronal mechanisms underlying the negative affective component of pain and may provide therapeutic targets for treating intractable pain accompanied by psychological factors.
Collapse
Affiliation(s)
- Tomoyuki Kaneko
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Katsuyuki Kaneda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Atsushi Ohno
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Daiki Takahashi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Taiki Hara
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Taiju Amano
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Soichiro Ide
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Mitsuhiro Yoshioka
- Department of Neuropharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| |
Collapse
|
41
|
Schmeltzer SN, Herman JP, Sah R. Neuropeptide Y (NPY) and posttraumatic stress disorder (PTSD): A translational update. Exp Neurol 2016; 284:196-210. [PMID: 27377319 PMCID: PMC8375392 DOI: 10.1016/j.expneurol.2016.06.020] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 12/12/2022]
Abstract
Posttraumatic stress disorder (PTSD) is a trauma-evoked syndrome, with variable prevalence within the human population due to individual differences in coping and resiliency. In this review, we discuss evidence supporting the relevance of neuropeptide Y (NPY), a stress regulatory transmitter in PTSD. We consolidate findings from preclinical, clinical, and translational studies of NPY that are of relevance to PTSD with an attempt to provide a current update of this area of research. NPY is abundantly expressed in forebrain limbic and brainstem areas that regulate stress and emotional behaviors. Studies in rodents demonstrate a role for NPY in stress responses, anxiety, fear, and autonomic regulation, all relevant to PTSD symptomology. Genetic studies support an association of NPY polymorphisms with stress coping and affect. Importantly, cerebrospinal fluid (CSF) measurements in combat veterans provide direct evidence of NPY association with PTSD diagnosis and symptomology. In addition, NPY involvement in pain, depression, addiction, and metabolism may be relevant to comorbidities associated with PTSD. Collectively, the literature supports the relevance of NPY to PTSD pathophysiology, although knowledge gaps remain. The NPY system is an attractive target in terms of understanding the physiological basis of PTSD as well as treatment of the disorder.
Collapse
Affiliation(s)
- Sarah N Schmeltzer
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States
| | - James P Herman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Renu Sah
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States; VA Medical Center, Cincinnati, OH, 45220, United States.
| |
Collapse
|
42
|
Challenges for the in vivo quantification of brain neuropeptides using microdialysis sampling and LC-MS. Bioanalysis 2016; 8:1965-85. [PMID: 27554986 DOI: 10.4155/bio-2016-0119] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In recent years, neuropeptides and their receptors have received an increased interest in neuropharmacological research. Although these molecules are considered relatively small compared with proteins, their in vivo quantification using microdialysis is more challenging than for small molecules. Low microdialysis recoveries, aspecific adsorption and the presence of various multiply charged precursor ions during ESI-MS/MS detection hampers the in vivo quantification of these low abundant biomolecules. Every step in the workflow, from sampling until analysis, has to be optimized to enable the sensitive analysis of these compounds in microdialysates.
Collapse
|
43
|
Waraczynski M. Toward a systems-oriented approach to the role of the extended amygdala in adaptive responding. Neurosci Biobehav Rev 2016; 68:177-194. [PMID: 27216212 DOI: 10.1016/j.neubiorev.2016.05.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 04/02/2016] [Accepted: 05/19/2016] [Indexed: 11/19/2022]
Abstract
Research into the structure and function of the basal forebrain macrostructure called the extended amygdala (EA) has recently seen considerable growth. This paper reviews that work, with the objectives of identifying underlying themes and developing a common goal towards which investigators of EA function might work. The paper begins with a brief review of the structure and the ontological and phylogenetic origins of the EA. It continues with a review of research into the role of the EA in both aversive and appetitive states, noting that these two seemingly disparate avenues of research converge on the concept of reinforcement - either negative or positive - of adaptive responding. These reviews lead to a proposal as to where the EA may fit in the organization of the basal forebrain, and an invitation to investigators to place their findings in a unifying conceptual framework of the EA as a collection of neural ensembles that mediate adaptive responding.
Collapse
Affiliation(s)
- Meg Waraczynski
- Department of Psychology, University of Wisconsin-Whitewater, 800 West Main Street, Whitewater, WI 53190, USA.
| |
Collapse
|
44
|
Tasan RO, Verma D, Wood J, Lach G, Hörmer B, de Lima TCM, Herzog H, Sperk G. The role of Neuropeptide Y in fear conditioning and extinction. Neuropeptides 2016; 55:111-26. [PMID: 26444585 DOI: 10.1016/j.npep.2015.09.007] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/10/2015] [Accepted: 09/10/2015] [Indexed: 12/23/2022]
Abstract
While anxiety disorders are the brain disorders with the highest prevalence and constitute a major burden for society, a considerable number of affected people are still treated insufficiently. Thus, in an attempt to identify potential new anxiolytic drug targets, neuropeptides have gained considerable attention in recent years. Compared to classical neurotransmitters they often have a regionally restricted distribution and may bind to several distinct receptor subtypes. Neuropeptide Y (NPY) is a highly conserved neuropeptide that is specifically concentrated in limbic brain areas and signals via at least 5 different G-protein-coupled receptors. It is involved in a variety of physiological processes including the modulation of emotional-affective behaviors. An anxiolytic and stress-reducing property of NPY is supported by many preclinical studies. Whether NPY may also interact with processing of learned fear and fear extinction is comparatively unknown. However, this has considerable relevance since pathological, inappropriate and generalized fear expression and impaired fear extinction are hallmarks of human post-traumatic stress disorder and a major reason for its treatment-resistance. Recent evidence from different laboratories emphasizes a fear-reducing role of NPY, predominantly mediated by exogenous NPY acting on Y1 receptors. Since a reduction of fear expression was also observed in Y1 receptor knockout mice, other Y receptors may be equally important. By acting on Y2 receptors, NPY promotes fear extinction and generates a long-term suppression of fear, two important preconditions that could support cognitive behavioral therapies in human patients. A similar effect has been demonstrated for the closely related pancreatic polypeptide (PP) when acting on Y4 receptors. Preliminary evidence suggests that NPY modulates fear in particular by activation of Y1 and Y2 receptors in the basolateral and central amygdala, respectively. In the basolateral amygdala, NPY signaling activates inhibitory G protein-coupled inwardly-rectifying potassium channels or suppresses hyperpolarization-induced I(h) currents in a Y1 receptor-dependent fashion, favoring a general suppression of neuronal activity. A more complex situation has been described for the central extended amygdala, where NPY reduces the frequency of inhibitory and excitatory postsynaptic currents. In particular the inhibition of long-range central amygdala output neurons may result in a Y2 receptor-dependent suppression of fear. The role of NPY in processes of learned fear and fear extinction is, however, only beginning to emerge, and multiple questions regarding the relevance of endogenous NPY and different receptor subtypes remain elusive. Y2 receptors may be of particular interest for future studies, since they are the most prominent Y receptor subtype in the human brain and thus among the most promising therapeutic drug targets when translating preclinical evidence to potential new therapies for human anxiety disorders.
Collapse
Affiliation(s)
- R O Tasan
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria.
| | - D Verma
- Institute of Physiology I, University of Münster, D-48149 Münster, Germany
| | - J Wood
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - G Lach
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria; Capes Foundation, Ministry of Education of Brazil, 70040-020 Brasília/DF, Brazil
| | - B Hörmer
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - T C M de Lima
- Department of Pharmacology, Federal University of Santa Catarina, 88049-970 Florianópolis, Brazil
| | - H Herzog
- Neuroscience Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - G Sperk
- Department of Pharmacology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
45
|
Daniel SE, Rainnie DG. Stress Modulation of Opposing Circuits in the Bed Nucleus of the Stria Terminalis. Neuropsychopharmacology 2016; 41:103-25. [PMID: 26096838 PMCID: PMC4677121 DOI: 10.1038/npp.2015.178] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/22/2015] [Accepted: 06/17/2015] [Indexed: 12/11/2022]
Abstract
The anterior bed nucleus of the stria terminalis (BNST) has been recognized as a critical structure in regulating trait anxiety, contextual fear memory, and appetitive behavior, and is known to be sensitive to stress manipulations. As one of the most complex structures in the central nervous system, the intrinsic circuitry of the BNST is largely unknown; however, recent technological developments have allowed researchers to begin to untangle the internal connections of the nucleus. This research has revealed the possibility of two opposing circuits, one anxiolytic and one anxiogenic, within the BNST, the relative strength of which determines the behavioral outcome. The balance of these pathways is critical in maintaining a normal physiological and behavioral state; however, stress and drugs of abuse can differentially affect the opposing circuitry within the nucleus to shift the balance to a pathological state. In this review, we will examine how stress interacts with the neuromodulators, corticotropin-releasing factor, norepinephrine, dopamine, and serotonin to affect the circuitry of the BNST as well as how synaptic plasticity in the BNST is modulated by stress, resulting in long-lasting changes in the circuit and behavioral state.
Collapse
Affiliation(s)
- Sarah E Daniel
- Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Donald G Rainnie
- Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
46
|
KIMURA YUSUKE, ISHIKAWA MASASHI, HORI YOKO, OKABE TADASHI, SAKAMOTO ATSUHIRO. Effect of electroconvulsive stimulation on messenger RNA expression in the prefrontal cortex in a rat pain model. Biomed Rep 2015; 3:802-806. [PMID: 26623019 PMCID: PMC4660606 DOI: 10.3892/br.2015.525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/04/2015] [Indexed: 11/05/2022] Open
Abstract
Previous reports have shown that electroconvulsive therapy (ECT) is efficacious in the treatment of neuropathic pain; however, its mechanism of action remains unclear. The present study aimed to understand these mechanisms by investigating the alterations in the expression of neuropeptide Y (NPY) and interleukin-1β (IL-1β) in the prefrontal cortex. A rat model of neuropathic pain produced by chronic constrictive injury of the sciatic nerve was used, and mechanical and thermal hyperalgesia were evaluated starting 2 days after the injury. Using a pulse generator, ECT was administered to the rodents for 6 days from days 7-12 after the injury. Thermal and mechanical stimulation were administered to assess pain thresholds. Quantitative polymerase chain reaction, used to measure gene expression levels in the prefrontal cortex, showed that NPY and IL-1β gene expression levels in the prefrontal cortex increased following the injury. The present results indicate that these gene expression level variations may be associated with the mechanisms underlying the effect of ECT in treating neuropathic pain.
Collapse
Affiliation(s)
- YUSUKE KIMURA
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - MASASHI ISHIKAWA
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - YOKO HORI
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - TADASHI OKABE
- Department of Anesthesiology, Hitachi, Ltd., Hitachinaka General Hospital, Ibaraki 312-0057, Japan
| | - ATSUHIRO SAKAMOTO
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| |
Collapse
|
47
|
Nagano Y, Kaneda K, Maruyama C, Ide S, Kato F, Minami M. Corticotropin-releasing factor enhances inhibitory synaptic transmission to type III neurons in the bed nucleus of the stria terminalis. Neurosci Lett 2015; 600:56-61. [DOI: 10.1016/j.neulet.2015.05.059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/18/2015] [Accepted: 05/29/2015] [Indexed: 11/27/2022]
|
48
|
|
49
|
Kash TL, Pleil KE, Marcinkiewcz CA, Lowery-Gionta EG, Crowley N, Mazzone C, Sugam J, Hardaway JA, McElligott ZA. Neuropeptide regulation of signaling and behavior in the BNST. Mol Cells 2015; 38:1-13. [PMID: 25475545 PMCID: PMC4314126 DOI: 10.14348/molcells.2015.2261] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 09/29/2014] [Indexed: 12/23/2022] Open
Abstract
Recent technical developments have transformed how neuroscientists can probe brain function. What was once thought to be difficult and perhaps impossible, stimulating a single set of long range inputs among many, is now relatively straight-forward using optogenetic approaches. This has provided an avalanche of data demonstrating causal roles for circuits in a variety of behaviors. However, despite the critical role that neuropeptide signaling plays in the regulation of behavior and physiology of the brain, there have been remarkably few studies demonstrating how peptide release is causally linked to behaviors. This is likely due to both the different time scale by which peptides act on and the modulatory nature of their actions. For example, while glutamate release can effectively transmit information between synapses in milliseconds, peptide release is potentially slower [See the excellent review by Van Den Pol on the time scales and mechanisms of release (van den Pol, 2012)] and it can only tune the existing signals via modulation. And while there have been some studies exploring mechanisms of release, it is still not as clearly known what is required for efficient peptide release. Furthermore, this analysis could be complicated by the fact that there are multiple peptides released, some of which may act in contrast. Despite these limitations, there are a number of groups making progress in this area. The goal of this review is to explore the role of peptide signaling in one specific structure, the bed nucleus of the stria terminalis, that has proven to be a fertile ground for peptide action.
Collapse
Affiliation(s)
- Thomas L. Kash
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Kristen E. Pleil
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Catherine A. Marcinkiewcz
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Emily G. Lowery-Gionta
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Nicole Crowley
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Christopher Mazzone
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Jonathan Sugam
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - J. Andrew Hardaway
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| | - Zoe A. McElligott
- Bowles Center for Alcohol Studies and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill,
USA
| |
Collapse
|
50
|
Su J, Tanaka Y, Muratsubaki T, Kano M, Kanazawa M, Fukudo S. Injection of corticotropin-releasing hormone into the amygdala aggravates visceral nociception and induces noradrenaline release in rats. Neurogastroenterol Motil 2015; 27:30-9. [PMID: 25359531 DOI: 10.1111/nmo.12462] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 09/26/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Corticotropin-releasing hormone (CRH) and its receptor 1 (CRH-R1) play an important role in the colonic response to stress. The central nucleus of the amygdala (CeA) is a major extrahypothalamic site that contains a large number of neurons expressing both CRH and CRH-R1. Here, we verified the hypothesis that CRH in the CeA sensitizes visceral nociception via CRH-R1 with release of noradrenaline, dopamine, and serotonin (5-HT) in the CeA. METHODS In male Wistar rats, visceral sensitivity was quantified by recording the visceromotor response to colorectal distension (CRD) with administration of vehicle, CRH, or the CRH-R1 antagonist CP-154526+ CRH or CRH-R1 antagonist CP-154526 alone into the CeA. Simultaneously, extracellular levels of noradrenaline, dopamine, and 5-HT were measured in the CeA using microdialysis. All data were obtained under restraint conditions. KEY RESULTS Administration of CRH into the CeA significantly increased the number of abdominal muscle contractions in response to CRD. CP-154526 significantly blocked the number of abdominal muscle contractions in response to CRD with the administration of CRH into the CeA. Noradrenaline in the CeA was increased by CRD, further increased by CRH, and inhibited by CRH-R1 antagonist. Dopamine in the CeA was also exaggerated by CRH but was not inhibited by CRH-R1 antagonist. 5-HT in the CeA was unchanged. CONCLUSIONS & INFERENCES These results suggest that CRH in the CeA sensitizes visceral nociception via CRH-R1 with release of noradrenaline.
Collapse
Affiliation(s)
- J Su
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | |
Collapse
|