1
|
Li JL, Zhu CH, Tian MM, Liu Y, Ma L, Tao LJ, Zheng P, Yu JQ, Liu N. Negative allosteric modulator of Group Ⅰ mGluRs: Recent advances and therapeutic perspective for neuropathic pain. Neuroscience 2024; 560:406-421. [PMID: 39368605 DOI: 10.1016/j.neuroscience.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Neuropathic pain (NP) is a widespread public health problem that existing therapeutic treatments cannot manage adequately; therefore, novel treatment strategies are urgently required. G-protein-coupled receptors are important for intracellular signal transduction, and widely participate in physiological and pathological processes, including pain perception. Group I metabotropic glutamate receptors (mGluRs), including mGluR1 and mGluR5, are predominantly implicated in central sensitization, which can lead to hyperalgesia and allodynia. Many orthosteric site antagonists targeting Group I mGluRs have been found to alleviate NP, but their poor efficacy, low selectivity, and numerous side effects limit their development in NP treatment. Here we reviewed the advantages of Group I mGluRs negative allosteric modulators (NAMs) over orthosteric site antagonists based on allosteric modulation mechanism, and the challenges and opportunities of Group I mGluRs NAMs in NP treatment. This article aims to elucidate the advantages and future development potential of Group I mGluRs NAMs in the treatment of NP.
Collapse
Affiliation(s)
- Jia-Ling Li
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Chun-Hao Zhu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Miao-Miao Tian
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Yue Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Lin Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China
| | - Li-Jun Tao
- Department of Pharmacy, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750000, China
| | - Ping Zheng
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China.
| | - Jian-Qiang Yu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China.
| | - Ning Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750000, China; School of Basic Medical Science, Ningxia Medical University, Yinchuan 750000, China.
| |
Collapse
|
2
|
Bretová K, Svobodová V, Dubový P. Changes in Cx43 and AQP4 Proteins, and the Capture of 3 kDa Dextran in Subpial Astrocytes of the Rat Medial Prefrontal Cortex after Both Sham Surgery and Sciatic Nerve Injury. Int J Mol Sci 2024; 25:10989. [PMID: 39456773 PMCID: PMC11507206 DOI: 10.3390/ijms252010989] [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: 09/17/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
A subpopulation of astrocytes on the brain's surface, known as subpial astrocytes, constitutes the "glia limitans superficialis" (GLS), which is an interface between the brain parenchyma and the cerebrospinal fluid (CSF) in the subpial space. Changes in connexin-43 (Cx43) and aquaporin-4 (AQP4) proteins in subpial astrocytes were examined in the medial prefrontal cortex at postoperative day 1, 3, 7, 14, and 21 after sham operation and sciatic nerve compression (SNC). In addition, we tested the altered uptake of TRITC-conjugated 3 kDa dextran by reactive subpial astrocytes. Cellular immunofluorescence (IF) detection and image analysis were used to examine changes in Cx43 and AQP4 protein levels, as well as TRITC-conjugated 3 kDa dextran, in subpial astrocytes. The intensity of Cx43-IF was significantly increased, but AQP4-IF decreased in subpial astrocytes of sham- and SNC-operated rats during all survival periods compared to naïve controls. Similarly, the uptake of 3 kDa dextran in the GLS was reduced following both sham and SNC operations. The results suggest that both sciatic nerve injury and peripheral tissue injury alone can induce changes in subpial astrocytes related to the spread of their reactivity across the cortical surface mediated by increased amounts of gap junctions. At the same time, water transport and solute uptake were impaired in subpial astrocytes.
Collapse
Affiliation(s)
| | | | - Petr Dubový
- Cellular and Molecular Neurobiology Research Group, Department of Anatomy, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic; (K.B.)
| |
Collapse
|
3
|
Shigetomi E, Suzuki H, Hirayama YJ, Sano F, Nagai Y, Yoshihara K, Koga K, Tateoka T, Yoshioka H, Shinozaki Y, Kinouchi H, Tanaka KF, Bito H, Tsuda M, Koizumi S. Disease-relevant upregulation of P2Y 1 receptor in astrocytes enhances neuronal excitability via IGFBP2. Nat Commun 2024; 15:6525. [PMID: 39117630 PMCID: PMC11310333 DOI: 10.1038/s41467-024-50190-7] [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: 08/18/2023] [Accepted: 06/26/2024] [Indexed: 08/10/2024] Open
Abstract
Reactive astrocytes play a pivotal role in the pathogenesis of neurological diseases; however, their functional phenotype and the downstream molecules by which they modify disease pathogenesis remain unclear. Here, we genetically increase P2Y1 receptor (P2Y1R) expression, which is upregulated in reactive astrocytes in several neurological diseases, in astrocytes of male mice to explore its function and the downstream molecule. This astrocyte-specific P2Y1R overexpression causes neuronal hyperexcitability by increasing both astrocytic and neuronal Ca2+ signals. We identify insulin-like growth factor-binding protein 2 (IGFBP2) as a downstream molecule of P2Y1R in astrocytes; IGFBP2 acts as an excitatory signal to cause neuronal excitation. In neurological disease models of epilepsy and stroke, reactive astrocytes upregulate P2Y1R and increase IGFBP2. The present findings identify a mechanism underlying astrocyte-driven neuronal hyperexcitability, which is likely to be shared by several neurological disorders, providing insights that might be relevant for intervention in diverse neurological disorders.
Collapse
Affiliation(s)
- Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan.
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan.
| | - Hideaki Suzuki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Yukiho J Hirayama
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Fumikazu Sano
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Yuki Nagai
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Kohei Yoshihara
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keisuke Koga
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Neurophysiology, Hyogo College of Medicine, Hyogo, 663-8501, Japan
| | - Toru Tateoka
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Hideyuki Yoshioka
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Hiroyuki Kinouchi
- Department of Neurosurgery, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, 409-3898, Japan.
- Yamanashi GLIA center, University of Yamanashi, Yamanashi, 409-3898, Japan.
| |
Collapse
|
4
|
Nanda U, Zhang G, Underhill D, Pangarkar S. Management of Pain and Headache After Traumatic Brain Injury. Phys Med Rehabil Clin N Am 2024; 35:573-591. [PMID: 38945652 DOI: 10.1016/j.pmr.2024.02.009] [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] [Indexed: 07/02/2024]
Abstract
This article will identify common causes of pain following traumatic brain injury (TBI), discuss current treatment strategies for these complaints, and help tailor treatments for both acute and chronic settings. We will also briefly discuss primary and secondary headache disorders, followed by common secondary pain disorders that may be related to trauma.
Collapse
Affiliation(s)
- Udai Nanda
- Department of Physical Medicine and Rehabilitation, Pain Management, Headache Center of Excellence, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA; Division of Physical Medicine and Rehabilitation, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Grace Zhang
- Division of Physical Medicine and Rehabilitation, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - David Underhill
- Division of Physical Medicine and Rehabilitation, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Sanjog Pangarkar
- Division of Physical Medicine and Rehabilitation, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA; Department of Physical Medicine and Rehabilitation, Pain Management, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
5
|
Luo R, Hu X, Li X, Lei F, Liao P, Yi L, Zhang X, Zhou B, Jiang R. Dysfunctional astrocyte glutamate uptake in the hypothalamic paraventricular nucleus contributes to visceral pain and anxiety-like behavior in mice with chronic pancreatitis. Glia 2024. [PMID: 39046219 DOI: 10.1002/glia.24595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/12/2024] [Accepted: 07/08/2024] [Indexed: 07/25/2024]
Abstract
Abdominal visceral pain is a predominant symptom in patients with chronic pancreatitis (CP); however, the underlying mechanism of pain in CP remains elusive. We hypothesized that astrocytes in the hypothalamic paraventricular nucleus (PVH) contribute to CP pain pathogenesis. A mouse model of CP was established by repeated intraperitoneal administration of caerulein to induce abdominal visceral pain. Abdominal mechanical stimulation, open field and elevated plus maze tests were performed to assess visceral pain and anxiety-like behavior. Fiber photometry, brain slice Ca2+ imaging, electrophysiology, and immunohistochemistry were used to investigate the underlying mechanisms. Mice with CP displayed long-term abdominal mechanical allodynia and comorbid anxiety, which was accompanied by astrocyte glial fibrillary acidic protein reactivity, elevated Ca2+ signaling, and astroglial glutamate transporter-1 (GLT-1) deficits in the PVH. Specifically, reducing astrocyte Ca2+ signaling in the PVH via chemogenetics significantly rescued GLT-1 deficits and alleviated mechanical allodynia and anxiety in mice with CP. Furthermore, we found that GLT-1 deficits directly contributed to the hyperexcitability of VGLUT2PVH neurons in mice with CP, and that pharmacological activation of GLT-1 alleviated the hyperexcitability of VGLUT2PVH neurons, abdominal visceral pain, and anxiety in these mice. Taken together, our data suggest that dysfunctional astrocyte glutamate uptake in the PVH contributes to visceral pain and anxiety in mice with CP, highlighting GLT-1 as a potential therapeutic target for chronic pain in patients experiencing CP.
Collapse
Affiliation(s)
- Rong Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojun Hu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Li
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Fan Lei
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Liao
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Limei Yi
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Zhang
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China
| | - Bin Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Ruotian Jiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
da Silva MDV, Martelossi-Cebinelli G, Yaekashi KM, Carvalho TT, Borghi SM, Casagrande R, Verri WA. A Narrative Review of the Dorsal Root Ganglia and Spinal Cord Mechanisms of Action of Neuromodulation Therapies in Neuropathic Pain. Brain Sci 2024; 14:589. [PMID: 38928589 PMCID: PMC11202229 DOI: 10.3390/brainsci14060589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Neuropathic pain arises from injuries to the nervous system in diseases such as diabetes, infections, toxicity, and traumas. The underlying mechanism of neuropathic pain involves peripheral and central pathological modifications. Peripheral mechanisms entail nerve damage, leading to neuronal hypersensitivity and ectopic action potentials. Central sensitization involves a neuropathological process with increased responsiveness of the nociceptive neurons in the central nervous system (CNS) to their normal or subthreshold input due to persistent stimuli, leading to sustained electrical discharge, synaptic plasticity, and aberrant processing in the CNS. Current treatments, both pharmacological and non-pharmacological, aim to alleviate symptoms but often face challenges due to the complexity of neuropathic pain. Neuromodulation is emerging as an important therapeutic approach for the treatment of neuropathic pain in patients unresponsive to common therapies, by promoting the normalization of neuronal and/or glial activity and by targeting cerebral cortical regions, spinal cord, dorsal root ganglia, and nerve endings. Having a better understanding of the efficacy, adverse events and applicability of neuromodulation through pre-clinical studies is of great importance. Unveiling the mechanisms and characteristics of neuromodulation to manage neuropathic pain is essential to understand how to use it. In the present article, we review the current understanding supporting dorsal root ganglia and spinal cord neuromodulation as a therapeutic approach for neuropathic pain.
Collapse
Affiliation(s)
- Matheus Deroco Veloso da Silva
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Immunology, Parasitology and General Pathology, Londrina State University, Londrina 86057-970, PR, Brazil; (M.D.V.d.S.); (G.M.-C.); (K.M.Y.); (S.M.B.)
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Immunology, Parasitology and General Pathology, Londrina State University, Londrina 86057-970, PR, Brazil; (M.D.V.d.S.); (G.M.-C.); (K.M.Y.); (S.M.B.)
| | - Kelly Megumi Yaekashi
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Immunology, Parasitology and General Pathology, Londrina State University, Londrina 86057-970, PR, Brazil; (M.D.V.d.S.); (G.M.-C.); (K.M.Y.); (S.M.B.)
| | - Thacyana T. Carvalho
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Sergio M. Borghi
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Immunology, Parasitology and General Pathology, Londrina State University, Londrina 86057-970, PR, Brazil; (M.D.V.d.S.); (G.M.-C.); (K.M.Y.); (S.M.B.)
- Center for Research in Health Sciences, University of Northern Paraná, Londrina 86041-140, PR, Brazil
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, PR, Brazil;
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy and Cancer, Department of Immunology, Parasitology and General Pathology, Londrina State University, Londrina 86057-970, PR, Brazil; (M.D.V.d.S.); (G.M.-C.); (K.M.Y.); (S.M.B.)
- Biological Sciences Center, State University of Londrina, Rod. Celso Garcia Cid Pr 445, KM 380, P.O. Box 10.011, Londrina 86057-970, PR, Brazil
| |
Collapse
|
7
|
Wu C, Li Y, He X, Sun H, Zhang S, Hou F, Hu M, Lan A, Zhang H, Qi L, Zhang H, Liao H. Chemogenetic activation of astrocytic Gi signaling promotes spinogenesis and motor functional recovery after stroke. Glia 2024; 72:1150-1164. [PMID: 38436489 DOI: 10.1002/glia.24521] [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: 02/21/2023] [Revised: 02/18/2024] [Accepted: 02/23/2024] [Indexed: 03/05/2024]
Abstract
Ischemic stroke is the leading cause of adult disability. The rewiring of surviving neurons is the fundamental process for functional recovery. Accumulating evidence implicates astrocytes in synapses and neural circuits formation, but few studies have further studied how to enhance the effects of astrocytes on synapse and circuits after stroke and its impacts on post-stroke functional recovery. In this study, we made use of chemogenetics to specifically activate astrocytic Gi signaling in the peri-infarcted sensorimotor cortex at different time epochs in a mouse model of photothrombotic stroke. We found that early activation of astrocytic hM4Di after stroke by CNO modulates astrocyte activity and upregulates synaptogenic molecules including thrombospondin-1 (TSP1) as revealed by bulk RNA-sequencing, but no significant improvement was observed in dendritic spine density and behavioral performance in grid walking test. Interestingly, when the manipulation was initiated at the subacute phase of stroke, the recovery of spine density and motor function could be effectively promoted, accompanied by increased TSP1 expression. Our data highlight the important role of astrocytes in synapse remodeling during the repair phase of stroke and suggest astrocytic Gi signaling activation as a potential strategy for synapse regeneration, circuit rewiring, and functional recovery.
Collapse
Affiliation(s)
- Chaoran Wu
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yu Li
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xinran He
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hao Sun
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Shiwen Zhang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Fengsheng Hou
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Mengqiu Hu
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Aili Lan
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hao Zhang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Long Qi
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Huibin Zhang
- Center for Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, China
| | - Hong Liao
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
8
|
Cheng Y, Zhai Y, Yuan Y, Wang Q, Li S, Sun H. The Contributions of Thrombospondin-1 to Epilepsy Formation. Neurosci Bull 2024; 40:658-672. [PMID: 38528256 PMCID: PMC11127911 DOI: 10.1007/s12264-024-01194-2] [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: 06/27/2023] [Accepted: 01/27/2024] [Indexed: 03/27/2024] Open
Abstract
Epilepsy is a neural network disorder caused by uncontrolled neuronal hyperexcitability induced by an imbalance between excitatory and inhibitory networks. Abnormal synaptogenesis plays a vital role in the formation of overexcited networks. Recent evidence has confirmed that thrombospondin-1 (TSP-1), mainly secreted by astrocytes, is a critical cytokine that regulates synaptogenesis during epileptogenesis. Furthermore, numerous studies have reported that TSP-1 is also involved in other processes, such as angiogenesis, neuroinflammation, and regulation of Ca2+ homeostasis, which are closely associated with the occurrence and development of epilepsy. In this review, we summarize the potential contributions of TSP-1 to epilepsy development.
Collapse
Affiliation(s)
- Yao Cheng
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yujie Zhai
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yi Yuan
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Qiaoyun Wang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Shucui Li
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| | - Hongliu Sun
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
9
|
Mazzitelli M, Ponomareva O, Presto P, John J, Neugebauer V. Impaired amygdala astrocytic signaling worsens neuropathic pain-associated neuronal functions and behaviors. Front Pharmacol 2024; 15:1368634. [PMID: 38576475 PMCID: PMC10991799 DOI: 10.3389/fphar.2024.1368634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/06/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction: Pain is a clinically relevant health care issue with limited therapeutic options, creating the need for new and improved analgesic strategies. The amygdala is a limbic brain region critically involved in the regulation of emotional-affective components of pain and in pain modulation. The central nucleus of amygdala (CeA) serves major output functions and receives nociceptive information via the external lateral parabrachial nucleus (PB). While amygdala neuroplasticity has been linked causally to pain behaviors, non-neuronal pain mechanisms in this region remain to be explored. As an essential part of the neuroimmune system, astrocytes that represent about 40-50% of glia cells within the central nervous system, are required for physiological neuronal functions, but their role in the amygdala remains to be determined for pain conditions. In this study, we measured time-specific astrocyte activation in the CeA in a neuropathic pain model (spinal nerve ligation, SNL) and assessed the effects of astrocyte inhibition on amygdala neuroplasticity and pain-like behaviors in the pain condition. Methods and Results: Glial fibrillary acidic protein (GFAP, astrocytic marker) immunoreactivity and mRNA expression were increased at the chronic (4 weeks post-SNL), but not acute (1 week post-SNL), stage of neuropathic pain. In order to determine the contribution of astrocytes to amygdala pain-mechanisms, we used fluorocitric acid (FCA), a selective inhibitor of astrocyte metabolism. Whole-cell patch-clamp recordings were performed from neurons in the laterocapsular division of the CeA (CeLC) obtained from chronic neuropathic rats. Pre-incubation of brain slices with FCA (100 µM, 1 h), increased excitability through altered hyperpolarization-activated current (Ih) functions, without significantly affecting synaptic responses at the PB-CeLC synapse. Intra-CeA injection of FCA (100 µM) had facilitatory effects on mechanical withdrawal thresholds (von Frey and paw pressure tests) and emotional-affective behaviors (evoked vocalizations), but not on facial grimace score and anxiety-like behaviors (open field test), in chronic neuropathic rats. Selective inhibition of astrocytes by FCA was confirmed with immunohistochemical analyses showing decreased astrocytic GFAP, but not NeuN, signal in the CeA. Discussion: Overall, these results suggest a complex modulation of amygdala pain functions by astrocytes and provide evidence for beneficial functions of astrocytes in CeA in chronic neuropathic pain.
Collapse
Affiliation(s)
- Mariacristina Mazzitelli
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Olga Ponomareva
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Peyton Presto
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Julia John
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
10
|
Han S, Jiang B, Ren J, Gao F, Wen J, Zhou T, Wang L, Wei X. Impaired Lactate Release in Dorsal CA1 Astrocytes Contributed to Nociceptive Sensitization and Comorbid Memory Deficits in Rodents. Anesthesiology 2024; 140:538-557. [PMID: 37651459 DOI: 10.1097/aln.0000000000004756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
BACKGROUND Memory deficits are a common comorbid disorder in patients suffering from neuropathic pain. The mechanisms underlying the comorbidities remain elusive. The hypothesis of this study was that impaired lactate release from dysfunctional astrocytes in dorsal hippocampal CA1 contributed to memory deficits. METHODS A spared nerve injury model was established to induce both pain and memory deficits in rats and mice of both sexes. von Frey tests, novel object recognition, and conditioned place preference tests were applied to evaluate the behaviors. Whole-cell recording, fiber photometry, Western blotting, and immunohistochemistry combined with intracranial injections were used to explore the underlying mechanisms. RESULTS Animals with spared sciatic nerve injury that had displayed nociception sensitization or memory deficit comorbidities demonstrated a reduction in the intrinsic excitability of pyramidal neurons, accompanied by reduced Ca2+ activation in astrocytes (ΔF/F, sham: 6 ± 2%; comorbidity: 2 ± 0.4%) and a decrease in the expression of glial fibrillary acidic protein and lactate levels in the dorsal CA1. Exogenous lactate supply or increasing endogenous lactate release by chemogenetic activation of astrocytes alleviated this comorbidity by enhancing the cell excitability (129 ± 4 vs. 88 ± 10 for 3.5 mM lactate) and potentiating N-methyl-d-aspartate receptor-mediated excitatory postsynaptic potentials of pyramidal neurons. In contrast, inhibition of lactate synthesis, blocking lactate transporters, or chemogenetic inhibition of astrocytes resulted in comorbidity-like behaviors in naive animals. Notably, β2-adrenergic receptors in astrocytes but not neurons were downregulated in dorsal CA1 after spared nerve injury. Microinjection of a β2 receptor agonist into dorsal CA1 or activation of the noradrenergic projections onto the hippocampus from the locus coeruleus alleviated the comorbidity, possibly by increasing lactate release. CONCLUSIONS Impaired lactate release from dysfunctional astrocytes, which could be rescued by activation of the locus coeruleus, led to nociception and memory deficits after peripheral nerve injury. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Shuang Han
- Department of Human Anatomy and Physiology, and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Bin Jiang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, and Department of Human Anatomy and Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jiale Ren
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Feng Gao
- Department of Human Anatomy and Physiology, and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Junjian Wen
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Taihe Zhou
- Department of Human Anatomy and Physiology, and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Laijian Wang
- Department of Human Anatomy and Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xuhong Wei
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Human Anatomy and Physiology, and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
11
|
Bak MS, Park H, Yoon H, Chung G, Shin H, Shin S, Kim TW, Lee K, Nägerl UV, Kim SJ, Kim SK. Machine learning-based evaluation of spontaneous pain and analgesics from cellular calcium signals in the mouse primary somatosensory cortex using explainable features. Front Mol Neurosci 2024; 17:1356453. [PMID: 38450042 PMCID: PMC10915002 DOI: 10.3389/fnmol.2024.1356453] [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: 12/15/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction Pain that arises spontaneously is considered more clinically relevant than pain evoked by external stimuli. However, measuring spontaneous pain in animal models in preclinical studies is challenging due to methodological limitations. To address this issue, recently we developed a deep learning (DL) model to assess spontaneous pain using cellular calcium signals of the primary somatosensory cortex (S1) in awake head-fixed mice. However, DL operate like a "black box", where their decision-making process is not transparent and is difficult to understand, which is especially evident when our DL model classifies different states of pain based on cellular calcium signals. In this study, we introduce a novel machine learning (ML) model that utilizes features that were manually extracted from S1 calcium signals, including the dynamic changes in calcium levels and the cell-to-cell activity correlations. Method We focused on observing neural activity patterns in the primary somatosensory cortex (S1) of mice using two-photon calcium imaging after injecting a calcium indicator (GCaMP6s) into the S1 cortex neurons. We extracted features related to the ratio of up and down-regulated cells in calcium activity and the correlation level of activity between cells as input data for the ML model. The ML model was validated using a Leave-One-Subject-Out Cross-Validation approach to distinguish between non-pain, pain, and drug-induced analgesic states. Results and discussion The ML model was designed to classify data into three distinct categories: non-pain, pain, and drug-induced analgesic states. Its versatility was demonstrated by successfully classifying different states across various pain models, including inflammatory and neuropathic pain, as well as confirming its utility in identifying the analgesic effects of drugs like ketoprofen, morphine, and the efficacy of magnolin, a candidate analgesic compound. In conclusion, our ML model surpasses the limitations of previous DL approaches by leveraging manually extracted features. This not only clarifies the decision-making process of the ML model but also yields insights into neuronal activity patterns associated with pain, facilitating preclinical studies of analgesics with higher potential for clinical translation.
Collapse
Affiliation(s)
- Myeong Seong Bak
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Division of AI and Data Analysis, Neurogrin Inc., Seoul, Republic of Korea
| | - Haney Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Division of Preclinical R&D, Neurogrin Inc., Seoul, Republic of Korea
| | - Heera Yoon
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Division of Preclinical R&D, Neurogrin Inc., Seoul, Republic of Korea
| | - Geehoon Chung
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyunjin Shin
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Soonho Shin
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tai Wan Kim
- Department of Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Kyungjoon Lee
- Department of East-West Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - U. Valentin Nägerl
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 and University of Bordeaux, Bordeaux, France
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sun Kwang Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Department of East-West Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
12
|
Saito K, Shigetomi E, Shinozaki Y, Kobayashi K, Parajuli B, Kubota Y, Sakai K, Miyakawa M, Horiuchi H, Nabekura J, Koizumi S. Microglia sense astrocyte dysfunction and prevent disease progression in an Alexander disease model. Brain 2024; 147:698-716. [PMID: 37955589 PMCID: PMC10834242 DOI: 10.1093/brain/awad358] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 11/14/2023] Open
Abstract
Alexander disease (AxD) is an intractable neurodegenerative disorder caused by GFAP mutations. It is a primary astrocyte disease with a pathological hallmark of Rosenthal fibres within astrocytes. AxD astrocytes show several abnormal phenotypes. Our previous study showed that AxD astrocytes in model mice exhibit aberrant Ca2+ signals that induce AxD aetiology. Here, we show that microglia have unique phenotypes with morphological and functional alterations, which are related to the pathogenesis of AxD. Immunohistochemical studies of 60TM mice (AxD model) showed that AxD microglia exhibited highly ramified morphology. Functional changes in microglia were assessed by Ca2+ imaging using hippocampal brain slices from Iba1-GCaMP6-60TM mice and two-photon microscopy. We found that AxD microglia showed aberrant Ca2+ signals, with high frequency Ca2+ signals in both the processes and cell bodies. These microglial Ca2+ signals were inhibited by pharmacological blockade or genetic knockdown of P2Y12 receptors but not by tetrodotoxin, indicating that these signals are independent of neuronal activity but dependent on extracellular ATP from non-neuronal cells. Our single-cell RNA sequencing data showed that the expression level of Entpd2, an astrocyte-specific gene encoding the ATP-degrading enzyme NTPDase2, was lower in AxD astrocytes than in wild-type astrocytes. In situ ATP imaging using the adeno-associated virus vector GfaABC1D ATP1.0 showed that exogenously applied ATP was present longer in 60TM mice than in wild-type mice. Thus, the increased ATP level caused by the decrease in its metabolizing enzyme in astrocytes could be responsible for the enhancement of microglial Ca2+ signals. To determine whether these P2Y12 receptor-mediated Ca2+ signals in AxD microglia play a significant role in the pathological mechanism, a P2Y12 receptor antagonist, clopidogrel, was administered. Clopidogrel significantly exacerbated pathological markers in AxD model mice and attenuated the morphological features of microglia, suggesting that microglia play a protective role against AxD pathology via P2Y12 receptors. Taken together, we demonstrated that microglia sense AxD astrocyte dysfunction via P2Y12 receptors as an increase in extracellular ATP and alter their morphology and Ca2+ signalling, thereby protecting against AxD pathology. Although AxD is a primary astrocyte disease, our study may facilitate understanding of the role of microglia as a disease modifier, which may contribute to the clinical diversity of AxD.
Collapse
Affiliation(s)
- Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kenji Kobayashi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Bijay Parajuli
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Yuto Kubota
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kent Sakai
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Miho Miyakawa
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Hiroshi Horiuchi
- Division of Homeostatic Development, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8585, Japan
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8585, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| |
Collapse
|
13
|
Shinozaki Y, Namekata K, Guo X, Harada T. Glial cells as a promising therapeutic target of glaucoma: beyond the IOP. FRONTIERS IN OPHTHALMOLOGY 2024; 3:1310226. [PMID: 38983026 PMCID: PMC11182302 DOI: 10.3389/fopht.2023.1310226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/18/2023] [Indexed: 07/11/2024]
Abstract
Glial cells, a type of non-neuronal cell found in the central nervous system (CNS), play a critical role in maintaining homeostasis and regulating CNS functions. Recent advancements in technology have paved the way for new therapeutic strategies in the fight against glaucoma. While intraocular pressure (IOP) is the most well-known modifiable risk factor, a significant number of glaucoma patients have normal IOP levels. Because glaucoma is a complex, multifactorial disease influenced by various factors that contribute to its onset and progression, it is imperative that we consider factors beyond IOP to effectively prevent or slow down the disease's advancement. In the realm of CNS neurodegenerative diseases, glial cells have emerged as key players due to their pivotal roles in initiating and hastening disease progression. The inhibition of dysregulated glial function holds the potential to protect neurons and restore brain function. Consequently, glial cells represent an enticing therapeutic candidate for glaucoma, even though the majority of glaucoma research has historically concentrated solely on retinal ganglion cells (RGCs). In addition to the neuroprotection of RGCs, the proper regulation of glial cell function can also facilitate structural and functional recovery in the retina. In this review, we offer an overview of recent advancements in understanding the non-cell-autonomous mechanisms underlying the pathogenesis of glaucoma. Furthermore, state-of-the-art technologies have opened up possibilities for regenerating the optic nerve, which was previously believed to be incapable of regeneration. We will also delve into the potential roles of glial cells in the regeneration of the optic nerve and the restoration of visual function.
Collapse
Affiliation(s)
- Youichi Shinozaki
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
14
|
Fujimura M. Gabapentin improves neuropathic pain in Minamata disease model rats. Environ Health Prev Med 2024; 29:31. [PMID: 38825526 PMCID: PMC11157338 DOI: 10.1265/ehpm.24-00035] [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: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Methylmercury (MeHg), the causative agent of Minamata disease, damages the cranial nervous system and causes specific sensory disturbances, especially hypoesthesia, in the extremities. However, recent reports demonstrate that patients with chronic Minamata disease conversely develop neuropathic pain in the lower extremities. Studies on our established Minamata disease model rats showed that MeHg-mediated neurodegeneration might induce neuropathic pain by over time through inducing rewiring with neuronal activation in the somatosensory cortex via microglial activation in the spinal dorsal horn. METHODS In this study, the effects of gabapentin, a potentially effective treatment for neuropathic pain, was evaluated using this Minamata disease model rats. To further elucidate the mechanism of its medicinal effects, histochemical and biochemical analyses of the nervous system of Minamata disease model rats were conducted. RESULTS Gabapentin treatment restored the reduction in the pain threshold caused by MeHg exposure in rats. Histochemical and biochemical analyses revealed that gabapentin showed no effect on MeHg-induced neurodegeneration in entire nervous system and microglial activation in the spinal dorsal horn. However, it was shown that gabapentin may reduce excessive synaptogenesis through its antagonist action on the alpha2-delta-1 subunit of calcium channels in the somatosensory cortex. CONCLUSIONS These results indicate that gabapentin may alleviated neuropathic pain in MeHg poisoning, as typified by Minamata disease, by reversibly modulation synaptic rewiring in the somatosensory cortex.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, Minamata, Japan
| |
Collapse
|
15
|
Yao D, Chen Y, Chen G. The role of pain modulation pathway and related brain regions in pain. Rev Neurosci 2023; 34:899-914. [PMID: 37288945 DOI: 10.1515/revneuro-2023-0037] [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: 03/25/2023] [Accepted: 05/18/2023] [Indexed: 06/09/2023]
Abstract
Pain is a multifaceted process that encompasses unpleasant sensory and emotional experiences. The essence of the pain process is aversion, or perceived negative emotion. Central sensitization plays a significant role in initiating and perpetuating of chronic pain. Melzack proposed the concept of the "pain matrix", in which brain regions associated with pain form an interconnected network, rather than being controlled by a singular brain region. This review aims to investigate distinct brain regions involved in pain and their interconnections. In addition, it also sheds light on the reciprocal connectivity between the ascending and descending pathways that participate in pain modulation. We review the involvement of various brain areas during pain and focus on understanding the connections among them, which can contribute to a better understanding of pain mechanisms and provide opportunities for further research on therapies for improved pain management.
Collapse
Affiliation(s)
- Dandan Yao
- Department of Anesthesiology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Gang Chen
- Department of Anesthesiology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| |
Collapse
|
16
|
de Lima IBQ, Cardozo PL, Fahel JS, Lacerda JPS, Miranda AS, Teixeira AL, Ribeiro FM. Blockade of mGluR5 in astrocytes derived from human iPSCs modulates astrocytic function and increases phagocytosis. Front Immunol 2023; 14:1283331. [PMID: 38146365 PMCID: PMC10749358 DOI: 10.3389/fimmu.2023.1283331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/23/2023] [Indexed: 12/27/2023] Open
Abstract
TNF-α is essential for induction and maintenance of inflammatory responses and its dysregulation is associated with susceptibility to various pathogens that infect the central nervous system. Activation of both microglia and astrocytes leads to TNF-α production, which in turn triggers further activation of these cells. Astrocytes have been implicated in the pathophysiology of a wide range of neurodegenerative diseases with either harmful or protective roles, as these cells are capable of secreting several inflammatory factors and also promote synapse elimination and remodeling. These responses are possible because they sense their surroundings via several receptors, including the metabotropic glutamate receptor 5 (mGluR5). Under neuroinflammatory conditions, mGluR5 activation in astrocytes can be neuroprotective or have the opposite effect. In the current study, we investigated the role of mGluR5 in hiPSC-derived astrocytes subjected to pro-inflammatory stimulation by recombinant TNF-α (rTNF-α). Our results show that mGluR5 blockade by CTEP decreases the secreted levels of pro-inflammatory cytokines (IL-6 and IL-8) following short rTNF-α stimulation, although this effect subsides with time. Additionally, CTEP enhances synaptoneurosome phagocytosis by astrocytes in both non-stimulated and rTNF-α-stimulated conditions, indicating that mGluR5 blockade alone is enough to drive synaptic material engulfment. Finally, mGluR5 antagonism as well as rTNF-α stimulation augment the expression of the reactivity marker SERPINA3 and reduces the expression of synaptogenic molecules. Altogether, these data suggest a complex role for mGluR5 in human astrocytes, since its blockade may have beneficial and detrimental effects under inflammatory conditions.
Collapse
Affiliation(s)
- Izabella B. Q. de Lima
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pablo L. Cardozo
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Julia S. Fahel
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana P. S. Lacerda
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Aline S. Miranda
- Department of Morphology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antônio L. Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Fabiola M. Ribeiro
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
17
|
Rupareliya VP, Singh AA, Butt AM, A H, Kumar H. The "molecular soldiers" of the CNS: Astrocytes, a comprehensive review on their roles and molecular signatures. Eur J Pharmacol 2023; 959:176048. [PMID: 37758010 DOI: 10.1016/j.ejphar.2023.176048] [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: 06/27/2023] [Revised: 08/24/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023]
Abstract
For a long time, neurons held the position of central players in the nervous system. Since there are far more astrocytes than neurons in the brain, it makes us wonder if these cells just take up space and support the neurons or if they are actively participating in central nervous system (CNS) homeostasis. Now, astrocytes' contribution to CNS physiology is appreciated as they are known to regulate ion and neurotransmitter levels, synapse formation and elimination, blood-brain barrier integrity, immune function, cerebral blood flow, and many more. In many neurological and psychiatric disorders, astrocyte functions are altered. Advancements in microscopic and transcriptomic tools revealed populations of astrocytes with varied morphology, electrophysiological properties, and transcriptomic profiles. Neuron-circuit-specific functions and neuron-specific interactions of astroglial subpopulations are found, which suggests that diversity is essential in carrying out diverse region-specific CNS functions. Investigations on heterogeneous astrocyte populations are revealing new astrocyte functions and their role in pathological conditions, opening a new therapeutic avenue for targeting neurological conditions. The true extent of astrocytic heterogeneity and its functional implications are yet to be fully explored. This review summarizes essential astrocytic functions and their relevance in pathological conditions and discusses astrocytic diversity in relation to morphology, function, and gene expression throughout the CNS.
Collapse
Affiliation(s)
- Vimal P Rupareliya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Aditya A Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Ayub Mohammed Butt
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hariharan A
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
18
|
Liu Z, Wang Z, Wei Y, Shi J, Shi T, Chen X, Li L. Transcriptomic Profiling of Tetrodotoxin-Induced Neurotoxicity in Human Cerebral Organoids. Mar Drugs 2023; 21:588. [PMID: 37999412 PMCID: PMC10672545 DOI: 10.3390/md21110588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Tetrodotoxin (TTX) is an exceedingly toxic non-protein biotoxin that demonstrates remarkable selectivity and affinity for sodium channels on the excitation membrane of nerves. This property allows TTX to effectively obstruct nerve conduction, resulting in nerve paralysis and fatality. Although the mechanistic aspects of its toxicity are well understood, there is a dearth of literature addressing alterations in the neural microenvironment subsequent to TTX poisoning. In this research endeavor, we harnessed human pluripotent induced stem cells to generate cerebral organoids-an innovative model closely mirroring the structural and functional intricacies of the human brain. This model was employed to scrutinize the comprehensive transcriptomic shifts induced by TTX exposure, thereby delving into the neurotoxic properties of TTX and its potential underlying mechanisms. Our findings revealed 455 differentially expressed mRNAs (DEmRNAs), 212 differentially expressed lncRNAs (DElncRNAs), and 18 differentially expressed miRNAs (DEmiRNAs) in the TTX-exposed group when juxtaposed with the control cohort. Through meticulous Gene Ontology (GO) annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and protein-protein interaction (PPI) analysis, we ascertained that these differential genes predominantly participate in the regulation of voltage-gated channels and synaptic homeostasis. A comprehensive ceRNA network analysis unveiled that DEmRNAs exert control over the expression of ion channels and neurocytokines, suggesting their potential role in mediating apoptosis.
Collapse
Affiliation(s)
- Zhanbiao Liu
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China (J.S.)
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Zhe Wang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China
| | - Yue Wei
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China (J.S.)
| | - Jingjing Shi
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China (J.S.)
| | - Tong Shi
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China (J.S.)
| | - Xuejun Chen
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China (J.S.)
| | - Liqin Li
- State Key Laboratory of NBC Protection for Civilian, Beijing 102205, China (J.S.)
| |
Collapse
|
19
|
Prokhorenko MA, Smyth JT. Astrocyte store-operated calcium entry is required for centrally mediated neuropathic pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544231. [PMID: 37333230 PMCID: PMC10274864 DOI: 10.1101/2023.06.08.544231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Central sensitization is a critical step in chronic neuropathic pain formation following acute nerve injury. Central sensitization is defined by nociceptive and somatosensory circuitry changes in the spinal cord leading to dysfunction of antinociceptive gamma-aminobutyric acid (GABA)ergic cells (Li et al., 2019), amplification of ascending nociceptive signals, and hypersensitivity (Woolf, 2011). Astrocytes are key mediators of the neurocircuitry changes that underlie central sensitization and neuropathic pain, and astrocytes respond to and regulate neuronal function through complex Ca2+ signaling mechanisms. Clear definition of the astrocyte Ca2+ signaling mechanisms involved in central sensitization may lead to new therapeutic targets for treatment of chronic neuropathic pain, as well as enhance our understanding of the complex central nervous system (CNS) adaptions that occur following nerve injury. Ca2+ release from astrocyte endoplasmic reticulum (ER) Ca2+ stores via the inositol 1,4,5-trisphosphate receptor (IP3R) is required for centrally mediated neuropathic pain (Kim et al, 2016); however recent evidence suggests the involvement of additional astrocyte Ca2+ signaling mechanisms. We therefore investigated the role of astrocyte store-operated Ca2+ entry (SOCE), which mediates Ca2+ influx in response to ER Ca2+ store depletion. Using an adult Drosophila melanogaster model of central sensitization based on thermal allodynia in response to leg amputation nerve injury (Khuong et al., 2019), we show that astrocytes exhibit SOCE-dependent Ca2+ signaling events three to four days following nerve injury. Astrocyte-specific suppression of Stim and Orai, the key mediators of SOCE Ca2+ influx, completely inhibited the development of thermal allodynia seven days following injury, and also inhibited the loss of ventral nerve cord (VNC) GABAergic neurons that is required for central sensitization in flies. We lastly show that constitutive SOCE in astrocytes results in thermal allodynia even in the absence of nerve injury. Our results collectively demonstrate that astrocyte SOCE is necessary and sufficient for central sensitization and development of hypersensitivity in Drosophila, adding key new understanding to the astrocyte Ca2+ signaling mechanisms involved in chronic pain.
Collapse
Affiliation(s)
- Mariya A. Prokhorenko
- Neuroscience Graduate Program and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD
| | - Jeremy T. Smyth
- Neuroscience Graduate Program and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD
- Department of Anatomy, Physiology, and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD
| |
Collapse
|
20
|
McGinnis A, Ji RR. The Similar and Distinct Roles of Satellite Glial Cells and Spinal Astrocytes in Neuropathic Pain. Cells 2023; 12:965. [PMID: 36980304 PMCID: PMC10047571 DOI: 10.3390/cells12060965] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Preclinical studies have identified glial cells as pivotal players in the genesis and maintenance of neuropathic pain after nerve injury associated with diabetes, chemotherapy, major surgeries, and virus infections. Satellite glial cells (SGCs) in the dorsal root and trigeminal ganglia of the peripheral nervous system (PNS) and astrocytes in the central nervous system (CNS) express similar molecular markers and are protective under physiological conditions. They also serve similar functions in the genesis and maintenance of neuropathic pain, downregulating some of their homeostatic functions and driving pro-inflammatory neuro-glial interactions in the PNS and CNS, i.e., "gliopathy". However, the role of SGCs in neuropathic pain is not simply as "peripheral astrocytes". We delineate how these peripheral and central glia participate in neuropathic pain by producing different mediators, engaging different parts of neurons, and becoming active at different stages following nerve injury. Finally, we highlight the recent findings that SGCs are enriched with proteins related to fatty acid metabolism and signaling such as Apo-E, FABP7, and LPAR1. Targeting SGCs and astrocytes may lead to novel therapeutics for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Aidan McGinnis
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
21
|
Chen C, Sun L, Adler A, Zhou H, Zhang L, Zhang L, Deng J, Bai Y, Zhang J, Yang G, Gan WB, Tang P. Synchronized activity of sensory neurons initiates cortical synchrony in a model of neuropathic pain. Nat Commun 2023; 14:689. [PMID: 36755026 PMCID: PMC9908980 DOI: 10.1038/s41467-023-36093-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023] Open
Abstract
Increased low frequency cortical oscillations are observed in people with neuropathic pain, but the cause of such elevated cortical oscillations and their impact on pain development remain unclear. By imaging neuronal activity in a spared nerve injury (SNI) mouse model of neuropathic pain, we show that neurons in dorsal root ganglia (DRG) and somatosensory cortex (S1) exhibit synchronized activity after peripheral nerve injury. Notably, synchronized activity of DRG neurons occurs within hours after injury and 1-2 days before increased cortical oscillations. This DRG synchrony is initiated by axotomized neurons and mediated by local purinergic signaling at the site of nerve injury. We further show that synchronized DRG activity after SNI is responsible for increasing low frequency cortical oscillations and synaptic remodeling in S1, as well as for inducing animals' pain-like behaviors. In naive mice, enhancing the synchrony, not the level, of DRG neuronal activity causes synaptic changes in S1 and pain-like behaviors similar to SNI mice. Taken together, these results reveal the critical role of synchronized DRG neuronal activity in increasing cortical plasticity and oscillations in a neuropathic pain model. These findings also suggest the potential importance of detection and suppression of elevated cortical oscillations in neuropathic pain states.
Collapse
Affiliation(s)
- Chao Chen
- Department of Orthopaedics, Peking 301 Hospital, Beijing, China
- Department of Hand Surgery, Shenzhen People's Hospital, Second Clinical Medicine College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Linlin Sun
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Neuroscience Research Institute, Peking University, Beijing, China
| | - Avital Adler
- Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Hang Zhou
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Licheng Zhang
- Department of Orthopaedics, Peking 301 Hospital, Beijing, China
| | - Lihai Zhang
- Department of Orthopaedics, Peking 301 Hospital, Beijing, China
| | - Junhao Deng
- Department of Orthopaedics, Peking 301 Hospital, Beijing, China
| | - Yang Bai
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Jinhui Zhang
- Department of Orthopaedics, the Affiliated Southeast Hospital of Xiamen University, Zhangzhou 175 Hospital, Zhangzhou, Fujian, China
| | - Guang Yang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA.
| | - Wen-Biao Gan
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China.
| | - Peifu Tang
- Department of Orthopaedics, Peking 301 Hospital, Beijing, China.
| |
Collapse
|
22
|
Astrocytes regulate neuronal network activity by mediating synapse remodeling. Neurosci Res 2023; 187:3-13. [PMID: 36170922 DOI: 10.1016/j.neures.2022.09.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 11/22/2022]
Abstract
Based on experience during our life, neuronal connectivity continuously changes through structural remodeling of synapses. Recent studies have shown that the complex interaction between astrocytes and synapses regulates structural synapse remodeling by inducing the formation and elimination of synapses, as well as their functional maturation. Defects in this astrocyte-mediated synapse remodeling cause problems in not only neuronal network activities but also animal behaviors. Moreover, in various neurological disorders, astrocytes have been shown to play central roles in the initiation and progression of synaptic pathophysiology through impaired interactions with synapses. In this review, we will discuss recent studies identifying the novel roles of astrocytes in neuronal circuit remodeling, focusing on synapse formation and elimination. We will also discuss the potential implication of defective astrocytic function in evoking various brain disorders.
Collapse
|
23
|
Chen G, Xu J, Luo H, Luo X, Singh SK, Ramirez JJ, James ML, Mathew JP, Berger M, Eroglu C, Ji RR. Hevin/Sparcl1 drives pathological pain through spinal cord astrocyte and NMDA receptor signaling. JCI Insight 2022; 7:161028. [PMID: 36256481 PMCID: PMC9746899 DOI: 10.1172/jci.insight.161028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 10/13/2022] [Indexed: 01/24/2023] Open
Abstract
High endothelial venule protein/SPARC-like 1 (hevin/Sparcl1) is an astrocyte-secreted protein that regulates synapse formation in the brain. Here we show that astrocytic hevin signaling plays a critical role in maintaining chronic pain. Compared with WT mice, hevin-null mice exhibited normal mechanical and heat sensitivity but reduced inflammatory pain. Interestingly, hevin-null mice have faster recovery than WT mice from neuropathic pain after nerve injury. Intrathecal injection of WT hevin was sufficient to induce persistent mechanical allodynia in naive mice. In hevin-null mice with nerve injury, adeno-associated-virus-mediated (AAV-mediated) re-expression of hevin in glial fibrillary acidic protein-expressing (GFAP-expressing) spinal cord astrocytes could reinstate neuropathic pain. Mechanistically, hevin is crucial for spinal cord NMDA receptor (NMDAR) signaling. Hevin-potentiated N-Methyl-D-aspartic acid (NMDA) currents are mediated by GluN2B-containing NMDARs. Furthermore, intrathecal injection of a neutralizing Ab against hevin alleviated acute and persistent inflammatory pain, postoperative pain, and neuropathic pain. Secreted hevin that was detected in mouse cerebrospinal fluid (CSF) and nerve injury significantly increased CSF hevin abundance. Finally, neurosurgery caused rapid and substantial increases in SPARCL1/HEVIN levels in human CSF. Collectively, our findings support a critical role of hevin and astrocytes in the maintenance of chronic pain. Neutralizing of secreted hevin with monoclonal Ab may provide a new therapeutic strategy for treating acute and chronic pain and NMDAR-medicated neurodegeneration.
Collapse
Affiliation(s)
- Gang Chen
- Center for Translational Pain Medicine, Department of Anesthesiology, and
| | - Jing Xu
- Center for Translational Pain Medicine, Department of Anesthesiology, and
| | - Hao Luo
- Center for Translational Pain Medicine, Department of Anesthesiology, and
| | - Xin Luo
- Center for Translational Pain Medicine, Department of Anesthesiology, and
| | - Sandeep K. Singh
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Juan J. Ramirez
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neurobiology
| | | | | | | | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neurobiology,,Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina, USA.,Duke Institute for Brain Sciences (DIBS), Durham, North Carolina, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, and,Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neurobiology,,Duke Institute for Brain Sciences (DIBS), Durham, North Carolina, USA
| |
Collapse
|
24
|
Chen YL, Feng XL, Cheung CW, Liu JA. Mode of action of astrocytes in pain: From the spinal cord to the brain. Prog Neurobiol 2022; 219:102365. [DOI: 10.1016/j.pneurobio.2022.102365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/09/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
|
25
|
Fujimura M. Fasudil, a ROCK inhibitor, prevents neuropathic pain in Minamata disease model rats. Toxicol Lett 2022; 371:38-45. [PMID: 36244566 DOI: 10.1016/j.toxlet.2022.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/24/2022] [Accepted: 10/11/2022] [Indexed: 02/13/2023]
Abstract
Methylmercury (MeHg), an environmental toxicant, is known to cause sensory impairment by inducing neurodegeneration of sensory nervous systems. However, in recent years, it has been revealed that neuropathic pain occurs in the chronic phase of MeHg poisoning, that is, in current Minamata disease patients. Our recent study using Minamata disease model rats demonstrated that MeHg-mediated neurodegeneration in the sensory nervous system may induce inflammatory microglia production in the dorsal horn of the spinal cord and subsequent somatosensory cortical rewiring, leading to neuropathic pain. We hypothesized that inhibition of the Rho-associated coiled coil-forming protein kinase (ROCK) pathway could prevent MeHg-induced neuropathic pain because the ROCK pathway is known to be involved in inducing the production of inflammatory microglia. Here, we showed for the first time that Fasudil, a ROCK inhibitor, can prevent neuropathic pain in Minamata disease model rats. In this model, Fasudil significantly suppressed nerve injury-induced inflammatory microglia production in the dorsal horn of the spinal cord and prevented subsequent somatosensory cortical rewiring. These results suggest that the ROCK pathway is involved in the onset and development of neuropathic pain in the chronic phase of Minamata disease, and that its inhibition is effective in pain prevention.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto 867-0008, Japan.
| |
Collapse
|
26
|
Astrocytes in Chronic Pain: Cellular and Molecular Mechanisms. Neurosci Bull 2022; 39:425-439. [PMID: 36376699 PMCID: PMC10043112 DOI: 10.1007/s12264-022-00961-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/17/2022] [Indexed: 11/15/2022] Open
Abstract
AbstractChronic pain is challenging to treat due to the limited therapeutic options and adverse side-effects of therapies. Astrocytes are the most abundant glial cells in the central nervous system and play important roles in different pathological conditions, including chronic pain. Astrocytes regulate nociceptive synaptic transmission and network function via neuron–glia and glia–glia interactions to exaggerate pain signals under chronic pain conditions. It is also becoming clear that astrocytes play active roles in brain regions important for the emotional and memory-related aspects of chronic pain. Therefore, this review presents our current understanding of the roles of astrocytes in chronic pain, how they regulate nociceptive responses, and their cellular and molecular mechanisms of action.
Collapse
|
27
|
Malange KF, Navia-Pelaez JM, Dias EV, Lemes JBP, Choi SH, Dos Santos GG, Yaksh TL, Corr M. Macrophages and glial cells: Innate immune drivers of inflammatory arthritic pain perception from peripheral joints to the central nervous system. FRONTIERS IN PAIN RESEARCH 2022; 3:1018800. [PMID: 36387416 PMCID: PMC9644179 DOI: 10.3389/fpain.2022.1018800] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/03/2022] [Indexed: 07/22/2023] Open
Abstract
Millions of people suffer from arthritis worldwide, consistently struggling with daily activities due to debilitating pain evoked by this disease. Perhaps the most intensively investigated type of inflammatory arthritis is rheumatoid arthritis (RA), where, despite considerable advances in research and clinical management, gaps regarding the neuroimmune interactions that guide inflammation and chronic pain in this disease remain to be clarified. The pain and inflammation associated with arthritis are not isolated to the joints, and inflammatory mechanisms induced by different immune and glial cells in other tissues may affect the development of chronic pain that results from the disease. This review aims to provide an overview of the state-of-the-art research on the roles that innate immune, and glial cells play in the onset and maintenance of arthritis-associated pain, reviewing nociceptive pathways from the joint through the dorsal root ganglion, spinal circuits, and different structures in the brain. We will focus on the cellular mechanisms related to neuroinflammation and pain, and treatments targeting these mechanisms from the periphery and the CNS. A comprehensive understanding of the role these cells play in peripheral inflammation and initiation of pain and the central pathways in the spinal cord and brain will facilitate identifying new targets and pathways to aide in developing therapeutic strategies to treat joint pain associated with RA.
Collapse
Affiliation(s)
- Kaue Franco Malange
- Department of Anesthesiology, University of California, San Diego, CA, United States
| | | | - Elayne Vieira Dias
- Department of Neurology, University of California, San Francisco, CA, United States
| | | | - Soo-Ho Choi
- Department of Medicine, University of California, San Diego, CA, United States
| | | | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, CA, United States
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, CA, United States
| |
Collapse
|
28
|
Kruyer A. Astrocyte Heterogeneity in Regulation of Synaptic Activity. Cells 2022; 11:cells11193135. [PMID: 36231097 PMCID: PMC9562199 DOI: 10.3390/cells11193135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/02/2022] [Accepted: 10/02/2022] [Indexed: 02/07/2023] Open
Abstract
Our awareness of the number of synapse regulatory functions performed by astroglia is rapidly expanding, raising interesting questions regarding astrocyte heterogeneity and specialization across brain regions. Whether all astrocytes are poised to signal in a multitude of ways, or are instead tuned to surrounding synapses and how astroglial signaling is altered in psychiatric and cognitive disorders are fundamental questions for the field. In recent years, molecular and morphological characterization of astroglial types has broadened our ability to design studies to better analyze and manipulate specific functions of astroglia. Recent data emerging from these studies will be discussed in depth in this review. I also highlight remaining questions emerging from new techniques recently applied toward understanding the roles of astrocytes in synapse regulation in the adult brain.
Collapse
Affiliation(s)
- Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
29
|
Choi S, Jang DC, Chung G, Kim SK. Transcutaneous Auricular Vagus Nerve Stimulation Enhances Cerebrospinal Fluid Circulation and Restores Cognitive Function in the Rodent Model of Vascular Cognitive Impairment. Cells 2022; 11:cells11193019. [PMID: 36230988 PMCID: PMC9564197 DOI: 10.3390/cells11193019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular cognitive impairment (VCI) is a common sequela of cerebrovascular disorders. Although transcutaneous auricular vagus nerve stimulation (taVNS) has been considered a complementary treatment for various cognitive disorders, preclinical data on the effect of taVNS on VCI and its mechanism remain ambiguous. To measure cerebrospinal fluid (CSF) circulation during taVNS, we used in vivo two-photon microscopy with CSF and vasculature tracers. VCI was induced by transient bilateral common carotid artery occlusion (tBCCAO) surgery in mice. The animals underwent anesthesia, off-site stimulation, or taVNS for 20 min. Cognitive tests, including the novel object recognition and the Y-maze tests, were performed 24 h after the last treatment. The long-term treatment group received 6 days of treatment and was tested on day 7; the short-term treatment group received 2 days of treatment and was tested 3 days after tBCCAO surgery. CSF circulation increased remarkably in the taVNS group, but not in the anesthesia-control or off-site-stimulation-control groups. The cognitive impairment induced by tBCCAO was significantly restored after both long- and short-term taVNS. In terms of effects, both long- and short-term stimulations showed similar recovery effects. Our findings provide evidence that taVNS can facilitate CSF circulation and that repetitive taVNS can ameliorate VCI symptoms.
Collapse
Affiliation(s)
- Seunghwan Choi
- Department of East-West Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Dong Cheol Jang
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Geehoon Chung
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Sun Kwang Kim
- Department of East-West Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
- Correspondence:
| |
Collapse
|
30
|
Dong Z, Zhan T, Sun H, Wang J, Duan G, Zhang Y, Chen Y, Huang Y, Xu S. Astrocytic ERK/STAT1 signaling contributes to maintenance of stress-related visceral hypersensitivity in rats. THE JOURNAL OF PAIN 2022; 23:1973-1988. [PMID: 35914642 DOI: 10.1016/j.jpain.2022.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 10/16/2022]
Abstract
The rostral anterior cingulate cortex (rACC) has been found to be an important brain region in mediating visceral hypersensitivity. However, the underlying mechanisms remain unclear. This study aimed to explore the role of astrocytes in the maintenance of visceral hypersensitivity induced by chronic water avoidance stress (WAS) as well as the potential signaling pathway that activates astrocytes in the rACC. We found that ACC-reactive astrogliosis resulted in the overexpression of c-fos, TSP-1, and BDNF in stress-related visceral hypersensitivity rats. Visceral hypersensitivity was reversed by pharmacological inhibition of astrocytic activation after WAS, as were the overexpression of c-fos, TSP-1 and BDNF. Activation of the astrocytic Gi-pathway increased the visceral sensitivity and expression of c-fos, TSP-1, and BDNF. Visceral hypersensitivity was also ameliorated by the pharmacological inhibition of ERK and STAT1 phosphorylation after WAS. Furthermore, inhibition of the ERK-STAT1 cascade reduced astrocytic activation. These findings suggest that astrocytic ERK/STAT1 signaling in the rACC contributes to the maintenance of stress-related visceral hypersensitivity. PERSPECTIVE: Visceral hypersensitivity is a key factor in the pathophysiology of irritable bowel syndrome. This study highlights the important role of astrocytic ERK/STAT1 signaling in activating astrocytes in the rostral anterior cingulate cortex, which contributes to visceral hypersensitivity.
Collapse
Affiliation(s)
- Zhiyu Dong
- Department of Gastroenterology, Tongji Institute of Digestive Diseases, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Tingting Zhan
- Department of Gastroenterology, Tongji Institute of Digestive Diseases, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Huihui Sun
- Department of Gastroenterology, Tongji Institute of Digestive Diseases, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Junwen Wang
- Department of Gastroenterology, Tongji Institute of Digestive Diseases, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Guangbing Duan
- Department of Gastroenterology, Tongji Institute of Digestive Diseases, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yan Zhang
- Department of Gastroenterology, Tongji Institute of Digestive Diseases, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Ying Chen
- Department of Gastroenterology, Tongji Institute of Digestive Diseases, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Ying Huang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Ministry of Education), Department of Physiology and Pharmacology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Shuchang Xu
- Department of Gastroenterology, Tongji Institute of Digestive Diseases, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| |
Collapse
|
31
|
Activation of NLRP3 Is Required for a Functional and Beneficial Microglia Response after Brain Trauma. Pharmaceutics 2022; 14:pharmaceutics14081550. [PMID: 35893807 PMCID: PMC9332196 DOI: 10.3390/pharmaceutics14081550] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/04/2022] Open
Abstract
Despite the numerous research studies on traumatic brain injury (TBI), many physiopathologic mechanisms remain unknown. TBI is a complex process, in which neuroinflammation and glial cells play an important role in exerting a functional immune and damage-repair response. The activation of the NLRP3 inflammasome is one of the first steps to initiate neuroinflammation and so its regulation is essential. Using a closed-head injury model and a pharmacological (MCC950; 3 mg/kg, pre- and post-injury) and genetical approach (NLRP3 knockout (KO) mice), we defined the transcriptional and behavioral profiles 24 h after TBI. Wild-type (WT) mice showed a strong pro-inflammatory response, with increased expression of inflammasome components, microglia and astrocytes markers, and cytokines. There was no difference in the IL1β production between WT and KO, nor compensatory mechanisms of other inflammasomes. However, some microglia and astrocyte markers were overexpressed in KO mice, resulting in an exacerbated cytokine expression. Pretreatment with MCC950 replicated the behavioral and blood-brain barrier results observed in KO mice and its administration 1 h after the lesion improved the damage. These findings highlight the importance of NLRP3 time-dependent activation and its role in the fine regulation of glial response.
Collapse
|
32
|
Boorman DC, Keay KA. Sex differences in morphine sensitivity are associated with differential glial expression in the brainstem of rats with neuropathic pain. J Neurosci Res 2022; 100:1890-1907. [PMID: 35853016 PMCID: PMC9543783 DOI: 10.1002/jnr.25103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/11/2022]
Abstract
Chronic pain is more prevalent and reported to be more severe in women. Opioid analgesics are less effective in women and result in stronger nauseant effects. The neurobiological mechanisms underlying these sex differences have yet to be clearly defined, though recent research has suggested neuronal–glial interactions are likely involved. We have previously shown that similar to people, morphine is less effective at reducing pain behaviors in female rats. In this study, we used the immunohistochemical detection of glial fibrillary acidic protein (GFAP) expression to investigate sex differences in astrocyte density and morphology in six medullary regions known to be modulated by pain and/or opioids. Morphine administration had small sex‐dependent effects on overall GFAP expression, but not on astrocyte morphology, in the rostral ventromedial medulla, the subnucleus reticularis dorsalis, and the area postrema. Significant sex differences in the density and morphology of GFAP immunopositive astrocytes were detected in all six regions. In general, GFAP‐positive cells in females showed smaller volumes and reduced complexity than those observed in males. Furthermore, females showed lower overall GFAP expression in all regions except for the area postrema, the critical medullary region responsible for opioid‐induced nausea and emesis. These data support the possibility that differences in astrocyte activity might underlie the sex differences seen in the processing of opioids in the context of chronic neuropathic pain.
Collapse
Affiliation(s)
- Damien C. Boorman
- School of Medical Sciences and the Brain and Mind Centre The University of Sydney Camperdown New South Wales Australia
| | - Kevin A. Keay
- School of Medical Sciences and the Brain and Mind Centre The University of Sydney Camperdown New South Wales Australia
| |
Collapse
|
33
|
Takeda I, Yoshihara K, Cheung DL, Kobayashi T, Agetsuma M, Tsuda M, Eto K, Koizumi S, Wake H, Moorhouse AJ, Nabekura J. Controlled activation of cortical astrocytes modulates neuropathic pain-like behaviour. Nat Commun 2022; 13:4100. [PMID: 35835747 PMCID: PMC9283422 DOI: 10.1038/s41467-022-31773-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/04/2022] [Indexed: 11/20/2022] Open
Abstract
Chronic pain is a major public health problem that currently lacks effective treatment options. Here, a method that can modulate chronic pain-like behaviour induced by nerve injury in mice is described. By combining a transient nerve block to inhibit noxious afferent input from injured peripheral nerves, with concurrent activation of astrocytes in the somatosensory cortex (S1) by either low intensity transcranial direct current stimulation (tDCS) or via the chemogenetic DREADD system, we could reverse allodynia-like behaviour previously established by partial sciatic nerve ligation (PSL). Such activation of astrocytes initiated spine plasticity to reduce those synapses formed shortly after PSL. This reversal from allodynia-like behaviour persisted well beyond the active treatment period. Thus, our study demonstrates a robust and potentially translational approach for modulating pain, that capitalizes on the interplay between noxious afferents, sensitized central neuronal circuits, and astrocyte-activation induced synaptic plasticity. Astrocytes may contribute to synaptic remodelling in the cortex in chronic pain states. Here the authors describe modulation of astrocyte activity to drive circuit reorganization in somatosensory cortex in mice, along with peripheral nerve block, which could be a potential therapeutic approach for the treatment of chronic pain.
Collapse
Affiliation(s)
- Ikuko Takeda
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Anatomy and Molecular Cell Biology Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, Okazaki, Japan
| | - Kohei Yoshihara
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Dennis L Cheung
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Japan
| | - Tomoko Kobayashi
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Japan
| | - Masakazu Agetsuma
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Japan.,Division of Molecular Design, Research Center for Systems Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kei Eto
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiology, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, Okazaki, Japan.,Center of Optical Scattering Image Science Department of Systems Science, Kobe University, Kobe, Japan
| | - Andrew J Moorhouse
- Department of Physiology, School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Japan. .,Graduate School of Medicine, Nagoya University, Nagoya, Japan. .,Department of Physiological Sciences, Graduate University for Advanced Studies, SOKENDAI, Hayama, Japan.
| |
Collapse
|
34
|
Chung G, Kim SK. Therapeutics for Chemotherapy-Induced Peripheral Neuropathy: Approaches with Natural Compounds from Traditional Eastern Medicine. Pharmaceutics 2022; 14:pharmaceutics14071407. [PMID: 35890302 PMCID: PMC9319448 DOI: 10.3390/pharmaceutics14071407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/23/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) often develops in patients with cancer treated with commonly used anti-cancer drugs. The symptoms of CIPN can occur acutely during chemotherapy or emerge after cessation, and often accompany long-lasting intractable pain. This adverse side effect not only affects the quality of life but also limits the use of chemotherapy, leading to a reduction in the survival rate of patients with cancer. Currently, effective treatments for CIPN are limited, and various interventions are being applied by clinicians and patients because of the unmet clinical need. Potential approaches to ameliorate CIPN include traditional Eastern medicine-based methods. Medicinal substances from traditional Eastern medicine have well-established analgesic effects and are generally safe. Furthermore, many substances can also improve other comorbid symptoms in patients. This article aims to provide information regarding traditional Eastern medicine-based plant extracts and natural compounds for CIPN. In this regard, we briefly summarized the development, mechanisms, and changes in the nervous system related to CIPN, and reviewed the substances of traditional Eastern medicine that have been exploited to treat CIPN in preclinical and clinical settings.
Collapse
Affiliation(s)
- Geehoon Chung
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Sun Kwang Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea;
- Department of Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Correspondence:
| |
Collapse
|
35
|
Hiraga SI, Itokazu T, Nishibe M, Yamashita T. Neuroplasticity related to chronic pain and its modulation by microglia. Inflamm Regen 2022; 42:15. [PMID: 35501933 PMCID: PMC9063368 DOI: 10.1186/s41232-022-00199-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/19/2022] [Indexed: 01/03/2023] Open
Abstract
Neuropathic pain is often chronic and can persist after overt tissue damage heals, suggesting that its underlying mechanism involves the alteration of neuronal function. Such an alteration can be a direct consequence of nerve damage or a result of neuroplasticity secondary to the damage to tissues or to neurons. Recent studies have shown that neuroplasticity is linked to causing neuropathic pain in response to nerve damage, which may occur adjacent to or remotely from the site of injury. Furthermore, studies have revealed that neuroplasticity relevant to chronic pain is modulated by microglia, resident immune cells of the central nervous system (CNS). Microglia may directly contribute to synaptic remodeling and altering pain circuits, or indirectly contribute to neuroplasticity through property changes, including the secretion of growth factors. We herein highlight the mechanisms underlying neuroplasticity that occur in the somatosensory circuit of the spinal dorsal horn, thalamus, and cortex associated with chronic pain following injury to the peripheral nervous system (PNS) or CNS. We also discuss the dynamic functions of microglia in shaping neuroplasticity related to chronic pain. We suggest further understanding of post-injury ectopic plasticity in the somatosensory circuits may shed light on the differential mechanisms underlying nociceptive, neuropathic, and nociplastic-type pain. While one of the prominent roles played by microglia appears to be the modulation of post-injury neuroplasticity. Therefore, future molecular- or genetics-based studies that address microglia-mediated post-injury neuroplasticity may contribute to the development of novel therapies for chronic pain.
Collapse
Affiliation(s)
- Shin-Ichiro Hiraga
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takahide Itokazu
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan. .,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Mariko Nishibe
- Center for Strategic Innovative Dentistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Toshihide Yamashita
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan. .,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan. .,WPI Immunology Frontier Research Center, Osaka, Japan. .,Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| |
Collapse
|
36
|
Danjo Y, Shigetomi E, Hirayama YJ, Kobayashi K, Ishikawa T, Fukazawa Y, Shibata K, Takanashi K, Parajuli B, Shinozaki Y, Kim SK, Nabekura J, Koizumi S. Transient astrocytic mGluR5 expression drives synaptic plasticity and subsequent chronic pain in mice. J Exp Med 2022; 219:213089. [PMID: 35319723 PMCID: PMC8952801 DOI: 10.1084/jem.20210989] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 01/03/2022] [Accepted: 01/31/2022] [Indexed: 01/02/2023] Open
Abstract
Activation of astrocytes has a profound effect on brain plasticity and is critical for the pathophysiology of several neurological disorders including neuropathic pain. Here, we show that metabotropic glutamate receptor 5 (mGluR5), which reemerges in astrocytes in a restricted time frame, is essential for these functions. Although mGluR5 is absent in healthy adult astrocytes, it transiently reemerges in astrocytes of the somatosensory cortex (S1). During a limited spatiotemporal time frame, astrocytic mGluR5 drives Ca2+ signals; upregulates multiple synaptogenic molecules such as Thrombospondin-1, Glypican-4, and Hevin; causes excess excitatory synaptogenesis; and produces persistent alteration of S1 neuronal activity, leading to mechanical allodynia. All of these events were abolished by the astrocyte-specific deletion of mGluR5. Astrocytes dynamically control synaptic plasticity by turning on and off a single molecule, mGluR5, which defines subsequent persistent brain functions, especially under pathological conditions.
Collapse
Affiliation(s)
- Yosuke Danjo
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Yukiho J Hirayama
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kenji Kobayashi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Tatsuya Ishikawa
- Department of Functional Anatomy, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Keisuke Shibata
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kenta Takanashi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Bijay Parajuli
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Sun Kwang Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan.,Department of Physiological Sciences, The Graduate School for Advanced Study, Hayama, Kanagawa, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
37
|
Miranpuri GS, Bali P, Nguyen J, Kim JJ, Modgil S, Mehra P, Buttar S, Brown G, Yutuc N, Singh H, Wood A, Singh J, Anand A. Role of Microglia and Astrocytes in Spinal Cord Injury Induced Neuropathic Pain. Ann Neurosci 2022; 28:219-228. [PMID: 35341227 PMCID: PMC8948321 DOI: 10.1177/09727531211046367] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/03/2021] [Indexed: 12/30/2022] Open
Abstract
Background: Spinal cord injuries incite varying degrees of symptoms in patients, ranging
from weakness and incoordination to paralysis. Common amongst spinal cord
injury (SCI) patients, neuropathic pain (NP) is a debilitating medical
condition. Unfortunately, there remain many clinical impediments in treating
NP because there is a lack of understanding regarding the mechanisms behind
SCI-induced NP (SCINP). Given that more than 450,000 people in the United
States alone suffer from SCI, it is unsatisfactory that current treatments
yield poor results in alleviating and treating NP. Summary: In this review, we briefly discussed the models of SCINP along with the
mechanisms of NP progression. Further, current treatment modalities are
herein explored for SCINP involving pharmacological interventions targeting
glia cells and astrocytes. Key message: The studies presented in this review provide insight for new directions
regarding SCINP alleviation. Given the severity and incapacitating effects
of SCINP, it is imperative to study the pathways involved and find new
therapeutic targets in coordination with stem cell research, and to develop
a new gold-standard in SCINP treatment.
Collapse
Affiliation(s)
- Gurwattan S Miranpuri
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Parul Bali
- Department of Biological Sciences, Indian Institute of Science Education & Research Mohali, India
| | - Justyn Nguyen
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Jason J Kim
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Shweta Modgil
- Neuroscience research lab, Department of Neurology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Priya Mehra
- Neuroscience research lab, Department of Neurology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.,Department of Biotechnology, Panjab University, Chandigarh, India
| | - Seah Buttar
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Greta Brown
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Noemi Yutuc
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Harpreet Singh
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Aleksandar Wood
- Department of Neurological Surgery, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Jagtar Singh
- Department of Biotechnology, Panjab University, Chandigarh, India
| | - Akshay Anand
- Neuroscience research lab, Department of Neurology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.,CCRYN- Collaborative Centre for Mind Body Intervention through Yoga.,Centre of Phenomenology and Cognitive Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
38
|
Kim YR, Kim SJ. Altered synaptic connections and inhibitory network of the primary somatosensory cortex in chronic pain. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:69-75. [PMID: 35203057 PMCID: PMC8890942 DOI: 10.4196/kjpp.2022.26.2.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
Chronic pain is induced by tissue or nerve damage and is accompanied by pain hypersensitivity (i.e., allodynia and hyperalgesia). Previous studies using in vivo two-photon microscopy have shown functional and structural changes in the primary somatosensory (S1) cortex at the cellular and synaptic levels in inflammatory and neuropathic chronic pain. Furthermore, alterations in local cortical circuits were revealed during the development of chronic pain. In this review, we summarize recent findings regarding functional and structural plastic changes of the S1 cortex and alteration of the S1 inhibitory network in chronic pain. Finally, we discuss potential neuromodulators driving modified cortical circuits and suggest further studies to understand the cortical mechanisms that induce pain hypersensitivity.
Collapse
Affiliation(s)
- Yoo Rim Kim
- Departments of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sang Jeong Kim
- Departments of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Departments of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
39
|
The Correlation between Functional Connectivity of the Primary Somatosensory Cortex and Cervical Spinal Cord Microstructural Injury in Patients with Cervical Spondylotic Myelopathy. DISEASE MARKERS 2022; 2022:2623179. [PMID: 35096201 PMCID: PMC8791726 DOI: 10.1155/2022/2623179] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/17/2021] [Indexed: 11/23/2022]
Abstract
Objectives To explore functional connectivity reorganization of the primary somatosensory cortex, the chronic microstructure damage of the cervical spinal cord, and their relationship in cervical spondylotic myelopathy (CSM) patients. Methods Thirty-three patients with CSM and 23 healthy controls (HCs) were recruited for rs-fMRI and cervical spinal cord diffusion tensor imaging (DTI) scans. Six subregions (including leg, back, chest, hand, finger and face) of bilateral primary somatosensory cortex (S1) were selected for seed-based whole-brain functional connectivity (FC). Then, we calculated the apparent diffusion coefficient (ADC) and fractional anisotropy (FA) values of the cervical spinal cord. Correlation analysis was conducted between FC values of brain regions and DTI parameters of cervical spinal cord (ADC, FA), and their relationship with each other and clinical parameters. Results Compared with the HC group, the CSM group showed decreased FC between areas of the left S1hand, the left S1leg, the right S1chest, and the right S1leg with brain regions. The mean FA values of the cervical spinal cord in CSM patients were positively correlated with JOA scores. Especially, the FApos values of bilateral posterior funiculus were positively correlated with JOA scores. The ADC and FA values of bilateral posterior funiculus in the cervical spinal cord were also positively correlated with the FC values. Conclusions There was synchronization between chronic cervical spinal cord microstructural injury and cerebral cortex sensory function compensatory recombination. DTI parameters of the posterior cervical spinal cord could objectively reflect the degree of cerebral cortex sensory function impairment to a certain extent.
Collapse
|
40
|
Koizumi S. Glial Purinergic Signals and Psychiatric Disorders. Front Cell Neurosci 2022; 15:822614. [PMID: 35069121 PMCID: PMC8766327 DOI: 10.3389/fncel.2021.822614] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 12/08/2021] [Indexed: 12/30/2022] Open
Abstract
Emotion-related neural networks are regulated in part by the activity of glial cells, and glial dysfunction can be directly related to emotional diseases such as depression. Here, we discuss three different therapeutic strategies involving astrocytes that are effective for treating depression. First, the antidepressant, fluoxetine, acts on astrocytes and increases exocytosis of ATP. This has therapeutic effects via brain-derived neurotrophic factor-dependent mechanisms. Second, electroconvulsive therapy is a well-known treatment for drug-resistant depression. Electroconvulsive therapy releases ATP from astrocytes to induce leukemia inhibitory factors and fibroblast growth factor 2, which leads to antidepressive actions. Finally, sleep deprivation therapy is well-known to cause antidepressive effects. Sleep deprivation also increases release of ATP, whose metabolite, adenosine, has antidepressive effects. These independent treatments share the same mechanism, i.e., ATP release from astrocytes, indicating an essential role of glial purinergic signals in the pathogenesis of depression.
Collapse
Affiliation(s)
- Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
- GLIA Center, University of Yamanashi, Yamanashi, Japan
- *Correspondence: Schuichi Koizumi
| |
Collapse
|
41
|
Iseppon F, Linley JE, Wood JN. Calcium imaging for analgesic drug discovery. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2022; 11:100083. [PMID: 35079661 PMCID: PMC8777277 DOI: 10.1016/j.ynpai.2021.100083] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/17/2021] [Accepted: 12/27/2021] [Indexed: 11/24/2022]
Abstract
Somatosensation and pain are complex phenomena involving a rangeofspecialised cell types forming different circuits within the peripheral and central nervous systems. In recent decades, advances in the investigation of these networks, as well as their function in sensation, resulted from the constant evolution of electrophysiology and imaging techniques to allow the observation of cellular activity at the population level both in vitro and in vivo. Genetically encoded indicators of neuronal activity, combined with recent advances in DNA engineering and modern microscopy, offer powerful tools to dissect and visualise the activity of specific neuronal subpopulations with high spatial and temporal resolution. In recent years various groups developed in vivo imaging techniques to image calcium transients in the dorsal root ganglia, the spinal cord and the brain of anesthetised and awake, behaving animals to address fundamental questions in both the physiology and pathophysiology of somatosensation and pain. This approach, besides giving unprecedented details on the circuitry of innocuous and painful sensation, can be a very powerful tool for pharmacological research, from the characterisation of new potential drugs to the discovery of new, druggable targets within specific neuronal subpopulations. Here we summarise recent developments in calcium imaging for pain research, discuss technical challenges and advances, and examine the potential positive impact of this technique in early preclinical phases of the analgesic drug discovery process.
Collapse
Affiliation(s)
- Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, WC1E 6BT London, UK
- Discovery UK, Neuroscience, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - John E. Linley
- Discovery UK, Neuroscience, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, WC1E 6BT London, UK
| |
Collapse
|
42
|
Koizumi S, Shigetomi E, Sano F, Saito K, Kim SK, Nabekura J. Abnormal Ca 2+ Signals in Reactive Astrocytes as a Common Cause of Brain Diseases. Int J Mol Sci 2021; 23:149. [PMID: 35008573 PMCID: PMC8745111 DOI: 10.3390/ijms23010149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/30/2022] Open
Abstract
In pathological brain conditions, glial cells become reactive and show a variety of responses. We examined Ca2+ signals in pathological brains and found that reactive astrocytes share abnormal Ca2+ signals, even in different types of diseases. In a neuropathic pain model, astrocytes in the primary sensory cortex became reactive and showed frequent Ca2+ signals, resulting in the production of synaptogenic molecules, which led to misconnections of tactile and pain networks in the sensory cortex, thus causing neuropathic pain. In an epileptogenic model, hippocampal astrocytes also became reactive and showed frequent Ca2+ signals. In an Alexander disease (AxD) model, hGFAP-R239H knock-in mice showed accumulation of Rosenthal fibers, a typical pathological marker of AxD, and excessively large Ca2+ signals. Because the abnormal astrocytic Ca2+ signals observed in the above three disease models are dependent on type II inositol 1,4,5-trisphosphate receptors (IP3RII), we reanalyzed these pathological events using IP3RII-deficient mice and found that all abnormal Ca2+ signals and pathologies were markedly reduced. These findings indicate that abnormal Ca2+ signaling is not only a consequence but may also be greatly involved in the cause of these diseases. Abnormal Ca2+ signals in reactive astrocytes may represent an underlying pathology common to multiple diseases.
Collapse
Affiliation(s)
- Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan; (E.S.); (F.S.); (K.S.)
- GLIA Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan; (E.S.); (F.S.); (K.S.)
- GLIA Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Fumikazu Sano
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan; (E.S.); (F.S.); (K.S.)
- GLIA Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan; (E.S.); (F.S.); (K.S.)
- GLIA Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Sun Kwang Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea;
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki 444-8585, Japan;
| |
Collapse
|
43
|
Wang Y, Wu Z, Wang D, Huang C, Xu J, Liu C, Yang C. Muscle-brain communication in pain: The key role of myokines. Brain Res Bull 2021; 179:25-35. [PMID: 34871710 DOI: 10.1016/j.brainresbull.2021.11.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 12/24/2022]
Abstract
Pain is the most common reason for a physician visit, which accounts for a considerable proportion of the global burden of disease and greatly affects patients' quality of life. Therefore, there is an urgent need to identify new therapeutic targets involved in pain. Exercise-induced hypoalgesia (EIH) is a well known phenomenon observed worldwide. However, the available evidence demonstrates that the mechanisms of EIH remain unclear. One of the most accepted hypotheses has been the activation of several endogenous systems in the brain. Recently, the concept that the muscle acts as a secretory organ has attracted increasing attention. Proteins secreted by the muscle are called myokines, playing a critical role in communicating with other organs, such as the brain. This review will focus on several myokines and discuss their roles in EIH.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zifeng Wu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Di Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chaoli Huang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Jiali Xu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
44
|
Koizumi S. Glial pharmacology in Asia & Beyond. Pharmacol Res Perspect 2021; 9:e00881. [PMID: 34676997 PMCID: PMC8532131 DOI: 10.1002/prp2.881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Schuichi Koizumi
- Department of NeuropharmacologyGraduate School of MedicineUniversity of YamanashiYamanashiJapan
- GLIA CenterUniversity of YamanashiYamanashiJapan
| |
Collapse
|
45
|
Takanashi K, Shibata K, Mizuno K, Komatsu R, Koizumi S. Goshajinkigan attenuates paclitaxel-induced neuropathic pain via cortical astrocytes. Pharmacol Res Perspect 2021; 9:e00850. [PMID: 34676996 PMCID: PMC8532134 DOI: 10.1002/prp2.850] [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: 07/12/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/31/2022] Open
Abstract
The anticancer agents platinum derivatives and taxanes such as paclitaxel (PCX) often cause neuropathy known as chemotherapy‐induced peripheral neuropathy with high frequency. However, the cellular and molecular mechanisms underlying such neuropathy largely remain unknown. Here, we show new findings that the effect of Goshajinkigan (GJG), a Japanese KAMPO medicine, inhibits PCX‐induced neuropathy by acting on astrocytes. The administration of PCX in mice caused the sustained neuropathy lasting at least 4 weeks, which included mechanical allodynia and thermal hyperalgesia but not cold allodynia. PCX‐evoked pain behaviors were associated with the sensitization of all primary afferent fibers. PCX did not activate microglia or astrocytes in the spinal cord. However, it significantly activated astrocytes in the primary sensory (S1) cortex without affecting S1 microglial activation there. GJG significantly inhibited the PCX‐induced mechanical allodynia by 50% and thermal hyperalgesia by 90%, which was in accordance with the abolishment of astrocytic activation in the S1 cortex. Finally, the inhibition of S1 astrocytes by an astrocyte‐toxin L‐alpha‐aminoadipic acid abolished the PCX‐induced neuropathy. Our findings suggest that astrocytes in the S1 cortex would play an important role in the pathogenesis of PCX‐induced neuropathy and are a potential target for its treatment.
Collapse
Affiliation(s)
- Kenta Takanashi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Keisuke Shibata
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Keita Mizuno
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan.,Tsumura Research Laboratories, Kampo Scientific Strategies Division, Tsumura & Co, Inashiki-gun, Japan
| | - Ryohei Komatsu
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, Japan.,Yamanashi GLIA Center, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
46
|
Twible C, Abdo R, Zhang Q. Astrocyte Role in Temporal Lobe Epilepsy and Development of Mossy Fiber Sprouting. Front Cell Neurosci 2021; 15:725693. [PMID: 34658792 PMCID: PMC8514632 DOI: 10.3389/fncel.2021.725693] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Epilepsy affects approximately 50 million people worldwide, with 60% of adult epilepsies presenting an onset of focal origin. The most common focal epilepsy is temporal lobe epilepsy (TLE). The role of astrocytes in the presentation and development of TLE has been increasingly studied and discussed within the literature. The most common histopathological diagnosis of TLE is hippocampal sclerosis. Hippocampal sclerosis is characterized by neuronal cell loss within the Cornu ammonis and reactive astrogliosis. In some cases, mossy fiber sprouting may be observed. Mossy fiber sprouting has been controversial in its contribution to epileptogenesis in TLE patients, and the mechanisms surrounding the phenomenon have yet to be elucidated. Several studies have reported that mossy fiber sprouting has an almost certain co-existence with reactive astrogliosis within the hippocampus under epileptic conditions. Astrocytes are known to play an important role in the survival and axonal outgrowth of central and peripheral nervous system neurons, pointing to a potential role of astrocytes in TLE and associated cellular alterations. Herein, we review the recent developments surrounding the role of astrocytes in the pathogenic process of TLE and mossy fiber sprouting, with a focus on proposed signaling pathways and cellular mechanisms, histological observations, and clinical correlations in human patients.
Collapse
Affiliation(s)
- Carolyn Twible
- Department of Pathology and Lab Medicine, Western University, London, ON, Canada
| | - Rober Abdo
- Department of Pathology and Lab Medicine, Western University, London, ON, Canada.,Department of Anatomy and Cell Biology, Western University, London, ON, Canada
| | - Qi Zhang
- Department of Pathology and Lab Medicine, Western University, London, ON, Canada.,Department of Pathology and Lab Medicine, London Health Sciences Centre, University Hospital, London, ON, Canada
| |
Collapse
|
47
|
Zhao B, Pan Y, Xu H, Song X. Wnt10a/β-catenin signalling is involved in kindlin-1-mediated astrocyte activation in a chronic construction injury rat model. Eur J Neurosci 2021; 54:7409-7421. [PMID: 34618385 DOI: 10.1111/ejn.15488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 11/28/2022]
Abstract
The activation of spinal astrocytes and release of neuroinflammatory mediators are important events in neuropathic pain (NP) pathogenesis. In this study, we investigated the role of Wnt10a/β-catenin signalling in kindlin-1-mediated astrocyte activation using a chronic constriction injury (CCI) NP rat model. Using kindlin-1 overexpression and knockdown plasmids, we assessed hyperalgesia, changes in spinal astrocyte activation and the release of inflammatory mediators in a NP rat model. We also performed coimmunoprecipitation, Western blotting and real-time polymerase chain reaction (PCR) to characterize the underlying mechanisms of kindlin-1 in astrocyte cultures in vitro. Kindlin-1 was significantly upregulated in CCI rats and promoted hyperalgesia. Moreover, we observed increased kindlin-1, Wnt10a and glial fibrillary acidic protein (GFAP; biomarker for astroglial injury) levels and the release of inflammatory mediators in NP rats (p < 0.05). Inhibiting GFAP in vitro led to decreased kindlin-1 levels, prevented astrocyte activation, decreased Wnt10a level and the release of inflammatory mediators (p < 0.05). Coimmunoprecipitation showed that kindlin-1 can interact with Wnt10a. We showed that kindlin-1-mediated astrocyte activation was associated with Wnt10a/β-catenin signalling and the downstream release of inflammatory mediators in a CCI NP rat model. Our findings provide novel insights into the molecular mechanisms of kindlin-1-mediated astrocyte activation after CCI.
Collapse
Affiliation(s)
- Baisong Zhao
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yongying Pan
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Haiping Xu
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xingrong Song
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
48
|
Mechanistic Insight into the Effects of Curcumin on Neuroinflammation-Driven Chronic Pain. Pharmaceuticals (Basel) 2021; 14:ph14080777. [PMID: 34451874 PMCID: PMC8397941 DOI: 10.3390/ph14080777] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/22/2022] Open
Abstract
Chronic pain is a persistent and unremitting condition that has immense effects on patients' quality of life. Studies have shown that neuroinflammation is associated with the induction and progression of chronic pain. The activation of microglia and astrocytes is the major hallmark of spinal neuroinflammation leading to neuronal excitability in the projection neurons. Excessive activation of microglia and astrocytes is one of the major contributing factors to the exacerbation of pain. However, the current chronic pain treatments, mainly by targeting the neuronal cells, remain ineffective and unable to meet the patients' needs. Curcumin, a natural plant product found in the Curcuma genus, improves chronic pain by diminishing the release of inflammatory mediators from the spinal glia. This review details the role of curcumin in microglia and astrocytes both in vitro and in vivo and how it improves pain. We also describe the mechanism of curcumin by highlighting the major glia-mediated cascades in pain. Moreover, the role of curcumin on inflammasome and epigenetic regulation is discussed. Furthermore, we discuss the strategies used to improve the efficacy of curcumin. This review illustrates that curcumin modulating microglia and astrocytes could assure the treatment of chronic pain by suppressing spinal neuroinflammation.
Collapse
|
49
|
Tanaka M, Shigetomi E, Parajuli B, Nagatomo H, Shinozaki Y, Hirayama Y, Saito K, Kubota Y, Danjo Y, Lee JH, Kim SK, Nabekura J, Koizumi S. Adenosine A 2B receptor down-regulates metabotropic glutamate receptor 5 in astrocytes during postnatal development. Glia 2021; 69:2546-2558. [PMID: 34339538 DOI: 10.1002/glia.24006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 11/07/2022]
Abstract
Metabotropic glutamate receptor 5 (mGluR5) in astrocytes is a key molecule for controlling synapse remodeling. Although mGluR5 is abundant in neonatal astrocytes, its level is gradually down-regulated during development and is almost absent in the adult. However, in several pathological conditions, mGluR5 re-emerges in adult astrocytes and contributes to disease pathogenesis by forming uncontrolled synapses. Thus, controlling mGluR5 expression in astrocyte is critical for several diseases, but the mechanism that regulates mGluR5 expression remains unknown. Here, we show that adenosine triphosphate (ATP)/adenosine-mediated signals down-regulate mGluR5 in astrocytes. First, in situ Ca2+ imaging of astrocytes in acute cerebral slices from post-natal day (P)7-P28 mice showed that Ca2+ responses evoked by (S)-3,5-dihydroxyphenylglycine (DHPG), a mGluR5 agonist, decreased during development, whereas those evoked by ATP or its metabolite, adenosine, increased. Second, ATP and adenosine suppressed expression of the mGluR5 gene, Grm5, in cultured astrocytes. Third, the decrease in the DHPG-evoked Ca2+ responses was associated with down-regulation of Grm5. Interestingly, among several adenosine (P1) receptor and ATP (P2) receptor genes, only the adenosine A2B receptor gene, Adora2b, was up-regulated in the course of development. Indeed, we observed that down-regulation of Grm5 was suppressed in Adora2b knockout astrocytes at P14 and in situ Ca2+ imaging from Adora2b knockout mice indicated that the A2B receptor inhibits mGluR5 expression in astrocytes. Furthermore, deletion of A2B receptor increased the number of excitatory synapse in developmental stage. Taken together, the A2B receptor is critical for down-regulation of mGluR5 in astrocytes, which would contribute to terminate excess synaptogenesis during development.
Collapse
Affiliation(s)
- Masayoshi Tanaka
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Bijay Parajuli
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Hiroaki Nagatomo
- Center for Life Science Research, University of Yamanashi, Yamanashi, Japan
| | - Youichi Shinozaki
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Yuri Hirayama
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Kozo Saito
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Yuto Kubota
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Yosuke Danjo
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| | - Ji Hwan Lee
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Sun Kwang Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, South Korea.,Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.,GLIA Center, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
50
|
Tanaka M, Török N, Tóth F, Szabó Á, Vécsei L. Co-Players in Chronic Pain: Neuroinflammation and the Tryptophan-Kynurenine Metabolic Pathway. Biomedicines 2021; 9:biomedicines9080897. [PMID: 34440101 PMCID: PMC8389666 DOI: 10.3390/biomedicines9080897] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 01/09/2023] Open
Abstract
Chronic pain is an unpleasant sensory and emotional experience that persists or recurs more than three months and may extend beyond the expected time of healing. Recently, nociplastic pain has been introduced as a descriptor of the mechanism of pain, which is due to the disturbance of neural processing without actual or potential tissue damage, appearing to replace a concept of psychogenic pain. An interdisciplinary task force of the International Association for the Study of Pain (IASP) compiled a systematic classification of clinical conditions associated with chronic pain, which was published in 2018 and will officially come into effect in 2022 in the 11th revision of the International Statistical Classification of Diseases and Related Health Problems (ICD-11) by the World Health Organization. ICD-11 offers the option for recording the presence of psychological or social factors in chronic pain; however, cognitive, emotional, and social dimensions in the pathogenesis of chronic pain are missing. Earlier pain disorder was defined as a condition with chronic pain associated with psychological factors, but it was replaced with somatic symptom disorder with predominant pain in the Diagnostic and Statistical Manual of Mental Disorders, 5th Edition (DSM-5) in 2013. Recently clinical nosology is trending toward highlighting neurological pathology of chronic pain, discounting psychological or social factors in the pathogenesis of pain. This review article discusses components of the pain pathway, the component-based mechanisms of pain, central and peripheral sensitization, roles of chronic inflammation, and the involvement of tryptophan-kynurenine pathway metabolites, exploring the participation of psychosocial and behavioral factors in central sensitization of diseases progressing into the development of chronic pain, comorbid diseases that commonly present a symptom of chronic pain, and psychiatric disorders that manifest chronic pain without obvious actual or potential tissue damage.
Collapse
Affiliation(s)
- Masaru Tanaka
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary; (M.T.); (N.T.); (F.T.)
- Interdisciplinary Excellence Centre, Department of Neurology, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary;
| | - Nóra Török
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary; (M.T.); (N.T.); (F.T.)
- Interdisciplinary Excellence Centre, Department of Neurology, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary;
| | - Fanni Tóth
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary; (M.T.); (N.T.); (F.T.)
| | - Ágnes Szabó
- Interdisciplinary Excellence Centre, Department of Neurology, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary;
| | - László Vécsei
- MTA-SZTE, Neuroscience Research Group, Semmelweis u. 6, H-6725 Szeged, Hungary; (M.T.); (N.T.); (F.T.)
- Interdisciplinary Excellence Centre, Department of Neurology, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary;
- Correspondence: ; Tel.: +36-62-545-351
| |
Collapse
|