1
|
Blank N, Weiner M, Patel S, Köhler S, Thaiss CA. Mind the GAPS: Glia associated with psychological stress. J Neuroendocrinol 2024:e13451. [PMID: 39384366 DOI: 10.1111/jne.13451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/01/2024] [Accepted: 09/05/2024] [Indexed: 10/11/2024]
Abstract
Glial cells are an integral component of the nervous system, performing crucial functions that extend beyond structural support, including modulation of the immune system, tissue repair, and maintaining tissue homeostasis. Recent studies have highlighted the importance of glial cells as key mediators of stress responses across different organs. This review focuses on the roles of glial cells in peripheral tissues in health and their involvement in diseases linked to psychological stress. Populations of glia associated with psychological stress ("GAPS") emerge as a promising target cell population in our basic understanding of stress-associated pathologies, highlighting their role as mediators of the deleterious effects of psychological stress on various health conditions. Ultimately, new insights into the impact of stress on glial cell populations in the periphery may support clinical efforts aimed at improving the psychological state of patients for improved health outcomes.
Collapse
Affiliation(s)
- Niklas Blank
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Molly Weiner
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shaan Patel
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sarah Köhler
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christoph A Thaiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Obesity, Diabetes and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Qiu X, Yang Y, Da X, Wang Y, Chen Z, Xu C. Satellite glial cells in sensory ganglia play a wider role in chronic pain via multiple mechanisms. Neural Regen Res 2024; 19:1056-1063. [PMID: 37862208 PMCID: PMC10749601 DOI: 10.4103/1673-5374.382986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/26/2023] [Accepted: 07/10/2023] [Indexed: 10/22/2023] Open
Abstract
Satellite glial cells are unique glial cells that surround the cell body of primary sensory neurons. An increasing body of evidence suggests that in the presence of inflammation and nerve damage, a significant number of satellite glial cells become activated, thus triggering a series of functional changes. This suggests that satellite glial cells are closely related to the occurrence of chronic pain. In this review, we first summarize the morphological structure, molecular markers, and physiological functions of satellite glial cells. Then, we clarify the multiple key roles of satellite glial cells in chronic pain, including gap junction hemichannel Cx43, membrane channel Pannexin1, K channel subunit 4.1, ATP, purinergic P2 receptors, and a series of additional factors and their receptors, including tumor necrosis factor, glutamate, endothelin, and bradykinin. Finally, we propose that future research should focus on the specific sorting of satellite glial cells, and identify genomic differences between physiological and pathological conditions. This review provides an important perspective for clarifying mechanisms underlying the peripheral regulation of chronic pain and will facilitate the formulation of new treatment plans for chronic pain.
Collapse
Affiliation(s)
- Xiaoyun Qiu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yuanzhi Yang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaoli Da
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
3
|
Bang S, Jiang C, Xu J, Chandra S, McGinnis A, Luo X, He Q, Li Y, Wang Z, Ao X, Parisien M, Fernandes de Araujo LO, Jahangiri Esfahani S, Zhang Q, Tonello R, Berta T, Diatchenko L, Ji RR. Satellite glial GPR37L1 and its ligand maresin 1 regulate potassium channel signaling and pain homeostasis. J Clin Invest 2024; 134:e173537. [PMID: 38530364 PMCID: PMC11060744 DOI: 10.1172/jci173537] [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: 06/29/2023] [Accepted: 03/12/2024] [Indexed: 03/27/2024] Open
Abstract
G protein-coupled receptor 37-like 1 (GPR37L1) is an orphan GPCR with largely unknown functions. Here, we report that Gpr37l1/GRP37L1 ranks among the most highly expressed GPCR transcripts in mouse and human dorsal root ganglia (DRGs) and is selectively expressed in satellite glial cells (SGCs). Peripheral neuropathy induced by streptozotoxin (STZ) and paclitaxel (PTX) led to reduced GPR37L1 expression on the plasma membrane in mouse and human DRGs. Transgenic mice with Gpr37l1 deficiency exhibited impaired resolution of neuropathic pain symptoms following PTX- and STZ-induced pain, whereas overexpression of Gpr37l1 in mouse DRGs reversed pain. GPR37L1 is coexpressed with potassium channels, including KCNJ10 (Kir4.1) in mouse SGCs and both KCNJ3 (Kir3.1) and KCNJ10 in human SGCs. GPR37L1 regulates the surface expression and function of the potassium channels. Notably, the proresolving lipid mediator maresin 1 (MaR1) serves as a ligand of GPR37L1 and enhances KCNJ10- or KCNJ3-mediated potassium influx in SGCs through GPR37L1. Chemotherapy suppressed KCNJ10 expression and function in SGCs, which MaR1 rescued through GPR37L1. Finally, genetic analysis revealed that the GPR37L1-E296K variant increased chronic pain risk by destabilizing the protein and impairing the protein's function. Thus, GPR37L1 in SGCs offers a therapeutic target for the protection of neuropathy and chronic pain.
Collapse
Affiliation(s)
- Sangsu Bang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Changyu Jiang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Jing Xu
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Sharat Chandra
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Aidan McGinnis
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Xin Luo
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Qianru He
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Yize Li
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Zilong Wang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Xiang Ao
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Marc Parisien
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Lorenna Oliveira Fernandes de Araujo
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Sahel Jahangiri Esfahani
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Qin Zhang
- Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Raquel Tonello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Luda Diatchenko
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurobiology and
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
4
|
Lai A, Iliff D, Zaheer K, Gansau J, Laudier DM, Zachariou V, Iatridis JC. Annulus Fibrosus Injury Induces Acute Neuroinflammation and Chronic Glial Response in Dorsal Root Ganglion and Spinal Cord-An In Vivo Rat Discogenic Pain Model. Int J Mol Sci 2024; 25:1762. [PMID: 38339040 PMCID: PMC10855200 DOI: 10.3390/ijms25031762] [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: 11/27/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Chronic painful intervertebral disc (IVD) degeneration (i.e., discogenic pain) is a major source of global disability needing improved knowledge on multiple-tissue interactions and how they progress in order improve treatment strategies. This study used an in vivo rat annulus fibrosus (AF) injury-driven discogenic pain model to investigate the acute and chronic changes in IVD degeneration and spinal inflammation, as well as sensitization, inflammation, and remodeling in dorsal root ganglion (DRG) and spinal cord (SC) dorsal horn. AF injury induced moderate IVD degeneration with acute and broad spinal inflammation that progressed to DRG to SC changes within days and weeks, respectively. Specifically, AF injury elevated macrophages in the spine (CD68) and DRGs (Iba1) that peaked at 3 days post-injury, and increased microglia (Iba1) in SC that peaked at 2 weeks post-injury. AF injury also triggered glial responses with elevated GFAP in DRGs and SC at least 8 weeks post-injury. Spinal CD68 and SC neuropeptide Substance P both remained elevated at 8 weeks, suggesting that slow and incomplete IVD healing provides a chronic source of inflammation with continued SC sensitization. We conclude that AF injury-driven IVD degeneration induces acute spinal, DRG, and SC inflammatory crosstalk with sustained glial responses in both DRGs and SC, leading to chronic SC sensitization and neural plasticity. The known association of these markers with neuropathic pain suggests that therapeutic strategies for discogenic pain need to target both spinal and nervous systems, with early strategies managing acute inflammatory processes, and late strategies targeting chronic IVD inflammation, SC sensitization, and remodeling.
Collapse
Affiliation(s)
- Alon Lai
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Denise Iliff
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kashaf Zaheer
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jennifer Gansau
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Damien M Laudier
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Venetia Zachariou
- Department of Pharmacology, Physiology and Biophysics, Chobanian and Avedisian School of Medicine at Boston University, Boston, MA 02118, USA
| | - James C Iatridis
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
5
|
Cazzaniga S, Real G, Finazzi S, Lorini LF, Forget P, Bugada D. How to Modulate Peripheral and Central Nervous System to Treat Acute Postoperative Pain and Prevent Pain Persistence. Curr Neuropharmacol 2024; 22:23-37. [PMID: 37563811 PMCID: PMC10716883 DOI: 10.2174/1570159x21666230810103508] [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: 10/31/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 08/12/2023] Open
Abstract
Chronic postoperative pain (CPSP) is a major issue after surgery, which may impact on patient's quality of life. Traditionally, CPSP is believed to rely on maladaptive hyperalgesia and risk factors have been identified that predispose to CPSP, including acute postoperative pain. Despite new models of prediction are emerging, acute pain is still a modifiable factor that can be challenged with perioperative analgesic strategies. In this review we present the issue of CPSP, focusing on molecular mechanism underlying the development of acute and chronic hyperalgesia. Also, we focus on how perioperative strategies can impact directly or indirectly (by reducing postoperative pain intensity) on the development of CPSP.
Collapse
Affiliation(s)
- Sara Cazzaniga
- Emergency and Intensive Care Department, ASST Papa Giovanni XXIII, 24127, Bergamo, Italy
| | - Giovanni Real
- Department of Health Sciences, University of Milan, 20122, Milan, Italy
| | - Simone Finazzi
- Department of Health Sciences, University of Milan, 20122, Milan, Italy
| | - Luca F Lorini
- Emergency and Intensive Care Department, ASST Papa Giovanni XXIII, 24127, Bergamo, Italy
| | - Patrice Forget
- School of Medicine, Medical Sciences and Nutrition, Epidemiology Group, Institute of Applied Health Sciences, University of Aberdeen, Scotland, United Kingdom
- Department of Anaesthesia, NHS Grampian, Aberdeen AB25 2ZD, Scotland, United Kingdom
| | - Dario Bugada
- Emergency and Intensive Care Department, ASST Papa Giovanni XXIII, 24127, Bergamo, Italy
| |
Collapse
|
6
|
Bang S, Jiang C, Xu J, Chandra S, McGinnis A, Luo X, He Q, Li Y, Wang Z, Ao X, Parisien M, Fernandes de Araujo LO, Esfahan SJ, Zhang Q, Tonello R, Berta T, Diatchenko L, Ji RR. Satellite glial GPR37L1 regulates maresin and potassium channel signaling for pain control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.03.569787. [PMID: 38106084 PMCID: PMC10723316 DOI: 10.1101/2023.12.03.569787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
G protein coupled receptor 37-like 1 (GPR37L1) is an orphan GPCR and its function remains largely unknown. Here we report that GPR37L1 transcript is highly expressed compared to all known GPCRs in mouse and human dorsal root ganglia (DRGs) and selectively expressed in satellite glial cells (SGCs). Peripheral neuropathy following diabetes and chemotherapy by streptozotocin and paclitaxel resulted in downregulations of surface GPR37L1 in mouse and human DRGs. Transgenic mice with Gpr37l1 deficiency exhibited impaired resolution of neuropathic pain symptom (mechanical allodynia), whereas overexpression of Gpr37l1 in mouse DRGs can reverse neuropathic pain. Notably, GPR37L1 is co-expressed and coupled with potassium channels in SGCs. We found striking species differences in potassium channel expression in SGCs, with predominant expression of KCNJ10 and KCNJ3 in mouse and human SGCs, respectively. GPR37L1 regulates the surface expression and function of KCNJ10 and KCNJ3. We identified the pro-resolving lipid mediator maresin 1 (MaR1) as a GPR37L1 ligand. MaR1 increases KCNJ10/KCNJ3-mediated potassium influx in SGCs via GPR37L1. MaR1 protected chemotherapy-induced suppression of KCNJ13/KCNJ10 expression and function in SGCs. Finally, genetic analysis revealed that the GPR37L1-E296K variant is associated with increased chronic pain risk by destabilizing the protein. Thus, GPR37L1 in SGCs offers a new target for neuropathy protection and pain control.
Collapse
|
7
|
Konnova EA, Deftu AF, Chu Sin Chung P, Pertin M, Kirschmann G, Decosterd I, Suter MR. Characterisation of GFAP-Expressing Glial Cells in the Dorsal Root Ganglion after Spared Nerve Injury. Int J Mol Sci 2023; 24:15559. [PMID: 37958541 PMCID: PMC10647921 DOI: 10.3390/ijms242115559] [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: 09/20/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Satellite glial cells (SGCs), enveloping primary sensory neurons' somas in the dorsal root ganglion (DRG), contribute to neuropathic pain upon nerve injury. Glial fibrillary acidic protein (GFAP) serves as an SGC activation marker, though its DRG satellite cell specificity is debated. We employed the hGFAP-CFP transgenic mouse line, designed for astrocyte studies, to explore its expression within the peripheral nervous system (PNS) after spared nerve injury (SNI). We used diverse immunostaining techniques, Western blot analysis, and electrophysiology to evaluate GFAP+ cell changes. Post-SNI, GFAP+ cell numbers increased without proliferation, and were found near injured ATF3+ neurons. GFAP+ FABP7+ SGCs increased, yet 75.5% of DRG GFAP+ cells lacked FABP7 expression. This suggests a significant subset of GFAP+ cells are non-myelinating Schwann cells (nmSC), indicated by their presence in the dorsal root but not in the ventral root which lacks unmyelinated fibres. Additionally, patch clamp recordings from GFAP+ FABP7-cells lacked SGC-specific Kir4.1 currents, instead displaying outward Kv currents expressing Kv1.1 and Kv1.6 channels specific to nmSCs. In conclusion, this study demonstrates increased GFAP expression in two DRG glial cell subpopulations post-SNI: GFAP+ FABP7+ SGCs and GFAP+ FABP7- nmSCs, shedding light on GFAP's specificity as an SGC marker after SNI.
Collapse
Affiliation(s)
- Elena A. Konnova
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), 1005 Lausanne, Switzerland
| | - Alexandru-Florian Deftu
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), 1005 Lausanne, Switzerland
| | - Paul Chu Sin Chung
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), 1005 Lausanne, Switzerland
| | - Marie Pertin
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), 1005 Lausanne, Switzerland
| | - Guylène Kirschmann
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), 1005 Lausanne, Switzerland
| | - Isabelle Decosterd
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), 1005 Lausanne, Switzerland
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| | - Marc R. Suter
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV), 1005 Lausanne, Switzerland
- Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
8
|
Ou M, Chen Y, Liu J, Zhang D, Yang Y, Shen J, Miao C, Tang SJ, Liu X, Mulkey DK, Zhu T, Zhou C. Spinal astrocytic MeCP2 regulates Kir4.1 for the maintenance of chronic hyperalgesia in neuropathic pain. Prog Neurobiol 2023; 224:102436. [PMID: 36931588 PMCID: PMC10372923 DOI: 10.1016/j.pneurobio.2023.102436] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Astrocyte activation in the spinal dorsal horn may play an important role in the development of chronic neuropathic pain, but the mechanisms involved in astrocyte activation and their modulatory effects remain unknown. The inward rectifying potassium channel protein 4.1 (Kir4.1) is the most important background K+ channel in astrocytes. However, how Kir4.1 is regulated and contributes to behavioral hyperalgesia in chronic pain is unknown. In this study, single-cell RNA sequencing analysis indicated that the expression levels of both Kir4.1 and Methyl-CpG-binding protein 2 (MeCP2) were decreased in spinal astrocytes after chronic constriction injury (CCI) in a mouse model. Conditional knockout of the Kir4.1 channel in spinal astrocytes led to hyperalgesia, and overexpression of the Kir4.1 channel in spinal cord relieved CCI-induced hyperalgesia. Expression of spinal Kir4.1 after CCI was regulated by MeCP2. Electrophysiological recording in spinal slices showed that knockdown of Kir4.1 significantly up-regulated the excitability of astrocytes and then functionally changed the firing patterns of neurons in dorsal spinal cord. Therefore, targeting spinal Kir4.1 may be a therapeutic approach for hyperalgesia in chronic neuropathic pain.
Collapse
Affiliation(s)
- Mengchan Ou
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yali Chen
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Donghang Zhang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yaoxin Yang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jiefei Shen
- Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases and Department of Prosthodontics, West China Stomatology Hospital of Sichuan University, Chengdu 610041, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shao-Jun Tang
- Department of Anesthesiology and The Stony Brook University Pain and Analgesia Research Center (SPARC), Stony Brook University, Stony Brook, NY 11794, USA
| | - Xin Liu
- Department of Anesthesiology and The Stony Brook University Pain and Analgesia Research Center (SPARC), Stony Brook University, Stony Brook, NY 11794, USA
| | - Daniel K Mulkey
- Departments of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269, USA
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital of Sichuan University, Chengdu 610041, China.
| |
Collapse
|
9
|
McGinnis A, Ji RR. The Similar and Distinct Roles of Satellite Glial Cells and Spinal Astrocytes in Neuropathic Pain. Cells 2023; 12:965. [PMID: 36980304 PMCID: PMC10047571 DOI: 10.3390/cells12060965] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Preclinical studies have identified glial cells as pivotal players in the genesis and maintenance of neuropathic pain after nerve injury associated with diabetes, chemotherapy, major surgeries, and virus infections. Satellite glial cells (SGCs) in the dorsal root and trigeminal ganglia of the peripheral nervous system (PNS) and astrocytes in the central nervous system (CNS) express similar molecular markers and are protective under physiological conditions. They also serve similar functions in the genesis and maintenance of neuropathic pain, downregulating some of their homeostatic functions and driving pro-inflammatory neuro-glial interactions in the PNS and CNS, i.e., "gliopathy". However, the role of SGCs in neuropathic pain is not simply as "peripheral astrocytes". We delineate how these peripheral and central glia participate in neuropathic pain by producing different mediators, engaging different parts of neurons, and becoming active at different stages following nerve injury. Finally, we highlight the recent findings that SGCs are enriched with proteins related to fatty acid metabolism and signaling such as Apo-E, FABP7, and LPAR1. Targeting SGCs and astrocytes may lead to novel therapeutics for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Aidan McGinnis
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
10
|
Reed CB, Feltri ML, Wilson ER. Peripheral glia diversity. J Anat 2022; 241:1219-1234. [PMID: 34131911 PMCID: PMC8671569 DOI: 10.1111/joa.13484] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Recent years have seen an evolving appreciation for the role of glial cells in the nervous system. As we move away from the typical neurocentric view of neuroscience, the complexity and variability of central nervous system glia is emerging, far beyond the three main subtypes: astrocytes, oligodendrocytes, and microglia. Yet the diversity of the glia found in the peripheral nervous system remains rarely discussed. In this review, we discuss the developmental origin, morphology, and function of the different populations of glia found in the peripheral nervous system, including: myelinating Schwann cells, Remak Schwann cells, repair Schwann cells, satellite glia, boundary cap-derived glia, perineurial glia, terminal Schwann cells, glia found in the skin, olfactory ensheathing cells, and enteric glia. The morphological and functional heterogeneity of glia found in the periphery reflects the diverse roles the nervous system performs throughout the body. Further, it highlights a complexity that should be appreciated and considered when it comes to a complete understanding of the peripheral nervous system in health and disease.
Collapse
Affiliation(s)
- Chelsey B. Reed
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of NeurologyJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| | - M. Laura Feltri
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of NeurologyJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
- Department of BiochemistryJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| | - Emma R. Wilson
- Hunter James Kelly Research InstituteJacobs School of Medicine and Biomedical Sciences StateUniversity of New York at BuffaloBuffaloNew YorkUSA
- Department of BiochemistryJacobs School of Medicine and Biomedical SciencesState University of New York at BuffaloBuffaloNew YorkUSA
| |
Collapse
|
11
|
Andreeva D, Murashova L, Burzak N, Dyachuk V. Satellite Glial Cells: Morphology, functional heterogeneity, and role in pain. Front Cell Neurosci 2022; 16:1019449. [PMID: 36274990 PMCID: PMC9583829 DOI: 10.3389/fncel.2022.1019449] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Neurons in the somatic, sympathetic, and parasympathetic ganglia are surrounded by envelopes consisting of satellite glial cells (SGCs). Recently, it has become clear that SGCs are highly altered after nerve injury, which influences neuronal excitability and, consequently, the development and maintenance of pain in different animal models of chronic pain. However, the exact mechanism underlying chronic pain is not fully understood yet because it is assumed that SGCs in different ganglia share many common peculiarities, making the process complex. Here, we review recent data on morphological and functional heterogeneity and changes in SGCs in various pain conditions and their role in response to injury. More research is required to decipher the role of SGCs in diseases, such as chronic pain, neuropathology, and neurodegenerative diseases.
Collapse
|
12
|
Su PYP, Zhang L, He L, Zhao N, Guan Z. The Role of Neuro-Immune Interactions in Chronic Pain: Implications for Clinical Practice. J Pain Res 2022; 15:2223-2248. [PMID: 35957964 PMCID: PMC9359791 DOI: 10.2147/jpr.s246883] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Chronic pain remains a public health problem and contributes to the ongoing opioid epidemic. Current pain management therapies still leave many patients with poorly controlled pain, thus new or improved treatments are desperately needed. One major challenge in pain research is the translation of preclinical findings into effective clinical practice. The local neuroimmune interface plays an important role in the initiation and maintenance of chronic pain and is therefore a promising target for novel therapeutic development. Neurons interface with immune and immunocompetent cells in many distinct microenvironments along the nociceptive circuitry. The local neuroimmune interface can modulate the activity and property of the neurons to affect peripheral and central sensitization. In this review, we highlight a specific subset of many neuroimmune interfaces. In the central nervous system, we examine the interface between neurons and microglia, astrocytes, and T lymphocytes. In the periphery, we profile the interface between neurons in the dorsal root ganglion with T lymphocytes, satellite glial cells, and macrophages. To bridge the gap between preclinical research and clinical practice, we review the preclinical studies of each neuroimmune interface, discuss current clinical treatments in pain medicine that may exert its action at the neuroimmune interface, and highlight opportunities for future clinical research efforts.
Collapse
Affiliation(s)
- Po-Yi Paul Su
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Lingyi Zhang
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Anesthesiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Liangliang He
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Pain Management, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Na Zhao
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
13
|
Wang Z, Martellucci S, Van Enoo A, Austin D, Gelber C, Campana WM. α1-Antitrypsin derived SP16 peptide demonstrates efficacy in rodent models of acute and neuropathic pain. FASEB J 2022; 36:e22093. [PMID: 34888951 PMCID: PMC8669735 DOI: 10.1096/fj.202101031rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/02/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023]
Abstract
SP16 is an innovative peptide derived from the carboxyl-terminus of α1-Antitrypsin (AAT), corresponding to residues 364-380, and contains recognition sequences for the low-density lipoprotein receptor-related protein-1 (LRP1). LRP1 is an endocytic and cell-signaling receptor that regulates inflammation. Deletion of Lrp1 in Schwann cells increases neuropathic pain; however, the role of LRP1 activation in nociceptive and neuropathic pain regulation remains unknown. Herein, we show that SP16 is bioactive in sensory neurons in vitro. Neurite length and regenerative gene expression were increased by SP16. In PC12 cells, SP16 activated Akt and ERK1/2 cell-signaling in an LRP1-dependent manner. When formalin was injected into mouse hind paws, to model inflammatory pain, SP16 dose-dependently attenuated nociceptive pain behaviors in the early and late phases. In a second model of acute pain using capsaicin, SP16 significantly reduced paw licking in both male and female mice (p < .01) similarly to enzymatically inactive tissue plasminogen activator, a known LRP1 interactor. SP16 also prevented development of tactile allodynia after partial nerve ligation and this response was sustained for nine days (p < .01). Immunoblot analysis of the injured nerve revealed decreased CD11b (p < .01) and Toll-like receptor-4 (p < .005). In injured dorsal root ganglia SP16 reduced CD11b+ cells (p < .05) and GFAP (p < .005), indicating that inflammatory cell recruitment and satellite cell activation were inhibited. In conclusion, administration of SP16 blocked pain-related responses in three distinct pain models, suggesting efficacy against acute nociceptive, inflammatory, and neuropathic pain. SP16 also attenuated innate immunity in the PNS. These studies identify SP16 as a potentially effective treatment for pain.
Collapse
Affiliation(s)
- Zixuan Wang
- Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla CA, 92093-0629 USA
| | - Stefano Martellucci
- Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla CA, 92093-0629 USA
| | - Alicia Van Enoo
- Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla CA, 92093-0629 USA;,Program in Neurosciences, University of California, San Diego, La Jolla CA 92093, USA
| | | | | | - Wendy M. Campana
- Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla CA, 92093-0629 USA;,Program in Neurosciences, University of California, San Diego, La Jolla CA 92093, USA;,San Diego Veterans Administration Health Care System, CA, 92161, USA
| |
Collapse
|
14
|
Li S, Zhao F, Tang Q, Xi C, He J, Wang Y, Zhu MX, Cao Z. Sarco/endoplasmic reticulum Ca 2+ -ATPase 2b mediates oxidation-induced endoplasmic reticulum stress to regulate neuropathic pain. Br J Pharmacol 2021; 179:2016-2036. [PMID: 34811737 DOI: 10.1111/bph.15744] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/24/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Neuropathic pain is a widespread health problem with limited curative treatment. Decreased sarco/endoplasmic reticulum Ca2+ -ATPase (SERCA) expression has been reported in dorsal root ganglion (DRG) of animals suffering from neuropathic pain. We aimed to establish the relationship between SERCA expression and the pain responses and to elucidate the underlying molecular mechanism. EXPERIMENTAL APPROACH Neuropathic pain was modeled using rat chronic constriction injury (CCI). Ca2+ imaging and current clamp patch-clamp were used to determine cytosolic Ca2+ levels and action potential firing, respectively. Western blots, immunofluorescence staining and RT-PCR were used to quantitatively assess protein and mRNA expression, respectively. H&E staining and coupled enzyme assay were used to evaluate the nerve injury and SERCA2b activity, respectively. KEY RESULTS SERCA2b is the predominant SERCA isoform in rat DRG and its expression is decreased after CCI at mRNA, protein and activity levels. Whereas inhibiting SERCA with thapsigargin causes neuronal hyperexcitation, nerve injury, ER stress, satellite glial cell activation and mechanical allodynia, activating SERCA by CDN1163 or overexpressing SERCA2b in DRG after CCI produces long-term relief of mechanical and thermal allodynia with accompanied morphological and functional restoration through alleviation of ER stress. Furthermore, the downregulation of DRG SERCA2b in CCI rats is caused by increased production of reactive oxygen species (ROS) through Sp1-dependent transcriptional inhibition. CONCLUSION AND IMPLICATIONS Our findings reveal a novel pathway centering around SERCA2b as the key molecule underlying the mechanism of development and maintenance of neuropathic pain, and SERCA2b activators have the potential for therapeutic treatment of neuropathic pain.
Collapse
Affiliation(s)
- Shaoheng Li
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fang Zhao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qinglian Tang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chuchu Xi
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jing He
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yujing Wang
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines and Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
15
|
Common and discrete mechanisms underlying chronic pain and itch: peripheral and central sensitization. Pflugers Arch 2021; 473:1603-1615. [PMID: 34245379 DOI: 10.1007/s00424-021-02599-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 05/26/2021] [Accepted: 06/22/2021] [Indexed: 12/30/2022]
Abstract
Normally, an obvious antagonism exists between pain and itch. In normal conditions, painful stimuli suppress itch sensation, whereas pain killers often generate itch. Although pain and itch are mediated by separate pathways under normal conditions, most chemicals are not highly specific to one sensation in chronic pathologic conditions. Notably, in patients with neuropathic pain, histamine primarily induces pain rather than itch, while in patients with atopic dermatitis, bradykinin triggers itch rather than pain. Accordingly, repetitive scratching even enhances itch sensation in chronic itch conditions. Physicians often prescribe pain relievers to patients with chronic itch, suggesting common mechanisms underlying chronic pain and itch, especially peripheral and central sensitization. Rather than separating itch and pain, studies should investigate chronic itch and pain including neuropathic and inflammatory conditions. Here, we reviewed chronic sensitization leading to chronic pain and itch at both peripheral and central levels. Studies investigating the connection between pain and itch facilitate the development of new therapeutics against both chronic dysesthesias based on the underlying pathophysiology.
Collapse
|
16
|
Kotliarova A, Sidorova YA. Glial Cell Line-Derived Neurotrophic Factor Family Ligands, Players at the Interface of Neuroinflammation and Neuroprotection: Focus Onto the Glia. Front Cell Neurosci 2021; 15:679034. [PMID: 34220453 PMCID: PMC8250866 DOI: 10.3389/fncel.2021.679034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/21/2021] [Indexed: 12/25/2022] Open
Abstract
Well-known effects of neurotrophic factors are related to supporting the survival and functioning of various neuronal populations in the body. However, these proteins seem to also play less well-documented roles in glial cells, thus, influencing neuroinflammation. This article summarizes available data on the effects of glial cell line derived neurotrophic factor (GDNF) family ligands (GFLs), proteins providing trophic support to dopaminergic, sensory, motor and many other neuronal populations, in non-neuronal cells contributing to the development and maintenance of neuropathic pain. The paper also contains our own limited data describing the effects of small molecules targeting GFL receptors on the expression of the satellite glial marker IBA1 in dorsal root ganglia of rats with surgery- and diabetes-induced neuropathy. In our experiments activation of GFLs receptors with either GFLs or small molecule agonists downregulated the expression of IBA1 in this tissue of experimental animals. While it can be a secondary effect due to a supportive role of GFLs in neuronal cells, growing body of evidence indicates that GFL receptors are expressed in glial and peripheral immune system cells. Thus, targeting GFL receptors with either proteins or small molecules may directly suppress the activation of glial and immune system cells and, therefore, reduce neuroinflammation. As neuroinflammation is considered to be an important contributor to the process of neurodegeneration these data further support research efforts to modulate the activity of GFL receptors in order to develop disease-modifying treatments for neurodegenerative disorders and neuropathic pain that target both neuronal and glial cells.
Collapse
Affiliation(s)
- Anastasiia Kotliarova
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Yulia A Sidorova
- Laboratory of Molecular Neuroscience, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
17
|
Huang B, Zdora I, de Buhr N, Lehmbecker A, Baumgärtner W, Leitzen E. Phenotypical peculiarities and species-specific differences of canine and murine satellite glial cells of spinal ganglia. J Cell Mol Med 2021; 25:6909-6924. [PMID: 34096171 PMCID: PMC8278083 DOI: 10.1111/jcmm.16701] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/16/2022] Open
Abstract
Satellite glial cells (SGCs) are located in the spinal ganglia (SG) of the peripheral nervous system and tightly envelop each neuron. They preserve tissue homeostasis, protect neurons and react in response to injury. This study comparatively characterizes the phenotype of murine (mSGCs) and canine SGCs (cSGCs). Immunohistochemistry and immunofluorescence as well as 2D and 3D imaging techniques were performed to describe a SGC-specific marker panel, identify potential functional subsets and other phenotypical, species-specific peculiarities. Glutamine synthetase (GS) and the potassium channel Kir 4.1 are SGC-specific markers in murine and canine SG. Furthermore, a subset of mSGCs showed CD45 immunoreactivity and the majority of mSGCs were immunopositive for neural/glial antigen 2 (NG2), indicating an immune and a progenitor cell character. The majority of cSGCs were immunopositive for glial fibrillary acidic protein (GFAP), 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNPase) and Sox2. Therefore, cSGCs resemble central nervous system glial cells and progenitor cells. SGCs lacked expression of macrophage markers CD107b, Iba1 and CD204. Double labelling with GS/Kir 4.1 highlights the unique anatomy of SGC-neuron units and emphasizes the indispensability of further staining and imaging techniques for closer insights into the specific distribution of markers and potential colocalizations.
Collapse
Affiliation(s)
- Bei Huang
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Isabel Zdora
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Nicole de Buhr
- Department of Biochemistry, University of Veterinary Medicine, Hannover, Germany.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine, Hannover, Germany
| | - Annika Lehmbecker
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany.,Center of Systems Neuroscience, Hannover, Germany
| | - Eva Leitzen
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany
| |
Collapse
|
18
|
Leo M, Schmitt LI, Kutritz A, Kleinschnitz C, Hagenacker T. Cisplatin-induced activation and functional modulation of satellite glial cells lead to cytokine-mediated modulation of sensory neuron excitability. Exp Neurol 2021; 341:113695. [PMID: 33727094 DOI: 10.1016/j.expneurol.2021.113695] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/01/2021] [Accepted: 03/11/2021] [Indexed: 12/23/2022]
Abstract
Cisplatin plays an essential role in the treatment of various cancers. Cisplatin exhibits high efficacy, but it often leads to severe neurotoxic side effects, such as chemotherapy-induced polyneuropathy (CIPN). The pathophysiology of CIPN is not fully understood. There is increasing evidence for damage to satellite glial cells (SGC) and dorsal root ganglion (DRG) neurons. We investigated the influence of cisplatin on the function of SGCs and the direct influence on DRGs. Satellite glial cells were isolated from DRG and exposed to 0.1, 1, 10, or 100 μM cisplatin for 2 h, 4 h, and 24 h. Using immunocytochemical staining and Western blot analysis, the expression of the glial fibrillary acid protein (GFAP), reactive oxygen species (ROS), and inward rectifier potassium channel 4.1 (Kir4.1) was determined. An increase in the immune reactivity (IR) and protein levels of GFAP and ROS was measured, and a reduction of IR and protein level of Kir4.1 was detected. A decrease in these channels' current density was observed using the whole-cell patch-clamp recording. The interleukin-6 (IL-6) and tumor necrosis factor α (TNFα) release of SGCs increased after cisplatin exposure as measured using ELISA, and interleukin-1β (IL-1β) decreased. The SGC-secreted factors in the supernatant after cisplatin treatment led to a modulation of cultured DRG neurons' excitability. Taken together, the modulation and function of different SGC proteins could be linked to a direct impact of cisplatin. Further, SGC-secreted factors influenced the excitability of sensory neurons. Overall, SGCs could be a potential target in preventing and treating chemotherapy-induced neuropathic pain.
Collapse
Affiliation(s)
- Markus Leo
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany.
| | - Linda-Isabell Schmitt
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Andrea Kutritz
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Tim Hagenacker
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| |
Collapse
|
19
|
Use of low-dose naltrexone in the management of chronic pain conditions: A systematic review. J Am Dent Assoc 2021; 151:891-902.e1. [PMID: 33228882 DOI: 10.1016/j.adaj.2020.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND The authors aimed to evaluate the efficacy of low-dose naltrexone in the management of chronic pain conditions and determine its potential use in orofacial pain management. METHODS A comprehensive literature review was completed in the PubMed/MEDLINE, Embase, Cumulated Index to Nursing and Allied Health Literature, Dentistry and Oral Sciences Source Library databases up through June 17, 2019, using terms such as neurogenic, inflammation, naltrexone, temporomandibular, and chronic pain. The primary outcome was reduction in pain intensity and, secondarily, improvement in quality of life. RESULTS A total of 793 studies were obtained with the initial search and 8 articles were selected for evaluation. Of these 8 articles, 4 were case reports, 3 were clinical studies, and 1 was a randomized controlled trial. Six studies included data on fibromyalgia, 2 studies included data on chronic regional pain syndrome, and 1 examined multiple diagnoses, including fibromyalgia, interstitial cystitis, and chronic pelvic pain. The primary outcome of all of the studies was pain intensity reduction. CONCLUSIONS AND PRACTICAL IMPLICATIONS Low-dose naltrexone provides an alternative in medical management of chronic pain disorders as a novel anti-inflammatory and immunomodulator. It can offer additional management options, as orofacial pain conditions share characteristics with other chronic pain disorders. Owing to the size and heterogeneity of the studies, more large-scale studies are needed, along with additional studies assessing orofacial pain response to low-dose naltrexone.
Collapse
|
20
|
Du E, Wang A, Fan R, Rong L, Yang R, Xing J, Shi X, Qiao B, Yu R, Xu C. Catestatin enhances ATP-induced activation of glial cells mediated by purinergic receptor P2X 4. J Recept Signal Transduct Res 2021; 42:160-168. [PMID: 33504266 DOI: 10.1080/10799893.2021.1878536] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The activation of glial cells and its possible mechanism play an extremely important role in understanding the pathophysiological process of some clinical diseases, and catestatin (CST) is involved in regulating this activation. In this project, we found that CST could enhance the activation of satellite glial cells (SGCs) and microglial cells and that the expression of P2X4 was increased; the co-expression of the P2X4 receptor with glial fibrillary acidic protein (GFAP) and the P2X4 receptor with CD11b was also increased significantly in glial cells of the ATP + CST group, and TNF-α and IL-1β also showed a rising trend; the expression of phosphorylated ERK1/2 was also increased in the ATP + CST group. In summary, we conclude that CST could enhance ATP-induced activation of SGCs and microglial cells mediated by the P2X4 receptor and that the ERK1/2 signaling pathway may be involved in this activation process.
Collapse
Affiliation(s)
- Errong Du
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, P.R. China
| | - Anhui Wang
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, P.R. China
| | - Rongping Fan
- The Fourth Clinical Medical College of Nanchang University, Nanchang, P.R. China
| | - Lilou Rong
- The Fourth Clinical Medical College of Nanchang University, Nanchang, P.R. China
| | - Runan Yang
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, P.R. China
| | - Juping Xing
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, P.R. China
| | - Xiangchao Shi
- Queen Mary School, Medical Department, Nanchang University-Queen Mary University of London, Nanchang, P.R. China
| | - Bao Qiao
- Queen Mary School, Medical Department, Nanchang University-Queen Mary University of London, Nanchang, P.R. China
| | - Ruoyang Yu
- Queen Mary School, Medical Department, Nanchang University-Queen Mary University of London, Nanchang, P.R. China
| | - Changshui Xu
- Department of Physiology, Basic Medical College of Nanchang University, Nanchang, P.R. China.,Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, P.R. China
| |
Collapse
|
21
|
Zhang Y, Standifer KM. Exacerbated Headache-Related Pain in the Single Prolonged Stress Preclinical Model of Post-traumatic Stress Disorder. Cell Mol Neurobiol 2020; 41:1009-1018. [PMID: 32930941 PMCID: PMC8159770 DOI: 10.1007/s10571-020-00962-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 09/03/2020] [Indexed: 11/29/2022]
Abstract
Chronic headache pain is one of the most commonly reported comorbid pain conditions with post-traumatic stress disorder (PTSD) patients and resistant to effective treatment, yet no combined preclinical model of the two disorders has been reported. Here, we used a modified chronic headache pain model to investigate the contribution of single prolonged stress (SPS) model of PTSD with sodium nitroprusside (SNP)-induced hyperalgesia. Injection of SNP (2 mg/kg, i.p.) occurred every other day from day 7 to day 15 after initiation of SPS in rats. Paw withdrawal threshold (PWT) to von Frey stimuli and tail flick latencies (TFL) dramatically decreased as early as 7 days after SPS and lasted until at least day 21. Basal PWT and TFL also significantly decreased during the SNP treatment period. The lower nociceptive thresholds recovered in 6 days following the final SNP injection in SNP group, but not in SPS + SNP group. Elevated nociceptin/OFQ (N/OFQ) levels observed in cerebrospinal fluid of SPS rats were even higher in SPS + SNP group. Glial fibrillary acidic protein (GFAP) and N/OFQ peptide (NOP) receptor mRNA expression increased in dorsal root ganglia (DRG) 21 days after SPS exposure; mRNA increases in the SPS/SNP group was more pronounced than SPS or SNP alone. GFAP protein expression was upregulated in trigeminal ganglia by SPS. Our results indicate that traumatic stress exaggerated chronic SNP-induced nociceptive hypersensitivity, and that N/OFQ and activated satellite glia cells may play an important role in the interaction between both conditions.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kelly M Standifer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA. .,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
22
|
Implication of Neuronal Versus Microglial P2X4 Receptors in Central Nervous System Disorders. Neurosci Bull 2020; 36:1327-1343. [PMID: 32889635 DOI: 10.1007/s12264-020-00570-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/06/2020] [Indexed: 02/08/2023] Open
Abstract
The P2X4 receptor (P2X4) is an ATP-gated cation channel that is highly permeable to Ca2+ and widely expressed in neuronal and glial cell types throughout the central nervous system (CNS). A growing body of evidence indicates that P2X4 plays key roles in numerous central disorders. P2X4 trafficking is highly regulated and consequently in normal situations, P2X4 is present on the plasma membrane at low density and found mostly within intracellular endosomal/lysosomal compartments. An increase in the de novo expression and/or surface density of P2X4 has been observed in microglia and/or neurons during pathological states. This review aims to summarize knowledge on P2X4 functions in CNS disorders and provide some insights into the relative contributions of neuronal and glial P2X4 in pathological contexts. However, determination of the cell-specific functions of P2X4 along with its intracellular and cell surface roles remain to be elucidated before its potential as a therapeutic target in multiple disorders can be defined.
Collapse
|
23
|
Schmitt LI, Leo M, Kutritz A, Kleinschnitz C, Hagenacker T. Activation and functional modulation of satellite glial cells by oxaliplatin lead to hyperexcitability of sensory neurons in vitro. Mol Cell Neurosci 2020; 105:103499. [PMID: 32389805 DOI: 10.1016/j.mcn.2020.103499] [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: 12/12/2019] [Revised: 03/28/2020] [Accepted: 05/03/2020] [Indexed: 01/24/2023] Open
Abstract
Platinum-based chemotherapeutics still play an important role in cancer therapy, however, severe side effects, such as painful neuropathy, occur frequently. The pathophysiologic mechanisms depend on the applied chemotherapeutic agent and are still controversial. In addition to neuronal damage, disturbance of glial cell activity may contribute to neurotoxicity. Here, we focused on the effect of oxaliplatin on satellite glial cell (SGC) function and on the activity of the dorsal root ganglion (DRG) neurons. SGCs were isolated as high-purity cultures and treated with 1 and 10 μM oxaliplatin for 2, 4 and 24 h. Subsequently, glial fibrillary acid protein (GFAP), reactive oxygen species (ROS), Connexin-43 (Cx-43), and inward rectifier potassium channel 4.1 (Kir4.1) expression was determined by immunocytochemical staining (ICC) and Western blot analyses. Immunochemical staining and Western blot analysis showed an increase in the immune reactivity (IR) and protein levels of ROS, GFAP, and Cx-43. Furthermore, reduction of the IR and protein levels and current density were demonstrated using patch-clamp measurements, of Kir4.1 channels after oxaliplatin exposure. Cytokine release in SGCs was measured using enzyme-linked immunosorbent assays (ELISA) after oxaliplatin exposure and indicated an increased release of IL-6 and TNFα, while IL-1β was decreased. The direct influence of SGC-secreted factors in the supernatant after oxaliplatin treatment led to the hyperexcitability of cultured DRG neurons. In summary, oxaliplatin has a direct impact on the modulation and function of different SGC proteins. Furthermore, SGC-released factors influence the excitability of sensory neurons, qualifying SGCs as potential targets for the prevention and treatment of oxaliplatin-induced polyneuropathy.
Collapse
Affiliation(s)
| | - Markus Leo
- Department of Neurology, Neuroscience Lab, University Hospital Essen, Germany
| | - Andrea Kutritz
- Department of Neurology, Neuroscience Lab, University Hospital Essen, Germany
| | | | - Tim Hagenacker
- Department of Neurology, Neuroscience Lab, University Hospital Essen, Germany.
| |
Collapse
|
24
|
Mandge D, Shukla PR, Bhatnagar A, Manchanda R. Computational Model for Cross-Depolarization in DRG Neurons via Satellite Glial Cells using [K] o: Role of Kir4.1 Channels and Extracellular Leakage. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:2320-2323. [PMID: 31946364 DOI: 10.1109/embc.2019.8857153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Satellite glial cells (SGCs) are glial cells found in the peripheral nervous system where they tightly envelop the somata of the primary sensory neurons such as dorsal root ganglion (DRG) neurons and nodose ganglion (NG) neurons. The somata of these neurons are generally compactly packed in their respective ganglia (DRG and NG). SGCs covering a neuron behave as an insulator of electrical activity from neighbouring neurons within the ganglion. Several studies have however shown that the somata show "cross-depolarization" (CD). Origin of CDs has been hypothesized to be chemical in nature: either from neurotransmitter release from both SGCs and somata or from elevation of extracellular potassium concentration ([K]o) in the vicinity of somata. Here, we investigate the role of Kir4.1 channels on SGC and diffusion/clearance factor (β) of [K]o from the space between SGC and DRG neuron somata to the bulk extracellular space in ganglion. We show using two "Soma-SGC Units" interacting via gap junction that a combination of Kir4.1 and β could be responsible for CD between DRG neuron somata in pathological conditions.
Collapse
|
25
|
Kumar S, Vinayak M. NADPH oxidase1 inhibition leads to regression of central sensitization during formalin induced acute nociception via attenuation of ERK1/2-NFκB signaling and glial activation. Neurochem Int 2019; 134:104652. [PMID: 31891736 DOI: 10.1016/j.neuint.2019.104652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/02/2019] [Accepted: 12/19/2019] [Indexed: 12/18/2022]
Abstract
Role of NADPH oxidase1 in the development of inflammatory pain has been demonstrated by gene knockout studies. Nevertheless, pharmacological inhibition of NOX1 is a requisite approach for therapeutic utility. Recently, we have reported the anti-nociceptive effect of newly identified NOX1 specific inhibitor ML171 (2-acetylphenothiazine). Inhibition of NOX1 resulted in attenuation of nociceptive sensitization during acute inflammatory pain via inhibition of ROS generation and its downstream ERK1/2 activation. However, glial activation accompanying inflammation is closely related to the initiation and maintenance of pain. Peripheral nociceptive inputs activate the primary afferents via release of various chemical mediators which are potentially capable of mediating signals from neuron to glia in DRG and subsequently in spinal cord dorsal horn. The subsequent interactions between neuron and glia contribute to pain hypersensitivity. Thus, the present study was focused to investigate the effect of ML171 on ERK1/2 signaling, glial activation, and crosstalk between neuron and glia in a mouse model of formalin induced acute nociception. Thus, the present study was focused to investigate the effect of ML171 on ERK1/2 signaling, glial activation, and crosstalk between neuron and glia in DRG and dorsal horn of the spinal cord of lumbar region (L3-L5) in a mouse model of formalin induced acute nociception. Intraperitoneal administration of ML171 decreased nociceptive behavioral responses, i.e. the flinch and lick counts, in formalin induced nociceptive mice. Immunofluorescence and Western blot analysis demonstrated decreased levels of nociceptive mediators like p-ERK1/2, p-NFκB p65, Iba1 and GFAP in DRG as well as in spinal cord dorsal horn; supporting anti-nociceptive potential of ML171. Further, co-localization studies showed the neuron-glia crosstalk in tissue dependent manner. ERK1/2 was found to be activated in glia and NFκB in neurons in DRG; whereas in case of spinal cord ERK1/2 was activated in neurons and NFκB in astrocytes. Decrease in nociceptive behavioral response and activation of nociceptive mediators after intraperitoneal administration of ML171 strongly advocate anti-nociceptive potential of ML171. This is the first report demonstrating modulation of ERK1/2-NFκB signaling pathway, glial activation and regulation of neuron-glia crosstalk by NADPH oxidase1 inhibition towards its anti-nociceptive action.
Collapse
Affiliation(s)
- Sanjay Kumar
- Department of Zoology, Biochemistry and Molecular Biology Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Manjula Vinayak
- Department of Zoology, Biochemistry and Molecular Biology Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
26
|
Propargylglycine decreases neuro-immune interaction inducing pain response in temporomandibular joint inflammation model. Nitric Oxide 2019; 93:90-101. [DOI: 10.1016/j.niox.2019.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 09/22/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022]
|
27
|
Zhang Q, Zhao J, Shen J, Zhang X, Ren R, Ma Z, He Y, Kang Q, Wang Y, Dong X, Sun J, Liu Z, Yi X. The ATP-P2X7 Signaling Pathway Participates in the Regulation of Slit1 Expression in Satellite Glial Cells. Front Cell Neurosci 2019; 13:420. [PMID: 31607866 PMCID: PMC6761959 DOI: 10.3389/fncel.2019.00420] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/02/2019] [Indexed: 11/17/2022] Open
Abstract
Slit1 is one of the known signaling factors of the slit family and can promote neurite growth by binding to its receptor, Robo2. Upregulation of Slit1 expression in dorsal root ganglia (DRG) after peripheral nerve injury plays an important role in nerve regeneration. Each sensory neuronal soma in the DRG is encapsulated by several surrounding satellite glial cells (SGCs) to form a neural structural unit. However, the temporal and spatial patterns of Slit1 upregulation in SGCs in DRG and its molecular mechanisms are not well understood. This study examined the spatial and temporal patterns of Slit1 expression in DRG after sciatic nerve crush by immunohistochemistry and western blotting. The effect of neuronal damage signaling on the expression of Slit1 in SGCs was studied in vivo by fluorescent gold retrograde tracing and double immunofluorescence staining. The relationship between the expression of Slit1 in SGCs and neuronal somas was also observed by culturing DRG cells and double immunofluorescence labeling. The molecular mechanism of Slit1 was further explored by immunohistochemistry and western blotting after intraperitoneal injection of Bright Blue G (BBG, P2X7R inhibitor). The results showed that after peripheral nerve injury, the expression of Slit1 in the neurons and SGCs of DRG increased. The expression of Slit1 was presented with a time lag in SGCs than in neurons. The expression of Slit1 in SGCs was induced by contact with surrounding neuronal somas. Through injured cell localization, it was found that the expression of Slit1 was stronger in SGCs surrounding injured neurons than in SGCs surrounding non-injured neurons. The expression of vesicular nucleotide transporter (VNUT) in DRG neurons was increased by injury signaling. After the inhibition of P2X7R, the expression of Slit1 in SGCs was downregulated, and the expression of VNUT in DRG neurons was upregulated. These results indicate that the ATP-P2X7R pathway is involved in signal transduction from peripheral nerve injury to SGCs, leading to the upregulation of Slit1 expression.
Collapse
Affiliation(s)
- Quanpeng Zhang
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Jiuhong Zhao
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Jing Shen
- Department of Ophthalmology, First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xianfang Zhang
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Rui Ren
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Zhijian Ma
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Yuebin He
- Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| | - Qian Kang
- Infection Control Department, People's Hospital of Xing'an County, Guilin, China
| | - Yanshan Wang
- Quality Inspection Department, Minghui Industry (Shenzhen) Co., Ltd., Shenzhen, China
| | - Xu Dong
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, China
| | - Jin Sun
- Department of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Zhuozhou Liu
- Department of Clinical Medicine, Hainan Medical University, Haikou, China
| | - Xinan Yi
- Department of Anatomy, Hainan Medical University, Haikou, China.,Joint Laboratory for Neuroscience, Hainan Medical University, Fourth Military Medical University, Haikou, China
| |
Collapse
|
28
|
Abstract
Permanent disabilities following CNS injuries result from the failure of injured axons to regenerate and rebuild functional connections with their original targets. By contrast, injury to peripheral nerves is followed by robust regeneration, which can lead to recovery of sensory and motor functions. This regenerative response requires the induction of widespread transcriptional and epigenetic changes in injured neurons. Considerable progress has been made in recent years in understanding how peripheral axon injury elicits these widespread changes through the coordinated actions of transcription factors, epigenetic modifiers and, to a lesser extent, microRNAs. Although many questions remain about the interplay between these mechanisms, these new findings provide important insights into the pivotal role of coordinated gene expression and chromatin remodelling in the neuronal response to injury.
Collapse
Affiliation(s)
- Marcus Mahar
- Department of Neuroscience, Hope Center for Neurological Disorders and Center of Regenerative Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Valeria Cavalli
- Department of Neuroscience, Hope Center for Neurological Disorders and Center of Regenerative Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
29
|
Zhu G, Dai B, Chen Z, He L, Guo J, Dan Y, Liang S, Li G. Effects of chronic lead exposure on the sympathoexcitatory response associated with the P2X7 receptor in rat superior cervical ganglia. Auton Neurosci 2019; 219:33-41. [DOI: 10.1016/j.autneu.2019.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 02/01/2019] [Accepted: 03/20/2019] [Indexed: 12/23/2022]
|
30
|
Neonatal vincristine administration modulates intrinsic neuronal excitability in the rat dorsal root ganglion and spinal dorsal horn during adolescence. Pain 2019; 160:645-657. [PMID: 30681983 DOI: 10.1097/j.pain.0000000000001444] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Our recent work has shown that the early-life administration of vincristine (VNC), commonly used to treat pediatric cancers, evokes mechanical pain hypersensitivity in rats that emerges during adolescence and persists into adulthood. However, the underlying mechanisms remain unclear, as nothing is known about how neonatal VNC treatment influences peripheral and central nociceptive processing at the cellular level. Here, we used in vitro intracellular microelectrode and whole-cell patch-clamp recordings to evaluate the consequences of early-life VNC administration on the intrinsic membrane properties of adolescent dorsal root ganglion and spinal superficial dorsal horn neurons. The results demonstrate that VNC treatment increased the prevalence and rate of repetitive firing in both large- and medium-diameter sensory neurons, while reducing repetitive firing in small-diameter neurons, in comparison with vehicle-treated littermate controls. By contrast, passive membrane properties and peripheral conduction velocities were similar between experimental groups across all classes of primary afferents. Within the adolescent superficial dorsal horn, neonatal VNC exposure significantly enhanced the intrinsic membrane excitability of lamina I spinoparabrachial neurons, as evidenced by a decrease in rheobase and elevation of repetitive firing frequency compared with controls. Meanwhile, putative interneurons within lamina I exhibited a reduction in repetitive action potential discharge after early-life chemotherapy. Collectively, these findings suggest that neonatal VNC treatment evokes cell type-specific changes in intrinsic excitability at multiple levels of the ascending pain pathway. Overall, this work lays an essential foundation for the future exploration of the ionic mechanisms that drive chemotherapy-induced chronic pain in children and adolescents.
Collapse
|
31
|
Arichi S, Sasaki-Hamada S, Kadoya Y, Ogata M, Ishibashi H. Excitatory effect of bradykinin on intrinsic neurons of the rat heart. Neuropeptides 2019; 75:65-74. [PMID: 31047706 DOI: 10.1016/j.npep.2019.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/07/2019] [Accepted: 04/23/2019] [Indexed: 01/16/2023]
Abstract
The heart receives sympathetic and parasympathetic innervation through the intrinsic cardiac nervous system. Although bradykinin (BK) has negative inotropic and chronotropic properties of cardiac contraction, the direct effect of BK on the intrinsic neural network of the heart is still unclear. In the present study, the effect of BK on the intracardiac ganglion neurons isolated from rats was investigated using the perforated patch-clamp technique. Under current-clamp conditions, application of 0.1 μM BK depolarized the membrane, accompanied by repetitive firing of action potentials. When BK was applied repeatedly, the second responses were considerably less intense than the first application. The BK action was fully inhibited by the B2 receptor antagonist Hoe-140, but not by the B1 receptor antagonist des-Arg9-[Leu8]-BK. The BK response was mimicked by the B2 agonist [Hyp3]-BK. The BK-induced depolarization was inhibited by the phospholipase C inhibitor U-73122. BK evoked inward currents under voltage-clamp conditions at a holding potential of -60 mV. Removal of extracellular Ca2+ markedly increased the BK-induced currents, suggesting an involvement of Ca2+-permeable non-selective cation channels. The muscarinic agonist oxotremorine-M (OxoM) also elicited the extracellular Ca2+-sensitive cationic currents. The OxoM response did not exhibit rundown with repeated agonist application. The amplitude of current evoked by 1 μM OxoM was comparable to that induced by 0.1 μM BK. Co-application of 0.1 μM BK and 1 μM OxoM elicited the current whose peak amplitude was almost the same as that elicited by OxoM alone, suggesting that BK and OxoM activate same cation channels. BK also reduced the amplitude of M-current, while the M-current inhibitor XE-991 affected neither resting membrane potential nor the BK-induced depolarization. From these results, we suggest that BK regulates excitability of intrinsic cardiac neurons by both an activation of non-selective cation channels and an inhibition of M-type K+ channels through B2 receptors.
Collapse
Affiliation(s)
- Shiho Arichi
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan; Department of Brain Science, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan
| | - Sachie Sasaki-Hamada
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan
| | - Yuichi Kadoya
- Department of Anatomical Science, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan
| | - Masanori Ogata
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan; Department of Brain Science, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan
| | - Hitoshi Ishibashi
- Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara 252-0373, Japan; Department of Brain Science, Kitasato University Graduate School of Medical Sciences, Sagamihara 252-0373, Japan.
| |
Collapse
|
32
|
Role of Na V1.6 and Na Vβ4 Sodium Channel Subunits in a Rat Model of Low Back Pain Induced by Compression of the Dorsal Root Ganglia. Neuroscience 2019; 402:51-65. [PMID: 30699332 DOI: 10.1016/j.neuroscience.2019.01.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 01/18/2023]
Abstract
Low back pain is a common cause of chronic pain and disability. It is modeled in rodents by chronically compressing the lumbar dorsal root ganglia (DRG) with small metal rods, resulting in ipsilateral mechanical and cold hypersensitivity, and hyperexcitability of sensory neurons. Sodium channels are implicated in this hyperexcitability, but the responsible isoforms are unknown. In this study, we used siRNA-mediated knockdown of the pore-forming NaV1.6 and regulatory NaVβ4 sodium channel isoforms that have been previously implicated in a different model of low back pain caused by locally inflaming the L5 DRG. Knockdown of either subunit markedly reduced spontaneous pain and mechanical and cold hypersensitivity induced by DRG compression, and reduced spontaneous activity and hyperexcitability of sensory neurons with action potentials <1.5 msec (predominately cells with myelinated axons, based on conduction velocities measured in a subset of cells) 4 days after DRG compression. These results were similar to those previously obtained in the DRG inflammation model and some neuropathic pain models, in which sensory neurons other than nociceptors seem to play key roles. The cytokine profiles induced by DRG compression and DRG inflammation were also very similar, with upregulation of several type 1 pro-inflammatory cytokines and downregulation of type 2 anti-inflammatory cytokines. Surprisingly, the cytokine profile was largely unaffected by NaVβ4 knockdown in either model. The NaV1.6 channel, and the NaVβ4 subunit that can regulate NaV1.6 to enhance repetitive firing, play key roles in both models of low back pain; targeting the abnormal spontaneous activity they generate may have therapeutic value.
Collapse
|
33
|
Zhang L, Xie R, Yang J, Zhao Y, Qi C, Bian G, Wang M, Shan J, Wang C, Wang D, Luo C, Wang Y, Wu S. Chronic pain induces nociceptive neurogenesis in dorsal root ganglia from Sox2‐positive satellite cells. Glia 2019; 67:1062-1075. [DOI: 10.1002/glia.23588] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/15/2018] [Accepted: 12/21/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Li Zhang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
- Department of Anatomy, Institute of Basic Medical Science Xi'an Medical University Xi'an Shaanxi China
| | - Rougang Xie
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
| | - Jiping Yang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
- Department of Anatomy, Institute of Basic Medical Science Xi'an Medical University Xi'an Shaanxi China
| | - Youyi Zhao
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
| | - Chuchu Qi
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
| | - Ganlan Bian
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
| | - Mengmeng Wang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
| | - Junjia Shan
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
| | - Chen Wang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
| | - Dong Wang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
| | - Ceng Luo
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
| | - Yazhou Wang
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
| | - Shengxi Wu
- Department of Neurobiology, Institute of Neurosciences, School of Basic Medicine Fourth Military Medical University Xi'an Shaanxi China
| |
Collapse
|
34
|
Sorkin LS, Eddinger KA, Woller SA, Yaksh TL. Origins of antidromic activity in sensory afferent fibers and neurogenic inflammation. Semin Immunopathol 2018; 40:237-247. [PMID: 29423889 PMCID: PMC7879713 DOI: 10.1007/s00281-017-0669-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/28/2017] [Indexed: 10/18/2022]
Abstract
Neurogenic inflammation results from the release of biologically active agents from the peripheral primary afferent terminal. This release reflects the presence of releasable pools of active product and depolarization-exocytotic coupling mechanisms in the distal afferent terminal and serves to alter the physiologic function of innervated organ systems ranging from the skin and meninges to muscle, bone, and viscera. Aside from direct stimulation, this biologically important release from the peripheral afferent terminal can be initiated by antidromic activity arising from five anatomically distinct points of origin: (i) afferent collaterals at the peripheral-target organ level, (ii) afferent collaterals arising proximal to the target organ, (iii) from mid-axon where afferents lacking myelin sheaths (C fibers and others following demyelinating injuries) may display crosstalk and respond to local irritation, (iv) the dorsal root ganglion itself, and (v) the central terminals of the afferent in the dorsal horn where local circuits and bulbospinal projections can initiate the so-called dorsal root reflexes, i.e., antidromic traffic in the sensory afferent.
Collapse
Affiliation(s)
- Linda S Sorkin
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, USA.
| | - Kelly A Eddinger
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, USA
| | - Sarah A Woller
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, USA
| | - Tony L Yaksh
- Department of Anesthesiology, University of California, San Diego, San Diego, CA, USA
| |
Collapse
|
35
|
Pannese E. Biology and Pathology of Perineuronal Satellite Cells in Sensory Ganglia. BIOLOGY AND PATHOLOGY OF PERINEURONAL SATELLITE CELLS IN SENSORY GANGLIA 2018. [DOI: 10.1007/978-3-319-60140-3_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
36
|
Chen Y, Huang LYM. A simple and fast method to image calcium activity of neurons from intact dorsal root ganglia using fluorescent chemical Ca 2+ indicators. Mol Pain 2017; 13:1744806917748051. [PMID: 29212403 PMCID: PMC5731619 DOI: 10.1177/1744806917748051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chemical calcium indicators have been commonly used to monitor calcium (Ca2+) activity in cell bodies, i.e., somata, of isolated dorsal root ganglion neurons. Recent studies have shown that dorsal root ganglion somata play an essential role in soma–glia interactions and actively participate in the transmission of nociceptive signals. It is therefore desirable to develop methods to study Ca2+ activity in neurons and glia in intact dorsal root ganglia. In our previous studies, we found that incubation of intact dorsal root ganglia with acetoxymethyl dye resulted in efficient Ca2+ dye loading into glial cells but limited dye loading into neurons. Here, we introduce a useful method to load Ca2+ dyes in intact dorsal root ganglion neurons through electroporation. We found that electroporation greatly facilitated loading of Fluo-4 acetoxymethyl, Oregon green bapta-1-488 acetoxymethyl, and Fluo-4 pentapotassium salt into dorsal root ganglion neurons. In contrast, electroporation did not further facilitate dye loading into glia. Using electroporation followed by incubation of acetoxymethyl form Ca2+ dye, we can load acetoxymethyl Ca2+ dye well in both neurons and glia. With this approach, we found that inflammation induced by complete Freund’s adjuvant significantly increased the incidence of neuron–glia interactions in dorsal root ganglia. We also confirmed the actions of capsaicin and morphine on Ca2+ responses in dorsal root ganglion neurons. Thus, by promoting the loading of Ca2+ dye in neurons and glia through electroporation and incubation, Ca2+ activities in neurons and neuron–glia interactions can be well studied in intact dorsal root ganglia.
Collapse
Affiliation(s)
- Yong Chen
- 1 Department of Neuroscience, Cell Biology and Anatomy, 12338 University of Texas Medical Branch, Galveston , TX, USA
| | - Li-Yen M Huang
- 1 Department of Neuroscience, Cell Biology and Anatomy, 12338 University of Texas Medical Branch, Galveston , TX, USA
| |
Collapse
|
37
|
Canine dorsal root ganglia satellite glial cells represent an exceptional cell population with astrocytic and oligodendrocytic properties. Sci Rep 2017; 7:13915. [PMID: 29066783 PMCID: PMC5654978 DOI: 10.1038/s41598-017-14246-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/06/2017] [Indexed: 12/21/2022] Open
Abstract
Dogs can be used as a translational animal model to close the gap between basic discoveries in rodents and clinical trials in humans. The present study compared the species-specific properties of satellite glial cells (SGCs) of canine and murine dorsal root ganglia (DRG) in situ and in vitro using light microscopy, electron microscopy, and immunostainings. The in situ expression of CNPase, GFAP, and glutamine synthetase (GS) has also been investigated in simian SGCs. In situ, most canine SGCs (>80%) expressed the neural progenitor cell markers nestin and Sox2. CNPase and GFAP were found in most canine and simian but not murine SGCs. GS was detected in 94% of simian and 71% of murine SGCs, whereas only 44% of canine SGCs expressed GS. In vitro, most canine (>84%) and murine (>96%) SGCs expressed CNPase, whereas GFAP expression was differentially affected by culture conditions and varied between 10% and 40%. However, GFAP expression was induced by bone morphogenetic protein 4 in SGCs of both species. Interestingly, canine SGCs also stimulated neurite formation of DRG neurons. These findings indicate that SGCs represent an exceptional, intermediate glial cell population with phenotypical characteristics of oligodendrocytes and astrocytes and might possess intrinsic regenerative capabilities in vivo.
Collapse
|
38
|
Adães S, Almeida L, Potes CS, Ferreira AR, Castro-Lopes JM, Ferreira-Gomes J, Neto FL. Glial activation in the collagenase model of nociception associated with osteoarthritis. Mol Pain 2017; 13:1744806916688219. [PMID: 28326927 PMCID: PMC5302176 DOI: 10.1177/1744806916688219] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background Experimental osteoarthritis entails neuropathic-like changes in dorsal root ganglia (DRG) neurons. Since glial activation has emerged as a key player in nociception, being reported in numerous models of neuropathic pain, we aimed at evaluating if glial cell activation may also occur in the DRG and spinal cord of rats with osteoarthritis induced by intra-articular injection of collagenase. Methods Osteoarthritis was induced by two injections, separated by three days, of 500 U of type II collagenase into the knee joint of rats. Movement-induced nociception was evaluated by the Knee-Bend and CatWalk tests during the following six weeks. Glial fibrillary acidic protein (GFAP) expression in satellite glial cells of the DRG was assessed by immunofluorescence and Western Blot analysis; the pattern of GFAP and activating transcription factor-3 (ATF-3) expression was also compared through double immunofluorescence analysis. GFAP expression in astrocytes and IBA-1 expression in microglia of the L3–L5 spinal cord segments was assessed by immunohistochemistry and Western Blot analysis. The effect of the intrathecal administration of fluorocitrate, an inhibitor of glial activation, on movement-induced nociception was evaluated six weeks after the first collagenase injection. Results GFAP expression in satellite glial cells of collagenase-injected animals was significantly increased six weeks after osteoarthritis induction. Double immunofluorescence showed GFAP upregulation in satellite glial cells surrounding ATF-3-positive neurons. In the spinal cord of collagenase-injected animals, an ipsilateral upregulation of GFAP and IBA-1 was also observed. The inhibition of glial activation with fluorocitrate decreased movement- and loading-induced nociception. Conclusion Collagenase-induced knee osteoarthritis leads to the development of nociception associated with movement of the affected joint and to the activation of glial cells in both the DRG and the spinal cord. Inhibition of glial cell activation by fluorocitrate decreases these osteoarthritis-associated nociceptive behaviours. These results suggest that glial cell activation may play a role in the development of chronic pain in this experimental model of osteoarthritis.
Collapse
Affiliation(s)
- Sara Adães
- 1 Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,2 Morphysiology of the Somatosensory System Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,3 Departamento de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Lígia Almeida
- 1 Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,2 Morphysiology of the Somatosensory System Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,3 Departamento de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Catarina S Potes
- 1 Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,2 Morphysiology of the Somatosensory System Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,3 Departamento de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Ana Rita Ferreira
- 1 Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - José M Castro-Lopes
- 1 Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,2 Morphysiology of the Somatosensory System Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,3 Departamento de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Joana Ferreira-Gomes
- 1 Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,2 Morphysiology of the Somatosensory System Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,3 Departamento de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Fani L Neto
- 1 Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,2 Morphysiology of the Somatosensory System Group, Instituto de Biologia Molecular e Celular, Porto, Portugal.,3 Departamento de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| |
Collapse
|
39
|
Neuronal-Glial Interactions Maintain Chronic Neuropathic Pain after Spinal Cord Injury. Neural Plast 2017; 2017:2480689. [PMID: 28951789 PMCID: PMC5603132 DOI: 10.1155/2017/2480689] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/26/2017] [Accepted: 07/05/2017] [Indexed: 02/01/2023] Open
Abstract
The hyperactive state of sensory neurons in the spinal cord enhances pain transmission. Spinal glial cells have also been implicated in enhanced excitability of spinal dorsal horn neurons, resulting in pain amplification and distortions. Traumatic injuries of the neural system such as spinal cord injury (SCI) induce neuronal hyperactivity and glial activation, causing maladaptive synaptic plasticity in the spinal cord. Recent studies demonstrate that SCI causes persistent glial activation with concomitant neuronal hyperactivity, thus providing the substrate for central neuropathic pain. Hyperactive sensory neurons and activated glial cells increase intracellular and extracellular glutamate, neuropeptides, adenosine triphosphates, proinflammatory cytokines, and reactive oxygen species concentrations, all of which enhance pain transmission. In addition, hyperactive sensory neurons and glial cells overexpress receptors and ion channels that maintain this enhanced pain transmission. Therefore, post-SCI neuronal-glial interactions create maladaptive synaptic circuits and activate intracellular signaling events that permanently contribute to enhanced neuropathic pain. In this review, we describe how hyperactivity of sensory neurons contributes to the maintenance of chronic neuropathic pain via neuronal-glial interactions following SCI.
Collapse
|
40
|
Yu Y, Huang X, Di Y, Qu L, Fan N. Effect of CXCL12/CXCR4 signaling on neuropathic pain after chronic compression of dorsal root ganglion. Sci Rep 2017; 7:5707. [PMID: 28720830 PMCID: PMC5515923 DOI: 10.1038/s41598-017-05954-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 06/07/2017] [Indexed: 01/16/2023] Open
Abstract
Neuropathic pain is a complex, chronic pain state that often accompanies tissue damage, inflammation or injury of the nervous system. However the underlying molecular mechanisms still remain unclear. Here, we showed that CXCL12 and CXCR4 were upregulated in the dorsal root ganglion (DRG) after chronic compression of DRG (CCD), and some CXCR4 immunopositive neurons were also immunopositive for the nociceptive neuronal markers IB4, TRPV1, CGRP, and substance P. The incidence and amplitude of CXCL12-induced Ca2+ response in primary sensory neurons from CCD mice was significantly increased compared to those from control animals. CXCL12 depolarized the resting membrane potential, decreased the rheobase, and increased the number of action potentials evoked by a depolarizing current at 2X rheobase in neurons from CCD mice. The mechanical and thermal hypernociception after CCD was attenuated by administration of a CXCR4 antagonist AMD3100. These findings suggest that CXCL12/CXCR4 signaling contributes to hypernociception after CCD, and targeting CXCL12/CXCR4 signaling pathway may alleviate neuropathic pain.
Collapse
Affiliation(s)
- Yang Yu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province, 510370, China
| | - Xini Huang
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province, 510370, China
| | - Yuwei Di
- Department of Pathology and Laboratory Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Lintao Qu
- Department of Neurosurgery, Neurosurgery Pain Research Institute, Johns Hopkins University School of Medicine, 725N. Wolfe St., Baltimore, MD, 21205, USA
| | - Ni Fan
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), 36 Mingxin Road, Liwan District, Guangzhou, Guangdong Province, 510370, China.
| |
Collapse
|
41
|
Li X, Jiang S, Yang H, Liao Q, Cao S, Yan X, Huang D. Breakthrough Cancer Pain Is Associated with Spinal Gap Junction Activation via Regulation of Connexin 43 in a Mouse Model. Front Cell Neurosci 2017; 11:207. [PMID: 28769766 PMCID: PMC5511832 DOI: 10.3389/fncel.2017.00207] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/30/2017] [Indexed: 12/21/2022] Open
Abstract
Breakthrough cancer pain (BTcP) is a high-intensity, short-duration, unpredictable and uncontrollable pain. Recent studies have shown that activation of gap junction (GJ) in spinal cord plays an important role in the pathogenesis of BTcP. We examined the expressions of Glial fibrillary acidic protein (GFAP), connexin (Cx) 43 protein and phosphorylation of Cx43 (p-Cx43) in the spinal cord of mice. In addition, we investigated the effects of Gap26, a selective GJ blocker, on the expressions of GFAP, Cx43 and p-Cx43 in BTcP mice. We found that the expressions of GFAP and Cx43 proteins were significantly upregulated while p-Cx43 was down-regulated in the spinal cord in a mouse model of BTcP. The overexpression of Cx43 protein in the spinal cord increased GJ formation and enhanced BTcP. The variation of the ratio of p-Cx43/T-Cx43 (total Cx43) affected the function of GJ to induce BTcP. Furthermore, BTcP was alleviated by Gap26 via reducing pain hypersensitivity. The inhibition of Cx43 and p-Cx43 by Gap26 attenuated BTcP but the p/T ratio of Cx43 remained unchanged in BTcP mice. We reveal that the expression and phosphorylation of Cx43 affected BTcP and GJ activation facilitated BTcP via a Cx43-mediated signaling in the spinal cord. The finding may provide a scientific rationale for discovery and development of novel therapeutic targets for the treatment of BTcP clinically.
Collapse
Affiliation(s)
- Xin Li
- Department of Pain, The Third Xiangya Hospital and Institute of Pain Medicine, Central South UniversityChangsha, China
| | - Siqing Jiang
- Department of Pain, The Third Xiangya Hospital and Institute of Pain Medicine, Central South UniversityChangsha, China
| | - Hui Yang
- Department of Pain, The Third Xiangya Hospital and Institute of Pain Medicine, Central South UniversityChangsha, China
| | - Qian Liao
- Department of Pain, The Third Xiangya Hospital and Institute of Pain Medicine, Central South UniversityChangsha, China
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Southwest Medical UniversityLuzhou, China
| | - Xuebin Yan
- Department of Pain, The Third Xiangya Hospital and Institute of Pain Medicine, Central South UniversityChangsha, China
| | - Dong Huang
- Department of Pain, The Third Xiangya Hospital and Institute of Pain Medicine, Central South UniversityChangsha, China
| |
Collapse
|
42
|
Wu JR, Chen H, Yao YY, Zhang MM, Jiang K, Zhou B, Zhang DX, Wang J. Local injection to sciatic nerve of dexmedetomidine reduces pain behaviors, SGCs activation, NGF expression and sympathetic sprouting in CCI rats. Brain Res Bull 2017; 132:118-128. [DOI: 10.1016/j.brainresbull.2017.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/27/2017] [Indexed: 12/17/2022]
|
43
|
Gong K, Ohara PT, Jasmin L. Patch Clamp Recordings on Intact Dorsal Root Ganglia from Adult Rats. J Vis Exp 2016. [PMID: 27768031 DOI: 10.3791/54287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Patch clamp studies from dorsal root ganglia (DRGs) neurons have increased our understanding of the peripheral nervous system. Currently, the majority of recordings are conducted on dissociated DRG neurons, which is a standard preparation for most laboratories. Neuronal properties, however, can be altered by axonal injury resulting from enzyme digestion used in acquiring dissociated neurons. Further, dissociated neuron preparations cannot fully represent the microenvironment of the DRG since loss of contact with satellite glial cells that surround the primary sensory neurons is an unavoidable consequence of this method. To overcome the limitations in using conventional dissociated DRG neurons for patch clamp recordings, in this report we describe a method to prepare intact DRGs and conduct patch clamp recordings on individual primary sensory neurons ex vivo. This approach permits the fast and straightforward preparation of intact DRGs, mimicking in vivo conditions by keeping DRG neurons associated with their surrounding satellite glial cells and basement membrane. Furthermore, the method avoids axonal injury from manipulation and enzyme digestion such as when dissociating DRGs. This ex vivo preparation can additionally be used to study the interaction between primary sensory neurons and satellite glial cells.
Collapse
Affiliation(s)
- Kerui Gong
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco;
| | - Peter T Ohara
- Department of Anatomy, University of California, San Francisco
| | - Luc Jasmin
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco;
| |
Collapse
|
44
|
Coupled Activation of Primary Sensory Neurons Contributes to Chronic Pain. Neuron 2016; 91:1085-1096. [PMID: 27568517 DOI: 10.1016/j.neuron.2016.07.044] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 06/21/2016] [Accepted: 07/19/2016] [Indexed: 11/20/2022]
Abstract
Primary sensory neurons in the DRG play an essential role in initiating pain by detecting painful stimuli in the periphery. Tissue injury can sensitize DRG neurons, causing heightened pain sensitivity, often leading to chronic pain. Despite the functional importance, how DRG neurons function at a population level is unclear due to the lack of suitable tools. Here we developed an imaging technique that allowed us to simultaneously monitor the activities of >1,600 neurons/DRG in live mice and discovered a striking neuronal coupling phenomenon that adjacent neurons tend to activate together following tissue injury. This coupled activation occurs among various neurons and is mediated by an injury-induced upregulation of gap junctions in glial cells surrounding DRG neurons. Blocking gap junctions attenuated neuronal coupling and mechanical hyperalgesia. Therefore, neuronal coupling represents a new form of neuronal plasticity in the DRG and contributes to pain hypersensitivity by "hijacking" neighboring neurons through gap junctions.
Collapse
|
45
|
Ibrahim SIA, Strong JA, Zhang JM. Mineralocorticoid Receptor, A Promising Target for Improving Management of Low Back Pain by Epidural Steroid Injections. ACTA ACUST UNITED AC 2016; 3:177-184. [PMID: 28956026 PMCID: PMC5611848 DOI: 10.24015/japm.2016.0023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIM OF REVIEW Low back pain is a major health problem in United States and worldwide. In this review, we aim to show that mineralocorticoid receptor (MR) activation has a critical role in the initiation of immune and inflammatory responses, which in turn can impact the effectiveness of the currently used steroids for epidural injections in low back pain management since most steroids activate MR in addition to the primary target, glucocorticoid receptor (GR). Moreover, we would like to determine some of the benefits of blocking the MR-induced negative effects. Overall, we propose a novel therapeutic approach for low back pain management by using a combination of a MR antagonist and a GR agonist in the epidural injections. METHOD We will first introduce the societal cost of low back pain and discuss how epidural steroid injections became a popular treatment for this condition. We will then describe several preclinical models used for the study of low back pain conditions and the findings with respect to the role of MR in the development of inflammatory low back pain. RECENT FINDINGS MR has pro-inflammatory effects in many tissues which can counteract the anti-inflammatory effects induced by GR activation. Blocking MR using the selective MR antagonist eplerenone can reduce pain and sensory neuron excitability in experimental models of low back pain. Moreover, combining the MR antagonist with clinically used steroids is more effective in reducing pain behaviors than using the steroids alone. SUMMARY MR antagonists are promising candidates to increase the effectiveness of currently used steroids. Since the activation of the MR is evident in preclinical models of low back pain, blocking its deleterious effects can be beneficial in managing inflammatory pain conditions.
Collapse
Affiliation(s)
- Shaimaa I A Ibrahim
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, USA.,Graduate Program in Molecular, Cellular, and Biochemical Pharmacology, University of Cincinnati, Cincinnati, USA
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, USA
| |
Collapse
|
46
|
Pittman SK, Gracias NG, Fehrenbacher JC. Nerve growth factor alters microtubule targeting agent-induced neurotransmitter release but not MTA-induced neurite retraction in sensory neurons. Exp Neurol 2016; 279:104-115. [PMID: 26883566 DOI: 10.1016/j.expneurol.2016.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/25/2016] [Accepted: 02/13/2016] [Indexed: 10/22/2022]
Abstract
Peripheral neuropathy is a dose-limiting side effect of anticancer treatment with the microtubule-targeted agents (MTAs), paclitaxel and epothilone B (EpoB); however, the mechanisms by which the MTAs alter neuronal function and morphology are unknown. We previously demonstrated that paclitaxel alters neuronal sensitivity, in vitro, in the presence of nerve growth factor (NGF). Evidence in the literature suggests that NGF may modulate the neurotoxic effects of paclitaxel. Here, we examine whether NGF modulates changes in neuronal sensitivity and morphology induced by paclitaxel and EpoB. Neuronal sensitivity was assessed using the stimulated release of calcitonin gene-related peptide (CGRP), whereas morphology of established neurites was evaluated using a high content screening system. Dorsal root ganglion cultures, maintained in the absence or presence of NGF, were treated from day 7 to day 12 in culture with paclitaxel (300nM) or EpoB (30nM). Following treatment, the release of CGRP was stimulated using capsaicin or high extracellular potassium. In the presence of NGF, EpoB mimicked the effects of paclitaxel: capsaicin-stimulated release was attenuated, potassium-stimulated release was slightly enhanced and the total peptide content was unchanged. In the absence of NGF, both paclitaxel and EpoB decreased capsaicin- and potassium-stimulated release and the total peptide content, suggesting that NGF may reverse MTA-induced hyposensitivity. Paclitaxel and EpoB both decreased neurite length and branching, and this attenuation was unaffected by NGF in the growth media. These differential effects of NGF on neuronal sensitivity and morphology suggest that neurite retraction is not a causative factor to alter neuronal sensitivity.
Collapse
Affiliation(s)
- Sherry K Pittman
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, United States.
| | - Neilia G Gracias
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, United States; Indiana University School of Medicine, Stark Neuroscience Research Institute, United States.
| | - Jill C Fehrenbacher
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, United States; Indiana University School of Medicine, Stark Neuroscience Research Institute, United States; Indiana University School of Medicine, Department of Anesthesiology, United States.
| |
Collapse
|
47
|
Peripheral Nociceptors as Immune Sensors in the Development of Pain and Itch. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 904:77-85. [PMID: 26900064 DOI: 10.1007/978-94-017-7537-3_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The peripheral nervous system and the immune system perform a series of similar functionalities such as recognizing, responding, and adapting to external or internal stimuli despite significant morphological differences. The peripheral nervous system actively communicates and coordinates with the immune system to function as a unified defense system. The peripheral nervous system is highly regulated by the immune system, especially under inflammatory conditions. On the other hand, the nervous system can modulate the immune system via neurotransmitters and chemokines released by the peripheral nerve endings, particularly from nociceptors. In both physiological and pathological conditions, peripheral nociceptive (including pruriceptive) neurons may express a variety of immune-related receptors, such as chemokine receptors and immunoglobulin (Fc) receptors that are usually found on immune cells. Certain ligands such as chemokines and immune complexes may induce abnormal neuronal hyperexcitability and even ectopic action potential discharges, therefore producing the sensation of pain and/or itch in immune-related diseases. The immune-sensing mechanisms of peripheral nociceptors may play an important role in the development of chronic pain and pruritus and may indicate novel therapeutic strategies for these pathological conditions.
Collapse
|
48
|
Li X, Yang H, Ouyang Q, Liu F, Li J, Xiang Z, Yuan H. Enhanced RAGE Expression in the Dorsal Root Ganglion May Contribute to Neuropathic Pain Induced by Spinal Nerve Ligation in Rats. PAIN MEDICINE 2015; 17:803-12. [PMID: 26814270 DOI: 10.1093/pm/pnv035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 09/12/2015] [Indexed: 01/02/2023]
Abstract
OBJECTIVE There is some evidence implicating receptor for advanced glycation end products (RAGE) signaling in the pathogenesis of neuropathic pain (NP). The objective was to investigate whether RAGE signaling in the dorsal root ganglion (DRG) might contribute to NP following peripheral nerve injury. DESIGN Experimental study before and after spinal nerve ligation (SNL) surgery. SETTING Caged in a controlled environment. SUBJECTS Male Sprague-Dawley rats. METHODS A SNL rat model of NP was used. Mechanical hyperalgesia was measured by the paw withdrawal threshold (PWT) to mechanical stimuli (1.4-15 g). Protein expressions of RAGE (immunofluorescence and western blotting), glial fibrillary acidic protein (GFAP; satellite glial cell [SGC] activation marker), IL-1β (ELISA), TNF-α (ELISA), and NF-κB (western blotting) in the DRG were determined. RAGE signaling was inhibited by intrathecal injection of anti-RAGE antibody. RESULTS After 7 days, SNL surgery reduced the PWT and upregulated the protein expression of RAGE, GFAP, NF-κB, TNF-α, and IL-1β. Intrathecal injection of RAGE-neutralizing antibody attenuated the SNL-induced mechanical hyperalgesia, activation of SGCs, and upregulation of NF-κB, TNF-α, and IL-1β in the DRG. CONCLUSION RAGE signaling may contribute to the pain hypersensitivity observed in the rat SNL model of NP. Although the precise mechanism remains to be established, NF-κB, TNF-α, and IL-1β likely play a role, together with the activation of SGCs.
Collapse
Affiliation(s)
- Xiangnan Li
- *Department of Anesthesiology, Changzheng Hospital, the Second Military Medical University, Shanghai, 200003, China; Department of Anesthesiology, the Third People's Hospital of Yancheng, Yancheng, 224001, China
| | - Haiqin Yang
- *Department of Anesthesiology, Changzheng Hospital, the Second Military Medical University, Shanghai, 200003, China
| | - Qing Ouyang
- *Department of Anesthesiology, Changzheng Hospital, the Second Military Medical University, Shanghai, 200003, China
| | - Fangting Liu
- *Department of Anesthesiology, Changzheng Hospital, the Second Military Medical University, Shanghai, 200003, China
| | - Jian Li
- *Department of Anesthesiology, Changzheng Hospital, the Second Military Medical University, Shanghai, 200003, China
| | - Zhenghua Xiang
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology, Ministry of Education, Second Military Medical University, Shanghai, 200433, China
| | - Hongbin Yuan
- *Department of Anesthesiology, Changzheng Hospital, the Second Military Medical University, Shanghai, 200003, China;
| |
Collapse
|
49
|
Rajasekhar P, Poole DP, Liedtke W, Bunnett NW, Veldhuis NA. P2Y1 Receptor Activation of the TRPV4 Ion Channel Enhances Purinergic Signaling in Satellite Glial Cells. J Biol Chem 2015; 290:29051-62. [PMID: 26475857 DOI: 10.1074/jbc.m115.689729] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Indexed: 01/07/2023] Open
Abstract
Transient receptor potential (TRP) ion channels of peripheral sensory pathways are important mediators of pain, itch, and neurogenic inflammation. They are expressed by primary sensory neurons and by glial cells in the central nervous system, but their expression and function in satellite glial cells (SGCs) of sensory ganglia have not been explored. SGCs tightly ensheath neurons of sensory ganglia and can regulate neuronal excitability in pain and inflammatory states. Using a modified dissociation protocol, we isolated neurons with attached SGCs from dorsal root ganglia of mice. SGCs, which were identified by expression of immunoreactive Kir4.1 and glutamine synthetase, were closely associated with neurons, identified using the pan-neuronal marker NeuN. A subpopulation of SGCs expressed immunoreactive TRP vanilloid 4 (TRPV4) and responded to the TRPV4-selective agonist GSK1016790A by an influx of Ca(2+) ions. SGCs did not express functional TRPV1, TRPV3, or TRP ankyrin 1 channels. Responses to GSK1016790A were abolished by the TRPV4 antagonist HC067047 and were absent in SGCs from Trpv4(-/-) mice. The P2Y1-selective agonist 2-methylthio-ADP increased [Ca(2+)]i in SGCs, and responses were prevented by the P2Y1-selective antagonist MRS2500. P2Y1 receptor-mediated responses were enhanced in TRPV4-expressing SGCs and HEK293 cells, suggesting that P2Y1 couples to and activates TRPV4. PKC inhibitors prevented P2Y1 receptor activation of TRPV4. Our results provide the first evidence for expression of TRPV4 in SGCs and demonstrate that TRPV4 is a purinergic receptor-operated channel in SGCs of sensory ganglia.
Collapse
Affiliation(s)
- Pradeep Rajasekhar
- From the Monash Institute of Pharmaceutical Sciences, Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, and
| | - Daniel P Poole
- From the Monash Institute of Pharmaceutical Sciences, Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Departments of Anatomy and Neuroscience
| | - Wolfgang Liedtke
- the Department of Neurology, School of Medicine, Duke University, Durham, North Carolina 27710
| | - Nigel W Bunnett
- From the Monash Institute of Pharmaceutical Sciences, Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, and Department of Anaesthesia and Peri-operative Medicine, Monash University, Victoria 3052, Australia, Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia, and
| | - Nicholas A Veldhuis
- From the Monash Institute of Pharmaceutical Sciences, Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, and Genetics, and
| |
Collapse
|
50
|
Magni G, Merli D, Verderio C, Abbracchio MP, Ceruti S. P2Y2 receptor antagonists as anti-allodynic agents in acute and sub-chronic trigeminal sensitization: role of satellite glial cells. Glia 2015; 63:1256-69. [PMID: 25779655 DOI: 10.1002/glia.22819] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 01/28/2015] [Accepted: 02/27/2015] [Indexed: 12/18/2022]
Abstract
Trigeminal (TG) pain often lacks a satisfactory pharmacological control. A better understanding of the molecular cross-talk between TG neurons and surrounding satellite glial cells (SGCs) could help identifying innovative targets for the development of more effective analgesics. We have previously demonstrated that neuronal pro-algogenic mediators upregulate G protein-coupled nucleotide P2Y receptors (P2YRs) expressed by TG SGCs in vitro. Here, we have identified the specific P2YR subtypes involved (i.e., the ADP-sensitive P2Y1 R and the UTP-responsive P2Y2 R subtypes), and demonstrated the contribution of neuron-derived prostaglandins to their upregulation. Next, we have translated these data to an in vivo model of TG pain (namely, rats injected with Complete Freund's adjuvant in the temporomandibular joint), by demonstrating activation of SGCs and upregulation of P2Y1 R and P2Y2 R in the ipsi-lateral TG. To unequivocally link P2YRs to the development of facial allodynia, we treated animals with various purinergic antagonists. The selective P2Y2 R antagonist AR-C118925 completely inhibited SGCs activation, exerted a potent anti-allodynic effect that lasted over time, and was still effective when administration was started 6-days post induction of allodynia, i.e. under subchronic pain conditions. Conversely, the selective P2Y1 R antagonist MRS2179 was completely ineffective. Moreover, similarly to the anti-inflammatory drug acetylsalicylic acid and the known anti-migraine agent sumatriptan, the P2X/P2Y nonselective antagonist PPADS was only partially effective, and completely lost its activity under sub-chronic conditions. Taken together, our results highlight glial P2Y2 Rs as potential "druggable" targets for the successful management of TG-related pain.
Collapse
Affiliation(s)
- Giulia Magni
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università Degli Studi Di Milano, via Balzaretti, 9, Milan, Italy; Department of Drug Discovery and Development, Italian Institute of Technology (IIT), via Morego, 30, Genoa, Italy
| | | | | | | | | |
Collapse
|