1
|
Yamaoka K, Nozaki K, Zhu M, Terai H, Kobayashi K, Ito H, Matsumata M, Takemoto H, Ikeda S, Sotomaru Y, Mori T, Aizawa H, Hashimoto K. Neuron-non-neuron electrical coupling networks are involved in chronic stress-induced electrophysiological changes in lateral habenular neurons. J Physiol 2025; 603:2713-2740. [PMID: 40168081 PMCID: PMC12072243 DOI: 10.1113/jp287286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/20/2025] [Indexed: 04/03/2025] Open
Abstract
The lateral habenula (LHb) is a key brain structure that receives input from higher brain regions and regulates monoaminergic activity. LHb hyperactivity has been implicated in the pathophysiology of depression, but the electrophysiological mechanisms underlying this hyperactivity remain poorly understood. To address this issue, we investigated how chronic stress alters the firing properties of LHb neurons in a mouse model of chronic social defeat. Whole-cell recordings were conducted from LHb neurons in the mouse acute brain slices. LHb neurons exhibited two types of rebound depolarizing potentials (RDPs) after the offset of hyperpolarization: short-RDPs (lasting <400 ms) and long-RDPs (order of seconds). Stress-susceptible mice showed a significantly reduced occurrence of long-RDPs, whereas spike firing in response to depolarizing current injections remained unchanged. Both short- and long-RDPs were triggered by T-type voltage-dependent Ca2+ channels and shortened by small-conductance Ca2+-activated K+ (SK) channels. The prolonged depolarizing phase of long-RDPs was mediated by cyclic nucleotide-gated (CNG) channels, which were activated via electrical coupling formed between neurons and non-neuronal cells. Whole-cell recording using an internal solution including a gap junction-permeable dye revealed that neurons formed dye coupling with non-neuronal cells, including oligodendrocytes and/or oligodendrocyte precursor cells. RNA-sequencing and genome editing experiments suggested that Cnga4, a CNG channel subtype, was the primary candidate for the long depolarizing phase of long-RDP, and its expression was decreased in the stress-susceptible mice. These findings suggest that stress-dependent changes in the firing activity of neurons are regulated by neuron-non-neuron networks formed in the LHb. KEY POINTS: Mouse lateral habenular (LHb) neurons exhibit short (<400 ms) rebound depolarizing potentials (short-RDPs) or long-RDPs (order of seconds) (long-RDPs) after the offset of hyperpolarization. The incidence of long-RDP neurons is significantly reduced in mice susceptible to chronic social defeat stress. The long depolarizing phase of long-RDPs is mediated by cyclic nucleotide-gated (CNG) channels, which are activated in non-neuronal cells via gap junctions. The expression of Cnga4, the gene encoding a subtype of the CNG channel, is decreased in the stress-susceptible mice. These results help us understand the mechanisms underlying stress-induced electrophysiological changes in LHb neurons and the functional roles of neuron-non-neuron networks for these neurons.
Collapse
Affiliation(s)
- Kenji Yamaoka
- Department of Neurophysiology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Kanako Nozaki
- Department of Neurobiology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Meina Zhu
- Department of Neurobiology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Haruhi Terai
- Department of Neurobiology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Kenta Kobayashi
- Section of Viral Vector DevelopmentNational Institute for Physiological SciencesOkazakiJapan
| | - Hikaru Ito
- Department of Neurobiology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Miho Matsumata
- Department of Neurobiology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Hidenori Takemoto
- Department of Neurobiology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Shinya Ikeda
- Natural Science Center for Basic Research and DevelopmentHiroshima UniversityHiroshimaJapan
| | - Yusuke Sotomaru
- Natural Science Center for Basic Research and DevelopmentHiroshima UniversityHiroshimaJapan
| | - Tetsuji Mori
- Department of Biological Regulation, School of Health Science, Faculty of MedicineTottori UniversityYonagoJapan
| | - Hidenori Aizawa
- Department of Neurobiology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Kouichi Hashimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| |
Collapse
|
2
|
Birren SJ, Goodrich LV, Segal RA. Satellite Glial Cells: No Longer the Most Overlooked Glia. Cold Spring Harb Perspect Biol 2025; 17:a041367. [PMID: 38768970 PMCID: PMC11694750 DOI: 10.1101/cshperspect.a041367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Many glial biologists consider glia the neglected cells of the nervous system. Among all the glia of the central and peripheral nervous system, satellite glia may be the most often overlooked. Satellite glial cells (SGCs) are located in ganglia of the cranial nerves and the peripheral nervous system. These small cells surround the cell bodies of neurons in the trigeminal ganglia (TG), spiral ganglia, nodose and petrosal ganglia, sympathetic ganglia, and dorsal root ganglia (DRG). Essential SGC features include their intimate connections with the associated neurons, their small size, and their derivation from neural crest cells. Yet SGCs also exhibit tissue-specific properties and can change rapidly, particularly in response to injury. To illustrate the range of SGC functions, we will focus on three types: those of the spiral, sympathetic, and DRG, and consider both their shared features and those that differ based on location.
Collapse
Affiliation(s)
- Susan J Birren
- Department of Biology, Brandeis University, Waltham, Massachusetts 02453, USA
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Rosalind A Segal
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| |
Collapse
|
3
|
Moss KR, Saxena S. Schwann Cells in Neuromuscular Disorders: A Spotlight on Amyotrophic Lateral Sclerosis. Cells 2025; 14:47. [PMID: 39791748 PMCID: PMC11719703 DOI: 10.3390/cells14010047] [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: 11/14/2024] [Revised: 12/23/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex neurodegenerative disease primarily affecting motor neurons, leading to progressive muscle atrophy and paralysis. This review explores the role of Schwann cells in ALS pathogenesis, highlighting their influence on disease progression through mechanisms involving demyelination, neuroinflammation, and impaired synaptic function. While Schwann cells have been traditionally viewed as peripheral supportive cells, especially in motor neuron disease, recent evidence indicates that they play a significant role in ALS by impacting motor neuron survival and plasticity, influencing inflammatory responses, and altering myelination processes. Furthermore, advancements in understanding Schwann cell pathology in ALS combined with lessons learned from studying Charcot-Marie-Tooth disease Type 1 (CMT1) suggest potential therapeutic strategies targeting these cells may support nerve repair and slow disease progression. Overall, this review aims to provide comprehensive insights into Schwann cell classification, physiology, and function, underscoring the critical pathological contributions of Schwann cells in ALS and suggests new avenues for targeted therapeutic interventions aimed at modulating Schwann cell function in ALS.
Collapse
Affiliation(s)
- Kathryn R. Moss
- Department of Physical Medicine and Rehabilitation, University of Missouri School of Medicine, Columbia, MO 65211, USA
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA
| | - Smita Saxena
- Department of Physical Medicine and Rehabilitation, University of Missouri School of Medicine, Columbia, MO 65211, USA
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
4
|
Starobova H, Alshammari A, Winkler IG, Vetter I. The role of the neuronal microenvironment in sensory function and pain pathophysiology. J Neurochem 2024; 168:3620-3643. [PMID: 36394416 DOI: 10.1111/jnc.15724] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022]
Abstract
The high prevalence of pain and the at times low efficacy of current treatments represent a significant challenge to healthcare systems worldwide. Effective treatment strategies require consideration of the diverse pathophysiologies that underlie various pain conditions. Indeed, our understanding of the mechanisms contributing to aberrant sensory neuron function has advanced considerably. However, sensory neurons operate in a complex dynamic microenvironment that is controlled by multidirectional interactions of neurons with non-neuronal cells, including immune cells, neuronal accessory cells, fibroblasts, adipocytes, and keratinocytes. Each of these cells constitute and control the microenvironment in which neurons operate, inevitably influencing sensory function and the pathology of pain. This review highlights the importance of the neuronal microenvironment for sensory function and pain, focusing on cellular interactions in the skin, nerves, dorsal root ganglia, and spinal cord. We discuss the current understanding of the mechanisms by which neurons and non-neuronal cells communicate to promote or resolve pain, and how this knowledge could be used for the development of mechanism-based treatments.
Collapse
Affiliation(s)
- Hana Starobova
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Ammar Alshammari
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Ingrid G Winkler
- Mater Research Institute, The University of Queensland, Queensland, South Brisbane, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
- The School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
| |
Collapse
|
5
|
Goto T, Kuramoto E, Iwai H, Yamanaka A. Cytoarchitecture and intercellular interactions in the trigeminal ganglion: Associations with neuropathic pain in the orofacial region. J Oral Biosci 2024; 66:485-490. [PMID: 39032827 DOI: 10.1016/j.job.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Disorders of the trigeminal nerve, a sensory nerve of the orofacial region, often lead to complications in dental practice, including neuropathic pain, allodynia, and ectopic pain. Management of these complications requires an understanding of the cytoarchitecture of the trigeminal ganglion, where the cell bodies of the trigeminal nerve are located, and the mechanisms of cell-cell interactions. HIGHLIGHTS In the trigeminal ganglion, ganglion, satellite, Schwann, and immune cells coexist and interact. Cell-cell interactions are complex and occur through direct contact via gap junctions or through mediators such as adenosine triphosphate, nitric oxide, peptides, and cytokines. Interactions between the nervous and immune systems within the trigeminal ganglion may have neuroprotective effects during nerve injury or may exacerbate inflammation and produce chronic pain. Under pathological conditions of the trigeminal nerve, cell-cell interactions can cause allodynia and ectopic pain. Although cell-cell interactions that occur via mediators can act at some distance, they are more effective when the cells are close together. Therefore, information on the three-dimensional topography of trigeminal ganglion cells is essential for understanding the pathophysiology of ectopic pain. CONCLUSIONS A three-dimensional map of the somatotopic localization of trigeminal ganglion neurons revealed that ganglion cells innervating distant orofacial regions are often apposed to each other, interacting with and potentially contributing to ectopic pain. Elucidation of the complex network of mediators and their receptors responsible for intercellular communication within the trigeminal ganglion is essential for understanding ectopic pain.
Collapse
Affiliation(s)
- Tetsuya Goto
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| | - Eriko Kuramoto
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| | - Haruki Iwai
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Atsushi Yamanaka
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
6
|
Hanani M. From Gut Motility to Chronic Pain: Studies on the Mammalian Peripheral Nervous System. Bioelectricity 2024; 6:207-220. [PMID: 39372092 PMCID: PMC11447483 DOI: 10.1089/bioe.2024.0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024] Open
Affiliation(s)
- Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
7
|
Wong CE, Liu W, Huang CC, Lee PH, Huang HW, Chang Y, Lo HT, Chen HF, Kuo LC, Lee JS. Sciatic nerve stimulation alleviates neuropathic pain and associated neuroinflammation in the dorsal root ganglia in a rodent model. J Transl Med 2024; 22:770. [PMID: 39143617 PMCID: PMC11325705 DOI: 10.1186/s12967-024-05573-1] [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: 06/06/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Satellite glial cells (SGCs) in the dorsal root ganglia (DRG) play a pivotal role in the formation of neuropathic pain (NP). Sciatic nerve stimulation (SNS) neuromodulation was reported to alleviate NP and reduce neuroinflammation. However, the mechanisms underlying SNS in the DRG remain unclear. This study aimed to elucidate the mechanism of electric stimulation in reducing NP, focusing on the DRG. METHODS L5 nerve root ligation (NRL) NP rat model was studied. Ipsilateral SNS performed 1 day after NRL. Behavioral tests were performed to assess pain phenotypes. NanoString Ncounter technology was used to explore the differentially expressed genes and cellular pathways. Activated SGCs were characterized in vivo and in vitro. The histochemical alterations of SGCs, macrophages, and neurons in DRG were examined in vivo on post-injury day 8. RESULTS NRL induced NP behaviors including decreased pain threshold and latency on von Frey and Hargreaves tests. We found that following nerve injury, SGCs were hyperactivated, neurotoxic and had increased expression of NP-related ion channels including TRPA1, Cx43, and SGC-neuron gap junctions. Mechanistically, nerve injury induced reciprocal activation of SGCs and M1 macrophages via cytokines including IL-6, CCL3, and TNF-α mediated by the HIF-1α-NF-κB pathways. SNS suppressed SGC hyperactivation, reduced the expression of NP-related ion channels, and induced M2 macrophage polarization, thereby alleviating NP and associated neuroinflammation in the DRG. CONCLUSIONS NRL induced hyperactivation of SGCs, which had increased expression of NP-related ion channels. Reciprocal activation of SGCs and M1 macrophages surrounding the primary sensory neurons was mediated by the HIF-1α and NF-κB pathways. SNS suppressed SGC hyperactivation and skewed M1 macrophage towards M2. Our findings establish SGC activation as a crucial pathomechanism in the gliopathic alterations in NP, which can be modulated by SNS neuromodulation.
Collapse
Affiliation(s)
- Chia-En Wong
- Division of Neurosurgery, Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li Road, Tainan, 70428, Taiwan
| | - Wentai Liu
- Department of Bioengineering, University of California, Los Angeles, CA, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, USA
- Department of Electrical and Computer Engineering, Los Angeles, CA, USA
- Brain Research Institute, University of California, Los Angeles, CA, USA
| | - Chi-Chen Huang
- Division of Neurosurgery, Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li Road, Tainan, 70428, Taiwan
| | - Po-Hsuan Lee
- Division of Neurosurgery, Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li Road, Tainan, 70428, Taiwan
| | - Han-Wei Huang
- Department of Neurology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Yu Chang
- Division of Neurosurgery, Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li Road, Tainan, 70428, Taiwan
| | - Hsin-Tien Lo
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Fang Chen
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Li-Chieh Kuo
- Department of Occupational Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jung-Shun Lee
- Division of Neurosurgery, Department of Surgery, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No. 138, Sheng-Li Road, Tainan, 70428, Taiwan.
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
8
|
Qiao LY. Satellite Glial Cells Bridge Sensory Neuron Crosstalk in Visceral Pain and Cross-Organ Sensitization. J Pharmacol Exp Ther 2024; 390:213-221. [PMID: 38777604 PMCID: PMC11264254 DOI: 10.1124/jpet.123.002061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Following colonic inflammation, the uninjured bladder afferent neurons are also activated. The mechanisms and pathways underlying this sensory neuron cross-activation (from injured neurons to uninjured neurons) are not fully understood. Colonic and bladder afferent neurons reside in the same spinal segments and are separated by satellite glial cells (SGCs) and extracellular matrix in dorsal root ganglia (DRG). SGCs communicate with sensory neurons in a bidirectional fashion. This review summarizes the differentially regulated genes/proteins in the injured and uninjured DRG neurons and explores the role of SGCs in regulation of sensory neuron crosstalk in visceral cross-organ sensitization. The review also highlights the paracrine pathways in mediating neuron-SGC and SGC-neuron coupling with an emphasis on the neurotrophins and purinergic systems. Finally, I discuss the results from recent RNAseq profiling of SGCs to reveal useful molecular markers for characterization, functional study, and therapeutic targets of SGCs. SIGNIFICANCE STATEMENT: Satellite glial cells (SGCs) are the largest glial subtypes in sensory ganglia and play a critical role in mediating sensory neuron crosstalk, an underlying mechanism in colon-bladder cross-sensitization. Identification of novel and unique molecular markers of SGCs can advance the discovery of therapeutic targets in treatment of chronic pain including visceral pain comorbidity.
Collapse
Affiliation(s)
- Liya Y Qiao
- Department of Physiology and Biophysics, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
9
|
Su Y, Verkhratsky A, Yi C. Targeting connexins: possible game changer in managing neuropathic pain? Trends Mol Med 2024; 30:642-659. [PMID: 38594094 DOI: 10.1016/j.molmed.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024]
Abstract
Neuropathic pain is a chronic debilitating condition caused by nerve injury or a variety of diseases. At the core of neuropathic pain lies the aberrant neuronal excitability in the peripheral and/or central nervous system (PNS and CNS). Enhanced connexin expression and abnormal activation of connexin-assembled gap junctional channels are prominent in neuropathic pain along with reactive gliosis, contributing to neuronal hypersensitivity and hyperexcitability. In this review, we delve into the current understanding of how connexin expression and function contribute to the pathogenesis and pathophysiology of neuropathic pain and argue for connexins as potential therapeutic targets for neuropathic pain management.
Collapse
Affiliation(s)
- Yixun Su
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, Bilbao, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China; Shenzhen Key Laboratory of Chinese Medicine Active substance screening and Translational Research, Shenzhen, China.
| |
Collapse
|
10
|
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: 5] [Impact Index Per Article: 5.0] [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
|
11
|
Xing Q, Cibelli A, Yang GL, Dohare P, Li QH, Scemes E, Guan FX, Spray DC. Neuronal Panx1 drives peripheral sensitization in experimental plantar inflammatory pain. Mil Med Res 2024; 11:27. [PMID: 38685116 PMCID: PMC11057180 DOI: 10.1186/s40779-024-00525-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 03/25/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The channel-forming protein Pannexin1 (Panx1) has been implicated in both human studies and animal models of chronic pain, but the underlying mechanisms remain incompletely understood. METHODS Wild-type (WT, n = 24), global Panx1 KO (n = 24), neuron-specific Panx1 KO (n = 20), and glia-specific Panx1 KO (n = 20) mice were used in this study at Albert Einstein College of Medicine. The von Frey test was used to quantify pain sensitivity in these mice following complete Freund's adjuvant (CFA) injection (7, 14, and 21 d). The qRT-PCR was employed to measure mRNA levels of Panx1, Panx2, Panx3, Cx43, Calhm1, and β-catenin. Laser scanning confocal microscopy imaging, Sholl analysis, and electrophysiology were utilized to evaluate the impact of Panx1 on neuronal excitability and morphology in Neuro2a and dorsal root ganglion neurons (DRGNs) in which Panx1 expression or function was manipulated. Ethidium bromide (EtBr) dye uptake assay and calcium imaging were employed to investigate the role of Panx1 in adenosine triphosphate (ATP) sensitivity. β-galactosidase (β-gal) staining was applied to determine the relative cellular expression levels of Panx1 in trigeminal ganglia (TG) and DRG of transgenic mice. RESULTS Global or neuron-specific Panx1 deletion markedly decreased pain thresholds after CFA stimuli (7, 14, and 21 d; P < 0.01 vs. WT group), indicating that Panx1 was positively correlated with pain sensitivity. In Neuro2a, global Panx1 deletion dramatically reduced neurite extension and inward currents compared to the WT group (P < 0.05), revealing that Panx1 enhanced neurogenesis and excitability. Similarly, global Panx1 deletion significantly suppressed Wnt/β-catenin dependent DRG neurogenesis following 5 d of nerve growth factor (NGF) treatment (P < 0.01 vs. WT group). Moreover, Panx1 channels enhanced DRG neuron response to ATP after CFA injection (P < 0.01 vs. Panx1 KO group). Furthermore, ATP release increased Ca2+ responses in DRGNs and satellite glial cells surrounding them following 7 d of CFA treatment (P < 0.01 vs. Panx1 KO group), suggesting that Panx1 in glia also impacts exaggerated neuronal excitability. Interestingly, neuron-specific Panx1 deletion was found to markedly reduce differentiation in cultured DRGNs, as evidenced by stunted neurite outgrowth (P < 0.05 vs. Panx1 KO group; P < 0.01 vs. WT group or GFAP-Cre group), blunted activation of Wnt/β-catenin signaling (P < 0.01 vs. WT, Panx1 KO and GFAP-Cre groups), and diminished cell excitability (P < 0.01 vs. GFAP-Cre group) and response to ATP stimulation (P < 0.01 vs. WT group). Analysis of β-gal staining showed that cellular expression levels of Panx1 in neurons are significantly higher (2.5-fold increase) in the DRG than in the TG. CONCLUSIONS The present study revealed that neuronal Panx1 is a prominent driver of peripheral sensitivity in the setting of inflammatory pain through cell-autonomous effects on neuronal excitability. This hyperexcitability dependence on neuronal Panx1 contrasts with inflammatory orofacial pain, where similar studies revealed a prominent role for glial Panx1. The apparent differences in Panx1 expression in neuronal and non-neuronal TG and DRG cells are likely responsible for the distinct impact of these cell types in the two pain models.
Collapse
Affiliation(s)
- Qu Xing
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Antonio Cibelli
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, 70125, Italy
| | - Greta Luyuan Yang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, 06459, USA
| | - Preeti Dohare
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, 12208, USA
| | - Qing-Hua Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Eliana Scemes
- Department of Anatomy and Cell Biology, New York Medical College, Valhalla, NY, 10595, USA
| | - Fang-Xia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450001, China.
| | - David C Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
12
|
Black BJ, Ghazal RE, Lojek N, Williams V, Rajput JS, Lawson JM. Phenotypic Screening of Prospective Analgesics Among FDA-Approved Compounds using an iPSC-Based Model of Acute and Chronic Inflammatory Nociception. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303724. [PMID: 38189546 PMCID: PMC10953557 DOI: 10.1002/advs.202303724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 11/26/2023] [Indexed: 01/09/2024]
Abstract
Classical target-based drug screening is low-throughput, largely subjective, and costly. Phenotypic screening based on in vitro models is increasingly being used to identify candidate compounds that modulate complex cell/tissue functions. Chronic inflammatory nociception, and subsequent chronic pain conditions, affect peripheral sensory neuron activity (e.g., firing of action potentials) through myriad pathways, and remain unaddressed in regard to effective, non-addictive management/treatment options. Here, a chronic inflammatory nociception model is demonstrated based on induced pluripotent stem cell (iPSC) sensory neurons and glia, co-cultured on microelectrode arrays (MEAs). iPSC sensory co-cultures exhibit coordinated spontaneous extracellular action potential (EAP) firing, reaching a stable baseline after ≈27 days in vitro (DIV). Spontaneous and evoked EAP metrics are significantly modulated by 24-h incubation with tumor necrosis factor-alpha (TNF-α), representing an inflammatory phenotype. Compared with positive controls (lidocaine), this model is identified as an "excellent" stand-alone assay based on a modified Z' assay quality metric. This model is then used to screen 15 cherry-picked, off-label, Food and Drug Administration (FDA)-approved compounds; 10 of 15 are identified as "hits". Both hits and "misses" are discussed in turn. In total, this data suggests that iPSC sensory co-cultures on MEAs may represent a moderate-to-high-throughput assay for drug discovery targeting inflammatory nociception.
Collapse
Affiliation(s)
- Bryan James Black
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Rasha El Ghazal
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Neal Lojek
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Victoria Williams
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Jai Singh Rajput
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Jennifer M. Lawson
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| |
Collapse
|
13
|
Kuramoto E, Fukushima M, Sendo R, Ohno S, Iwai H, Yamanaka A, Sugimura M, Goto T. Three-dimensional topography of rat trigeminal ganglion neurons using a combination of retrograde labeling and tissue-clearing techniques. J Comp Neurol 2024; 532:e25584. [PMID: 38341648 DOI: 10.1002/cne.25584] [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: 09/07/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024]
Abstract
The trigeminal nerve is the sensory afferent of the orofacial regions and divided into three major branches. Cell bodies of the trigeminal nerve lie in the trigeminal ganglion and are surrounded by satellite cells. There is a close interaction between ganglion cells via satellite cells, but the function is not fully understood. In the present study, we clarified the ganglion cells' three-dimensional (3D) localization, which is essential to understand the functions of cell-cell interactions in the trigeminal ganglion. Fast blue was injected into 12 sites of the rat orofacial regions, and ganglion cells were retrogradely labeled. The labeled trigeminal ganglia were cleared by modified 3DISCO, imaged with confocal laser-scanning microscopy, and reconstructed in 3D. Histograms of the major axes of the fast blue-positive somata revealed that the peak major axes of the cells innervating the skin/mucosa were smaller than those of cells innervating the deep structures. Ganglion cells innervating the ophthalmic, maxillary, and mandibular divisions were distributed in the anterodorsal, central, and posterolateral portions of the trigeminal ganglion, respectively, with considerable overlap in the border region. The intermingling in the distribution of ganglion cells within each division was also high, in particular, within the mandibular division. Specifically, intermingling was observed in combinations of tongue and masseter/temporal muscles, maxillary/mandibular molars and masseter/temporal muscles, and tongue and mandibular molars. Double retrograde labeling confirmed that some ganglion cells innervating these combinations were closely apposed. Our data provide essential information for understanding the function of ganglion cell-cell interactions via satellite cells.
Collapse
Grants
- JP23H03119 Grants-in-Aid from The Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP23K09316 Grants-in-Aid from The Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP19K10058 Grants-in-Aid from The Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP19K10336 Grants-in-Aid from The Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP19KK0419 Grants-in-Aid from The Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP22H05162 Grants-in-Aid from The Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP22K09916 Grants-in-Aid from The Ministry of Education, Culture, Sports, Science and Technology (MEXT)
Collapse
Affiliation(s)
- Eriko Kuramoto
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Makoto Fukushima
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Ryozo Sendo
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Department of Dental Anesthesiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Sachi Ohno
- Department of Dental Anesthesiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Haruki Iwai
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Atsushi Yamanaka
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Mitsutaka Sugimura
- Department of Dental Anesthesiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Tetsuya Goto
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
14
|
Zhao Q, Jiang C, Zhao L, Dai X, Yi S. Unleashing Axonal Regeneration Capacities: Neuronal and Non-neuronal Changes After Injuries to Dorsal Root Ganglion Neuron Central and Peripheral Axonal Branches. Mol Neurobiol 2024; 61:423-433. [PMID: 37620687 DOI: 10.1007/s12035-023-03590-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Peripheral nerves obtain remarkable regenerative capacity while central nerves can hardly regenerate following nerve injury. Sensory neurons in the dorsal root ganglion (DRG) are widely used to decipher the dissimilarity between central and peripheral axonal regeneration as axons of DRG neurons bifurcate into the regeneration-incompetent central projections and the regeneration-competent peripheral projections. A conditioning peripheral branch injury facilitates central axonal regeneration and enables the growth and elongation of central axons. Peripheral axonal injury stimulates neuronal calcium influx, alters the start-point chromatin states, increases chromatin accessibility, upregulates the expressions of regeneration-promoting genes and the synthesis of proteins, and supports axonal regeneration. Following central axonal injury, the responses of DRG neurons are modest, resulting in poor intrinsic growth ability. Some non-neuronal cells in DRGs, for instance satellite glial cells, also exhibit diminished injury responses to central axon injury as compared with peripheral axon injury. Moreover, DRG central and peripheral axonal branches are respectively surrounded by inhibitory glial scars generated by central glial cells and a permissive microenvironment generated by Schwann cells and macrophages. The aim of this review is to look at changes of DRG neurons and non-neuronal cells after peripheral and central axon injuries and summarize the contributing roles of both neuronal intrinsic regenerative capacities and surrounding microenvironments in axonal regeneration.
Collapse
Affiliation(s)
- Qian Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Chunyi Jiang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Li Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Xiu Dai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China.
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
15
|
Jia S, Mai L, Yang H, Huang F, He H, Fan W. Cross-species gene expression patterns of purinergic signaling in the human and mouse trigeminal ganglion. Life Sci 2023; 332:122130. [PMID: 37769809 DOI: 10.1016/j.lfs.2023.122130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
Purinergic signaling system plays a pivotal role in the trigeminal ganglion (TG) which is a primary sensory tissue in vertebrate nervous systems involving orofacial nociception and peripheral sensitization. Despite previous efforts to reveal the expression patterns of purinergic components in the mouse TG, it is still unknown the interspecies differences between human and mouse. In this study, we provide a comprehensive transcriptome profile of the purinergic signaling system across diverse cell types and neuronal subpopulations within the human TG, systematically comparing it with mouse TG. In addition, the evolutionary conservation and species-specific expression patterns of the purinergic components are also discussed. We propose that the data can improve our understanding of purinergic signaling in the peripheral nervous system and facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Shilin Jia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Lijia Mai
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Hui Yang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Fang Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Hongwen He
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Wenguo Fan
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
| |
Collapse
|
16
|
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: 10] [Impact Index Per Article: 5.0] [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
|
17
|
Qi C, Lavriha P, Bayraktar E, Vaithia A, Schuster D, Pannella M, Sala V, Picotti P, Bortolozzi M, Korkhov VM. Structures of wild-type and selected CMT1X mutant connexin 32 gap junction channels and hemichannels. SCIENCE ADVANCES 2023; 9:eadh4890. [PMID: 37647412 PMCID: PMC10468125 DOI: 10.1126/sciadv.adh4890] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/27/2023] [Indexed: 09/01/2023]
Abstract
In myelinating Schwann cells, connection between myelin layers is mediated by gap junction channels (GJCs) formed by docked connexin 32 (Cx32) hemichannels (HCs). Mutations in Cx32 cause the X-linked Charcot-Marie-Tooth disease (CMT1X), a degenerative neuropathy without a cure. A molecular link between Cx32 dysfunction and CMT1X pathogenesis is still missing. Here, we describe the high-resolution cryo-electron cryo-myography (cryo-EM) structures of the Cx32 GJC and HC, along with two CMT1X-linked mutants, W3S and R22G. While the structures of wild-type and mutant GJCs are virtually identical, the HCs show a major difference: In the W3S and R22G mutant HCs, the amino-terminal gating helix partially occludes the pore, consistent with a diminished HC activity. Our results suggest that HC dysfunction may be involved in the pathogenesis of CMT1X.
Collapse
Affiliation(s)
- Chao Qi
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Pia Lavriha
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Erva Bayraktar
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Padua, Italy
| | - Anand Vaithia
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Dina Schuster
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Micaela Pannella
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Padua, Italy
| | - Valentina Sala
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
| | - Paola Picotti
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Mario Bortolozzi
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Physics and Astronomy “G. Galilei”, University of Padua, Padua, Italy
| | - Volodymyr M. Korkhov
- Institute of Molecular Biology and Biophysics, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| |
Collapse
|
18
|
Zhao J, Harrison S, Levy D. Meningeal P2X7 Signaling Mediates Migraine-Related Intracranial Mechanical Hypersensitivity. J Neurosci 2023; 43:5975-5985. [PMID: 37487740 PMCID: PMC10436684 DOI: 10.1523/jneurosci.0368-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/11/2023] [Accepted: 06/02/2023] [Indexed: 07/26/2023] Open
Abstract
Cortical spreading depolarization (CSD) is a key pathophysiological event that underlies visual and sensory auras in migraine. CSD is also thought to drive the headache phase in migraine by promoting the activation and mechanical sensitization of trigeminal primary afferent nociceptive neurons that innervate the cranial meninges. The factors underlying meningeal nociception in the wake of CSD remain poorly understood but potentially involve the parenchymal release of algesic mediators and damage-associated molecular patterns, particularly ATP. Here, we explored the role of ATP-P2X purinergic receptor signaling in mediating CSD-evoked meningeal afferent activation and mechanical sensitization. Male rats were subjected to a single CSD episode. In vivo, extracellular single-unit recording was used to measure meningeal afferent ongoing activity changes. Quantitative mechanical stimuli using a servomotor force-controlled stimulator assessed changes in the afferent's mechanosensitivity. Manipulation of meningeal P2X receptors was achieved via local administration of pharmacological agents. Broad-spectrum P2X receptor inhibition, selective blockade of the P2X7 receptor, and its related Pannexin 1 channel suppressed CSD-evoked afferent mechanical sensitization but did not affect the accompanying afferent activation response. Surprisingly, inhibition of the pronociceptive P2X2/3 receptor did not affect the activation or sensitization of meningeal afferents post-CSD. P2X7 signaling underlying afferent mechanosensitization was localized to the meninges and did not affect CSD susceptibility. We propose that meningeal P2X7 and Pannexin 1 signaling, potentially in meningeal macrophages or neutrophils, mediates the mechanical sensitization of meningeal afferents, which contributes to migraine pain by exacerbating the headache during normally innocuous physical activities.SIGNIFICANCE STATEMENT Activation and sensitization of meningeal afferents play a key role in migraine headache, but the underlying mechanisms remain unclear. Here, using a rat model of migraine with aura involving cortical spreading depolarization (CSD), we demonstrate that meningeal purinergic P2X7 signaling and its related Pannexin 1 pore, but not nociceptive P2X2/3 receptors, mediate prolonged meningeal afferent sensitization. Additionally, we show that meningeal P2X signaling does not contribute to the increased afferent ongoing activity in the wake of CSD. Our finding points to meningeal P2X7 signaling as a critical mechanism underlying meningeal nociception in migraine, the presence of distinct mechanisms underlying the activation and sensitization of meningeal afferents in migraine, and highlight the need to target both processes for effective migraine therapy.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02115
| | - Samantha Harrison
- Center for Anesthesia Research Excellence, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115
| | - Dan Levy
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
19
|
Wang M, Ho MS. Profiling neurotransmitter-evoked glial responses by RNA-sequencing analysis. Front Neural Circuits 2023; 17:1252759. [PMID: 37645568 PMCID: PMC10461064 DOI: 10.3389/fncir.2023.1252759] [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: 07/04/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023] Open
Abstract
Fundamental properties of neurons and glia are distinctively different. Neurons are excitable cells that transmit information, whereas glia have long been considered as passive bystanders. Recently, the concept of tripartite synapse is proposed that glia are structurally and functionally incorporated into the synapse, the basic unit of information processing in the brains. It has then become intriguing how glia actively communicate with the presynaptic and postsynaptic compartments to influence the signal transmission. Here we present a thorough analysis at the transcriptional level on how glia respond to different types of neurotransmitters. Adult fly glia were purified from brains incubated with different types of neurotransmitters ex vivo. Subsequent RNA-sequencing analyses reveal distinct and overlapping patterns for these transcriptomes. Whereas Acetylcholine (ACh) and Glutamate (Glu) more vigorously activate glial gene expression, GABA retains its inhibitory effect. All neurotransmitters fail to trigger a significant change in the expression of their synthesis enzymes, yet Glu triggers increased expression of neurotransmitter receptors including its own and nAChRs. Expressions of transporters for GABA and Glutamate are under diverse controls from DA, GABA, and Glu, suggesting that the evoked intracellular pathways by these neurotransmitters are interconnected. Furthermore, changes in the expression of genes involved in calcium signaling also functionally predict the change in the glial activity. Finally, neurotransmitters also trigger a general metabolic suppression in glia except the DA, which upregulates a number of genes involved in transporting nutrients and amino acids. Our findings fundamentally dissect the transcriptional change in glia facing neuronal challenges; these results provide insights on how glia and neurons crosstalk in a synaptic context and underlie the mechanism of brain function and behavior.
Collapse
Affiliation(s)
| | - Margaret S. Ho
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
20
|
Cao A, Gao W, Sawada T, Yoshimoto RU, Aijima R, Ohsaki Y, Kido MA. Transient Receptor Potential Channel Vanilloid 1 Contributes to Facial Mechanical Hypersensitivity in a Mouse Model of Atopic Asthma. J Transl Med 2023; 103:100149. [PMID: 37059266 DOI: 10.1016/j.labinv.2023.100149] [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: 11/15/2022] [Revised: 03/22/2023] [Accepted: 03/29/2023] [Indexed: 04/16/2023] Open
Abstract
Sensitive skin, a common pathophysiological feature of allergic diseases, is defined as an unpleasant sensation in response to stimuli that normally should not provoke such sensations. However, the relationship between allergic inflammation and hypersensitive skin in the trigeminal system remains to be elucidated. To explore whether bronchial allergic inflammation affects facial skin and primary sensory neurons, we used an ovalbumin (OVA)-induced asthma mouse model. Significant mechanical hypersensitivity was observed in the facial skin of mice with pulmonary inflammation induced by OVA sensitization compared to mice treated with adjuvant or vehicle as controls. The skin of OVA-treated mice showed an increased number of nerve fibers, especially rich intraepithelial nerves, compared to controls. Transient receptor potential channel vanilloid 1 (TRPV1)-immunoreactive nerves were enriched in the skin of OVA-treated mice. Moreover, epithelial TRPV1 expression was higher in OVA-treated mice than in controls. Trigeminal ganglia of OVA-treated mice displayed larger numbers of activated microglia/macrophages and satellite glia. In addition, more TRPV1 immunoreactive neurons were found in the trigeminal ganglia of OVA-treated mice than in controls. Mechanical hypersensitivity was suppressed in OVA-treated Trpv1-deficient mice, while topical skin application of a TRPV1 antagonist before behavioral testing reduced the reaction induced by mechanical stimulation. Our findings reveal that mice with allergic inflammation of the bronchi had mechanical hypersensitivity in the facial skin that may have resulted from TRPV1-mediated neuronal plasticity and glial activation in the trigeminal ganglion.
Collapse
Affiliation(s)
- Ailin Cao
- Division of Histology and Neuroanatomy, Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan; Department of Oral Pathology, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Weiqi Gao
- Division of Histology and Neuroanatomy, Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Takeshi Sawada
- Division of Histology and Neuroanatomy, Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Reiko U Yoshimoto
- Division of Histology and Neuroanatomy, Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan; Department of Oral Pathology, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Reona Aijima
- Department of Oral Maxillofacial Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Yasuyoshi Ohsaki
- Division of Histology and Neuroanatomy, Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Mizuho A Kido
- Division of Histology and Neuroanatomy, Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan; Department of Oral Pathology, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
21
|
Yang Q, Jiang M, Xu S, Yang L, Yang P, Song Y, Zhu H, Wang Y, Sun Y, Yan C, Yuan Z, Liu X, Bai Z. Mirror image pain mediated by D2 receptor regulation of astrocytic Cx43 phosphorylation and channel opening. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166657. [PMID: 36716897 DOI: 10.1016/j.bbadis.2023.166657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/21/2022] [Accepted: 01/20/2023] [Indexed: 01/29/2023]
Abstract
Mirror image pain (MIP), a clinical syndrome of contralateral pain hypersensitivity caused by unilateral injury, has been identified in various neuropathological conditions. Gap junctional protein Connexin 43 (Cx43), its phosphorylation levels and dopamine D2 receptor (DRD2) play key integrating roles in pain processing. We presume D2DR activity may affect Cx43 hemichannel opening via Cx43 phosphorylation levels to regulate MIP. This study shows that spinal astrocytic Cx43 directly interacts with DRD2 to mediate MIP. DRD2 and Cx43 expression levels were asymmetrically elevated in bilateral spinal during MIP, and DRD2 modulated the opening of primary astrocytic Cx43 hemichannels. Furthermore, Cx43 phosphorylation at Ser373 was increased during MIP, but decreased in DRD2 knockout (KO) mice. Finally, activation of spinal protein kinase A (PKA) altered the expression of Cx43 and its phosphorylation bilaterally, thus reversing the analgesic effect in DRD2 KO mice. Together, these data reveal that spinal Cx43 phosphorylation and channel opening are regulated by DRD2 via PKA activation, and that spinal Cx43 and DRD2 are key molecular sensors mediating mirror image pain.
Collapse
Affiliation(s)
- Qinghu Yang
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China; Yanan Engineering & Technological Research Centre for Resource Peptide Drugs, Yanan 716000, China; Yanan Key Laboratory for Neural Immuno-Tumor and Stem Cell, Yanan 716000, China
| | - Ming Jiang
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China; Yanan Engineering & Technological Research Centre for Resource Peptide Drugs, Yanan 716000, China; Yanan Key Laboratory for Neural Immuno-Tumor and Stem Cell, Yanan 716000, China
| | - Sen Xu
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China
| | - Liang Yang
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China; Yanan Engineering & Technological Research Centre for Resource Peptide Drugs, Yanan 716000, China; Yanan Key Laboratory for Neural Immuno-Tumor and Stem Cell, Yanan 716000, China
| | - Pan Yang
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China
| | - Yutian Song
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China
| | - Hongni Zhu
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China
| | - Yu Wang
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China
| | - Yahan Sun
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China
| | - Chengxiang Yan
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China
| | - Zhaoyue Yuan
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China
| | - Xia Liu
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China; Yanan Engineering & Technological Research Centre for Resource Peptide Drugs, Yanan 716000, China; Yanan Key Laboratory for Neural Immuno-Tumor and Stem Cell, Yanan 716000, China.
| | - Zhantao Bai
- School of Life Science & Research Center for Natural Peptide Drugs, Shaanxi Engineering & Technological Research Centre for Conservation & Utilization of Regional Biological Resources, Yanan University, Yanan 716000, China; Yanan Engineering & Technological Research Centre for Resource Peptide Drugs, Yanan 716000, China; Yanan Key Laboratory for Neural Immuno-Tumor and Stem Cell, Yanan 716000, China.
| |
Collapse
|
22
|
Lawson J, LaVancher E, DeAlmeida M, Black BJ. Electrically-evoked oscillating calcium transients in mono- and co-cultures of iPSC glia and sensory neurons. Front Cell Neurosci 2023; 17:1094070. [PMID: 37006467 PMCID: PMC10060658 DOI: 10.3389/fncel.2023.1094070] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/17/2023] [Indexed: 03/18/2023] Open
Abstract
Activated glia are known to exhibit either neuroprotective or neurodegenerative effects, depending on their phenotype, while participating in chronic pain regulation. Until recently, it has been believed that satellite glial cells and astrocytes are electrically slight and process stimuli only through intracellular calcium flux that triggers downstream signaling mechanisms. Though glia do not exhibit action potentials, they do express both voltage- and ligand-gated ion channels that facilitate measurable calcium transients, a measure of their own phenotypic excitability, and support and modulate sensory neuron excitability through ion buffering and secretion of excitatory or inhibitory neuropeptides (i.e., paracrine signaling). We recently developed a model of acute and chronic nociception using co-cultures of iPSC sensory neurons (SN) and spinal astrocytes on microelectrode arrays (MEAs). Until recently, only neuronal extracellular activity has been recorded using MEAs with a high signal-to-noise ratio and in a non-invasive manner. Unfortunately, this method has limited compatibility with simultaneous calcium transient imaging techniques, which is the most common method for monitoring the phenotypic activity of astrocytes. Moreover, both dye-based and genetically encoded calcium indicator imaging rely on calcium chelation, affecting the culture's long-term physiology. Therefore, it would be ideal to allow continuous and simultaneous direct phenotypic monitoring of both SNs and astrocytes in a high-to-moderate throughput non-invasive manner and would significantly advance the field of electrophysiology. Here, we characterize astrocytic oscillating calcium transients (OCa2+Ts) in mono- and co-cultures of iPSC astrocytes as well as iPSC SN-astrocyte co-cultures on 48 well plate MEAs. We demonstrate that astrocytes exhibit OCa2+Ts in an electrical stimulus amplitude- and duration-dependent manner. We show that OCa2+Ts can be pharmacologically inhibited with the gap junction antagonist, carbenoxolone (100 μM). Most importantly, we demonstrate that both neurons and glia can be phenotypically characterized in real time, repeatedly, over the duration of the culture. In total, our findings suggest that calcium transients in glial populations may serve as a stand-alone or supplemental screening technique for identifying potential analgesics or compounds targeting other glia-mediated pathologies.
Collapse
Affiliation(s)
| | | | | | - Bryan James Black
- Department of Biomedical Engineering, Francis College of Engineering, University of Massachusetts Lowell, Lowell, MA, United States
| |
Collapse
|
23
|
Abstract
Satellite glial cells (SGCs) that surround sensory neurons in the peripheral nervous system ganglia originate from neural crest cells. Although several studies have focused on SGCs, the origin and characteristics of SGCs are unknown, and their lineage remains unidentified. Traditionally, it has been considered that SGCs regulate the environment around neurons under pathological conditions, and perform functions of supporting, nourishing, and protecting neurons. However, recent studies demonstrated that SGCs may have the characteristics of stem cells. After nerve injury, SGCs up-regulate the expression of stem cell markers and can differentiate into functional sensory neurons. Moreover, SGCs express several markers of Schwann cell precursors and Schwann cells, such as CDH19, MPZ, PLP1, SOX10, ERBB3, and FABP7. Schwann cell precursors have also been proposed as a potential source of neurons in the peripheral nervous system. The similarity in function and markers suggests that SGCs may represent a subgroup of Schwann cell precursors. Herein, we discuss the roles and functions of SGCs, and the lineage relationship between SGCs and Schwann cell precursors. We also describe a new perspective on the roles and functions of SGCs. In the DRG located on the posterior root of spinal nerves, satellite glial cells wrap around each sensory neuron to form an anatomically and functionally distinct unit with the sensory neurons. Following nerve injury, satellite glial cells up-regulate the expression of progenitor markers, and can differentiate into neurons.
Collapse
|
24
|
Age-Related Changes in Neurons and Satellite Glial Cells in Mouse Dorsal Root Ganglia. Int J Mol Sci 2023; 24:ijms24032677. [PMID: 36769006 PMCID: PMC9916822 DOI: 10.3390/ijms24032677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
The effects of aging on the nervous system are well documented. However, most previous studies on this topic were performed on the central nervous system. The present study was carried out on the dorsal root ganglia (DRGs) of mice, and focused on age-related changes in DRG neurons and satellite glial cells (SGCs). Intracellular electrodes were used for dye injection to examine the gap junction-mediated coupling between neurons and SGCs, and for intracellular electrical recordings from the neurons. Tactile sensitivity was assessed with von Frey hairs. We found that 3-23% of DRG neurons were dye-coupled to SGCs surrounding neighboring neurons in 8-24-month (Mo)-old mice, whereas in young adult (3 Mo) mice, the figure was 0%. The threshold current for firing an action potential in sensory neurons was significantly lower in DRGs from 12 Mo mice compared with those from 3 Mo mice. The percentage of neurons with spontaneous subthreshold membrane potential oscillation was greater by two-fold in 12 Mo mice. The withdrawal threshold was lower by 22% in 12 Mo mice compared with 3 Mo ones. These results show that in the aged mice, a proportion of DRG neurons is coupled to SGCs, and that the membrane excitability of the DRG neurons increases with age. We propose that augmented neuron-SGC communications via gap junctions are caused by low-grade inflammation associated with aging, and this may contribute to pain behavior.
Collapse
|
25
|
Pozzi E, Ballarini E, Rodriguez-Menendez V, Canta A, Chiorazzi A, Monza L, Bossi M, Alberti P, Malacrida A, Meregalli C, Scuteri A, Cavaletti G, Carozzi VA. Paclitaxel, but Not Cisplatin, Affects Satellite Glial Cells in Dorsal Root Ganglia of Rats with Chemotherapy-Induced Peripheral Neurotoxicity. TOXICS 2023; 11:93. [PMID: 36850969 PMCID: PMC9961471 DOI: 10.3390/toxics11020093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/13/2023] [Accepted: 01/13/2023] [Indexed: 06/18/2023]
Abstract
Chemotherapy-induced peripheral neurotoxicity is one of the most common dose-limiting toxicities of several widely used anticancer drugs such as platinum derivatives (cisplatin) and taxanes (paclitaxel). Several molecular mechanisms related to the onset of neurotoxicity have already been proposed, most of them having the sensory neurons of the dorsal root ganglia (DRG) and the peripheral nerve fibers as principal targets. In this study we explore chemotherapy-induced peripheral neurotoxicity beyond the neuronocentric view, investigating the changes induced by paclitaxel (PTX) and cisplatin (CDDP) on satellite glial cells (SGC) in the DRG and their crosstalk. Rats were chronically treated with PTX (10 mg/Kg, 1qwx4) or CDDP (2 mg/Kg 2qwx4) or respective vehicles. Morpho-functional analyses were performed to verify the features of drug-induced peripheral neurotoxicity. Qualitative and quantitative immunohistochemistry, 3D immunofluorescence, immunoblotting, and transmission electron microscopy analyses were also performed to detect alterations in SGCs and their interconnections. We demonstrated that PTX, but not CDDP, produces a strong activation of SGCs in the DRG, by altering their interconnections and their physical contact with sensory neurons. SGCs may act as principal actors in PTX-induced peripheral neurotoxicity, paving the way for the identification of new druggable targets for the treatment and prevention of chemotherapy-induced peripheral neurotoxicity.
Collapse
Affiliation(s)
- Eleonora Pozzi
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Elisa Ballarini
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Virginia Rodriguez-Menendez
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Annalisa Canta
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Alessia Chiorazzi
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Laura Monza
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Mario Bossi
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Paola Alberti
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Alessio Malacrida
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Cristina Meregalli
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Arianna Scuteri
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Guido Cavaletti
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| | - Valentina Alda Carozzi
- School of Medicine and Surgery, University of Milano-Bicocca, 20216 Monza, Italy
- NeuroMI (Milan Center for Neuroscience), 20126 Milan, Italy
| |
Collapse
|
26
|
FM1-43 Dye Memorizes Piezo1 Activation in the Trigeminal Nociceptive System Implicated in Migraine Pain. Int J Mol Sci 2023; 24:ijms24021688. [PMID: 36675204 PMCID: PMC9861983 DOI: 10.3390/ijms24021688] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
It has been proposed that mechanosensitive Piezo1 channels trigger migraine pain in trigeminal nociceptive neurons, but the mechanosensitivity of satellite glial cells (SGCs) supporting neuronal sensitization has not been tested before. Moreover, tools to monitor previous Piezo1 activation are not available. Therefore, by using live calcium imaging with Fluo-4 AM and labeling with FM1-43 dye, we explored a new strategy to identify Piezo channels' activity in mouse trigeminal neurons, SGCs, and isolated meninges. The specific Piezo1 agonist Yoda1 induced calcium transients in both neurons and SGCs, suggesting the functional expression of Piezo1 channels in both types of cells. In Piezo1-transfected HEK cells, FM1-43 produced only a transient fluorescent response, whereas co-application with Yoda1 provided higher transient signals and a remarkable long-lasting FM1-43 'tail response'. A similar Piezo1-related FM1-43 trapping was observed in neurons and SGCs. The non-specific Piezo channel blocker, Gadolinium, inhibited the transient peak, confirming the involvement of Piezo1 receptors. Finally, FM1-43 labeling demonstrated previous activity in meningeal tissues 3.5 h after Yoda1 washout. Our data indicated that trigeminal neurons and SGCs express functional Piezo channels, and their activation provides sustained labeling with FM1-43. This long-lasting labelling can be used to monitor the ongoing and previous activation of Piezo1 channels in the trigeminal nociceptive system, which is implicated in migraine pain.
Collapse
|
27
|
Schwann cell functions in peripheral nerve development and repair. Neurobiol Dis 2023; 176:105952. [PMID: 36493976 DOI: 10.1016/j.nbd.2022.105952] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
The glial cell of the peripheral nervous system (PNS), the Schwann cell (SC), counts among the most multifaceted cells of the body. During development, SCs secure neuronal survival and participate in axonal path finding. Simultaneously, they orchestrate the architectural set up of the developing nerves, including the blood vessels and the endo-, peri- and epineurial layers. Perinatally, in rodents, SCs radially sort and subsequently myelinate individual axons larger than 1 μm in diameter, while small calibre axons become organised in non-myelinating Remak bundles. SCs have a vital role in maintaining axonal health throughout life and several specialized SC types perform essential functions at specific locations, such as terminal SC at the neuromuscular junction (NMJ) or SC within cutaneous sensory end organs. In addition, neural crest derived satellite glia maintain a tight communication with the soma of sensory, sympathetic, and parasympathetic neurons and neural crest derivatives are furthermore an indispensable part of the enteric nervous system. The remarkable plasticity of SCs becomes evident in the context of a nerve injury, where SC transdifferentiate into intriguing repair cells, which orchestrate a regenerative response that promotes nerve repair. Indeed, the multiple adaptations of SCs are captivating, but remain often ill-resolved on the molecular level. Here, we summarize and discuss the knowns and unknowns of the vast array of functions that this single cell type can cover in peripheral nervous system development, maintenance, and repair.
Collapse
|
28
|
Tanaka M, Zhang Y. Preclinical Studies of Posttraumatic Headache and the Potential Therapeutics. Cells 2022; 12:cells12010155. [PMID: 36611947 PMCID: PMC9818317 DOI: 10.3390/cells12010155] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/13/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Posttraumatic headache (PTH) attributed to traumatic brain injury (TBI) is a secondary headache developed within 7 days after head injury, and in a substantial number of patients PTH becomes chronic and lasts for more than 3 months. Current medications are almost entirely relied on the treatment of primary headache such as migraine, due to its migraine-like phenotype and the limited understanding on the PTH pathogenic mechanisms. To this end, increasing preclinical studies have been conducted in the last decade. We focus in this review on the trigeminovascular system from the animal studies since it provides the primary nociceptive sensory afferents innervating the head and face region, and the pathological changes in the trigeminal pathway are thought to play a key role in the development of PTH. In addition to the pathologies, PTH-like behaviors induced by TBI and further exacerbated by nitroglycerin, a general headache inducer through vasodilation are reviewed. We will overview the current pharmacotherapies including calcitonin gene-related peptide (CGRP) monoclonal antibody and sumatriptan in the PTH animal models. Given that modulation of the endocannabinoid (eCB) system has been well-documented in the treatment of migraine and TBI, the therapeutic potential of eCB in PTH will also be discussed.
Collapse
|
29
|
Calvin-Cejudo L, Martin F, Mendez LR, Coya R, Castañeda-Sampedro A, Gomez-Diaz C, Alcorta E. Neuron-glia interaction at the receptor level affects olfactory perception in adult Drosophila. iScience 2022; 26:105837. [PMID: 36624835 PMCID: PMC9823236 DOI: 10.1016/j.isci.2022.105837] [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: 08/09/2022] [Revised: 11/17/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Some types of glia play an active role in neuronal signaling by modifying their activity although little is known about their role in sensory information signaling at the receptor level. In this research, we report a functional role for the glia that surround the soma of the olfactory receptor neurons (OSNs) in adult Drosophila. Specific genetic modifications have been targeted to this cell type to obtain live individuals who are tested for olfactory preference and display changes both increasing and reducing sensitivity. A closer look at the antenna by Ca2+ imaging shows that odor activates the OSNs, which subsequently produce an opposite and smaller effect in the glia that partially counterbalances neuronal activation. Therefore, these glia may play a dual role in preventing excessive activation of the OSNs at high odorant concentrations and tuning the chemosensory window for the individual according to the network structure in the receptor organ.
Collapse
Affiliation(s)
- Laura Calvin-Cejudo
- Group of Neurobiology of the Sensory Systems (NEUROSEN), Department of Functional Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | - Fernando Martin
- Group of Neurobiology of the Sensory Systems (NEUROSEN), Department of Functional Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | - Luis R. Mendez
- Group of Neurobiology of the Sensory Systems (NEUROSEN), Department of Functional Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | - Ruth Coya
- Group of Neurobiology of the Sensory Systems (NEUROSEN), Department of Functional Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | - Ana Castañeda-Sampedro
- Group of Neurobiology of the Sensory Systems (NEUROSEN), Department of Functional Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | - Carolina Gomez-Diaz
- Group of Neurobiology of the Sensory Systems (NEUROSEN), Department of Functional Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
| | - Esther Alcorta
- Group of Neurobiology of the Sensory Systems (NEUROSEN), Department of Functional Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Corresponding author
| |
Collapse
|
30
|
Avraham O, Chamessian A, Feng R, Yang L, Halevi AE, Moore AM, Gereau RW, Cavalli V. Profiling the molecular signature of satellite glial cells at the single cell level reveals high similarities between rodents and humans. Pain 2022; 163:2348-2364. [PMID: 35503034 PMCID: PMC9522926 DOI: 10.1097/j.pain.0000000000002628] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 02/23/2022] [Indexed: 11/25/2022]
Abstract
ABSTRACT Peripheral sensory neurons located in dorsal root ganglia relay sensory information from the peripheral tissue to the brain. Satellite glial cells (SGCs) are unique glial cells that form an envelope completely surrounding each sensory neuron soma. This organization allows for close bidirectional communication between the neuron and its surrounding glial coat. Morphological and molecular changes in SGC have been observed in multiple pathological conditions such as inflammation, chemotherapy-induced neuropathy, viral infection, and nerve injuries. There is evidence that changes in SGC contribute to chronic pain by augmenting the neuronal activity in various rodent pain models. Satellite glial cells also play a critical role in axon regeneration. Whether findings made in rodent model systems are relevant to human physiology have not been investigated. Here, we present a detailed characterization of the transcriptional profile of SGC in mice, rats, and humans at the single cell level. Our findings suggest that key features of SGC in rodent models are conserved in humans. Our study provides the potential to leverage rodent SGC properties and identify potential targets in humans for the treatment of nerve injuries and alleviation of painful conditions.
Collapse
Affiliation(s)
- Oshri Avraham
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Alexander Chamessian
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Rui Feng
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Lite Yang
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Neuroscience Program, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Alexandra E. Halevi
- Department of Plastic and Reconstructive Surgery, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Amy M. Moore
- Department of Plastic and Reconstructive Surgery, The Ohio State University, Columbus Ohio, USA
| | - Robert W. Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
31
|
Jia S, Liu J, Chu Y, Liu Q, Mai L, Fan W. Single-cell RNA sequencing reveals distinct transcriptional features of the purinergic signaling in mouse trigeminal ganglion. Front Mol Neurosci 2022; 15:1038539. [PMID: 36311028 PMCID: PMC9606672 DOI: 10.3389/fnmol.2022.1038539] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/27/2022] [Indexed: 11/30/2022] Open
Abstract
Trigeminal ganglion (TG) is the first station of sensory pathways in the orofacial region. The TG neurons communicate with satellite glial cells (SGCs), macrophages and other cells forming a functional unit that is responsible for processing of orofacial sensory information. Purinergic signaling, one of the most widespread autocrine and paracrine pathways, plays a crucial role in intercellular communication. The multidirectional action of purinergic signaling in different cell types contributes to the neuromodulation and orofacial sensation. To fully understand the purinergic signaling in these processes, it is essential to determine the shared and unique expression patterns of genes associated with purinergic signaling in different cell types. Here, we performed single-cell RNA sequencing of 22,969 cells isolated from normal mouse TGs. We identified 18 distinct cell populations, including 6 neuron subpopulations, 3 glial subpopulations, 7 immune cell subpopulations, fibroblasts, and endothelial cells. We also revealed the transcriptional features of genes associated with purinergic signaling, including purinergic receptors, extracellular adenosine triphosphate (eATP) release channels, eATP metabolism-associated enzymes, and eATP transporters in each cell type. Our results have important implications for understanding and predicting the cell type-specific roles of the purinergic signaling in orofacial signal processing in the trigeminal primary sensory system.
Collapse
Affiliation(s)
- Shilin Jia
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - JinYue Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yanhao Chu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Qing Liu
- Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Lijia Mai
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Wenguo Fan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- *Correspondence: Wenguo Fan,
| |
Collapse
|
32
|
BzATP Activates Satellite Glial Cells and Increases the Excitability of Dorsal Root Ganglia Neurons In Vivo. Cells 2022; 11:cells11152280. [PMID: 35892578 PMCID: PMC9330736 DOI: 10.3390/cells11152280] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 12/14/2022] Open
Abstract
The purinergic system plays an important role in pain transmission. Recent studies have suggested that activation of P2-purinergic receptors (P2Rs) may be involved in neuron-satellite glial cell (SGC) interactions in the dorsal root ganglia (DRG), but the details remain unclear. In DRG, P2X7R is selectively expressed in SGCs, which closely surround neurons, and is highly sensitive to 3’-O-(4-Benzoyl) benzoyl-ATP (BzATP). Using calcium imaging in intact mice to survey a large number of DRG neurons and SGCs, we examined how intra-ganglionic purinergic signaling initiated by BzATP affects neuronal activities in vivo. We developed GFAP-GCaMP6s and Pirt-GCaMP6s mice to express the genetically encoded calcium indicator GGCaM6s in SGCs and DRG neurons, respectively. The application of BzATP to the ganglion induced concentration-dependent activation of SGCs in GFAP-GCaMP6s mice. In Pirt-GCaMP6s mice, BzATP initially activated more large-size neurons than small-size ones. Both glial and neuronal responses to BzATP were blocked by A438079, a P2X7R-selective antagonist. Moreover, blockers to pannexin1 channels (probenecid) and P2X3R (A317491) also reduced the actions of BzATP, suggesting that P2X7R stimulation may induce the opening of pannexin1 channels, leading to paracrine ATP release, which could further excite neurons by acting on P2X3Rs. Importantly, BzATP increased the responses of small-size DRG neurons and wide-dynamic range spinal neurons to subsequent peripheral stimuli. Our findings suggest that intra-ganglionic purinergic signaling initiated by P2X7R activation could trigger SGC-neuron interaction in vivo and increase DRG neuron excitability.
Collapse
|
33
|
Zheng Q, Dong X, Green DP, Dong X. Peripheral mechanisms of chronic pain. MEDICAL REVIEW 2022; 2:251-270. [PMID: 36067122 PMCID: PMC9381002 DOI: 10.1515/mr-2022-0013] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/13/2022] [Indexed: 11/15/2022]
Abstract
Abstract
Acutely, pain serves to protect us from potentially harmful stimuli, however damage to the somatosensory system can cause maladaptive changes in neurons leading to chronic pain. Although acute pain is fairly well controlled, chronic pain remains difficult to treat. Chronic pain is primarily a neuropathic condition, but studies examining the mechanisms underlying chronic pain are now looking beyond afferent nerve lesions and exploring new receptor targets, immune cells, and the role of the autonomic nervous system in contributing chronic pain conditions. The studies outlined in this review reveal how chronic pain is not only confined to alterations in the nervous system and presents findings on new treatment targets and for this debilitating disease.
Collapse
Affiliation(s)
- Qin Zheng
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xintong Dong
- The Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Dustin P. Green
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Howard Hughes Medical Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
34
|
Tang G, Pi L, Guo H, Hu Z, Zhou C, Hu Q, Peng H, Xiao Z, Zhang Z, Wang M, Peng T, Huang J, Liang S, Li G. Naringin Relieves Diabetic Cardiac Autonomic Neuropathy Mediated by P2Y14 Receptor in Superior Cervical Ganglion. Front Pharmacol 2022; 13:873090. [PMID: 35529431 PMCID: PMC9068893 DOI: 10.3389/fphar.2022.873090] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/23/2022] [Indexed: 12/30/2022] Open
Abstract
Diabetes mellitus (DM), an emerging chronic epidemic, contributes to mortality and morbidity around the world. Diabetic cardiac autonomic neuropathy (DCAN) is one of the most common complications associated with DM. Previous studies have shown that satellite glial cells (SGCs) in the superior cervical ganglia (SCG) play an indispensable role in DCAN progression. In addition, it has been shown that purinergic neurotransmitters, as well as metabotropic GPCRs, are involved in the pathophysiological process of DCAN. Furthermore, one traditional Chinese medicine, naringin may potently alleviate the effects of DCAN. Ferroptosis may be involved in DCAN progression. However, the role of naringin in DCAN as well as its detailed mechanism requires further investigation. In this research, we attempted to identify the effect and relevant mechanism of naringin in DCAN mitigation. We observed that compared with those of normal subjects, there were significantly elevated expression levels of P2Y14 and IL-1β in diabetic rats, both of which were remarkably diminished by treatment with either P2Y14 shRNA or naringin. In addition, abnormalities in blood pressure (BP), heart rate (HR), heart rate variability (HRV), sympathetic nerve discharge (SND), and cardiac structure in the diabetic model can also be partially returned to normal through the use of those treatments. Furthermore, a reduced expression of NRF2 and GPX4, as well as an elevated level of ROS, were detected in diabetic cases, which can also be improved with those treatments. Our results showed that naringin can effectively relieve DCAN mediated by the P2Y14 receptor of SGCs in the SCG. Moreover, the NRF2/GPX4 pathway involved in ferroptosis may become one of the principal mechanisms participating in DCAN progression, which can be modulated by P2Y14-targeted naringin and thus relieve DCAN. Hopefully, our research can supply one novel therapeutic target and provide a brilliant perspective for the treatment of DCAN.
Collapse
Affiliation(s)
- Gan Tang
- Queen Mary School, Medical School of Nanchang University, Nanchang, China
| | - Lingzhi Pi
- School of Basic Medicine, Medical School of Nanchang University, Nanchang, China
| | - Hongmin Guo
- Department of Physiology, Medical School of Nanchang University, Nanchang, China
| | - Zihui Hu
- Department of Physiology, Medical School of Nanchang University, Nanchang, China
| | - Congfa Zhou
- Department of Anatomy, Medical School of Nanchang University, Nanchang, China
| | - Qixing Hu
- Department of Physiology, Medical School of Nanchang University, Nanchang, China
| | - Hao Peng
- School of Basic Medicine, Medical School of Nanchang University, Nanchang, China
| | - Zehao Xiao
- Queen Mary School, Medical School of Nanchang University, Nanchang, China
| | - Zhihua Zhang
- Queen Mary School, Medical School of Nanchang University, Nanchang, China
| | - Miaomiao Wang
- Queen Mary School, Medical School of Nanchang University, Nanchang, China
| | - Taotao Peng
- School of Basic Medicine, Medical School of Nanchang University, Nanchang, China
| | - Jiaqi Huang
- Queen Mary School, Medical School of Nanchang University, Nanchang, China
| | - Shangdong Liang
- Department of Physiology, Medical School of Nanchang University, Nanchang, China
| | - Guilin Li
- Department of Physiology, Medical School of Nanchang University, Nanchang, China
- *Correspondence: Guilin Li,
| |
Collapse
|
35
|
Sinegubov A, Andreeva D, Burzak N, Vasyutina M, Murashova L, Dyachuk V. Heterogeneity and Potency of Peripheral Glial Cells in Embryonic Development and Adults. Front Mol Neurosci 2022; 15:737949. [PMID: 35401107 PMCID: PMC8990813 DOI: 10.3389/fnmol.2022.737949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
This review describes the heterogeneity of peripheral glial cell populations, from the emergence of Schwann cells (SCs) in early development, to their involvement, and that of their derivatives in adult glial populations. We focus on the origin of the first glial precursors from neural crest cells (NCCs), and their ability to differentiate into several cell types during development. We also discuss the heterogeneity of embryonic glia in light of the latest data from genetic tracing and transcriptome analysis. Special attention has been paid to the biology of glial populations in adult animals, by highlighting common features of different glial cell types and molecular differences that modulate their functions. Finally, we consider the communication of glial cells with axons of neurons in normal and pathological conditions. In conclusion, the present review details how information available on glial cell types and their functions in normal and pathological conditions may be utilized in the development of novel therapeutic strategies for the treatment of patients with neurodiseases.
Collapse
|
36
|
Khaitin A. Calcium in Neuronal and Glial Response to Axotomy. Int J Mol Sci 2021; 22:ijms222413344. [PMID: 34948141 PMCID: PMC8706492 DOI: 10.3390/ijms222413344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022] Open
Abstract
Neurotrauma assumes an instant or delayed disconnection of axons (axotomy), which affects not only neurons, but surrounding glia as well. Not only mechanically injured glia near the site of disconnection, especially transection, is subjected to the damage, but also glia that is remote from the lesion site. Glial cells, which surround the neuronal body, in turn, support neuron survival, so there is a mutual protection between neuron and glia. Calcium signaling is a central mediator of all post-axotomy events, both in neuron and glia, playing a critical role in their survival/regeneration or death/degeneration. The involvement of calcium in post-axotomy survival of the remote, mechanically intact glia is poorly studied. The purpose of this review is to sum up the calcium-involving mechanisms in responses of neurons and glial cells to axotomy to show their importance and to give some suggestions for future research of remote glia in this context.
Collapse
Affiliation(s)
- Andrey Khaitin
- Academy of Biology and Biotechnology, Southern Federal University, 344090 Rostov-on-Don, Russia
| |
Collapse
|
37
|
Rawat A, Morrison BM. Metabolic Transporters in the Peripheral Nerve-What, Where, and Why? Neurotherapeutics 2021; 18:2185-2199. [PMID: 34773210 PMCID: PMC8804006 DOI: 10.1007/s13311-021-01150-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 12/18/2022] Open
Abstract
Cellular metabolism is critical not only for cell survival, but also for cell fate, function, and intercellular communication. There are several different metabolic transporters expressed in the peripheral nervous system, and they each play important roles in maintaining cellular energy. The major source of energy in the peripheral nervous system is glucose, and glucose transporters 1 and 3 are expressed and allow blood glucose to be imported and utilized by peripheral nerves. There is also increasing evidence that other sources of energy, particularly monocarboxylates such as lactate that are transported primarily by monocarboxylate transporters 1 and 2 in peripheral nerves, can be efficiently utilized by peripheral nerves. Finally, emerging evidence supports an important role for connexins and possibly pannexins in the supply and regulation of metabolic energy. In this review, we will first define these critical metabolic transporter subtypes and then examine their localization in the peripheral nervous system. We will subsequently discuss the evidence, which comes both from experiments in animal models and observations from human diseases, supporting critical roles played by these metabolic transporters in the peripheral nervous system. Despite progress made in understanding the function of these transporters, many questions and some discrepancies remain, and these will also be addressed throughout this review. Peripheral nerve metabolism is fundamentally important and renewed interest in these pathways should help to answer many of these questions and potentially provide new treatments for neurologic diseases that are partly, or completely, caused by disruption of metabolism.
Collapse
Affiliation(s)
- Atul Rawat
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brett M Morrison
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
38
|
Goebel A, Krock E, Gentry C, Israel MR, Jurczak A, Urbina CM, Sandor K, Vastani N, Maurer M, Cuhadar U, Sensi S, Nomura Y, Menezes J, Baharpoor A, Brieskorn L, Sandström A, Tour J, Kadetoff D, Haglund L, Kosek E, Bevan S, Svensson CI, Andersson DA. Passive transfer of fibromyalgia symptoms from patients to mice. J Clin Invest 2021; 131:e144201. [PMID: 34196305 DOI: 10.1172/jci144201] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 05/11/2021] [Indexed: 12/25/2022] Open
Abstract
Fibromyalgia syndrome (FMS) is characterized by widespread pain and tenderness, and patients typically experience fatigue and emotional distress. The etiology and pathophysiology of fibromyalgia are not fully explained and there are no effective drug treatments. Here we show that IgG from FMS patients produced sensory hypersensitivity by sensitizing nociceptive neurons. Mice treated with IgG from FMS patients displayed increased sensitivity to noxious mechanical and cold stimulation, and nociceptive fibers in skin-nerve preparations from mice treated with FMS IgG displayed an increased responsiveness to cold and mechanical stimulation. These mice also displayed reduced locomotor activity, reduced paw grip strength, and a loss of intraepidermal innervation. In contrast, transfer of IgG-depleted serum from FMS patients or IgG from healthy control subjects had no effect. Patient IgG did not activate naive sensory neurons directly. IgG from FMS patients labeled satellite glial cells and neurons in vivo and in vitro, as well as myelinated fiber tracts and a small number of macrophages and endothelial cells in mouse dorsal root ganglia (DRG), but no cells in the spinal cord. Furthermore, FMS IgG bound to human DRG. Our results demonstrate that IgG from FMS patients produces painful sensory hypersensitivities by sensitizing peripheral nociceptive afferents and suggest that therapies reducing patient IgG titers may be effective for fibromyalgia.
Collapse
Affiliation(s)
- Andreas Goebel
- Walton Centre NHS Foundation Trust, Liverpool, United Kingdom.,Pain Research Institute, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Emerson Krock
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Clive Gentry
- King's College London, Wolfson CARD, Institute of Psychiatry, Psychology & Neuroscience, Guy's Campus, London, United Kingdom
| | - Mathilde R Israel
- King's College London, Wolfson CARD, Institute of Psychiatry, Psychology & Neuroscience, Guy's Campus, London, United Kingdom
| | - Alexandra Jurczak
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Carlos Morado Urbina
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Nisha Vastani
- King's College London, Wolfson CARD, Institute of Psychiatry, Psychology & Neuroscience, Guy's Campus, London, United Kingdom
| | - Margot Maurer
- King's College London, Wolfson CARD, Institute of Psychiatry, Psychology & Neuroscience, Guy's Campus, London, United Kingdom
| | - Ulku Cuhadar
- King's College London, Wolfson CARD, Institute of Psychiatry, Psychology & Neuroscience, Guy's Campus, London, United Kingdom
| | - Serena Sensi
- Pain Research Institute, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Yuki Nomura
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Joana Menezes
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Azar Baharpoor
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Louisa Brieskorn
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Angelica Sandström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jeanette Tour
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Diana Kadetoff
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Stockholm Spine Center, Upplands Väsby, Sweden
| | - Lisbet Haglund
- Department of Surgery, Division of Orthopaedic Surgery, McGill University, Montreal, Quebec, Canada
| | - Eva Kosek
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Stuart Bevan
- King's College London, Wolfson CARD, Institute of Psychiatry, Psychology & Neuroscience, Guy's Campus, London, United Kingdom
| | - Camilla I Svensson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - David A Andersson
- King's College London, Wolfson CARD, Institute of Psychiatry, Psychology & Neuroscience, Guy's Campus, London, United Kingdom
| |
Collapse
|
39
|
Affiliation(s)
- Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center and Faculty of Medicine, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Robert W Banks
- Department of Biosciences, and Biophysical Sciences Institute, Durham University, Durham, UK
| |
Collapse
|
40
|
Ren Y, Liu Y, Luo M. Gap Junctions Between Striatal D1 Neurons and Cholinergic Interneurons. Front Cell Neurosci 2021; 15:674399. [PMID: 34168539 PMCID: PMC8217616 DOI: 10.3389/fncel.2021.674399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/29/2021] [Indexed: 01/15/2023] Open
Abstract
The striatum participates in numerous important behaviors. Its principal projection neurons use GABA and peptides as neurotransmitters and interact extensively with interneurons, including cholinergic interneurons (ChIs) that are tonically active. Dissecting the interactions between projection neurons and ChIs is important for uncovering the role and mechanisms of the striatal microcircuits. Here, by combining several optogenetic tools with cell type-specific electrophysiological recordings, we uncovered direct electrical coupling between D1-type projection neurons and ChIs, in addition to the chemical transmission between these two major cell types. Optogenetic stimulation or inhibition led to bilateral current exchanges between D1 neurons and ChIs, which can be abolished by gap junction blockers. We further confirmed the presence of gap junctions through paired electrophysiological recordings and dye microinjections. Finally, we found that activating D1 neurons promotes basal activity of ChIs via gap junctions. Collectively, these results reveal the coexistence of the chemical synapse and gap junctions between D1 neurons and ChIs, which contributes to maintaining the tonically active firing patterns of ChIs.
Collapse
Affiliation(s)
- Yuqi Ren
- School of Life Sciences, Peking University, Beijing, China.,Peking University-Tsinghua University-NIBS Joint Graduate Program, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Yang Liu
- School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
| | - Minmin Luo
- National Institute of Biological Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Beijing, China
| |
Collapse
|
41
|
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: 0.8] [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
|
42
|
Wang D, Lu J, Xu X, Yuan Y, Zhang Y, Xu J, Chen H, Liu J, Shen Y, Zhang H. Satellite Glial Cells Give Rise to Nociceptive Sensory Neurons. Stem Cell Rev Rep 2021; 17:999-1013. [PMID: 33389681 DOI: 10.1007/s12015-020-10102-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 12/27/2022]
Abstract
Dorsal root ganglia (DRG) sensory neurons can transmit information about noxious stimulus to cerebral cortex via spinal cord, and play an important role in the pain pathway. Alterations of the pain pathway lead to CIPA (congenital insensitivity to pain with anhidrosis) or chronic pain. Accumulating evidence demonstrates that nerve damage leads to the regeneration of neurons in DRG, which may contribute to pain modulation in feedback. Therefore, exploring the regeneration process of DRG neurons would provide a new understanding to the persistent pathological stimulation and contribute to reshape the somatosensory function. It has been reported that a subpopulation of satellite glial cells (SGCs) express Nestin and p75, and could differentiate into glial cells and neurons, suggesting that SGCs may have differentiation plasticity. Our results in the present study show that DRG-derived SGCs (DRG-SGCs) highly express neural crest cell markers Nestin, Sox2, Sox10, and p75, and differentiate into nociceptive sensory neurons in the presence of histone deacetylase inhibitor VPA, Wnt pathway activator CHIR99021, Notch pathway inhibitor RO4929097, and FGF pathway inhibitor SU5402. The nociceptive sensory neurons express multiple functionally-related genes (SCN9A, SCN10A, SP, Trpv1, and TrpA1) and are able to generate action potentials and voltage-gated Na+ currents. Moreover, we found that these cells exhibited rapid calcium transients in response to capsaicin through binding to the Trpv1 vanilloid receptor, confirming that the DRG-SGC-derived cells are nociceptive sensory neurons. Further, we show that Wnt signaling promotes the differentiation of DRG-SGCs into nociceptive sensory neurons by regulating the expression of specific transcription factor Runx1, while Notch and FGF signaling pathways are involved in the expression of SCN9A. These results demonstrate that DRG-SGCs have stem cell characteristics and can efficiently differentiate into functional nociceptive sensory neurons, shedding light on the clinical treatment of sensory neuron-related diseases.
Collapse
Affiliation(s)
- Dongyan Wang
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Junhou Lu
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Xiaojing Xu
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Ye Yuan
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Yu Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jianwei Xu
- National Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Center for Tissue Engineering and Stem Cell Research, Guizhou Province Key Laboratory of Regenerative Medicine, Guizhou Medical University, Guiyang, 550004, China
| | - Huanhuan Chen
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Jinming Liu
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China
| | - Yixin Shen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Huanxiang Zhang
- Department of Cell Biology, Medical College of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
43
|
Ceruti S. From astrocytes to satellite glial cells and back: A 25 year-long journey through the purinergic modulation of glial functions in pain and more. Biochem Pharmacol 2020; 187:114397. [PMID: 33382970 DOI: 10.1016/j.bcp.2020.114397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/16/2022]
Abstract
Fundamental progresses have been made in pain research with a comprehensive understanding of the neuronal pathways which convey painful sensations from the periphery and viscera to the central nervous system and of the descending modulating pathways. Nevertheless, many patients still suffer from various painful conditions, which are often associated to other primary pathologies, and get no or poor relief from available painkillers. Thus, the interest of many researchers has concentrated on new and promising cellular targets and biochemical pathways. This is the case of glia cells, both in the peripheral and in the central nervous system, and of purinergic receptors. Starting from many intuitions and hypotheses raised by Prof. Geoffrey Burnstock, data have accumulated which clearly highlight the fundamental role exerted by several nucleotide and nucleoside receptors in the modulation of glial cell reaction to pain triggers and of their cross-talk with sensory neurons which significantly contributes to the transition from acute to chronic pain. The purinergic system has therefore become an appealing pharmacological target in pain research, also based on the quite unexpected discovery that purines are involved in ancient analgesic techniques such as acupuncture. A more in-depth understanding of the complex and intricated purine-orchestrated scenario in pain conditions will hopefully lead to the identification and clinical development of new and effective analgesics.
Collapse
Affiliation(s)
- Stefania Ceruti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti, 9, 20133 Milan, Italy.
| |
Collapse
|
44
|
Li Q, Ma TL, Qiu YQ, Cui WQ, Chen T, Zhang WW, Wang J, Mao-Ying QL, Mi WL, Wang YQ, Chu YX. Connexin 36 Mediates Orofacial Pain Hypersensitivity Through GluK2 and TRPA1. Neurosci Bull 2020; 36:1484-1499. [PMID: 33067780 PMCID: PMC7719140 DOI: 10.1007/s12264-020-00594-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 09/06/2020] [Indexed: 12/15/2022] Open
Abstract
Trigeminal neuralgia is a debilitating condition, and the pain easily spreads to other parts of the face. Here, we established a mouse model of partial transection of the infraorbital nerve (pT-ION) and found that the Connexin 36 (Cx36) inhibitor mefloquine caused greater alleviation of pT-ION-induced cold allodynia compared to the reduction of mechanical allodynia. Mefloquine reversed the pT-ION-induced upregulation of Cx36, glutamate receptor ionotropic kainate 2 (GluK2), transient receptor potential ankyrin 1 (TRPA1), and phosphorylated extracellular signal regulated kinase (p-ERK) in the trigeminal ganglion. Cold allodynia but not mechanical allodynia induced by pT-ION or by virus-mediated overexpression of Cx36 in the trigeminal ganglion was reversed by the GluK2 antagonist NS102, and knocking down Cx36 expression in Nav1.8-expressing nociceptors by injecting virus into the orofacial skin area of Nav1.8-Cre mice attenuated cold allodynia but not mechanical allodynia. In conclusion, we show that Cx36 contributes greatly to the development of orofacial pain hypersensitivity through GluK2, TRPA1, and p-ERK signaling.
Collapse
Affiliation(s)
- Qian Li
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China
| | - Tian-Le Ma
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China
| | - You-Qi Qiu
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China
| | - Wen-Qiang Cui
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China
- Department of Pain Management, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, 250000, China
| | - Teng Chen
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China
| | - Wen-Wen Zhang
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China
| | - Jing Wang
- Department of Nephropathy, The Third Affiliated Hospital of Shenzhen University, Luohu Hospital Group, Shenzhen, 518001, China
| | - Qi-Liang Mao-Ying
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China
| | - Wen-Li Mi
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China
| | - Yu-Xia Chu
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
45
|
Li Q, Wang YQ, Chu YX. The role of connexins and pannexins in orofacial pain. Life Sci 2020; 258:118198. [PMID: 32758624 DOI: 10.1016/j.lfs.2020.118198] [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: 05/13/2020] [Revised: 07/18/2020] [Accepted: 07/31/2020] [Indexed: 10/23/2022]
Abstract
Trigeminal neuralgia is characterized by extensive spreading of pain, referred to as ectopic pain, which describes the phenomenon of the pain passing from the injured regions to uninjured regions. Patients with orofacial pain often show no response to commonly used analgesics, and the exact mechanism of ectopic pain remains unclear, which restricts the development of specific drugs. The present review aims to summarize the contribution of the two families of transmembrane proteins, connexins (Cxs) and pannexins (Panxs), to the induction and spreading of orofacial pain and to provide potential targets for orofacial pain treatment. Cxs and Panxs have recently been shown to play essential roles in intercellular signal propagation in sensory ganglia, and previous studies have provided evidence for the contribution of several subtypes of Cxs and Panxs in various orofacial pain models. Upregulation of the expression of Cxs and Panxs in the trigeminal ganglia is observed in most cases after trigeminal injury, and regulating their expression or activity can improve pain-like behaviors in animals. It is speculated that after trigeminal injury, pain-related signals are transmitted to adjacent neurons and satellite glial cells in the trigeminal ganglia directly through gap junctions and simultaneously through hemichannels and pannexons through both autocrine and paracrine mechanisms. This review highlights recent discoveries in the regulation of Cxs and Panxs in different orofacial pain models and presents a hypothetical mechanism of ectopic pain in trigeminal neuralgia. In addition, the existing problems in current research are discussed.
Collapse
Affiliation(s)
- Qian Li
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Yu-Xia Chu
- Department of Integrative Medicine and Neurobiology, Institutes of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
46
|
Are glia targets for neuropathic orofacial pain therapy? J Am Dent Assoc 2020; 152:774-779. [PMID: 32921390 DOI: 10.1016/j.adaj.2020.05.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 01/06/2023]
|
47
|
Hanani M, Spray DC. Emerging importance of satellite glia in nervous system function and dysfunction. Nat Rev Neurosci 2020; 21:485-498. [PMID: 32699292 PMCID: PMC7374656 DOI: 10.1038/s41583-020-0333-z] [Citation(s) in RCA: 221] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2020] [Indexed: 02/08/2023]
Abstract
Satellite glial cells (SGCs) closely envelop cell bodies of neurons in sensory, sympathetic and parasympathetic ganglia. This unique organization is not found elsewhere in the nervous system. SGCs in sensory ganglia are activated by numerous types of nerve injury and inflammation. The activation includes upregulation of glial fibrillary acidic protein, stronger gap junction-mediated SGC-SGC and neuron-SGC coupling, increased sensitivity to ATP, downregulation of Kir4.1 potassium channels and increased cytokine synthesis and release. There is evidence that these changes in SGCs contribute to chronic pain by augmenting neuronal activity and that these changes are consistent in various rodent pain models and likely also in human pain. Therefore, understanding these changes and the resulting abnormal interactions of SGCs with sensory neurons could provide a mechanistic approach that might be exploited therapeutically in alleviation and prevention of pain. We describe how SGCs are altered in rodent models of four common types of pain: systemic inflammation (sickness behaviour), post-surgical pain, diabetic neuropathic pain and post-herpetic pain.
Collapse
Affiliation(s)
- Menachem Hanani
- Laboratory of Experimental Surgery, Hadassah-Hebrew University Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - David C Spray
- Dominick Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| |
Collapse
|
48
|
Abstract
Neuropathic pain caused by a lesion or disease of the somatosensory nervous system is a common chronic pain condition with major impact on quality of life. Examples include trigeminal neuralgia, painful polyneuropathy, postherpetic neuralgia, and central poststroke pain. Most patients complain of an ongoing or intermittent spontaneous pain of, for example, burning, pricking, squeezing quality, which may be accompanied by evoked pain, particular to light touch and cold. Ectopic activity in, for example, nerve-end neuroma, compressed nerves or nerve roots, dorsal root ganglia, and the thalamus may in different conditions underlie the spontaneous pain. Evoked pain may spread to neighboring areas, and the underlying pathophysiology involves peripheral and central sensitization. Maladaptive structural changes and a number of cell-cell interactions and molecular signaling underlie the sensitization of nociceptive pathways. These include alteration in ion channels, activation of immune cells, glial-derived mediators, and epigenetic regulation. The major classes of therapeutics include drugs acting on α2δ subunits of calcium channels, sodium channels, and descending modulatory inhibitory pathways.
Collapse
Affiliation(s)
- Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Rohini Kuner
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Aarhus University Hospital, Aarhus, Denmark; and Department of Pharmacology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
49
|
Liu W, Cui Y, Wei J, Sun J, Zheng L, Xie J. Gap junction-mediated cell-to-cell communication in oral development and oral diseases: a concise review of research progress. Int J Oral Sci 2020; 12:17. [PMID: 32532966 PMCID: PMC7293327 DOI: 10.1038/s41368-020-0086-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/30/2020] [Accepted: 05/19/2020] [Indexed: 02/05/2023] Open
Abstract
Homoeostasis depends on the close connection and intimate molecular exchange between extracellular, intracellular and intercellular networks. Intercellular communication is largely mediated by gap junctions (GJs), a type of specialized membrane contact composed of variable number of channels that enable direct communication between cells by allowing small molecules to pass directly into the cytoplasm of neighbouring cells. Although considerable evidence indicates that gap junctions contribute to the functions of many organs, such as the bone, intestine, kidney, heart, brain and nerve, less is known about their role in oral development and disease. In this review, the current progress in understanding the background of connexins and the functions of gap junctions in oral development and diseases is discussed. The homoeostasis of tooth and periodontal tissues, normal tooth and maxillofacial development, saliva secretion and the integrity of the oral mucosa depend on the proper function of gap junctions. Knowledge of this pattern of cell-cell communication is required for a better understanding of oral diseases. With the ever-increasing understanding of connexins in oral diseases, therapeutic strategies could be developed to target these membrane channels in various oral diseases and maxillofacial dysplasia.
Collapse
Affiliation(s)
- Wenjing Liu
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yujia Cui
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jieya Wei
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jianxun Sun
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases & National Clinical Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
50
|
Giaume C, Naus CC, Sáez JC, Leybaert L. Glial Connexins and Pannexins in the Healthy and Diseased Brain. Physiol Rev 2020; 101:93-145. [PMID: 32326824 DOI: 10.1152/physrev.00043.2018] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Over the past several decades a large amount of data have established that glial cells, the main cell population in the brain, dynamically interact with neurons and thus impact their activity and survival. One typical feature of glia is their marked expression of several connexins, the membrane proteins forming intercellular gap junction channels and hemichannels. Pannexins, which have a tetraspan membrane topology as connexins, are also detected in glial cells. Here, we review the evidence that connexin and pannexin channels are actively involved in dynamic and metabolic neuroglial interactions in physiological as well as in pathological situations. These features of neuroglial interactions open the way to identify novel non-neuronal aspects that allow for a better understanding of behavior and information processing performed by neurons. This will also complement the "neurocentric" view by facilitating the development of glia-targeted therapeutic strategies in brain disease.
Collapse
Affiliation(s)
- Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Christian C Naus
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Juan C Sáez
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Luc Leybaert
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; University Pierre et Marie Curie, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, Paris, France; Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile; Instituo de Neurociencias, Centro Interdisciplinario de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile; Physiology Group, Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|