1
|
Lorsung R, Cramer N, Alipio JB, Ji Y, Han S, Masri R, Keller A. Sex Differences in Central Amygdala Glutamate Responses to Calcitonin Gene-Related Peptide. J Neurosci 2025; 45:e1898242024. [PMID: 39663115 PMCID: PMC11714345 DOI: 10.1523/jneurosci.1898-24.2024] [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: 10/04/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024] Open
Abstract
Women are disproportionately affected by chronic pain compared with men. While societal and environmental factors contribute to this disparity, sex-based biological differences in the processing of pain are also believed to play significant roles. The central lateral nucleus of the amygdala (CeLC) is a key region for the emotional-affective dimension of pain, and a prime target for exploring sex differences in pain processing since a recent study demonstrated sex differences in CGRP actions in this region. Inputs to CeLC from the parabrachial nucleus (PB) play a causal role in aversive processing and release both glutamate and calcitonin gene-related peptide (CGRP). CGRP is thought to play a crucial role in chronic pain by potentiating glutamatergic signaling in CeLC. However, it is not known if this CGRP-mediated synaptic plasticity occurs similarly in males and females. Here, we tested the hypothesis that female CeLC neurons experience greater potentiation of glutamatergic signaling than males following endogenous CGRP exposure. Using trains of optical stimuli to evoke transient CGRP release from PB terminals in CeLC, we find that subsequent glutamatergic responses are preferentially potentiated in CeLC neurons from female mice. This potentiation was CGRP dependent and involved a postsynaptic mechanism. This sex difference in CGRP sensitivity may explain sex differences in affective pain processing.
Collapse
Affiliation(s)
- Rebecca Lorsung
- Department of Neurobiology and UM-MIND, School of Medicine, University of Maryland, Baltimore, Maryland 21201
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Nathan Cramer
- Department of Neurobiology and UM-MIND, School of Medicine, University of Maryland, Baltimore, Maryland 21201
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Jason Bondoc Alipio
- Department of Neurobiology and UM-MIND, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Yadong Ji
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, Maryland 21201
- Faculty of Dentistry, University of Jordan, Amman 11942, Jordan
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037
| | - Radi Masri
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, Maryland 21201
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, Maryland 21201
- Faculty of Dentistry, University of Jordan, Amman 11942, Jordan
| | - Asaf Keller
- Department of Neurobiology and UM-MIND, School of Medicine, University of Maryland, Baltimore, Maryland 21201
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
2
|
Lynch N, De Luca R, Spinieli RL, Rillosi E, Thomas RC, Sailesh S, Gangeddula N, Lima JD, Bandaru S, Arrigoni E, Burstein R, Thankachan S, Kaur S. Identifying the Brain Circuits that Regulate Pain-Induced Sleep Disturbances. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.20.629596. [PMID: 39763835 PMCID: PMC11702673 DOI: 10.1101/2024.12.20.629596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Pain therapies that alleviate both pain and sleep disturbances may be the most effective for pain relief, as both chronic pain and sleep loss render the opioidergic system, targeted by opioids, less sensitive and effective for analgesia. Therefore, we first studied the link between sleep disturbances and the activation of nociceptors in two acute pain models. Activation of nociceptors in both acute inflammatory (AIP) and opto-pain models led to sleep loss, decreased sleep spindle density, and increased sleep fragmentation that lasted 3 to 6 hours. This relationship is facilitated by the transmission of nociceptive signals through the spino-parabrachial pathways, converging at the wake-active PBelCGRP (parabrachial nucleus expressing Calcitonin Gene-Related Peptide) neurons, known to gate aversive stimuli. However, it has never been tested whether the targeted blocking of this wake pathway can alleviate pain-induced sleep disturbances without increasing sleepiness. Therefore, we next used selective ablations or optogenetic silencing and identified the key role played by the glutamatergic PBelCGRP in pain-induced sleep disturbances. Inactivating the PBelCGRP neurons by genetic deletion or optogenetic silencing prevented these sleep disturbances in both pain models. Furthermore, to understand the wake pathways underlying the pain-induced sleep disturbances, we silenced the PBelCGRP terminals at four key sites in the substantia innominata of the basal forebrain (SI-BF), the central nucleus of Amygdala (CeA), the bed nucleus of stria terminalis (BNST), or the lateral hypothalamus (LH). Silencing of the SI-BF and CeA also significantly reversed pain-induced sleep loss, specifically through the action on the CGRP and NMDA receptors. This was also confirmed by site-specific blockade of these receptors pharmacologically. Our results highlight the significant potential for selectively targeting the wake pathway to effectively treat pain and sleep disturbances, which will minimize risks associated with traditional analgesics.
Collapse
Affiliation(s)
- Nicole Lynch
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Roberto De Luca
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Richard L Spinieli
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Enrico Rillosi
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Renner C Thomas
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Samuel Sailesh
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Nishta Gangeddula
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Janayna D Lima
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Sathyajit Bandaru
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Elda Arrigoni
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Rami Burstein
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| | - Stephen Thankachan
- Boston VA Research Institute, Veterans Affairs Boston Healthcare System & Department of Psychiatry, Harvard Medical School, West Roxbury, MA 02132, USA
| | - Satvinder Kaur
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, 02215, USA
| |
Collapse
|
3
|
Lorsung R, Cramer N, Alipio JB, Ji Y, Han S, Masri R, Keller A. Sex differences in central amygdala glutamate responses to calcitonin gene-related peptide. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.09.622728. [PMID: 39574632 PMCID: PMC11581022 DOI: 10.1101/2024.11.09.622728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Women are disproportionately affected by chronic pain compared to men. While societal and environmental factors contribute to this disparity, sex-based biological differences in the processing of pain are also believed to play significant roles. The central lateral nucleus of the amygdala (CeLC) is a key region for the emotional-affective dimension of pain, and a prime target for exploring sex differences in pain processing since a recent study demonstrated sex differences in CGRP actions in this region. Inputs to CeLC from the parabrachial nucleus (PB) play a causal role in aversive processing, and release both glutamate and calcitonin gene-related peptide (CGRP). CGRP is thought to play a crucial role in chronic pain by potentiating glutamatergic signaling in CeLC. However, it is not known if this CGRP-mediated synaptic plasticity occurs similarly in males and females. Here, we tested the hypothesis that female CeLC neurons experience greater potentiation of glutamatergic signaling than males following endogenous CGRP exposure. Using trains of optical stimuli to evoke transient CGRP release from PB terminals in CeLC, we find that subsequent glutamatergic responses are preferentially potentiated in CeLC neurons from female mice. This potentiation was CGRP-dependent and involved a postsynaptic mechanism. This sex difference in CGRP sensitivity may explain sex differences in affective pain processing.
Collapse
Affiliation(s)
- Rebecca Lorsung
- Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201, USA
| | - Nathan Cramer
- Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201, USA
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jason Bondoc Alipio
- Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201, USA
| | - Yadong Ji
- Department of Advanced Oral Sciences and Therapeutics, School of Dentistry, University of Maryland, Baltimore, Maryland 21201, USA, and Faculty of Dentistry, University of Jordan, Amman, Jordan
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Radi Masri
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Advanced Oral Sciences and Therapeutics, School of Dentistry, University of Maryland, Baltimore, Maryland 21201, USA, and Faculty of Dentistry, University of Jordan, Amman, Jordan
| | - Asaf Keller
- Department of Neurobiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201, USA
- Center to Advance Chronic Pain Research, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
4
|
Eller MT, Frank F, Kaltseis K, Karisik A, Knoflach M, Broessner G. Novel Calcitonin Gene-Related Peptide (CGRP) Interfering Migraine Therapies and Stroke-A Review. Int J Mol Sci 2024; 25:11685. [PMID: 39519240 PMCID: PMC11546425 DOI: 10.3390/ijms252111685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Migraine and stroke are neurological disorders with significant global prevalence and impact. Recent advances in migraine therapy have focused on the calcitonin gene-related peptide (CGRP) pathway. This review examines the shared pathomechanisms between migraine and stroke, with emphasis on the role of CGRP. We analyze the current literature on CGRP's functions in cerebrovascular regulation, edema formation, neuroinflammation, and neuroprotection. CGRP acts as a potent vasodilator and plays a crucial role in trigeminovascular activation during migraine attacks. In stroke, CGRP has demonstrated neuroprotective effects by improving collateral circulation and reducing ischemia-reperfusion injury. Concerns have been raised about the potential impact of CGRP inhibitors on stroke risk and outcomes. Studies in animals suggest that CGRP receptor antagonists may worsen cerebral ischemia by impairing collateral flow. We discuss the implications of these findings for the use of CGRP-targeting therapies in migraine patients, especially those at increased risk of stroke. Additionally, we explore the complex interplay between CGRP, endothelial function, and platelet activity in both conditions. This review highlights the need for further research to elucidate the long-term cerebrovascular safety of CGRP pathway inhibitors and to identify potential subgroups of migraine patients who may be at higher risk of adverse cerebrovascular events with these novel therapies.
Collapse
Affiliation(s)
- Michael Thomas Eller
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.T.E.)
| | - Florian Frank
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.T.E.)
| | - Katharina Kaltseis
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.T.E.)
| | - Anel Karisik
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.T.E.)
- VASCage—Centre on Clinical Stroke Research, 6020 Innsbruck, Austria
| | - Michael Knoflach
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.T.E.)
- VASCage—Centre on Clinical Stroke Research, 6020 Innsbruck, Austria
| | - Gregor Broessner
- Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (M.T.E.)
| |
Collapse
|
5
|
Wulff BS, Kuhre RE, Selvaraj M, Rehfeld JF, Niss K, Fels JJ, Anna S, Raun K, Gerstenberg MK. Improved leptin sensitivity and increased soluble leptin receptor concentrations may underlie the additive effects of combining PYY [, , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , ] and exendin-4 on body weight lowering in diet-induced obese mice. Heliyon 2024; 10:e32009. [PMID: 39183855 PMCID: PMC11341243 DOI: 10.1016/j.heliyon.2024.e32009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 08/27/2024] Open
Abstract
Objective Co-treatment with long acting PYY and the GLP-1 receptor agonists has potential as an efficient obesity treatment. This study investigates whether the mechanisms behind additive reduction of food intake and weight loss depends on complementary effects in brain areas regulating food intake and if restoration of leptin sensitivity is involved. Methods Diet-induced obese (DIO) mice were co-treated with PYY(3-36) and exendin-4 (Ex4, GLP-1R agonist) for 14 days using minipumps. Leptin responsiveness was evaluated by measuring food intake and body weight after leptin injection, and gene expression profile was investigated in various of brain regions and liver. Results We show that weight loss associated with co-treatment of PYY(3-36) and Ex4 and Ex4 mono-treatment in DIO mice increased expression of several genes in area postrema (AP) known to be involved in appetite regulation and Cart, Pdyn, Bdnf and Klb were synergistically upregulated by the co-treatment. The upregulations were independent of weight loss, as shown by inclusion of a weight matched control. Moreover, PYY(3-36) and Ex4 co-treatment resulted in synergistically upregulated plasma concentrations of soluble leptin receptor (SLR) and improved sensitivity to exogenous leptin demonstrated by food intake lowering. Conclusion The study results suggest that synergistic upregulation of appetite-regulating genes in AP and improved leptin sensitivity are important mediators for the additive weight loss resulting from PYY and Ex4 co-treatment.
Collapse
Affiliation(s)
| | | | - Madhan Selvaraj
- Translational Research, Global Translation, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Jens F. Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Kristoffer Niss
- Biomarker Discovery, R&ED Digital Science and Innovation, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Johannes J. Fels
- Research Bioanalysis, Global Research Technologies, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Secher Anna
- Global Drug Discovery, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Kirsten Raun
- Global Drug Discovery, Novo Nordisk A/S, 2760, Måløv, Denmark
| | | |
Collapse
|
6
|
Sato N, Takahashi Y, Sugimura YK, Kato F. Presynaptic inhibition of excitatory synaptic transmission from the calcitonin gene-related peptide-containing parabrachial neurons to the central amygdala in mice - unexpected influence of systemic inflammation thereon. J Pharmacol Sci 2024; 154:264-273. [PMID: 38485344 DOI: 10.1016/j.jphs.2024.02.004] [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: 01/09/2024] [Accepted: 02/02/2024] [Indexed: 03/19/2024] Open
Abstract
The monosynaptic connection from the lateral parabrachial nucleus (LPB) to the central amygdala (CeA) serves as a fundamental pathway for transmitting nociceptive signals to the brain. The LPB receives nociceptive information from the dorsal horn and spinal trigeminal nucleus and sends it to the "nociceptive" CeA, which modulates pain-associated emotions and nociceptive sensitivity. To elucidate the role of densely expressed mu-opioid receptors (MORs) within this pathway, we investigated the effects of exogenously applied opioids on LPB-CeA synaptic transmission, employing optogenetics in mice expressing channelrhodopsin-2 in LPB neurons with calcitonin gene-related peptide (CGRP). A MOR agonist ([D-Ala2,N-Me-Phe4,Glycinol5]-enkephalin, DAMGO) significantly reduced the amplitude of light-evoked excitatory postsynaptic currents (leEPSCs), in a manner negatively correlated with an increase in the paired-pulse ratio. An antagonist of MORs significantly attenuated these effects. Notably, this antagonist significantly increased leEPSC amplitude when applied alone, an effect further amplified in mice subjected to lipopolysaccharide injection 2 h before brain isolation, yet not observed at the 24-h mark. We conclude that opioids could shut off the ascending nociceptive signal at the LPB-CeA synapse through presynaptic mechanisms. Moreover, this gating process might be modulated by endogenous opioids, and the innate immune system influences this modulation.
Collapse
Affiliation(s)
- Naoko Sato
- Department of Neuroscience, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan; Center for Neuroscience of Pain, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | - Yukari Takahashi
- Department of Neuroscience, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan; Center for Neuroscience of Pain, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | - Yae K Sugimura
- Department of Neuroscience, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan; Center for Neuroscience of Pain, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan
| | - Fusao Kato
- Department of Neuroscience, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan; Center for Neuroscience of Pain, The Jikei University School of Medicine, Minato-ku, Tokyo, 105-8461, Japan.
| |
Collapse
|
7
|
Torruella-Suárez ML, Neugebauer B, Flores-Felix K, Keller A, Carrasquillo Y, Cramer N. Divergent changes in PBN excitability in a mouse model of neuropathic pain. eNeuro 2024; 11:ENEURO.0416-23.2024. [PMID: 38331576 PMCID: PMC10921257 DOI: 10.1523/eneuro.0416-23.2024] [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: 10/16/2023] [Revised: 12/19/2023] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
The transition from acute to chronic pain involves maladaptive plasticity in central nociceptive pathways. Growing evidence suggests that changes within the parabrachial nucleus (PBN), an important component of the spino-parabrachio-amygdaloid pain pathway, are key contributors to the development and maintenance of chronic pain. In animal models of chronic pain, PBN neurons become sensitive to normally innocuous stimuli and responses to noxious stimuli become amplified and more often produce after-discharges that outlast the stimulus. Using ex vivo slice electrophysiology and two mouse models of neuropathic pain, sciatic cuff and chronic constriction of the infraorbital nerve (CCI-ION), we find that changes in the firing properties of PBN neurons and a shift in inhibitory synaptic transmission may underlie this phenomenon. Compared to PBN neurons from shams, a larger proportion of PBN neurons from mice with a sciatic cuff were spontaneously active at rest, and these same neurons showed increased excitability relative to shams. In contrast, quiescent PBN neurons from cuff mice were less excitable than those from shams. Despite an increase in excitability in a subset of PBN neurons, the presence of after-discharges frequently observed in vivo were largely absent ex vivo in both injury models. However, GABAB-mediated presynaptic inhibition of GABAergic terminals is enhanced in PBN neurons after CCI-ION. These data suggest that the amplified activity of PBN neurons observed in rodent models of chronic pain arise through a combination of changes in firing properties and network excitability.Significance Statement Hyperactivity of neurons in the parabrachial nucleus (PBN) is causally linked to exaggerated pain behaviors in rodent models of chronic pain but the underlying mechanisms remain unknown. Using two mouse models of neuropathic pain, we show the intrinsic properties of PBN neurons are largely unaltered following injury. However, subsets of PBN neurons become more excitable and GABAB receptor mediated suppression of inhibitory terminals is enhanced after injury. Thus, shifts in network excitability may be a contributing factor in injury induced potentiation of PBN activity.
Collapse
Affiliation(s)
- María L Torruella-Suárez
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Benjamin Neugebauer
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Krystal Flores-Felix
- Department of Neurobiology and UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland
| | - Asaf Keller
- Department of Neurobiology and UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland
| | - Yarimar Carrasquillo
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| | - Nathan Cramer
- Department of Neurobiology and UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
8
|
Torruella-Suárez ML, Neugebauer B, Flores-Felix K, Keller A, Carrasquillo Y, Cramer N. Divergent changes in PBN excitability in a mouse model of neuropathic pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.11.561891. [PMID: 37905065 PMCID: PMC10614750 DOI: 10.1101/2023.10.11.561891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
The transition from acute to chronic pain involves maladaptive plasticity in central nociceptive pathways. Growing evidence suggests that changes within the parabrachial nucleus (PBN), an important component of the spino-parabrachio-amygdaloid pain pathway, are key contributors to the development and maintenance of chronic pain. In animal models of chronic pain, PBN neurons become sensitive to normally innocuous stimuli and responses to noxious stimuli become amplified and more often produce after-discharges that outlast the stimulus. Using ex vivo slice electrophysiology and two mouse models of neuropathic pain, sciatic cuff and chronic constriction of the infraorbital nerve (CCI-ION), we find that changes in the firing properties of PBN neurons and a shift in inhibitory synaptic transmission may underlie this phenomenon. Compared to PBN neurons from shams, a larger proportion of PBN neurons from mice with a sciatic cuff were spontaneously active at rest, and these same neurons showed increased excitability relative to shams. In contrast, quiescent PBN neurons from cuff mice were less excitable than those from shams. Despite an increase in excitability in a subset of PBN neurons, the presence of after-discharges frequently observed in vivo were largely absent ex vivo in both injury models. However, GABAB-mediated presynaptic inhibition of GABAergic terminals is enhanced in PBN neurons after CCIION. These data suggest that the amplified activity of PBN neurons observed in rodent models of chronic pain arise through a combination of changes in firing properties and network excitability.
Collapse
Affiliation(s)
- María L Torruella-Suárez
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Benjamin Neugebauer
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Krystal Flores-Felix
- Department of Neurobiology and UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland
| | - Asaf Keller
- Department of Neurobiology and UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland
| | - Yarimar Carrasquillo
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, Maryland
| | - Nathan Cramer
- Department of Neurobiology and UM-MIND, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
9
|
Russo AF, Hay DL. CGRP physiology, pharmacology, and therapeutic targets: migraine and beyond. Physiol Rev 2023; 103:1565-1644. [PMID: 36454715 PMCID: PMC9988538 DOI: 10.1152/physrev.00059.2021] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP) is a neuropeptide with diverse physiological functions. Its two isoforms (α and β) are widely expressed throughout the body in sensory neurons as well as in other cell types, such as motor neurons and neuroendocrine cells. CGRP acts via at least two G protein-coupled receptors that form unusual complexes with receptor activity-modifying proteins. These are the CGRP receptor and the AMY1 receptor; in rodents, additional receptors come into play. Although CGRP is known to produce many effects, the precise molecular identity of the receptor(s) that mediates CGRP effects is seldom clear. Despite the many enigmas still in CGRP biology, therapeutics that target the CGRP axis to treat or prevent migraine are a bench-to-bedside success story. This review provides a contextual background on the regulation and sites of CGRP expression and CGRP receptor pharmacology. The physiological actions of CGRP in the nervous system are discussed, along with updates on CGRP actions in the cardiovascular, pulmonary, gastrointestinal, immune, hematopoietic, and reproductive systems and metabolic effects of CGRP in muscle and adipose tissues. We cover how CGRP in these systems is associated with disease states, most notably migraine. In this context, we discuss how CGRP actions in both the peripheral and central nervous systems provide a basis for therapeutic targeting of CGRP in migraine. Finally, we highlight potentially fertile ground for the development of additional therapeutics and combinatorial strategies that could be designed to modulate CGRP signaling for migraine and other diseases.
Collapse
Affiliation(s)
- Andrew F Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
- Department of Neurology, University of Iowa, Iowa City, Iowa
- Center for the Prevention and Treatment of Visual Loss, Department of Veterans Affairs Health Center, Iowa City, Iowa
| | - Debbie L Hay
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
10
|
Allen HN, Chaudhry S, Hong VM, Lewter LA, Sinha GP, Carrasquillo Y, Taylor BK, Kolber BJ. A Parabrachial-to-Amygdala Circuit That Determines Hemispheric Lateralization of Somatosensory Processing. Biol Psychiatry 2023; 93:370-381. [PMID: 36473754 PMCID: PMC9852076 DOI: 10.1016/j.biopsych.2022.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 09/07/2022] [Accepted: 09/10/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND The central amygdala (CeA) is a bilateral hub of pain and emotional processing with well-established functional lateralization. We reported that optogenetic manipulation of neural activity in the left and right CeA has opposing effects on bladder pain. METHODS To determine the influence of calcitonin gene-related peptide (CGRP) signaling from the parabrachial nucleus on this diametrically opposed lateralization, we administered CGRP and evaluated the activity of CeA neurons in acute brain slices as well as the behavioral signs of bladder pain in the mouse. RESULTS We found that CGRP increased firing in both the right and left CeA neurons. Furthermore, we found that CGRP administration in the right CeA increased behavioral signs of bladder pain and decreased bladder pain-like behavior when administered in the left CeA. CONCLUSIONS These studies reveal a parabrachial-to-amygdala circuit driven by opposing actions of CGRP that determines hemispheric lateralization of visceral pain.
Collapse
Affiliation(s)
- Heather N Allen
- Department of Biological Sciences, Duquesne University, Pittsburgh, Pennsylvania; Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, Texas; Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Anesthesiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sarah Chaudhry
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Veronica M Hong
- Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, Texas
| | - Lakeisha A Lewter
- Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, Texas
| | - Ghanshyam P Sinha
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yarimar Carrasquillo
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, Maryland
| | - Bradley K Taylor
- Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Anesthesiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Benedict J Kolber
- Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, Texas.
| |
Collapse
|
11
|
Wang Y, Krabbe S, Eddison M, Henry FE, Fleishman G, Lemire AL, Wang L, Korff W, Tillberg PW, Lüthi A, Sternson SM. Multimodal mapping of cell types and projections in the central nucleus of the amygdala. eLife 2023; 12:e84262. [PMID: 36661218 PMCID: PMC9977318 DOI: 10.7554/elife.84262] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
The central nucleus of the amygdala (CEA) is a brain region that integrates external and internal sensory information and executes innate and adaptive behaviors through distinct output pathways. Despite its complex functions, the diversity of molecularly defined neuronal types in the CEA and their contributions to major axonal projection targets have not been examined systematically. Here, we performed single-cell RNA-sequencing (scRNA-seq) to classify molecularly defined cell types in the CEA and identified marker genes to map the location of these neuronal types using expansion-assisted iterative fluorescence in situ hybridization (EASI-FISH). We developed new methods to integrate EASI-FISH with 5-plex retrograde axonal labeling to determine the spatial, morphological, and connectivity properties of ~30,000 molecularly defined CEA neurons. Our study revealed spatiomolecular organization of the CEA, with medial and lateral CEA associated with distinct molecularly defined cell families. We also found a long-range axon projection network from the CEA, where target regions receive inputs from multiple molecularly defined cell types. Axon collateralization was found primarily among projections to hindbrain targets, which are distinct from forebrain projections. This resource reports marker gene combinations for molecularly defined cell types and axon-projection types, which will be useful for selective interrogation of these neuronal populations to study their contributions to the diverse functions of the CEA.
Collapse
Affiliation(s)
- Yuhan Wang
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Sabine Krabbe
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Mark Eddison
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Fredrick E Henry
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Greg Fleishman
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Andrew L Lemire
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Lihua Wang
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Wyatt Korff
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Paul W Tillberg
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Andreas Lüthi
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Scott M Sternson
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
- Howard Hughes Medical Institute & Department of Neurosciences, University of California, San DiegoSan DiegoUnited States
| |
Collapse
|
12
|
Chou TM, Lee ZF, Wang SJ, Lien CC, Chen SP. CGRP-dependent sensitization of PKC-δ positive neurons in central amygdala mediates chronic migraine. J Headache Pain 2022; 23:157. [PMID: 36510143 PMCID: PMC9746101 DOI: 10.1186/s10194-022-01531-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND To investigate specific brain regions and neural circuits that are responsible for migraine chronification. METHODS We established a mouse model of chronic migraine with intermittent injections of clinically-relevant dose of nitroglycerin (0.1 mg/kg for 9 days) and validated the model with cephalic and extracephalic mechanical sensitivity, calcitonin gene-related peptide (CGRP) expression in trigeminal ganglion, and responsiveness to sumatriptan or central CGRP blockade. We explored the neurons that were sensitized along with migraine chronification and investigated their roles on migraine phenotypes with chemogenetics. RESULTS After repetitive nitroglycerin injections, mice displayed sustained supraorbital and hind paw mechanical hyperalgesia, which lasted beyond discontinuation of nitroglycerin infusion and could be transiently reversed by sumatriptan. The CGRP expression in trigeminal ganglion was also upregulated. We found the pERK positive cells were significantly increased in the central nucleus of the amygdala (CeA), and these sensitized cells in the CeA were predominantly protein kinase C-delta (PKC-δ) positive neurons co-expressing CGRP receptors. Remarkably, blockade of the parabrachial nucleus (PBN)-CeA CGRP neurotransmission by CGRP8-37 microinjection to the CeA attenuated the sustained cephalic and extracephalic mechanical hyperalgesia. Furthermore, chemogenetic silencing of the sensitized CeA PKC-δ positive neurons reversed the mechanical hyperalgesia and CGRP expression in the trigeminal ganglion. In contrast, repetitive chemogenetic activation of the CeA PKC-δ positive neurons recapitulated chronic migraine-like phenotypes in naïve mice. CONCLUSIONS Our data suggest that CeA PKC-δ positive neurons innervated by PBN CGRP positive neurons might contribute to the chronification of migraine, which may serve as future therapeutic targets for chronic migraine.
Collapse
Affiliation(s)
- Tse-Ming Chou
- grid.260539.b0000 0001 2059 7017Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.28665.3f0000 0001 2287 1366Interdisciplinary Neuroscience Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 115 Taiwan
| | - Zhung-Fu Lee
- grid.260539.b0000 0001 2059 7017Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.39382.330000 0001 2160 926XDevelopment, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030 USA
| | - Shuu-Jiun Wang
- grid.260539.b0000 0001 2059 7017Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.260539.b0000 0001 2059 7017Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.260539.b0000 0001 2059 7017College of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.278247.c0000 0004 0604 5314Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, 112 Taiwan
| | - Cheng-Chang Lien
- grid.260539.b0000 0001 2059 7017Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.28665.3f0000 0001 2287 1366Interdisciplinary Neuroscience Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 115 Taiwan ,grid.260539.b0000 0001 2059 7017Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan
| | - Shih-Pin Chen
- grid.28665.3f0000 0001 2287 1366Interdisciplinary Neuroscience Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 115 Taiwan ,grid.260539.b0000 0001 2059 7017Brain Research Center, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.260539.b0000 0001 2059 7017College of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.278247.c0000 0004 0604 5314Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, 112 Taiwan ,grid.260539.b0000 0001 2059 7017Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,grid.278247.c0000 0004 0604 5314Division of Translational Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, 112 Taiwan
| |
Collapse
|
13
|
Presto P, Neugebauer V. Sex Differences in CGRP Regulation and Function in the Amygdala in a Rat Model of Neuropathic Pain. Front Mol Neurosci 2022; 15:928587. [PMID: 35726298 PMCID: PMC9206543 DOI: 10.3389/fnmol.2022.928587] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/12/2022] [Indexed: 12/02/2022] Open
Abstract
The amygdala has emerged as a key player in the emotional response to pain and pain modulation. The lateral and capsular regions of the central nucleus of the amygdala (CeA) represent the “nociceptive amygdala” due to their high content of neurons that process pain-related information. These CeA divisions are the targets of the spino-parabrachio-amygdaloid pain pathway, which is the predominant source of calcitonin gene-related peptide (CGRP) within the amygdala. Changes in lateral and capsular CeA neurons have previously been observed in pain models, and synaptic plasticity in these areas has been linked to pain-related behavior. CGRP has been demonstrated to play an important role in peripheral and spinal mechanisms, and in pain-related amygdala plasticity in male rats in an acute arthritis pain model. However, the role of CGRP in chronic neuropathic pain-related amygdala function and behaviors remains to be determined for both male and female rats. Here we tested the hypothesis that the CGRP1 receptor is involved in neuropathic pain-related amygdala activity, and that blockade of this receptor can inhibit neuropathic pain behaviors in both sexes. CGRP mRNA expression levels in the CeA of male rats were upregulated at the acute stage of the spinal nerve ligation (SNL) model of neuropathic pain, whereas female rats had significantly higher CGRP and CGRP receptor component expression at the chronic stage. A CGRP1 receptor antagonist (CGRP 8-37) administered into the CeA in chronic neuropathic rats reduced mechanical hypersensitivity (von Frey and paw compression tests) in both sexes but showed female-predominant effects on emotional-affective responses (ultrasonic vocalizations) and anxiety-like behaviors (open field test). CGRP 8-37 inhibited the activity of CeA output neurons assessed with calcium imaging in brain slices from chronic neuropathic pain rats. Together, these findings may suggest that CGRP1 receptors in the CeA are involved in neuropathic pain-related amygdala activity and contribute to sensory aspects in both sexes but to emotional-affective pain responses predominantly in females. The sexually dimorphic function of CGRP in the amygdala would make CGRP1 receptors a potential therapeutic target for neuropathic pain relief, particularly in females in chronic pain conditions.
Collapse
Affiliation(s)
- Peyton Presto
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- *Correspondence: Volker Neugebauer
| |
Collapse
|
14
|
Seidel MF, Hügle T, Morlion B, Koltzenburg M, Chapman V, MaassenVanDenBrink A, Lane NE, Perrot S, Zieglgänsberger W. Neurogenic inflammation as a novel treatment target for chronic pain syndromes. Exp Neurol 2022; 356:114108. [PMID: 35551902 DOI: 10.1016/j.expneurol.2022.114108] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 11/24/2022]
Abstract
Chronic pain syndrome is a heterogeneous group of diseases characterized by several pathological mechanisms. One in five adults in Europe may experience chronic pain. In addition to the individual burden, chronic pain has a significant societal impact because of work and school absences, loss of work, early retirement, and high social and healthcare costs. Several anti-inflammatory treatments are available for patients with inflammatory or autoimmune diseases to control their symptoms, including pain. However, patients with degenerative chronic pain conditions, some with 10-fold or more elevated incidence relative to these manageable diseases, have few long-term pharmacological treatment options, limited mainly to non-steroidal anti-inflammatory drugs or opioids. For this review, we performed multiple PubMed searches using keywords such as "pain," "neurogenic inflammation," "NGF," "substance P," "nociception," "BDNF," "inflammation," "CGRP," "osteoarthritis," and "migraine." Many treatments, most with limited scientific evidence of efficacy, are available for the management of chronic pain through a trial-and-error approach. Although basic science and pre-clinical pain research have elucidated many biomolecular mechanisms of pain and identified promising novel targets, little of this work has translated into better clinical management of these conditions. This state-of-the-art review summarizes concepts of chronic pain syndromes and describes potential novel treatment strategies.
Collapse
Affiliation(s)
- Matthias F Seidel
- Department of Rheumatology, Spitalzentrum Biel-Centre Hospitalier Bienne, 2501 Biel-Bienne, Switzerland.
| | - Thomas Hügle
- Department of Rheumatology, University Hospital Lausanne, 1011 Lausanne, Switzerland
| | - Barton Morlion
- The Leuven Center for Algology and Pain Management, University of Leuven, Leuven, Belgium
| | - Martin Koltzenburg
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Clinical Neurophysiology, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Victoria Chapman
- Pain Centre Versus Arthritis, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom
| | - Antoinette MaassenVanDenBrink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Nancy E Lane
- Center for Musculoskeletal Health, University of California Davis School of Medicine, Sacramento, CA, USA; Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Serge Perrot
- Unité INSERM U987, Hôpital Ambroise Paré, Paris Descartes University, Boulogne Billancourt, France; Centre d'Evaluation et Traitement de la Douleur, Hôpital Cochin, Paris Descartes University, Paris, France
| | | |
Collapse
|
15
|
Zieglgänsberger W, Brenneisen R, Berthele A, Wotjak CT, Bandelow B, Tölle TR, Lutz B. Chronic Pain and the Endocannabinoid System: Smart Lipids - A Novel Therapeutic Option? Med Cannabis Cannabinoids 2022; 5:61-75. [PMID: 35702403 PMCID: PMC9149512 DOI: 10.1159/000522432] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 01/21/2022] [Indexed: 08/05/2023] Open
Abstract
The development of a high-end cannabinoid-based therapy is the result of intense translational research, aiming to convert recent discoveries in the laboratory into better treatments for patients. Novel compounds and new regimes for drug treatment are emerging. Given that previously unreported signaling mechanisms for cannabinoids have been uncovered, clinical studies detailing their high therapeutic potential are mandatory. The advent of novel genomic, optogenetic, and viral tracing and imaging techniques will help to further detail therapeutically relevant functional and structural features. An evolutionarily highly conserved group of neuromodulatory lipids, their receptors, and anabolic and catabolic enzymes are involved in a remarkable variety of physiological and pathological processes and has been termed the endocannabinoid system (ECS). A large body of data has emerged in recent years, pointing to a crucial role of this system in the regulation of the behavioral domains of acquired fear, anxiety, and stress-coping. Besides neurons, also glia cells and components of the immune system can differentially fine-tune patterns of neuronal activity. Dysregulation of ECS signaling can lead to a lowering of stress resilience and increased incidence of psychiatric disorders. Chronic pain may be understood as a disease process evoked by fear-conditioned nociceptive input and appears as the dark side of neuronal plasticity. By taking a toll on every part of your life, this abnormal persistent memory of an aversive state can be more damaging than its initial experience. All strategies for the treatment of chronic pain conditions must consider stress-related comorbid conditions since cognitive factors such as beliefs, expectations, and prior experience (memory of pain) are key modulators of the perception of pain. The anxiolytic and anti-stress effects of medical cannabinoids can substantially modulate the efficacy and tolerability of therapeutic interventions and will help to pave the way to a successful multimodal therapy. Why some individuals are more susceptible to the effects of stress remains to be uncovered. The development of personalized prevention or treatment strategies for anxiety and depression related to chronic pain must also consider gender differences. An emotional basis of chronic pain opens a new horizon of opportunities for developing treatment strategies beyond the repeated sole use of acutely acting analgesics. A phase I trial to determine the pharmacokinetics, psychotropic effects, and safety profile of a novel nanoparticle-based cannabinoid spray for oromucosal delivery highlights a remarkable innovation in galenic technology and urges clinical studies further detailing the huge therapeutic potential of medical cannabis (Lorenzl et al.; this issue).
Collapse
Affiliation(s)
| | | | | | | | - Borwin Bandelow
- Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany
| | | | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
16
|
Tian R, Zhang Y, Pan Q, Wang Y, Wen Q, Fan X, Qin G, Zhang D, Chen L, Zhang Y, Zhou J. Calcitonin gene-related peptide receptor antagonist BIBN4096BS regulates synaptic transmission in the vestibular nucleus and improves vestibular function via PKC/ERK/CREB pathway in an experimental chronic migraine rat model. J Headache Pain 2022; 23:35. [PMID: 35260079 PMCID: PMC8903578 DOI: 10.1186/s10194-022-01403-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023] Open
Abstract
Background Vestibular symptoms are frequently reported in patients with chronic migraine (CM). However, whether vestibular symptoms arise through overlapping neurobiology of migraine remains to be elucidated. The neuropeptide calcitonin gene-related peptide (CGRP) and CGRP1 receptor play important pathological roles in facilitating central sensitization in CM. Therefore, we aimed to investigate whether CGRP1 receptor contributes to vestibular dysfunction after CM by improving synaptic transmission in the vestibular nucleus (VN). Methods A CM rat model was established by recurrent intermittent administration of nitroglycerin (NTG). Migraine- and vestibular-related behaviors were assessed. CGRP1 receptor specific antagonist, BIBN4096BS, and protein kinase C (PKC) inhibitor chelerythrine chloride (CHE) were administered intracerebroventricularly. The expressions of CGRP and CGRP1 receptor components, calcitonin receptor-like receptor (CLR) and receptor activity modifying protein 1 (RAMP1) were evaluated by western blot, immunofluorescent staining and quantitative real-time polymerase chain reaction in the vestibular nucleus (VN). Synaptic associated proteins and synaptic morphological characteristics were explored by western blot, transmission electron microscope, and Golgi-cox staining. The expressions of PKC, phosphorylated extracellular signal regulated kinase (p-ERK), phosphorylated cAMP response element-binding protein at serine 133 site (p-CREB-S133) and c-Fos were detected using western blot or immunofluorescent staining. Results The expressions of CGRP, CLR and RAMP1 were significantly upregulated in CM rats. CLR and RAMP1 were expressed mainly in neurons. BIBN4096BS treatment and PKC inhibition alleviated mechanical allodynia, thermal hyperalgesia and vestibular dysfunction in CM rats. Additionally, BIBN4096BS treatment and PKC inhibition markedly inhibited the overexpression of synaptic associated proteins and restored the abnormal synaptic structure in VN after CM. Furthermore, BIBN4096BS treatment dysregulated the expression levels of PKC, p-ERK and p-CREB-S133, and attenuated neuronal activation in VN after CM. Conclusions The present study demonstrated that CGRP1 receptor inhibition improved vestibular function after CM by reversing the aberrant synaptic transmission via downregulating PKC/ERK/CREB signaling pathway. Therapeutic interventions by inhibiting CGRP/CGRP1 signaling may be a new target for the treatment of vestibular symptoms in CM.
Collapse
|
17
|
Mercer Lindsay N, Chen C, Gilam G, Mackey S, Scherrer G. Brain circuits for pain and its treatment. Sci Transl Med 2021; 13:eabj7360. [PMID: 34757810 DOI: 10.1126/scitranslmed.abj7360] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Nicole Mercer Lindsay
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Biology, CNC Program, Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Chong Chen
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gadi Gilam
- Division of Pain Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sean Mackey
- Division of Pain Medicine, Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,New York Stem Cell Foundation-Robertson Investigator, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
18
|
Jaramillo AA, Brown JA, Winder DG. Danger and distress: Parabrachial-extended amygdala circuits. Neuropharmacology 2021; 198:108757. [PMID: 34461068 DOI: 10.1016/j.neuropharm.2021.108757] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/04/2021] [Accepted: 08/18/2021] [Indexed: 12/21/2022]
Abstract
Our understanding of the role of the parabrachial nucleus (PBN) has evolved as technology has advanced, in part due to cell-specific studies and complex behavioral assays. This is reflected in the heterogeneous neuronal populations within the PBN to the extended amygdala (EA) circuits which encompass the bed nucleus of the stria terminalis (BNST) and central amygdala (CeA) circuitry, as they differentially modulate aspects of behavior in response to diverse threat-like contexts necessary for survival. Here we review how the PBN→CeA and PBN→BNST pathways differentially modulate fear-like behavior, innate and conditioned, through unique changes in neurotransmission in response to stress-inducing contexts. Furthermore, we hypothesize how in specific instances the PBN→CeA and PBN→BNST circuits are redundant and in part intertwined with their respective reciprocal projections. By deconstructing the interoceptive and exteroceptive components of affect- and stress related behavioral paradigms, evidence suggests that the PBN→CeA circuit modulates innate response to physical stimuli and fear conditioning. Conversely, the PBN→BNST circuit modulates distress-like stress in unpredictable contexts. Thereby, the PBN provides a pathway for alarming interoceptive and exteroceptive stimuli to be processed and relayed to the EA to induce stress-relevant affect. Additionally, we provide a framework for future studies to detail the cell-type specific intricacies of PBN→EA circuits in mediating behavioral responses to threats, and the relevance of the PBN in drug-use as it relates to threat and negative reinforcement. This article is part of the special Issue on 'Neurocircuitry Modulating Drug and Alcohol Abuse'.
Collapse
Affiliation(s)
- A A Jaramillo
- Vanderbilt University School of Medicine, Nashville, TN, USA; Dept. Mol. Phys. & Biophysics, USA; Vanderbilt Brain Institute, USA; Vanderbilt Center for Addiction Research, USA
| | - J A Brown
- Vanderbilt University School of Medicine, Nashville, TN, USA; Dept. Mol. Phys. & Biophysics, USA; Vanderbilt Brain Institute, USA; Vanderbilt Center for Addiction Research, USA; Department of Pharmacology, USA
| | - D G Winder
- Vanderbilt University School of Medicine, Nashville, TN, USA; Dept. Mol. Phys. & Biophysics, USA; Vanderbilt Brain Institute, USA; Vanderbilt Center for Addiction Research, USA; Department of Pharmacology, USA; Vanderbilt Kennedy Center, USA; Department of Psychiatry & Behavioral Sciences, USA.
| |
Collapse
|
19
|
Sugimoto M, Takahashi Y, Sugimura YK, Tokunaga R, Yajima M, Kato F. Active role of the central amygdala in widespread mechanical sensitization in rats with facial inflammatory pain. Pain 2021; 162:2273-2286. [PMID: 33900711 PMCID: PMC8280967 DOI: 10.1097/j.pain.0000000000002224] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/01/2021] [Accepted: 01/26/2021] [Indexed: 01/19/2023]
Abstract
ABSTRACT Widespread or ectopic sensitization is a hallmark symptom of chronic pain, characterized by aberrantly enhanced pain sensitivity in multiple body regions remote from the site of original injury or inflammation. The central mechanism underlying widespread sensitization remains unidentified. The central nucleus of the amygdala (also called the central amygdala, CeA) is well situated for this role because it receives nociceptive information from diverse body sites and modulates pain sensitivity in various body regions. In this study, we examined the role of the CeA in a novel model of ectopic sensitization of rats. Injection of formalin into the left upper lip resulted in latent bilateral sensitization in the hind paw lasting >13 days in male Wistar rats. Chemogenetic inhibition of gamma-aminobutyric acid-ergic neurons or blockade of calcitonin gene-related peptide receptors in the right CeA, but not in the left, significantly attenuated this sensitization. Furthermore, chemogenetic excitation of gamma-aminobutyric acid-ergic neurons in the right CeA induced de novo bilateral hind paw sensitization in the rats without inflammation. These results indicate that the CeA neuronal activity determines hind paw tactile sensitivity in rats with remote inflammatory pain. They also suggest that the hind paw sensitization used in a large number of preclinical studies might not be simply a sign of the pain at the site of injury but rather a representation of the augmented CeA activity resulting from inflammation/pain in any part of the body or from activities of other brain regions, which has an active role of promoting defensive/protective behaviors to avoid further bodily damage.
Collapse
Affiliation(s)
- Mariko Sugimoto
- Center for Neuroscience of Pain and Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
- Department of Anesthesiology, Teikyo University School of Medicine, Tokyo, Japan
| | - Yukari Takahashi
- Center for Neuroscience of Pain and Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
| | - Yae K. Sugimura
- Center for Neuroscience of Pain and Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
| | - Ryota Tokunaga
- Center for Neuroscience of Pain and Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
| | - Manami Yajima
- Center for Neuroscience of Pain and Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
- Department of Dental Anesthesiology, School of Dental Medicine, Tsurumi University, Yokohama, Japan
| | - Fusao Kato
- Center for Neuroscience of Pain and Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
Shibata M, Tang C. Implications of Transient Receptor Potential Cation Channels in Migraine Pathophysiology. Neurosci Bull 2021; 37:103-116. [PMID: 32870468 PMCID: PMC7811976 DOI: 10.1007/s12264-020-00569-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022] Open
Abstract
Migraine is a common and debilitating headache disorder. Although its pathogenesis remains elusive, abnormal trigeminal and central nervous system activity is likely to play an important role. Transient receptor potential (TRP) channels, which transduce noxious stimuli into pain signals, are expressed in trigeminal ganglion neurons and brain regions closely associated with the pathophysiology of migraine. In the trigeminal ganglion, TRP channels co-localize with calcitonin gene-related peptide, a neuropeptide crucially implicated in migraine pathophysiology. Many preclinical and clinical data support the roles of TRP channels in migraine. In particular, activation of TRP cation channel V1 has been shown to regulate calcitonin gene-related peptide release from trigeminal nerves. Intriguingly, several effective anti-migraine therapies, including botulinum neurotoxin type A, affect the functions of TRP cation channels. Here, we discuss currently available data regarding the roles of major TRP cation channels in the pathophysiology of migraine and the therapeutic applicability thereof.
Collapse
Affiliation(s)
- Mamoru Shibata
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
- Department of Neurology, Tokyo Dental College Ichikawa General Hospital, Chiba, 272-8513, Japan.
| | - Chunhua Tang
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| |
Collapse
|
21
|
Shibata M. [Novel migraine treatment with CGRP-related monoclonal antibodies]. Rinsho Shinkeigaku 2020; 60:668-676. [PMID: 32893246 DOI: 10.5692/clinicalneurol.cn-001469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Migraine is a common and debilitating neurological disorder characterized by recurrent attacks of moderate to severe throbbing headache accompanied by nausea, vomiting and photophobia/phonophobia. Because of its high prevalence, migraine causes a considerable financial burden on the society as well as impaired quality of life in individual patients. Scientific evidence shows that migraine is a quite complex neurological disorder that involves not only the trigeminovascular and autonomic systems but also the hypothalamus and cerebral cortex. Calcitonin gene-related peptide (CGRP) was originally discovered as a 37-amino acid neuropeptide derived from a calcitonin gene splicing variant. CGRP is found to be expressed in trigeminal ganglion neurons. Much attention has been attracted to this molecule since CGRP was found to be released from trigeminal terminals in animal migraine models. Subsequent studies demonstrated that CGRP administration induced migraine-like headache attacks specifically in migraineurs, thus highlighting a pivotal role of CGRP in the development of migraine attacks. Several CGRP receptor antagonists were shown to be efficacious for the treatment of acute migraine. Among them, telcagepant, was shown to exert a significant migraine prophylactic action as well. Nevertheless, the development of most of these agents were discontinued due to hepatotoxicity. Currently, newer CGRP receptor antagonists are being developed. On the other hand, monoclonal antibodies targeting CGRP and its receptor showed consistent efficacy for migraine prophylaxis with excellent safety profiles in Phase III clinical trials. Furthermore, emerging data support the long-term safety and efficacy of these antibodies. In this review article, the development and perspective of anti-migraine therapeutic strategies using CGRP-related antibodies are discussed.
Collapse
Affiliation(s)
- Mamoru Shibata
- Department of Neurology, Tokyo Dental College Ichikawa General Hospital
| |
Collapse
|
22
|
Bowen AJ, Chen JY, Huang YW, Baertsch NA, Park S, Palmiter RD. Dissociable control of unconditioned responses and associative fear learning by parabrachial CGRP neurons. eLife 2020; 9:e59799. [PMID: 32856589 PMCID: PMC7556873 DOI: 10.7554/elife.59799] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022] Open
Abstract
Parabrachial CGRP neurons receive diverse threat-related signals and contribute to multiple phases of adaptive threat responses in mice, with their inactivation attenuating both unconditioned behavioral responses to somatic pain and fear-memory formation. Because CGRPPBN neurons respond broadly to multi-modal threats, it remains unknown how these distinct adaptive processes are individually engaged. We show that while three partially separable subsets of CGRPPBN neurons broadly collateralize to their respective downstream partners, individual projections accomplish distinct functions: hypothalamic and extended amygdalar projections elicit assorted unconditioned threat responses including autonomic arousal, anxiety, and freezing behavior, while thalamic and basal forebrain projections generate freezing behavior and, unexpectedly, contribute to associative fear learning. Moreover, the unconditioned responses generated by individual projections are complementary, with simultaneous activation of multiple sites driving profound freezing behavior and bradycardia that are not elicited by any individual projection. This semi-parallel, scalable connectivity schema likely contributes to flexible control of threat responses in unpredictable environments.
Collapse
Affiliation(s)
- Anna J Bowen
- Department of Biochemistry, University of WashingtonSeattleUnited States
- Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
- Graduate Program in Neuroscience, University of WashingtonSeattleUnited States
| | - Jane Y Chen
- Department of Biochemistry, University of WashingtonSeattleUnited States
- Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
| | - Y Waterlily Huang
- Department of Biochemistry, University of WashingtonSeattleUnited States
- Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
| | - Nathan A Baertsch
- Center for Integrative Brain Research, Seattle Children’s Research InstituteSeattleUnited States
| | - Sekun Park
- Department of Biochemistry, University of WashingtonSeattleUnited States
- Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
| | - Richard D Palmiter
- Department of Biochemistry, University of WashingtonSeattleUnited States
- Howard Hughes Medical Institute, University of WashingtonSeattleUnited States
| |
Collapse
|
23
|
Allen HN, Bobnar HJ, Kolber BJ. Left and right hemispheric lateralization of the amygdala in pain. Prog Neurobiol 2020; 196:101891. [PMID: 32730859 DOI: 10.1016/j.pneurobio.2020.101891] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/29/2020] [Accepted: 07/22/2020] [Indexed: 02/04/2023]
Abstract
Hemispheric asymmetries within the brain have been identified across taxa and have been extensively studied since the early 19th century. Here, we discuss lateralization of a brain structure, the amygdala, and how this lateralization is reshaping how we understand the role of the amygdala in pain processing. The amygdala is an almond-shaped, bilateral brain structure located within the limbic system. Historically, the amygdala was known to have a role in the processing of emotions and attaching emotional valence to memories and other experiences. The amygdala has been extensively studied in fear conditioning and affect but recently has been shown to have an important role in processing noxious information and impacting pain. The amygdala is composed of multiple nuclei; of special interest is the central nucleus of the amygdala (CeA). The CeA receives direct nociceptive inputs from the parabrachial nucleus (PBN) through the spino-parabrachio-amygdaloid pathway as well as more highly processed cortical and thalamic input via the lateral and basolateral amygdala. Although the amygdala is a bilateral brain region, most data investigating the amygdala's role in pain have been generated from the right CeA, which has an overwhelmingly pro-nociceptive function across pain models. The left CeA has often been characterized to have no effect on pain modulation, a dampened pro-nociceptive function, or most recently an anti-nociceptive function. This review explores the current literature on CeA lateralization and the hemispheres' respective roles in the processing and modulation of different forms of pain.
Collapse
Affiliation(s)
- Heather N Allen
- Department of Biological Sciences and Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA, 15282, United States
| | - Harley J Bobnar
- Department of Biological Sciences and Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA, 15282, United States
| | - Benedict J Kolber
- Department of Biological Sciences and Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA, 15282, United States; Department of Neuroscience and Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, 75080, United States.
| |
Collapse
|
24
|
Kuner R, Kuner T. Cellular Circuits in the Brain and Their Modulation in Acute and Chronic Pain. Physiol Rev 2020; 101:213-258. [PMID: 32525759 DOI: 10.1152/physrev.00040.2019] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic, pathological pain remains a global health problem and a challenge to basic and clinical sciences. A major obstacle to preventing, treating, or reverting chronic pain has been that the nature of neural circuits underlying the diverse components of the complex, multidimensional experience of pain is not well understood. Moreover, chronic pain involves diverse maladaptive plasticity processes, which have not been decoded mechanistically in terms of involvement of specific circuits and cause-effect relationships. This review aims to discuss recent advances in our understanding of circuit connectivity in the mammalian brain at the level of regional contributions and specific cell types in acute and chronic pain. A major focus is placed on functional dissection of sub-neocortical brain circuits using optogenetics, chemogenetics, and imaging technological tools in rodent models with a view towards decoding sensory, affective, and motivational-cognitive dimensions of pain. The review summarizes recent breakthroughs and insights on structure-function properties in nociceptive circuits and higher order sub-neocortical modulatory circuits involved in aversion, learning, reward, and mood and their modulation by endogenous GABAergic inhibition, noradrenergic, cholinergic, dopaminergic, serotonergic, and peptidergic pathways. The knowledge of neural circuits and their dynamic regulation via functional and structural plasticity will be beneficial towards designing and improving targeted therapies.
Collapse
Affiliation(s)
- Rohini Kuner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; and Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Thomas Kuner
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; and Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
25
|
Neugebauer V, Mazzitelli M, Cragg B, Ji G, Navratilova E, Porreca F. Amygdala, neuropeptides, and chronic pain-related affective behaviors. Neuropharmacology 2020; 170:108052. [PMID: 32188569 DOI: 10.1016/j.neuropharm.2020.108052] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/04/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022]
Abstract
Neuropeptides play important modulatory roles throughout the nervous system, functioning as direct effectors or as interacting partners with other neuropeptide and neurotransmitter systems. Limbic brain areas involved in learning, memory and emotions are particularly rich in neuropeptides. This review will focus on the amygdala, a limbic region that plays a key role in emotional-affective behaviors and pain modulation. The amygdala is comprised of different nuclei; the basolateral (BLA) and central (CeA) nuclei and in between, the intercalated cells (ITC), have been linked to pain-related functions. A wide range of neuropeptides are found in the amygdala, particularly in the CeA, but this review will discuss those neuropeptides that have been explored for their role in pain modulation. Calcitonin gene-related peptide (CGRP) is a key peptide in the afferent nociceptive pathway from the parabrachial area and mediates excitatory drive of CeA neurons. CeA neurons containing corticotropin releasing factor (CRF) and/or somatostatin (SOM) are a source of long-range projections and serve major output functions, but CRF also acts locally to excite neurons in the CeA and BLA. Neuropeptide S (NPS) is associated with inhibitory ITC neurons that gate amygdala output. Oxytocin and vasopressin exert opposite (inhibitory and excitatory, respectively) effects on amygdala output. The opioid system of mu, delta and kappa receptors (MOR, DOR, KOR) and their peptide ligands (β-endorphin, enkephalin, dynorphin) have complex and partially opposing effects on amygdala function. Neuropeptides therefore serve as valuable targets to regulate amygdala function in pain conditions. This article is part of the special issue on Neuropeptides.
Collapse
Affiliation(s)
- Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| | - Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Bryce Cragg
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Edita Navratilova
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Frank Porreca
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
26
|
Liu Y, Chen QY, Lee JH, Li XH, Yu S, Zhuo M. Cortical potentiation induced by calcitonin gene-related peptide (CGRP) in the insular cortex of adult mice. Mol Brain 2020; 13:36. [PMID: 32151282 PMCID: PMC7063738 DOI: 10.1186/s13041-020-00580-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023] Open
Abstract
Recent studies demonstrate that calcitonin gene-related peptide (CGRP) plays critical roles in migraine. Immunohistochemistry and in situ hybridization studies have shown that CGRP and its receptors are expressed in cortical areas that are critical for pain perception including the anterior cingulate cortex (ACC) and insular cortex (IC). Recent studies reported that CGRP enhanced excitatory transmission in the ACC. However, little is known about the possible effect of CGRP on excitatory transmission in the IC. In the present study, we investigated the role of CGRP on synaptic transmission in the IC slices of adult male mice. Bath application of CGRP produced dose-dependent potentiation of evoked excitatory postsynaptic currents (eEPSCs). This potentiation was NMDA receptor (NMDAR) independent. After application of CGRP1 receptor antagonist CGRP8–37 or BIBN 4096, CGRP produced potentiation was significantly reduced. Paired-pulse facilitation was significantly decreased by CGRP, suggesting possible presynaptic mechanisms. Consistently, bath application of CGRP significantly increased the frequency of spontaneous and miniature excitatory postsynaptic currents (sEPSCs and mEPSCs). By contrast, amplitudes of sEPSCs and mEPSCs were not significantly affected. Finally, adenylyl cyclase subtype 1 (AC1) and protein kinase A (PKA) are critical for CGRP-produced potentiation, since both selective AC1 inhibitor NB001 and the PKA inhibitor KT5720 completely blocked the potentiation. Our results provide direct evidence that CGRP contributes to synaptic potentiation in the IC, and the AC1 inhibitor NB001 may be beneficial for the treatment of migraine in the future.
Collapse
Affiliation(s)
- Yinglu Liu
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.,Medical School of Chinese PLA and Department of Neurology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Qi-Yu Chen
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.,Institute for Brain Research, QingDao International Academician Park, Qing Dao, China
| | - Jung Hyun Lee
- Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Xu-Hui Li
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.,Institute for Brain Research, QingDao International Academician Park, Qing Dao, China
| | - Shengyuan Yu
- Medical School of Chinese PLA and Department of Neurology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Min Zhuo
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China. .,Department of Physiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada. .,Institute for Brain Research, QingDao International Academician Park, Qing Dao, China.
| |
Collapse
|
27
|
Borkum JM. CGRP and Brain Functioning: Cautions for Migraine Treatment. Headache 2019; 59:1339-1357. [DOI: 10.1111/head.13591] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Jonathan M. Borkum
- Department of Psychology University of Maine Orono ME USA
- Health Psych Maine Waterville ME USA
| |
Collapse
|
28
|
Li XH, Matsuura T, Liu RH, Xue M, Zhuo M. Calcitonin gene-related peptide potentiated the excitatory transmission and network propagation in the anterior cingulate cortex of adult mice. Mol Pain 2019; 15:1744806919832718. [PMID: 30717631 PMCID: PMC6396051 DOI: 10.1177/1744806919832718] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The neuropeptide of calcitonin gene-related peptide (CGRP) plays critical roles in chronic pain, especially in migraine. Immunohistochemistry and in situ hybridization studies have shown that CGRP and its receptors are expressed in cortical areas including pain perception-related prefrontal anterior cingulate cortex. However, less information is available for the functional roles of CGRP in cortical regions such as the anterior cingulate cortex (ACC). Recent studies have consistently demonstrated that long-term potentiation is a key cellular mechanism for chronic pain in the ACC. In the present study, we used 64-electrode array field recording system to investigate the effect of CGRP on excitatory transmission in the ACC. We found that CGRP induced potentiation of synaptic transmission in a dose-dependently manner (1, 10, 50, and 100 nM). CGRP also recruited inactive circuit in the ACC. An application of the calcitonin receptor-like receptor antagonist CGRP8-37 blocked CGRP-induced chemical long-term potentiation and the recruitment of inactive channels. CGRP-induced long-term potentiation was also blocked by N-methyl-D-aspartate (NMDA) receptor antagonist AP-5. Consistently, the application of CGRP increased NMDA receptor-mediated excitatory postsynaptic currents. Finally, we found that CGRP-induced long-term potentiation required the activation of calcium-stimulated adenylyl cyclase subtype 1 (AC1) and protein kinase A. Genetic deletion of AC1 using AC1−/− mice, an AC1 inhibitor NB001 or a protein kinase A inhibitor KT5720, all reduced or blocked CGRP-induced potentiation. Our results provide direct evidence that CGRP may contribute to synaptic potentiation in important physiological and pathological conditions in the ACC, an AC1 inhibitor NB001 may be beneficial for the treatment of chronic headache.
Collapse
Affiliation(s)
- Xu-Hui Li
- 1 Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.,2 Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, Ontario, Canada
| | - Takanori Matsuura
- 2 Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, Ontario, Canada.,3 Department of Orthopaedics, School of Medicine, University of Occupational and Environmental Health, Yahatanishi-ku, Kitakyushu, Japan
| | - Ren-Hao Liu
- 1 Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Man Xue
- 1 Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Min Zhuo
- 1 Center for Neuron and Disease, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.,2 Department of Physiology, Faculty of Medicine, University of Toronto, Medical Science Building, 1 King's College Circle, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Abstract
Pain has a strong emotional component and is defined by its unpleasantness. Chronic pain represents a complex disorder with anxio-depressive symptoms and cognitive deficits. Underlying mechanisms are still not well understood but an important role for interactions between prefrontal cortical areas and subcortical limbic structures has emerged. Evidence from preclinical studies in the rodent brain suggests that neuroplastic changes in prefrontal (anterior cingulate, prelimbic and infralimbic) cortical and subcortical (amygdala and nucleus accumbens) brain areas and their interactions (corticolimbic circuitry) contribute to the complexity and persistence of pain and may be predetermining factors as has been proposed in recent human neuroimaging studies.
Collapse
Affiliation(s)
- Jeremy M Thompson
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, United States
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX, United States; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States.
| |
Collapse
|
30
|
Yin D, Chen Y, Lu R, Fan B, Zhu S, Xu X, Xu Z. Translocation Associated Membrane Protein 1 Contributes to Chronic Constriction Injury-Induced Neuropathic Pain in the Dorsal Root Ganglion and Spinal Cord in Rats. J Mol Neurosci 2018; 66:535-546. [PMID: 30338452 DOI: 10.1007/s12031-018-1187-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 09/25/2018] [Indexed: 11/30/2022]
Abstract
Neuropathic pain is a severe debilitating state caused by injury or dysfunction of somatosensory nervous system, and the clinical treatment is still challenging. Translocation associated membrane protein 1 (TRAM1), an adapter protein, participates in a variety of transduction pathways and mediates the biological functions such as cell proliferation, activation, and differentiation. However, whether TRAM1 is involved in the pathogenesis of neuropathic pain is still unclear. In our study, we reported the role of TRAM1 in the maintenance of neuropathic pain induced by chronic constriction injury (CCI) on rats. By western blot and staining, we found that TRAM1 increased in the dorsal root ganglion (DRG) neurons and spinal cord (SC) neurons after CCI. Being similar to IB4-, CGRP-positive expressed area, TRAM1 also expressed in the superficial laminae of the spinal cord dorsal horn (SCDH), suggesting it was related to the innervations of the primary afferents. Moreover, intrathecal injection of TRAM1 siRNA or Toll-like receptor 4 (TLR4) inhibitor induced low expression of TRAM1 in SC, which alleviated the pain response induced by CCI. The upregulation of p-NF-κB expression was reversed by TRAM1 siRNA in SC and DRG, and intrathecal injection of p-NF-κB inhibitor relieved neuropathic pain. All the data indicated that TRAM1 could take part in CCI-induced pain and might be a potential treatment for chronic neuropathic pain.
Collapse
Affiliation(s)
- Dekun Yin
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, 226001, China
- Department of Anesthesiology, Funing People's Hospital of Jiangsu, Funing County, 224400, Jiangsu Province, China
| | - Yonglin Chen
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Rongxiang Lu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Bingbing Fan
- Department of Radiology, Zhongshan Hospital and Shanghai Institute of Medical Imaging, Department of Medical Imaging, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shunxing Zhu
- Laboratory Animal Center, Nantong University, Nantong, 226001, China
| | - Xingguo Xu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, 226001, China.
| | - Zhongling Xu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, 226001, China.
| |
Collapse
|
31
|
Miyazawa Y, Takahashi Y, Watabe AM, Kato F. Predominant synaptic potentiation and activation in the right central amygdala are independent of bilateral parabrachial activation in the hemilateral trigeminal inflammatory pain model of rats. Mol Pain 2018; 14:1744806918807102. [PMID: 30270724 PMCID: PMC6243415 DOI: 10.1177/1744806918807102] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nociceptive signals originating in the periphery are conveyed to the brain through specific afferent and ascending pathways. The spino-(trigemino-)parabrachio-amygdaloid pathway is one of the principal pathways mediating signals from nociception-specific ascending neurons to the central amygdala, a limbic structure involved in aversive signal-associated emotional responses, including the emotional aspects of pain. Recent studies suggest that the right and left central amygdala play distinct roles in the regulation of nociceptive responses. Using a latent formalin inflammatory pain model of the rat, we analyzed the right-left differences in synaptic potentiation at the synapses formed between the fibers from the lateral parabrachial nucleus and central amygdala neurons as well as those in the c-Fos expression in the lateral parabrachial nucleus, central amygdala, and the basolateral/lateral amygdala after formalin injection to either the right or left side of the rat upper lip. Although the single-sided formalin injection caused a significant bilateral increase in c-Fos-expressing neurons in the lateral parabrachial nucleus with slight projection-side dependence, the increase in the amplitude of postsynaptic excitatory currents and the number of c-Fos-expressing neurons in the central amygdala occurred predominantly on the right side regardless of the side of the inflammation. Although there was no significant correlation in the number of c-Fos-expressing neurons between the lateral parabrachial nucleus and central amygdala in the formalin-injected animals, these numbers were significantly correlated between the basolateral amygdala and central amygdala. It is thus concluded that the lateral parabrachial nucleus-central amygdala synaptic potentiation reported in various pain models is not a simple Hebbian plasticity in which raised inputs from the lateral parabrachial nucleus cause lateral parabrachial nucleus-central amygdala potentiation but rather an integrative and adaptive response involving specific mechanisms in the right central amygdala.
Collapse
Affiliation(s)
- Yuta Miyazawa
- 1 Department of Neuroscience, Jikei University School of Medicine, Tokyo, Japan.,2 Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan
| | - Yukari Takahashi
- 1 Department of Neuroscience, Jikei University School of Medicine, Tokyo, Japan.,2 Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan
| | - Ayako M Watabe
- 2 Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan.,3 Institute of Clinical Medicine and Research, Jikei University School of Medicine, Tokyo, Japan
| | - Fusao Kato
- 1 Department of Neuroscience, Jikei University School of Medicine, Tokyo, Japan.,2 Center for Neuroscience of Pain, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
32
|
Harris NA, Winder DG. Synaptic Plasticity in the Bed Nucleus of the Stria Terminalis: Underlying Mechanisms and Potential Ramifications for Reinstatement of Drug- and Alcohol-Seeking Behaviors. ACS Chem Neurosci 2018; 9:2173-2187. [PMID: 29851347 PMCID: PMC6146063 DOI: 10.1021/acschemneuro.8b00169] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The bed nucleus of the stria terminalis (BNST) is a component of the extended amygdala that shows significant changes in activity and plasticity through chronic exposure to drugs and stress. The region is critical for stress- and cue-induced reinstatement of drug-seeking behaviors and is thus a candidate region for the plastic changes that occur in abstinence that prime addicted patients for reinstatement behaviors. Here, we discuss the various forms of long-term potentiation (LTP) and long-term depression (LTD) in the rodent BNST and highlight the way that these changes in excitatory transmission interact with exposure to alcohol and other drugs of abuse, as well as other stressors. In addition, we highlight potential areas for future research in this area, including investigating input- and cell-specific bidirectional changes in activity. As we continue to accrue foundational knowledge in the mechanisms and effects of plasticity in the BNST, molecular targets and treatment strategies that are relevant to reinstatement behaviors will also begin to emerge. Here, we briefly discuss the effects of catecholamine receptor modulators on synaptic plasticity in the BNST due to the role of norepinephrine in LTD and dopamine on the short-term component of LTP as well as the role that signaling at these receptors plays in reinstatement of drug- and alcohol-seeking behaviors. We hope that insights gained on the specific changes in plasticity that occur within the BNST during abstinence from alcohol and other drugs of abuse will provide insight into the biological underpinnings of relapse behavior in human addicts and inform future treatment modalities for addiction that tackle this complex biological problem.
Collapse
Affiliation(s)
- Nicholas A. Harris
- Vanderbilt Center for Addiction Research
- Department of Molecular Physiology & Biophysics
| | - Danny G. Winder
- Vanderbilt Center for Addiction Research
- Department of Molecular Physiology & Biophysics
- Vanderbilt J.F. Kennedy Center for Research on Human Development
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
33
|
Vila-Pueyo M, Hoffmann J, Romero-Reyes M, Akerman S. Brain structure and function related to headache: Brainstem structure and function in headache. Cephalalgia 2018; 39:1635-1660. [PMID: 29969040 DOI: 10.1177/0333102418784698] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To review and discuss the literature relevant to the role of brainstem structure and function in headache. BACKGROUND Primary headache disorders, such as migraine and cluster headache, are considered disorders of the brain. As well as head-related pain, these headache disorders are also associated with other neurological symptoms, such as those related to sensory, homeostatic, autonomic, cognitive and affective processing that can all occur before, during or even after headache has ceased. Many imaging studies demonstrate activation in brainstem areas that appear specifically associated with headache disorders, especially migraine, which may be related to the mechanisms of many of these symptoms. This is further supported by preclinical studies, which demonstrate that modulation of specific brainstem nuclei alters sensory processing relevant to these symptoms, including headache, cranial autonomic responses and homeostatic mechanisms. REVIEW FOCUS This review will specifically focus on the role of brainstem structures relevant to primary headaches, including medullary, pontine, and midbrain, and describe their functional role and how they relate to mechanisms of primary headaches, especially migraine.
Collapse
Affiliation(s)
- Marta Vila-Pueyo
- Headache Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Jan Hoffmann
- Department of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcela Romero-Reyes
- Department of Neural and Pain Sciences, University of Maryland Baltimore, Baltimore, MD, USA
| | - Simon Akerman
- Department of Neural and Pain Sciences, University of Maryland Baltimore, Baltimore, MD, USA
| |
Collapse
|
34
|
Shinohara K, Watabe AM, Nagase M, Okutsu Y, Takahashi Y, Kurihara H, Kato F. Essential role of endogenous calcitonin gene-related peptide in pain-associated plasticity in the central amygdala. Eur J Neurosci 2017; 46:2149-2160. [PMID: 28833700 PMCID: PMC5698701 DOI: 10.1111/ejn.13662] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/04/2017] [Accepted: 08/14/2017] [Indexed: 12/16/2022]
Abstract
The role of the neuropeptide calcitonin gene‐related peptide (CGRP) is well established in nociceptive behaviors. CGRP is highly expressed in the projection pathway from the parabrachial nucleus to the laterocapsular region of the central amygdala (CeC), which plays a critical role in relaying nociceptive information. The CeC is a key structure in pain behavior because it integrates and modulates nociceptive information along with other sensory signals. Previous studies have demonstrated that blockade of the amygdalar CGRP‐signaling cascade attenuates nociceptive behaviors in pain models, while CGRP application facilitates amygdalar synaptic transmission and induces pain behaviors. Despite these lines of evidence, it remains unclear whether endogenous CGRP is involved in the development of nociceptive behaviors accompanied with amygdalar plasticity in a peripheral inflammation model in vivo. To directly address this, we utilized a previously generated CGRP knockout (KO) mouse to longitudinally study formalin‐induced plasticity and nociceptive behavior. We found that synaptic potentiation in the right PB‐CeC pathway that was observed in wild‐type mice was drastically attenuated in the CGRP KO mice 6 h post‐inflammation, when acute nociceptive behavior was no longer observed. Furthermore, the bilateral tactile allodynia 6 h post‐inflammation was significantly decreased in the CGRP KO mice. In contrast, the acute nociceptive behavior immediately after the formalin injection was reduced only at 20–25 min post‐injection in the CGRP KO mice. These results suggest that endogenous CGRP contributes to peripheral inflammation‐induced synaptic plasticity in the amygdala, and this plasticity may underlie the exaggerated nociception–emotion linkage in pain chronification.
Collapse
Affiliation(s)
- Kei Shinohara
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan.,Department of Orthopaedic Surgery, Jikei University School of Medicine, Minato-ku, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Ayako M Watabe
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Masashi Nagase
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Yuya Okutsu
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan.,Department of Orthopaedic Surgery, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Yukari Takahashi
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Hiroki Kurihara
- Department of Molecular Cell Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Fusao Kato
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan.,Center for Neuroscience of Pain, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| |
Collapse
|
35
|
Shinohara K, Watabe AM, Nagase M, Okutsu Y, Takahashi Y, Kurihara H, Kato F. Essential role of endogenous calcitonin gene-related peptide in pain-associated plasticity in the central amygdala. Eur J Neurosci 2017. [PMID: 28833700 DOI: 10.1111/ejn.2017.46.issue-610.1111/ejn.13662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
The role of the neuropeptide calcitonin gene-related peptide (CGRP) is well established in nociceptive behaviors. CGRP is highly expressed in the projection pathway from the parabrachial nucleus to the laterocapsular region of the central amygdala (CeC), which plays a critical role in relaying nociceptive information. The CeC is a key structure in pain behavior because it integrates and modulates nociceptive information along with other sensory signals. Previous studies have demonstrated that blockade of the amygdalar CGRP-signaling cascade attenuates nociceptive behaviors in pain models, while CGRP application facilitates amygdalar synaptic transmission and induces pain behaviors. Despite these lines of evidence, it remains unclear whether endogenous CGRP is involved in the development of nociceptive behaviors accompanied with amygdalar plasticity in a peripheral inflammation model in vivo. To directly address this, we utilized a previously generated CGRP knockout (KO) mouse to longitudinally study formalin-induced plasticity and nociceptive behavior. We found that synaptic potentiation in the right PB-CeC pathway that was observed in wild-type mice was drastically attenuated in the CGRP KO mice 6 h post-inflammation, when acute nociceptive behavior was no longer observed. Furthermore, the bilateral tactile allodynia 6 h post-inflammation was significantly decreased in the CGRP KO mice. In contrast, the acute nociceptive behavior immediately after the formalin injection was reduced only at 20-25 min post-injection in the CGRP KO mice. These results suggest that endogenous CGRP contributes to peripheral inflammation-induced synaptic plasticity in the amygdala, and this plasticity may underlie the exaggerated nociception-emotion linkage in pain chronification.
Collapse
Affiliation(s)
- Kei Shinohara
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
- Department of Orthopaedic Surgery, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
- Center for Neuroscience of Pain, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Ayako M Watabe
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
- Center for Neuroscience of Pain, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Masashi Nagase
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
- Center for Neuroscience of Pain, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Yuya Okutsu
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
- Department of Orthopaedic Surgery, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Yukari Takahashi
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
- Center for Neuroscience of Pain, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Hiroki Kurihara
- Department of Molecular Cell Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Fusao Kato
- Department of Neuroscience, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
- Center for Neuroscience of Pain, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| |
Collapse
|