151
|
Microglial P2Y12 Receptor Regulates Seizure-Induced Neurogenesis and Immature Neuronal Projections. J Neurosci 2019; 39:9453-9464. [PMID: 31597724 DOI: 10.1523/jneurosci.0487-19.2019] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 11/21/2022] Open
Abstract
Seizures are common in humans with various etiologies ranging from congenital aberrations to acute injuries that alter the normal balance of brain excitation and inhibition. A notable consequence of seizures is the induction of aberrant neurogenesis and increased immature neuronal projections. However, regulatory mechanisms governing these features during epilepsy development are not fully understood. Recent studies show that microglia, the brain's resident immune cell, contribute to normal neurogenesis and regulate seizure phenotypes. However, the role of microglia in aberrant neurogenic seizure contexts has not been adequately investigated. To address this question, we coupled the intracerebroventricular kainic acid model with current pharmacogenetic approaches to eliminate microglia in male mice. We show that microglia promote seizure-induced neurogenesis and subsequent seizure-induced immature neuronal projections above and below the pyramidal neurons between the DG and the CA3 regions. Furthermore, we identify microglial P2Y12 receptors (P2Y12R) as a participant in this neurogenic process. Together, our results implicate microglial P2Y12R signaling in epileptogenesis and provide further evidence for targeting microglia in general and microglial P2Y12R in specific to ameliorate proepileptogenic processes.SIGNIFICANCE STATEMENT Epileptogenesis is a process by which the brain develops epilepsy. Several processes have been identified that confer the brain with such epileptic characteristics, including aberrant neurogenesis and increased immature neuronal projections. Understanding the mechanisms that promote such changes is critical in developing therapies to adequately restrain epileptogenesis. We investigated the role of purinergic P2Y12 receptors selectively expressed by microglia, the resident brain immune cells. We report, for the first time, that microglia in general and microglial P2Y12 receptors in specific promote both aberrant neurogenesis and increased immature neuronal projections. These results indicate that microglia enhance epileptogenesis by promoting these processes and suggest that targeting this immune axis could be a novel therapeutic strategy in the clinic.
Collapse
|
152
|
Krishnan A, Bhavanam S, Zochodne D. An Intimate Role for Adult Dorsal Root Ganglia Resident Cycling Cells in the Generation of Local Macrophages and Satellite Glial Cells. J Neuropathol Exp Neurol 2019; 77:929-941. [PMID: 30169768 DOI: 10.1093/jnen/nly072] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The intricate interactions between neurons, glial, and inflammatory cells within peripheral ganglia are physiologically important, but not well explored. Here, we show that adult dorsal root ganglia (DRG) contain populations of self-renewing cells, collectively referred as DRG resident cycling cells (DRCCs), that are active not only in "quiescent" ganglia but also accelerate their turnover in response to distal axotomy. An unexpected proportion of DRCCs were resident macrophages. These cells closely accompanied, and aligned with recycling satellite glial cells (SGCs) that were juxtaposed to sensory neurons and possessed stem cell-like properties. Selective inhibition of colony stimulating factor 1 receptor prevented the local proliferation of macrophages. Interestingly, DRCC turnover was accompanied by apoptosis at later intervals indicating a balanced cellular milieu in the DRGs. These findings identify a complex interactive multicellular DRG microenvironment supporting self-renewal of both macrophages and SGCs and its potential implications in the overall response of adult DRGs to injury.
Collapse
Affiliation(s)
- Anand Krishnan
- Neuroscience and Mental Health Institute.,Division of Neurology, Department of Medicine.,Alberta Diabetes Institute
| | - Sudha Bhavanam
- Division of Laboratory Medicine and Pathology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Douglas Zochodne
- Neuroscience and Mental Health Institute.,Division of Neurology, Department of Medicine.,Alberta Diabetes Institute
| |
Collapse
|
153
|
Abstract
There are growing interests to study the molecular and cellular interactions among immune cells and sensory neurons in the dorsal root ganglia after peripheral nerve injury. Peripheral monocytic cells, including macrophages, are known to respond to a tissue injury through phagocytosis, antigen presentation, and cytokine release. Emerging evidence has implicated the contribution of dorsal root ganglia macrophages to neuropathic pain development and axonal repair in the context of nerve injury. Rapidly phenotyping (or "rapid isolation of") the response of dorsal root ganglia macrophages in the context of nerve injury is desired to identify the unknown neuroimmune factors. Here we demonstrate how our lab rapidly and effectively isolates macrophages from the dorsal root ganglia using an enzyme-free mechanical dissociation protocol. The samples are kept on ice throughout to limit cellular stress. This protocol is far less time consuming compared to the standard enzymatic protocol and has been routinely used for our Fluorescence-activated Cell Sorting analysis.
Collapse
Affiliation(s)
- Xiaobing Yu
- Department of Anesthesia and Perioperative Care, University of California San Francisco;
| | - Jacqueline Leff
- Department of Anesthesia and Perioperative Care, University of California San Francisco
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California San Francisco;
| |
Collapse
|
154
|
Zhang X, Li G. P2Y receptors in neuropathic pain. Pharmacol Biochem Behav 2019; 186:172788. [PMID: 31494119 DOI: 10.1016/j.pbb.2019.172788] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/15/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022]
Abstract
This review summarizes and evaluates the relationship between neuropathic pain and P2Y receptors from inception to 2019. Purinergic receptors have been well studied in recent years using various molecular biological methods. The main research objective of this review is to determine the association of P2Y1, P2Y2, P2Y6, P2Y12 and P2Y13 receptors with neuropathic pain. This review includes the most comprehensive subtypes of P2Y that related to neuropathic pain and the current therapeutic method of neuropathic pain. G protein-coupled P2Y receptors are located on neurons, astrocytes, oligodendrocytes and microglial cells and regulate neurotransmission. Nerve injury is the prime reason for abnormal regulation of P2Y receptor mRNA expression, subsequently, inducing neuropathic pain. Neuropathic pain is a type of chronic pain that is divided into peripheral, central and mixed. Numerous studies demonstrated a positive correlation between the expression level of P2Y receptors and neuropathic pain generation. Also, several reports showed that P2Y short hairpin RNA (shRNA) and P2Y antagonist can be used as an analgesic to relieve neuropathic pain via decreasing P2Y receptor expression level and neural cell activation. However, the transformation process from basic experiments to clinical applications is a long process. Current deficiencies and future research directions are discussed at the end of this review.
Collapse
Affiliation(s)
- Xinge Zhang
- Queen Mary, the University of London, Medical College of Nanchang University, Nanchang 330006, PR China
| | - Guilin Li
- Department of Physiology, Medical College of Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
155
|
Wong SSC, Lee UM, Wang XM, Chung SK, Cheung CW. Role of DLC2 and RhoA/ROCK pathway in formalin induced inflammatory pain in mice. Neurosci Lett 2019; 709:134379. [DOI: 10.1016/j.neulet.2019.134379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/01/2019] [Accepted: 07/15/2019] [Indexed: 12/30/2022]
|
156
|
Peng J, Liu Y, Umpierre AD, Xie M, Tian DS, Richardson JR, Wu LJ. Microglial P2Y12 receptor regulates ventral hippocampal CA1 neuronal excitability and innate fear in mice. Mol Brain 2019; 12:71. [PMID: 31426845 PMCID: PMC6700820 DOI: 10.1186/s13041-019-0492-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 08/13/2019] [Indexed: 12/31/2022] Open
Abstract
The P2Y12 receptor (P2Y12R) is a purinoceptor that is selectively expressed in microglia in the central nervous system. As a signature receptor, microglial P2Y12R mediates process chemotaxis towards ADP/ATP gradients and is engaged in several neurological diseases including chronic pain, stroke and seizures. However, the role of microglial P2Y12R in regulating neuronal excitability and innate behaviors is not fully understood. Here, we generated P2Y12-floxed mice to delete microglial P2Y12R beginning in development (CX3CR1Cre/+:P2Y12f/f; "constitutive knockout"), or after normal development in adult mice (CX3CR1CreER/+:P2Y12f/f; "induced knockout"). Using a battery of behavioral tests, we found that both constitutive and induced P2Y12R knockout mice exhibited innate fear but not learned fear behaviors. After mice were exposed to the elevated plus maze, the c-fos expression in ventral hippocampus CA1 neurons was robustly increased in P2Y12R knockout mice compared with wild-type mice. Consistently, using whole cell patch clamp recording, we found the excitability of ventral hippocampus CA1 neurons was increased in the P2Y12R knockout mice. The results suggest that microglial P2Y12R regulates neuronal excitability and innate fear behaviors in developing and adult mice.
Collapse
Affiliation(s)
- Jiyun Peng
- Institute of Life Science, Nanchang University, Nanchang, 330031, China.,Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Yong Liu
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Anthony D Umpierre
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Manling Xie
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Dai-Shi Tian
- Department of Neurology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jason R Richardson
- Departments of Environmental Health Sciences, Florida International University, Miami, FL, 33199, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA. .,Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA. .,Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
157
|
Chen G, Zhang YQ, Qadri YJ, Serhan CN, Ji RR. Microglia in Pain: Detrimental and Protective Roles in Pathogenesis and Resolution of Pain. Neuron 2019; 100:1292-1311. [PMID: 30571942 DOI: 10.1016/j.neuron.2018.11.009] [Citation(s) in RCA: 481] [Impact Index Per Article: 96.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 10/22/2018] [Accepted: 11/02/2018] [Indexed: 12/18/2022]
Abstract
The previous decade has seen a rapid increase in microglial studies on pain, with a unique focus on microgliosis in the spinal cord after nerve injury and neuropathic pain. Numerous signaling molecules are altered in microglia and contribute to the pathogenesis of pain. Here, we discuss how microglial signaling regulates spinal cord synaptic plasticity in acute and chronic pain conditions with different degrees and variations of microgliosis. We highlight that microglial mediators such as pro- and anti-inflammatory cytokines are powerful neuromodulators that regulate synaptic transmission and pain via neuron-glial interactions. We also reveal an emerging role of microglia in the resolution of pain, in part via specialized pro-resolving mediators including resolvins, protectins, and maresins. We also discuss a possible role of microglia in chronic itch.
Collapse
Affiliation(s)
- Gang Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Yu-Qiu Zhang
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yawar J Qadri
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Hale Transformative Medicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
158
|
Ma G, Yang J, Zhao B, Huang C, Wang R. Correlation between CCL2, CALCA, and CX3CL1 gene polymorphisms and chronic pain after cesarean section in Chinese Han women: A case control study. Medicine (Baltimore) 2019; 98:e16706. [PMID: 31441843 PMCID: PMC6716682 DOI: 10.1097/md.0000000000016706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Postoperative chronic pain is characterized by high incidence, long duration, and complex pathogenesis. The purpose of this study was to investigate the correlation between the single nucleotide polymorphisms of the CCL2 gene rs4586 (g.5974T>C), CALCA rs3781719 (-692T>C), CX3CL1 rs614230 (2342C>T), and the risk of postoperative chronic pain in Chinese Han women. METHODS We analyzed the CCL2 gene rs4586, CALCA rs3781719, CX3CL1 rs614230 single nucleotide polymorphism (SNPs) of 350 Chinese Han women with chronic postsurgical pain (CPSP) 6 months after cesarean section and 350 healthy women without chronic pain (HC). The levels of CCL2, CALCA, and CX3CL1 in serum were detected by enzyme-linked immunosorbent assay (ELISA). RESULTS The CCL2 rs4586 T allele and the CX3CL1 gene rs614230C allele were protective factors for CPSP risk (adjusted OR = 0.766, 95% CI: 0.675-0.865 and OR = 0.336, 95% CI: 0.644-0.835). The CALCA gene rs3781719C allele was a risk factor for CPSP (adjusted OR = 1.273, 95% CI: 1.125-1.424). CCL2 rs4586, CX3CL1 gene rs614230, and CALCA gene rs3781719 locus gene polymorphisms were associated with serum CCL2, CX3CL1, and CALCA protein levels. CONCLUSION Our results support that CCL2 gene rs4586, CALCA rs3781719, CX3CL1 rs614230 gene polymorphism are associated with the occurrence of chronic pain after cesarean section in Chinese Han women.
Collapse
Affiliation(s)
- Guoping Ma
- Department of Anesthesiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai
| | - Jingli Yang
- Department of Anesthesiology, The Affiliated Shanghai Pudong Hospital of Fudan University Shanghai
| | - Bange Zhao
- Department of Anesthesiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai
| | - Chengquan Huang
- Department of Anesthesiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai
| | - Rui Wang
- Department of Anesthesiology, HanDan Central Hospital, Hebei, China
| |
Collapse
|
159
|
Targeting Microglia Using Cx3cr1-Cre Lines: Revisiting the Specificity. eNeuro 2019; 6:ENEURO.0114-19.2019. [PMID: 31201215 PMCID: PMC6620394 DOI: 10.1523/eneuro.0114-19.2019] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/11/2019] [Accepted: 05/28/2019] [Indexed: 12/30/2022] Open
Abstract
Microglia play a pivotal role in maintaining homeostasis of the CNS. There is growing interest in understanding how microglia influence normal brain function and disease progression. Several microglia-specific Cx3cr1-Cre lines have been developed and have become indispensable tools in many investigations of microglial function. However, some recent studies have reported that these lines may have significant leakage into neurons. Other studies have reported that Cx3cr1 is expressed in non-microglial cells, including neurons and astrocytes, in vitro or in vivo either during brain development or upon neurological insult. All these reports raise serious concerns about the trustworthiness of these Cre-lines and whether the conclusions drawn from previous studies are valid. Here, we found that a floxed fluorescent reporter mouse line which has been frequently used to verify Cre lines displayed spontaneous expression of the GFP reporter, independent of Cre recombinase, thus revealing a potential caveat in assessing cre lines. We further confirmed that two Cx3cr1-Cre mouse lines can drive fluorescent reporter expression largely restrictively in microglia. Finally, we clarified that these two mouse lines maintain microglia-specific expression even following excitatory injury. Together, our findings confirm that two previously created Cx3cr1-Cre lines remain as invaluable tools for studying microglia. Moreover, to ensure the quality of data generated and the soundness of conclusions drawn from such data, it should be compulsory to thoroughly examine reporter lines for spontaneous leakiness when labeling cells to study CNS function and diseases.
Collapse
|
160
|
Sun R, Liu Y, Hou B, Lei Y, Bo J, Zhang W, Sun Y, Zhang Y, Zhang Z, Liu Z, Huo W, Mao Y, Ma Z, Gu X. Perioperative activation of spinal α7 nAChR promotes recovery from preoperative stress-induced prolongation of postsurgical pain. Brain Behav Immun 2019; 79:294-308. [PMID: 30797046 DOI: 10.1016/j.bbi.2019.02.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 01/20/2019] [Accepted: 02/20/2019] [Indexed: 12/01/2022] Open
Abstract
Preoperative stress could delay the recovery of postoperative pain and has been reported to be a risk factor for chronic postsurgical pain. As stress could facilitate the proinflammatory activation of microglia, we hypothesized that these cells may play a vital role in the development of preoperative stress-induced pain chronification after surgery. Our experiments were conducted in a rat model that consists of a single prolonged stress (SPS) procedure and plantar incision. A previous SPS exposure induced anxiety-like behaviors, prolonged incision-induced mechanical allodynia, and potentiated the activation of spinal microglia. Based on the results from ex vivo experiments, spinal microglia isolated from SPS-exposed rats secreted more proinflammatory cytokines upon challenge with LPS. Our results also demonstrated that microglia played a more important role than astrocytes in the initiation of SPS-induced prolongation of postsurgical pain. We further explored the therapeutic potential of agonism of α7 nAChR, an emerging anti-inflammatory target, for SPS-induced prolongation of postsurgical pain. Multiple intrathecal (i.t.) injections of PHA-543613 (an α7 nAChR agonist) or PNU-120596 (a type II positive allosteric modulator) during the perioperative period shortened the duration of postsurgical pain after SPS and suppressed SPS-potentiated microglia activation, but their effects were abolished by pretreatment with methyllycaconitine (an α7 nAChR antagonist; i.t.). Based on the results from ex vivo experiments, the anti-inflammatory effects of PHA-543613 and PNU-120596 may have been achieved by the direct modulation of microglia. In conclusion, stress-induced priming of spinal microglia played a key role in the initiation of preoperative stress-induced prolongation of postsurgical pain, and PHA-543613 and PNU-120596 may be potential candidates for preventing pain chronification after surgery.
Collapse
Affiliation(s)
- Rao Sun
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China; Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Liu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Bailing Hou
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Yishan Lei
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Jinhua Bo
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Wei Zhang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Yu'E Sun
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Ying Zhang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Zuoxia Zhang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Zhe Liu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Wenwen Huo
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Yanting Mao
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Zhengliang Ma
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China.
| |
Collapse
|
161
|
Huo W, Liu Y, Lei Y, Zhang Y, Huang Y, Mao Y, Wang C, Sun Y, Zhang W, Ma Z, Gu X. Imbalanced spinal infiltration of Th17/Treg cells contributes to bone cancer pain via promoting microglial activation. Brain Behav Immun 2019; 79:139-151. [PMID: 30685532 DOI: 10.1016/j.bbi.2019.01.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/16/2019] [Accepted: 01/23/2019] [Indexed: 12/30/2022] Open
Abstract
Increasing evidence suggests that T cells participate in the pathology of neuropathic pain, as well as the activation of microglia. However, whether T cells infiltrate into the spinal cord and contribute to the development of bone cancer pain (BCP) remains unknown. Here, we used a mouse model of BCP to show that numbers of T cells infiltrated into the spinal cord after sarcoma cell implantation with increased BCP, and most infiltrating T cells in the spinal cord were CD3+CD4+ T cells. Both Th17 and Treg subpopulations were analyzed by immunofluorescence. Treg cells in the spinal cord were transiently up-regulated, followed by an imbalance towards Th17 afterwards, and elevated IL-17/IL-17A levels were observed in both blood and spinal cord. Meanwhile, TGF-β, IL-6, and IL-23, the factors which regulate Th17/Treg differentiation, increased their expressions during the development of BCP. Additionally, IL-17A receptor (IL-17AR) was found to be expressed on microglia, and the level of IL-17AR increased with activated microglia during BCP development. Furthermore, BCP was ameliorated when IL-17/IL-17A neutralizing antibodies were intrathecally injected, accompanied with inhibited Th17/Treg infiltration and suppressed microglial activation. In conclusion, T cells infiltrated into the spinal cord with the imbalance of Th17/Treg towards Th17 during the development of BCP, which could promote the microglial activation and further increased BCP, while neutralizing IL-17/IL-17A in the spinal cord could ameliorate BCP. Our results suggest that targeting the imbalanced Th17/Treg infiltration in the spinal cord could be a novel strategy for BCP therapy.
Collapse
Affiliation(s)
- Wenwen Huo
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Yue Liu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Yishan Lei
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Ying Zhang
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Yulin Huang
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Yanting Mao
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Chenchen Wang
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Yu'e Sun
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Wei Zhang
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Zhengliang Ma
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing 210008, Jiangsu Province, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing 210008, Jiangsu Province, China.
| |
Collapse
|
162
|
Cowie AM, Dittel BN, Stucky CL. A Novel Sex-Dependent Target for the Treatment of Postoperative Pain: The NLRP3 Inflammasome. Front Neurol 2019; 10:622. [PMID: 31244767 PMCID: PMC6581722 DOI: 10.3389/fneur.2019.00622] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022] Open
Abstract
In recent years the innate immune system has been shown to be crucial for the pathogenesis of postoperative pain. The mediators released by innate immune cells drive the sensitization of sensory neurons following injury by directly acting on peripheral nerve terminals at the injury site. The predominate sensitization signaling pathway involves the proinflammatory cytokine interleukin-1β (IL-1β). IL-1β is known to cause pain by directly acting on sensory neurons. Evidence demonstrates that blockade of IL-1β signaling decreases postoperative pain, however complete blockade of IL-1β signaling increases the risk of infection and decreases effective wound healing. IL-1β requires activation by an inflammasome; inflammasomes are cytosolic receptors of the innate immune system. NOD-like receptor protein 3 (NLRP3) is the predominant inflammasome activated by endogenous molecules that are released by tissue injury such as that which occurs during neuropathic and inflammatory pain disorders. Given that selective inhibition of NLRP3 alleviates postoperative mechanical pain, its selective targeting may be a novel and effective strategy for the treatment of pain that would avoid complications of global IL-1β inhibition. Moreover, NLRP3 is activated in pain in a sex-dependent and cell type-dependent manner. Sex differences in the innate immune system have been shown to drive pain and sensitization through different mechanisms in inflammatory and neuropathic pain disorders, indicating that it is imperative that both sexes are studied when researchers investigate and identify new targets for pain therapeutics. This review will highlight the roles of the innate immune response, the NLRP3 inflammasome, and sex differences in neuropathic and inflammatory pain.
Collapse
Affiliation(s)
- Ashley M Cowie
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bonnie N Dittel
- Blood Research Institute, Versiti, Milwaukee, WI, United States.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
163
|
Long Noncoding RNA Expression Profile in BV2 Microglial Cells Exposed to Lipopolysaccharide. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5387407. [PMID: 31309106 PMCID: PMC6594345 DOI: 10.1155/2019/5387407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/26/2019] [Indexed: 02/07/2023]
Abstract
Neuropathic pain, which is one of the most common forms of chronic pain, seriously increases healthcare costs and impairs patients' quality of life with an incidence of 7–10% worldwide. Microglia cell activation plays a key role in the progression of neuropathic pain. Better understanding of novel molecules modulating microglia cell activation and these underlying functions will extremely benefit the exploration of new treatment. Recent studies suggested long noncoding RNAs may be involved in neuropathic pain. However, its underlying functions and mechanisms in microglia cell activation remain unclear. To identify the differentially expressed lncRNAs and predict their functions in the progression of microglia cell activation, GSE103156 was analyzed using integrated bioinformatics methods. The expression levels of selected lncRNAs and mRNAs were determined by real-time PCR. In the present study, a total of 56 lncRNAs and 298 mRNAs were significantly differentially expressed. The differentially expressed mRNAs were mainly enriched in NF-kappa B signaling pathway, TNF signaling pathway, Toll-like receptor signaling pathway, and NOD-like receptor signaling pathway. The top 10 hub genes were Tnf, Il6, Stat1, Cxcl10, Il1b, Tlr2, Irf1, Ccl2, Irf7, and Ccl5 in the PPI network. Our results showed that Gm8989, Gm8979, and AV051173 may be involved in the progression of microglia cell activation. Taken together, our findings suggest that lots of lncRNAs may be involved in BV2 microglia cell activation in vitro. The findings may provide relevant information for the development of promising targets for the microglial cells activation of neuropathic pain in vivo in the future.
Collapse
|
164
|
Zhao P, Chao W, Li W. FBXW5 reduction alleviates spinal cord injury (SCI) by blocking microglia activity: A mechanism involving p38 and JNK. Biochem Biophys Res Commun 2019; 514:558-564. [DOI: 10.1016/j.bbrc.2019.04.086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 04/12/2019] [Indexed: 12/12/2022]
|
165
|
Guimarães RM, Davoli-Ferreira M, Fonseca MM, Damasceno LEA, Santa-Cecilia FV, Kusuda R, Menezes GB, Cunha FQ, Alves-Filho JC, Cunha TM. Frontline Science: Blood-circulating leukocytes fail to infiltrate the spinal cord parenchyma after spared nerve injury. J Leukoc Biol 2019; 106:541-551. [PMID: 31150565 DOI: 10.1002/jlb.hi1118-458r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 12/12/2022] Open
Abstract
The development of neuropathic pain after peripheral nerve injury involves neuroimmune-glial interactions in the spinal cord. However, whether the development of neuropathic pain depends on the infiltration of peripheral immune cells, such as monocytes, into the spinal cord parenchyma after peripheral nerve damage remains unclear. Here, we used a combination of different techniques such as transgenic reporter mouse (Cx3cr1GFP/+ and Ccr2RFP/+ mice), bone marrow chimeric mice, and parabiosis to investigate this issue in spared nerve injury (SNI) model. Herein, we provided robust evidence that, although microglial cells are activated/proliferate at the dorsal horn of the spinal cord after SNI, peripheral hematopoietic cells (including monocytes) are not able to infiltrate into the spinal cord parenchyma. Furthermore, there was no evidence of CCR2 expression in intrinsic cells of the spinal cord. However, microglial cells activation/proliferation in the spinal cord and mechanical allodynia after SNI were reduced in Ccr2-deficient mice. These results suggest that blood-circulating leukocytes cells are not able to infiltrate the spinal cord parenchyma after distal peripheral nerve injury. Nevertheless, they indicate that CCR2-expressing cells might be indirectly regulating microglia activation/proliferation in the spinal cord after SNI. In conclusion, our study supports that CCR2 inhibition could be explored as an interventional approach to reduce microglia activation and consequently neuropathic pain development after peripheral nerve injury.
Collapse
Affiliation(s)
- Rafaela M Guimarães
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Graduate Program in Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marcela Davoli-Ferreira
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Graduate Program in Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Miriam M Fonseca
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Luis Eduardo A Damasceno
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Graduate Program in Basic and Applied Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Flavia V Santa-Cecilia
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ricardo Kusuda
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Gustavo B Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernando Q Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jose C Alves-Filho
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thiago M Cunha
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
166
|
Abstract
Tramadol-an atypical opioid analgesic-has a unique pharmacokinetic and pharmacodynamic profile, with opioidergic, noradrenergic, and serotonergic actions. Tramadol has long been used as a well-tolerated alternative to other drugs in moderate pain because of its opioidergic and monoaminergic activities. However, cumulative evidence has been gathered over the last few years that supports other likely mechanisms and uses of tramadol in pain management. Tramadol has modulatory effects on several mediators involved in pain signaling, such as voltage-gated sodium ion channels, transient receptor potential V1 channels, glutamate receptors, α2-adrenoceptors, adenosine receptors, and mechanisms involving substance P, calcitonin gene-related peptide, prostaglandin E2, and proinflammatory cytokines. Tramadol also modifies the crosstalk between neuronal and non-neuronal cells in peripheral and central sites. Through these molecular effects, tramadol could modulate peripheral and central neuronal hyperexcitability. Given the broad spectrum of molecular targets, tramadol as a unimodal analgesic relieves a broad range of pain types, such as postoperative, low back, and neuropathic pain and that associated with labor, osteoarthritis, fibromyalgia, and cancer. Moreover, tramadol has anxiolytic, antidepressant, and anti-shivering activities that could improve pain management outcomes. The aim of this review was to address these issues in the context of maladaptive physiological and psychological processes that are associated with different pain types.
Collapse
Affiliation(s)
- Ahmed Barakat
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt.
| |
Collapse
|
167
|
He W, Long T, Pan Q, Zhang S, Zhang Y, Zhang D, Qin G, Chen L, Zhou J. Microglial NLRP3 inflammasome activation mediates IL-1β release and contributes to central sensitization in a recurrent nitroglycerin-induced migraine model. J Neuroinflammation 2019; 16:78. [PMID: 30971286 PMCID: PMC6456991 DOI: 10.1186/s12974-019-1459-7] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 03/22/2019] [Indexed: 12/12/2022] Open
Abstract
Background Central sensitization is an important mechanism of chronic migraine (CM) and is related to the inflammatory response of microglia. The NOD-like receptor protein 3 (NLRP3) inflammasome may regulate the inflammatory process of microglia in several neurological diseases, but its role in CM is largely unknown. Therefore, the aim of this study was to identify the precise role of microglial NLRP3 in CM. Methods An experimental CM mouse model was established by repeated intraperitoneal (i.p) injection with nitroglycerin (NTG). We evaluated the expression levels of NLRP3 and its downstream interleukin (IL)-1β protein in the trigeminal nucleus caudalis (TNC; which is a central area relevant to migraine pain) at different time points. To further examine the effects of the NLRP3 inflammasome pathway on central sensitization of CM, we examined MCC950, an NLRP3 inflammasome-specific inhibitor, and IL-1ra, an IL-1β antagonist, whether altered NTG-induced mechanical hyperalgesia of the periorbital area and hind paw. The effect of MCC950 and IL-1ra on c-Fos, phosphorylated extracellular signal-regulated kinase (p-ERK) and calcitonin gene-related peptide (CGRP) expression in the TNC were also analyzed. The cell localization of NLRP3 and IL-1β in the TNC was evaluated by immunofluorescence staining. Results Repeated NTG administration induced acute and chronic mechanical hyperalgesia and increased expression of NLRP3 and IL-1β. Blockade of NLRP3 or IL-1β reduced NTG-induced hyperalgesia, and this effect was accompanied by a significant inhibition of the NTG-induced increase in p-ERK, c-Fos and CGRP levels in the TNC. Immunofluorescence staining revealed that NLRP3 and IL-1β were mainly expressed in microglia in the TNC, and the IL-1β receptor, IL-1R, was mainly expressed in neurons in the TNC. Conclusions These results indicate that NLRP3 activation in the TNC participates in the microglial-neuronal signal by mediating the inflammatory response. This process contributes to the central sensitization observed in CM. Electronic supplementary material The online version of this article (10.1186/s12974-019-1459-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei He
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, People's Republic of China
| | - Ting Long
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, People's Republic of China
| | - Qi Pan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, People's Republic of China
| | - Shanshan Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, People's Republic of China
| | - Yixin Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, People's Republic of China
| | - Dunke Zhang
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, People's Republic of China
| | - Guangcheng Qin
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, People's Republic of China
| | - Lixue Chen
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, People's Republic of China
| | - Jiying Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st You Yi Road, Yu Zhong District, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
168
|
Feng L, Murugan M, Bosco DB, Liu Y, Peng J, Worrell GA, Wang HL, Ta LE, Richardson JR, Shen Y, Wu LJ. Microglial proliferation and monocyte infiltration contribute to microgliosis following status epilepticus. Glia 2019; 67:1434-1448. [PMID: 31179602 DOI: 10.1002/glia.23616] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022]
Abstract
Microglial activation has been recognized as a major contributor to inflammation of the epileptic brain. Seizures are commonly accompanied by remarkable microgliosis and loss of neurons. In this study, we utilize the CX3CR1GFP/+ CCR2RFP/+ genetic mouse model, in which CX3CR1+ resident microglia and CCR2+ monocytes are labeled with GFP and RFP, respectively. Using a combination of time-lapse two-photon imaging and whole-cell patch clamp recording, we determined the distinct morphological, dynamic, and electrophysiological characteristics of infiltrated monocytes and resident microglia, and the evolution of their behavior at different time points following kainic acid-induced seizures. Seizure activated microglia presented enlarged somas with less ramified processes, whereas, infiltrated monocytes were smaller, highly motile cells that lacked processes. Moreover, resident microglia, but not infiltrated monocytes, proliferate locally in the hippocampus after seizure. Microglial proliferation was dependent on the colony-stimulating factor 1 receptor (CSF-1R) pathway. Pharmacological inhibition of CSF-1R reduced seizure-induced microglial proliferation, which correlated with attenuation of neuronal death without altering acute seizure behaviors. Taken together, we demonstrated that proliferation of activated resident microglia contributes to neuronal death in the hippocampus via CSF-1R after status epilepticus, providing potential therapeutic targets for neuroprotection in epilepsy.
Collapse
Affiliation(s)
- Lijie Feng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.,Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey
| | - Madhuvika Murugan
- Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey.,Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Yong Liu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Jiyun Peng
- Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey.,Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | - Hai-Long Wang
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Lauren E Ta
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Jason R Richardson
- Department of Environmental Health Sciences, Robert Stempel School of Public Health and Social Work, Florida International University, Miami, Florida
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Long-Jun Wu
- Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey.,Department of Neurology, Mayo Clinic, Rochester, Minnesota.,Department of Neuroscience, Mayo Clinic, Jacksonville, Florida.,Department of Immunology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
169
|
Ha JW, You MJ, Park HS, Kim JW, Kwon MS. Differential effect of LPS and paclitaxel on microglial functional phenotypes and circulating cytokines: the possible role of CX3CR1 and IL-4/10 in blocking persistent inflammation. Arch Pharm Res 2019; 42:359-368. [PMID: 30852731 DOI: 10.1007/s12272-019-01137-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/23/2019] [Indexed: 10/27/2022]
Abstract
Neuroinflammation plays a role in cancer chemotherapy-induced chronic pain. Thus far, most studies have focused on neuroinflammation suppression. However, there are limited reports of which factor is involved in the transition from acute inflammation to chronic inflammation, resulting in neuroinflammation and chronic pain. Here, we compared the inflammatory reaction and pain response induced by LPS and paclitaxel. LPS (0.5 mg/kg) or paclitaxel (2 mg/kg/day for 5 days) was administered intraperitoneally to mice, and mechanical allodynia was examined by von Frey test. LPS induced transient mechanical allodynia, whereas paclitaxel induced persistent mechanical allodynia. The CD86/CX3CR1 ratio remained unchanged due to CX3CR1 elevation following LPS injection, whereas the ratio was increased on day 1 after paclitaxel injection. LPS also increased CD45, CCL2, and CCL5 mRNA in the spinal cord and circulating pro- and anti-inflammatory cytokines 1 day after injection; however, the pattern was not consistent. Paclitaxel gradually increased inflammatory cytokines in the spinal cord. CX3CR1 might be involved in blocking the transition from acute pain to persistent pain in the LPS group. In addition, serum IL-4 and IL-10 elevation in the LPS group may be associated with chronic pain prevention. Therefore, targeting CX3CR1, IL-4, and IL-10 might be an alternative therapeutic strategy.
Collapse
Affiliation(s)
- Joong-Won Ha
- Department of Pharmacology, School of Medicine, CHA University, CHA Bio Complex, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.,Department of Orthopedic Surgery, National Health Insurance Service Ilsan Hospital, Ilsan, 10444, Republic of Korea
| | - Min-Jung You
- Department of Pharmacology, School of Medicine, CHA University, CHA Bio Complex, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hyun-Sun Park
- Department of Pharmacology, School of Medicine, CHA University, CHA Bio Complex, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Jong Wan Kim
- Department of Surgery, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, 40 Sukwoo-Dong, Hwaseong-Si, Gyeonggi-do, 14068, Republic of Korea.
| | - Min-Soo Kwon
- Department of Pharmacology, School of Medicine, CHA University, CHA Bio Complex, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
170
|
Green LA, Nebiolo JC, Smith CJ. Microglia exit the CNS in spinal root avulsion. PLoS Biol 2019; 17:e3000159. [PMID: 30794533 PMCID: PMC6402705 DOI: 10.1371/journal.pbio.3000159] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 03/06/2019] [Accepted: 02/06/2019] [Indexed: 12/31/2022] Open
Abstract
Microglia are central nervous system (CNS)-resident cells. Their ability to migrate outside of the CNS, however, is not understood. Using time-lapse imaging in an obstetrical brachial plexus injury (OBPI) model, we show that microglia squeeze through the spinal boundary and emigrate to peripheral spinal roots. Although both macrophages and microglia respond, microglia are the debris-clearing cell. Once outside the CNS, microglia re-enter the spinal cord in an altered state. These peripheral nervous system (PNS)-experienced microglia can travel to distal CNS areas from the injury site, including the brain, with debris. This emigration is balanced by two mechanisms—induced emigration via N-methyl-D-aspartate receptor (NMDA) dependence and restriction via contact-dependent cellular repulsion with macrophages. These discoveries open the possibility that microglia can migrate outside of their textbook-defined regions in disease states. Microglia are normally assumed to be confined to the central nervous system (CNS), but this study shows show that after spinal root injury, microglia can exit the CNS to clear debris. Upon re-entry, the emigrated microglia are altered and can travel to distal areas such as the brain. Cells are precisely organized in specific anatomical domains to ensure normal functioning of the nervous system. One such cell type, microglia, is usually considered to be confined to the central nervous system (CNS). Using time-lapse imaging to capture microglia as they migrate, we show that their characteristic CNS-residency can be altered after spinal root injury. After such injury, the microglia exit the spinal root to the periphery, where they clear debris at the injury site and then carry that debris back into the CNS. In addition, microglia that leave the CNS after spinal root injury become distinct from those that remain within the CNS. This emigration event of microglia after injury is driven by two mechanisms—dependence on glutamatergic signaling that induces their emigration to the injury and interactions with macrophages that prevent their ectopic exit from the spinal cord. Together, these discoveries raise the possibility that microglia could override their CNS-residency in certain disease contexts.
Collapse
Affiliation(s)
- Lauren A Green
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Julia C Nebiolo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Cody J Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| |
Collapse
|
171
|
Amygdaloid administration of tetrapentylammonium attenuates development of pain and anxiety-like behavior following peripheral nerve injury. Pharmacol Rep 2019; 71:54-60. [DOI: 10.1016/j.pharep.2018.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/28/2018] [Accepted: 08/16/2018] [Indexed: 02/07/2023]
|
172
|
Beazley-Long N, Moss CE, Ashby WR, Bestall SM, Almahasneh F, Durrant AM, Benest AV, Blackley Z, Ballmer-Hofer K, Hirashima M, Hulse RP, Bates DO, Donaldson LF. VEGFR2 promotes central endothelial activation and the spread of pain in inflammatory arthritis. Brain Behav Immun 2018; 74:49-67. [PMID: 29548992 PMCID: PMC6302073 DOI: 10.1016/j.bbi.2018.03.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/02/2018] [Accepted: 03/11/2018] [Indexed: 02/01/2023] Open
Abstract
Chronic pain can develop in response to conditions such as inflammatory arthritis. The central mechanisms underlying the development and maintenance of chronic pain in humans are not well elucidated although there is evidence for a role of microglia and astrocytes. However in pre-clinical models of pain, including models of inflammatory arthritis, there is a wealth of evidence indicating roles for pathological glial reactivity within the CNS. In the spinal dorsal horn of rats with painful inflammatory arthritis we found both a significant increase in CD11b+ microglia-like cells and GFAP+ astrocytes associated with blood vessels, and the number of activated blood vessels expressing the adhesion molecule ICAM-1, indicating potential glio-vascular activation. Using pharmacological interventions targeting VEGFR2 in arthritic rats, to inhibit endothelial cell activation, the number of dorsal horn ICAM-1+ blood vessels, CD11b+ microglia and the development of secondary mechanical allodynia, an indicator of central sensitization, were all prevented. Targeting endothelial VEGFR2 by inducible Tie2-specific VEGFR2 knock-out also prevented secondary allodynia in mice and glio-vascular activation in the dorsal horn in response to inflammatory arthritis. Inhibition of VEGFR2 in vitro significantly blocked ICAM-1-dependent monocyte adhesion to brain microvascular endothelial cells, when stimulated with inflammatory mediators TNF-α and VEGF-A165a. Taken together our findings suggest that a novel VEGFR2-mediated spinal cord glio-vascular mechanism may promote peripheral CD11b+ circulating cell transmigration into the CNS parenchyma and contribute to the development of chronic pain in inflammatory arthritis. We hypothesise that preventing this glio-vascular activation and circulating cell translocation into the spinal cord could be a new therapeutic strategy for pain caused by rheumatoid arthritis.
Collapse
Affiliation(s)
- Nicholas Beazley-Long
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK.
| | - Catherine Elizabeth Moss
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK
| | - William Robert Ashby
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK
| | - Samuel Marcus Bestall
- Cancer Biology, School of Medicine, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | - Fatimah Almahasneh
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK
| | - Alexandra Margaret Durrant
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK
| | - Andrew Vaughan Benest
- Cancer Biology, School of Medicine, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | - Zoe Blackley
- Cancer Biology, School of Medicine, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | | | - Masanori Hirashima
- Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Japan
| | - Richard Phillip Hulse
- Cancer Biology, School of Medicine, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | - David Owen Bates
- Cancer Biology, School of Medicine, QMC, University of Nottingham, Nottingham NG7 2UH, UK,COMPARE University of Birmingham and University of Nottingham Midlands, UK
| | - Lucy Frances Donaldson
- Arthritis Research UK Pain Centre & School of Life Sciences, Medical School, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
173
|
Han J, Zhu K, Zhang X, Harris RA. Enforced microglial depletion and repopulation as a promising strategy for the treatment of neurological disorders. Glia 2018; 67:217-231. [PMID: 30378163 PMCID: PMC6635749 DOI: 10.1002/glia.23529] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/17/2018] [Accepted: 08/22/2018] [Indexed: 01/18/2023]
Abstract
Microglia are prominent immune cells in the central nervous system (CNS) and are critical players in both neurological development and homeostasis, and in neurological diseases when dysfunctional. Our previous understanding of the phenotypes and functions of microglia has been greatly extended by a dearth of recent investigations. Distinct genetically defined subsets of microglia are now recognized to perform their own independent functions in specific conditions. The molecular profiling of single microglial cells indicates extensively heterogeneous reactions in different neurological disorders, resulting in multiple potentials for crosstalk with other kinds of CNS cells such as astrocytes and neurons. In settings of neurological diseases it could thus be prudent to establish effective cell‐based therapies by targeting entire microglial networks. Notably, activated microglial depletion through genetic targeting or pharmacological therapies within a suitable time window can stimulate replenishment of the CNS niche with new microglia. Additionally, enforced repopulation through provision of replacement cells also represents a potential means of exchanging dysfunctional with functional microglia. In each setting the newly repopulated microglia might have the potential to resolve ongoing neuroinflammation. In this review, we aim to summarize the most recent knowledge of microglia and to highlight microglial depletion and subsequent repopulation as a promising cell replacement therapy. Although glial cell replacement therapy is still in its infancy and future translational studies are still required, the approach is scientifically sound and provides new optimism for managing the neurotoxicity and neuroinflammation induced by activated microglia.
Collapse
Affiliation(s)
- Jinming Han
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Keying Zhu
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Xing‐Mei Zhang
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Robert A. Harris
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| |
Collapse
|
174
|
Zhang X, Gao Y, Wang Q, Du S, He X, Gu N, Lu Y. Riluzole induces LTD of spinal nociceptive signaling via postsynaptic GluR2 receptors. J Pain Res 2018; 11:2577-2586. [PMID: 30464577 PMCID: PMC6209077 DOI: 10.2147/jpr.s169686] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Riluzole - a major therapeutic medicine for patients with amyotrophic lateral sclerosis - reportedly has anti-nociceptive and anti-allodynic efficacies in neuropathic pain models. However, little is known about its effect on neurotransmission in the spinal superficial dorsal horn (SDH). The present study aims to investigate the effects of riluzole on the synaptic transmission of SDH nociceptive pathways in both physiological and pathological conditions. Materials and methods Spinal nerve ligation was used to produce a neuropathic pain model. Mechanical allodynia behavior was assessed with Von Frey filaments. Riluzole's effects on nociceptive synaptic transmission under both physiological and pathological conditions were examined by patch-clamp recordings in rat SDH neurons. Results The principal findings of the present study are three-fold. First, we affirm that riluzole has a remarkable long-lasting analgesic effect on both in vitro and in vivo pathological pain models. Second, the prolonged inhibitory effects of riluzole on spinal nociceptive signaling are mediated by both presynaptic and postsynaptic mechanisms. Finally, endocytosis of post-synaptic GluR2 contributes to the riluzole-induced long-term depression (LTD) of the spinal nociceptive pathway. Conclusion The present study finds that riluzole induces LTD of nociceptive signaling in the SDH and produces long-lasting anti-allodynia effects in nerve injury-induced neuropathic pain conditions via postsynaptic AMPA receptors associated with the endocytosis of GluR2.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Pain Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China, ;
| | - Yandong Gao
- Department of Anesthesiology, First Hospital of Yulin City, Yulin 719000, China
| | - Qun Wang
- Department of Pain Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China, ;
| | - Shibin Du
- Department of Pain Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China, ;
| | - Xiaolan He
- Department of Pain Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China, ;
| | - Nan Gu
- Department of Pain Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China, ;
| | - Yan Lu
- Department of Pain Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China, ;
| |
Collapse
|
175
|
Huo W, Zhang Y, Liu Y, Lei Y, Sun R, Zhang W, Huang Y, Mao Y, Wang C, Ma Z, Gu X. Dehydrocorydaline attenuates bone cancer pain by shifting microglial M1/M2 polarization toward the M2 phenotype. Mol Pain 2018; 14:1744806918781733. [PMID: 29882480 PMCID: PMC6009085 DOI: 10.1177/1744806918781733] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Bone cancer pain remains a major challenge in patients with primary or metastatic bone cancer due to a lack of understanding the mechanisms. Previous studies have revealed the two distinct functional polarization states of microglia (classically activated M1 and alternatively activated M2) in the spinal cord in nerve injury–induced neuropathic pain. However, whether microglia in the spinal cord polarize to M1 and M2 phenotypes and contribute to the development of bone cancer pain remains unclear. In this study, we used a mouse model with bone cancer to characterize the M1/M2 polarization of microglia in the spinal cord during the development of bone cancer pain, and investigated the antinociceptive effects of dehydrocorydaline, an alkaloidal component isolated from Rhizoma corydalis on bone cancer pain. Our results show that microglia in the spinal cord presented increased M1 polarization and decreased M2 polarization, while overproduction of IL-1β and inhibited expression of IL-10 was detected during bone cancer pain development. Intraperitoneal administration of dehydrocorydaline (10 mg/kg) had significant antinociceptive effects on day 14 after osteosarcoma cell implantation, accompanied by suppressed M1 phenotype and upregulated M2 phenotype of microglia in the spinal cord, while alleviated inflammatory response was observed then. These results suggest that the imbalanced polarization of microglia toward the M1 phenotype in the spinal cord may contribute to the development of bone cancer pain, while dehydrocorydaline helps to attenuate bone cancer pain, with microglial polarization shifting toward the M2 phenotype in the spinal cord.
Collapse
Affiliation(s)
- Wenwen Huo
- 1 Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Ying Zhang
- 1 Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Yue Liu
- 1 Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Yishan Lei
- 1 Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Rao Sun
- 1 Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Wei Zhang
- 1 Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Yulin Huang
- 1 Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Yanting Mao
- 1 Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Chenchen Wang
- 1 Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Zhengliang Ma
- 1 Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| | - Xiaoping Gu
- 1 Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical Department of Nanjing University, Nanjing, China
| |
Collapse
|
176
|
Yang G, Chen L, Gao Z, Wang Y. Implication of microglia activation and CSF-1/CSF-1Rpathway in lumbar disc degeneration-related back pain. Mol Pain 2018; 14:1744806918811238. [PMID: 30326776 PMCID: PMC6243401 DOI: 10.1177/1744806918811238] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Back pain is common and costly. Although lumbar disc degeneration has long been regarded as a major contributor to back pain, how disc degeneration leads to back pain remains unclear. Recent studies observed microglia activation in the spinal cord after disc degeneration, suggesting activated microglia may be involved in discogenic back pain. To determine whether microglia activation participates in disc degeneration-induced back pain, we used a modified disc puncture-induced degeneration-related back pain mouse model to examine the changes in spinal microglia and investigate the potential link between microglia activation and discogenic back pain. In this study, 46 CX3CR1GFP/+ male mice were used in experimental and sham groups. A modified posterolateral retroperitoneal approach was used to expose the L3/L4 disc to induce the needle puncture in the experimental group. Behavioral tests, including grip force and physical function, were used to measure back pain at pre- and postsurgery. The L3 dorsal root ganglions and lumbar spinal cord were obtained at postoperative weeks 1 to 4 followed by immunofluorescence with different antibodies. Micrographs were obtained by confocal microscopy, and morphometric measurements of microglia were analyzed using Imaris. The punctured disc underwent progressive degeneration and mice with disc degeneration showed impaired grip force and physical function. Compared to the control mice, the number of microglia in the lumbar spinal cord was significantly increased in the disc-punctured animals. Moreover, accumulated microglia exhibited larger soma size and lesser ramification in the disc-injured mice. Immunofluorescence demonstrated colony-stimulating factor 1, a cytokine that promotes microglia repopulation, was significantly increased in L3 dorsal root ganglions, whereas its receptor colony-stimulating factor 1 receptor was upregulated on microglia in the disc-injured mice. In summary, lumbar disc puncture caused progressive disc degeneration which induced microglia activation and back pain in mice. Increased colony-stimulating factor 1/colony-stimulating factor 1 receptor signaling is involved in the disc degeneration-induced microglia activation and back pain.
Collapse
Affiliation(s)
- Ge Yang
- 1 Spine Lab, Department of Orthopedic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lunhao Chen
- 1 Spine Lab, Department of Orthopedic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Gao
- 2 Department of Neurobiology, Institute of Neuroscience, National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Wang
- 1 Spine Lab, Department of Orthopedic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
177
|
Yang M, Xu W, Wang Y, Jiang X, Li Y, Yang Y, Yuan H. CD11b-activated Src signal attenuates neuroinflammatory pain by orchestrating inflammatory and anti-inflammatory cytokines in microglia. Mol Pain 2018; 14:1744806918808150. [PMID: 30280656 PMCID: PMC6311569 DOI: 10.1177/1744806918808150] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Neuroinflammation plays an important role in the induction and maintenance of chronic pain. Orchestra of pattern-recognition receptor-induced pro-inflammatory and anti-inflammatory cytokines is critical for inflammation homeostasis. CD11b on macrophages could inhibit toll-like receptor (TLR) activation-induced inflammatory responses. However, the function of CD11b on microglia remains unknown. In the current study, we demonstrated that CD11b-deficient microglia cells produced more inflammatory cytokines, such as interleukin-6 and tumor necrosis factor alpha, while less anti-inflammatory cytokines. Signal transduction assay confirmed that nuclear factor-κB activation was increased in CD11b-deficient microglia cells, which resulted from decreased activation of Src. Inhibition of Src by PP1 increased inflammation in wild-type microglia cells significantly, but not in CD11b-deficient microglia cells. In vivo, CD11b-deficient mice were more susceptible to chronic constrictive injury-induced allodynia and hyperalgesia with significantly more inflammatory cytokines expression. All these results indicated that the regulatory function of CD11b-Src signal pathway on both inflammatory and anti-inflammatory cytokines in microglia cells is a potential target in neuropathic pain treatment.
Collapse
Affiliation(s)
- Mei Yang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Wenyun Xu
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Yiru Wang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Xin Jiang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Yingke Li
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Yajuan Yang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| |
Collapse
|
178
|
Santa-Cecília FV, Ferreira DW, Guimaraes RM, Cecilio NT, Fonseca MM, Lopes AH, Davoli-Ferreira M, Kusuda R, Souza GR, Nachbur U, Alves-Filho JC, Teixeira MM, Zamboni DS, Cunha FQ, Cunha TM. The NOD2 signaling in peripheral macrophages contributes to neuropathic pain development. Pain 2018; 160:102-116. [DOI: 10.1097/j.pain.0000000000001383] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
179
|
Wang C, Gu L, Ruan Y, Geng X, Xu M, Yang N, Yu L, Jiang Y, Zhu C, Yang Y, Zhou Y, Guan X, Luo W, Liu Q, Dong X, Yu G, Lan L, Tang Z. Facilitation of MrgprD by TRP-A1 promotes neuropathic pain. FASEB J 2018; 33:1360-1373. [PMID: 30148678 DOI: 10.1096/fj.201800615rr] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Neuropathic pain remains a therapeutic challenge because of its complicated mechanisms. Mas-related GPCR D (MrgprD) is specifically expressed in small-diameter, nociceptive neurons of dorsal root ganglia (DRGs) and is implicated in pain modulation. However, the underlying mechanism of MrgprD involved in neuropathic pain remains elusive. In this study, we used behavioral experiments and physiologic examination methods to investigate the role of MrgprD in chronic constriction injury (CCI)-induced neuropathic pain. We found that MrgprD is necessary for the initiation of mechanical hypersensitivity and cold allodynia, but not for heat allodynia. Moreover, we demonstrated that transient receptor potential cation channel (TRP)-A1 was the ion channel downstream of MrgprD, and the β-alanine-induced calcium signal was attributed mostly to TRP-A1 function. We further showed that PKA serves as a downstream mediator of β-alanine-activated MrgprD signaling to activate TRP-A1 in DRG neurons and in human embryonic kidney 293 cells, to coexpress MrgprD and TRP-A1 plasmids. Finally, we found that the β-alanine-induced pain behavior was increased, whereas the itching behavior was unchanged in CCI models compared with sham-injured animals. Knockout of TRPA1 also attenuated the β-alanine-induced pain behavior in CCI models. In conclusion, MrgprD is essential in cold allodynia in CCI-induced neuropathic pain through the PKA-TRP-A1 pathway. TRP-A1 facilitates MrgprD to development of neuropathic pain. Our findings reveal a novel mechanism of neuropathic pain formation and highlight MrgprD as a promising drug target for the treatment of neuropathic pain.-Wang, C., Gu, L., Ruan, Y., Geng, X., Xu, M., Yang, N., Yu, L., Jiang, Y., Zhu, C., Yang, Y., Zhou, Y., Guan, X., Luo, W., Liu, Q., Dong, X., Yu, G., Lan, L., Tang, Z. Facilitation of MrgprD by TRP-A1 promotes neuropathic pain.
Collapse
Affiliation(s)
- Changming Wang
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory Cultivation Base for Traditional Chinese Medicine Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Drug Target and Drug for Degenerative Disease of Jiangsu Province, Nanjing University of Chinese Medicine, Nanjing, China
| | - Leying Gu
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory Cultivation Base for Traditional Chinese Medicine Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Drug Target and Drug for Degenerative Disease of Jiangsu Province, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yonglan Ruan
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory Cultivation Base for Traditional Chinese Medicine Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Drug Target and Drug for Degenerative Disease of Jiangsu Province, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiao Geng
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory Cultivation Base for Traditional Chinese Medicine Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Drug Target and Drug for Degenerative Disease of Jiangsu Province, Nanjing University of Chinese Medicine, Nanjing, China
| | - Miao Xu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Niuniu Yang
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Department of Traditional Chinese and Western Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Lei Yu
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yucui Jiang
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chan Zhu
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory Cultivation Base for Traditional Chinese Medicine Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Drug Target and Drug for Degenerative Disease of Jiangsu Province, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Yang
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory Cultivation Base for Traditional Chinese Medicine Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Drug Target and Drug for Degenerative Disease of Jiangsu Province, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuan Zhou
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory Cultivation Base for Traditional Chinese Medicine Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Drug Target and Drug for Degenerative Disease of Jiangsu Province, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaowei Guan
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory Cultivation Base for Traditional Chinese Medicine Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Drug Target and Drug for Degenerative Disease of Jiangsu Province, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenqin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Qin Liu
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, USA.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Center for Sensory Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA; and.,Howard Hughes Medical Institute, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Guang Yu
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory Cultivation Base for Traditional Chinese Medicine Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Drug Target and Drug for Degenerative Disease of Jiangsu Province, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Lan
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zongxiang Tang
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, China.,State Key Laboratory Cultivation Base for Traditional Chinese Medicine Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China.,Key Laboratory of Drug Target and Drug for Degenerative Disease of Jiangsu Province, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
180
|
Laflamme N, Cisbani G, Préfontaine P, Srour Y, Bernier J, St-Pierre MK, Tremblay MÈ, Rivest S. mCSF-Induced Microglial Activation Prevents Myelin Loss and Promotes Its Repair in a Mouse Model of Multiple Sclerosis. Front Cell Neurosci 2018; 12:178. [PMID: 30018535 PMCID: PMC6037698 DOI: 10.3389/fncel.2018.00178] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/07/2018] [Indexed: 11/16/2022] Open
Abstract
A pathological hallmark of multiple sclerosis (MS) is myelin loss in brain white matter accompanied by compromised remyelination. Demyelinated lesions are deeply associated with oligodendrocyte apoptosis and a robust inflammatory response. Although various studies point towards a noxious role of inflammation in MS, others emphasize a positive role for the innate immune cells in disease progression. A cytokine well-known to stimulate cell survival, proliferation and differentiation of myeloid cells, macrophage colony-stimulating factor (mCSF), was administered to mice during a 5 week-long cuprizone diet. Treated mice exhibited reduced myelin loss during the demyelination phase, together with an increased number of microglia and oligodendrocyte precursor cells in lesion sites. Tamoxifen-induced conditional deletion of the mCSF receptor in microglia from cuprizone-fed mice caused aberrant myelin debris accumulation in the corpus callosum and reduced microglial phagocytic response. mCSF therefore plays a key role in stimulating myelin clearance by the brain innate immune cells, which is a prerequisite for proper remyelination and myelin repair processes.
Collapse
Affiliation(s)
- Nathalie Laflamme
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Giulia Cisbani
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Paul Préfontaine
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Younes Srour
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Jordan Bernier
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Marie-Kim St-Pierre
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Marie-Ève Tremblay
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC, Canada
| |
Collapse
|
181
|
Kohno K, Kitano J, Kohro Y, Tozaki-Saitoh H, Inoue K, Tsuda M. Temporal Kinetics of Microgliosis in the Spinal Dorsal Horn after Peripheral Nerve Injury in Rodents. Biol Pharm Bull 2018; 41:1096-1102. [DOI: 10.1248/bpb.b18-00278] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Keita Kohno
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University
| | - Junko Kitano
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University
| | - Yuta Kohro
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University
| | - Hidetoshi Tozaki-Saitoh
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University
| | - Kazuhide Inoue
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University
| | - Makoto Tsuda
- Department of Life Innovation, Graduate School of Pharmaceutical Sciences, Kyushu University
| |
Collapse
|
182
|
Yang M, Chen W, Zhang Y, Yang R, Wang Y, Yuan H. EphrinB/EphB signaling contributes to spinal nociceptive processing via calpain‑1 and caspase‑3. Mol Med Rep 2018; 18:268-278. [PMID: 29749521 PMCID: PMC6059679 DOI: 10.3892/mmr.2018.8996] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 03/26/2018] [Indexed: 12/26/2022] Open
Abstract
Previous studies have indicated that an important subfamily of receptor tyrosine kinases, ephrins and their receptors, are important in pain signaling, particularly in spinal nociceptive processing. In the present study, the role of the ephrin/Eph signaling pathway was confirmed, and it was shown that this signaling was also involved in spinal nociceptive processing through the actions of calpain‑1 and caspase‑3. First, the ephrinB ligands, ephrinB1‑Fc or ephrinB2‑Fc, were introduced into experimental mice via intrathecal injection, and it was found that this injection induced marked time‑ and dose‑dependent mechanical allodynia and thermal hyperalgesia, accompanied by increased levels of calpain‑1 and caspase‑3 in the spinal cord. MDL28170, an inhibitor of calpain‑1, reversed the behavioral effects and ameliorated the increases in calpain‑1 and caspase‑3. Second, it was found that the administration of EphB1 between L5 and L6 in mice inhibited the mechanical allodynia and thermal hyperalgesia induced by chronic constrictive injury. In addition, to demonstrate the cell phenotypes responsible for the increased levels of calpain‑1 and caspase‑3 in the spinal cord following injection with ephrinB2‑Fc, double immunofluorescent labeling was performed, which indicated that calpain‑1 and caspase‑3 were localized in neurons, but not in astrocytes or microglial cells. In conclusion, the present study suggested that ephrinB/EphB signaling contributes to spinal nociceptive processing via the actions of calpain‑1 and caspase‑3.
Collapse
Affiliation(s)
- Mei Yang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Wei Chen
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Yu Zhang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Rui Yang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Yiru Wang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Hongbin Yuan
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
183
|
Zhao X, Eyo UB, Murguan M, Wu LJ. Microglial interactions with the neurovascular system in physiology and pathology. Dev Neurobiol 2018; 78:604-617. [PMID: 29318762 PMCID: PMC5980686 DOI: 10.1002/dneu.22576] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 01/01/2018] [Accepted: 01/06/2018] [Indexed: 01/11/2023]
Abstract
Microglia as immune cells of the central nervous system (CNS) play significant roles not only in pathology but also in physiology, such as shaping of the CNS during development and its proper maintenance in maturity. Emerging research is showing a close association between microglia and the neurovasculature that is critical for brain energy supply. In this review, we summarize the current literature on microglial interaction with the vascular system in the normal and diseased brain. First, we highlight data that indicate interesting potential involvement of microglia in developmental angiogenesis. Then we discuss the evidence for microglial participation with the vasculature in neuropathologies from brain tumors to acute injuries such as ischemic stroke to chronic neurodegenerative conditions. We conclude by suggesting future areas of research to advance the field in light of current technical progress and outstanding questions. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 78: 604-617, 2018.
Collapse
Affiliation(s)
- Xiaoliang Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
| | - Ukpong B. Eyo
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| | - Madhuvika Murguan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854
| |
Collapse
|
184
|
Inquimbert P, Moll M, Latremoliere A, Tong CK, Whang J, Sheehan GF, Smith BM, Korb E, Athié MCP, Babaniyi O, Ghasemlou N, Yanagawa Y, Allis CD, Hof PR, Scholz J. NMDA Receptor Activation Underlies the Loss of Spinal Dorsal Horn Neurons and the Transition to Persistent Pain after Peripheral Nerve Injury. Cell Rep 2018; 23:2678-2689. [PMID: 29847798 PMCID: PMC6276118 DOI: 10.1016/j.celrep.2018.04.107] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/12/2018] [Accepted: 04/26/2018] [Indexed: 12/31/2022] Open
Abstract
Peripheral nerve lesions provoke apoptosis in the dorsal horn of the spinal cord. The cause of cell death, the involvement of neurons, and the relevance for the processing of somatosensory information are controversial. Here, we demonstrate in a mouse model of sciatic nerve injury that glutamate-induced neurodegeneration and loss of γ-aminobutyric acid (GABA)ergic interneurons in the superficial dorsal horn promote the transition from acute to chronic neuropathic pain. Conditional deletion of Grin1, the essential subunit of N-methyl-d-aspartate-type glutamate receptors (NMDARs), protects dorsal horn neurons from excitotoxicity and preserves GABAergic inhibition. Mice deficient in functional NMDARs exhibit normal nociceptive responses and acute pain after nerve injury, but this initial increase in pain sensitivity is reversible. Eliminating NMDARs fully prevents persistent pain-like behavior. Reduced pain in mice lacking proapoptotic Bax confirmed the significance of neurodegeneration. We conclude that NMDAR-mediated neuron death contributes to the development of chronic neuropathic pain.
Collapse
Affiliation(s)
- Perrine Inquimbert
- Centre National de la Recherche Scientifique, UPR 3212, Institut des Neurosciences Cellulaires et Intégratives and Université de Strasbourg, 67084 Strasbourg, France; F.M. Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Moll
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA; Institute of Pharmacology, Heidelberg University, 69120 Heidelberg, Germany
| | - Alban Latremoliere
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Department of Neurology and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Chi-Kun Tong
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - John Whang
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - Gregory F Sheehan
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - Brendan M Smith
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - Erica Korb
- Laboratory of Chromatin Biology and Epigenetics, Rockefeller University, New York, NY 10065, USA
| | - Maria C P Athié
- Department of Structural and Functional Biology, State University of Campinas, Campinas, SP 13083-865, Brazil
| | - Olusegun Babaniyi
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Nader Ghasemlou
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - C David Allis
- Laboratory of Chromatin Biology and Epigenetics, Rockefeller University, New York, NY 10065, USA
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joachim Scholz
- Departments of Anesthesiology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
185
|
Abstract
While spinal microglia play a role in early stages of neuropathic pain etiology, whether they are useful targets to reverse chronic pain at late stages remains unknown. Here, we show that microglia activation in the spinal cord persists for >3 months following nerve injury in rodents, beyond involvement of proinflammatory cytokine and chemokine signalling. In this chronic phase, selective depletion of spinal microglia in male rats with the targeted immunotoxin Mac1-saporin and blockade of brain-derived neurotrophic factor-TrkB signalling with intrathecal TrkB Fc chimera, but not cytokine inhibition, almost completely reversed pain hypersensitivity. By contrast, local spinal administration of Mac1-saporin did not affect nociceptive withdrawal threshold in control animals nor did it affect the strength of afferent-evoked synaptic activity in the spinal dorsal horn in normal conditions. These findings show that the long-term, chronic phase of nerve injury-induced pain hypersensitivity is maintained by microglia-neuron interactions. The findings also effectively separate the central signalling pathways underlying the maintenance phase of the pathology from the early and peripheral inflammatory reactions to injury, pointing to different targets for the treatment of acute vs chronic injury-induced pain.
Collapse
|
186
|
Cortical remodeling after electroacupuncture therapy in peripheral nerve repairing model. Brain Res 2018; 1690:61-73. [PMID: 29654733 DOI: 10.1016/j.brainres.2018.04.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/23/2018] [Accepted: 04/06/2018] [Indexed: 01/23/2023]
Abstract
Electroacupuncture (EA) is an alternative therapy for peripheral nerve injury (PNI). The treatment relies on post-therapeutic effect rather than real-time effect. We utilized fMRI to clarify the resting-state alteration caused by sustained effect of EA on peripheral nerve repairing model. Twenty-four rats were divided equally into three groups: normal group, model group and intervention group. Rats of the model and intervention group underwent sciatic nerve transection and direct anastomosis. EA intervention at ST-36 and GB-30 was conducted continuously for 4 months on the intervention group. Behavioral assessments and fMRI were performed 1 month and 4 months after surgery. Intervention group showed significant improvement on the gait parameters max contact mean intensity (MCMI) and thermal withdrawal latency (TWL) than model group. EA-related sustained effects of amplitude of low frequency fluctuations (ALFF) could be described as a remolding pattern of somatosensory area and sensorimotor integration regions which presented higher ALFF in the contralateral hemisphere and lower in the ipsilateral hemisphere than model group. Interhemispheric functional connectivity (FC) analysis showed a significantly lower FC after EA therapy between the largest significantly different clusters in bilateral somatosensory cortices than the model group 4 months after surgery(p < 0.05). And the model group presented significantly higher FC than the normal group at both two time-points (p < 0.01). The sustained effect of EA on peripheral nerve repairing rats appeared to induce both regional and extensive neuroplasticity in bilateral hemispheres. We proposed that such EA-related effect was a reverse of maladaptive plasticity caused by PNI.
Collapse
|
187
|
Pirt Together with TRPV1 Is Involved in the Regulation of Neuropathic Pain. Neural Plast 2018; 2018:4861491. [PMID: 29808083 PMCID: PMC5902011 DOI: 10.1155/2018/4861491] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/21/2018] [Indexed: 12/11/2022] Open
Abstract
Neuropathic pain is a chronic pain and reduces the life quality of patients substantially. Transient receptor potential vanilloid channel 1 (TRPV1), a nonselective cation channel, has been shown to play a crucial role in neuropathic pain. Although TRPV1 plays an important role in neuropathic pain, the mechanism of how TRPV1 was regulated in neuropathic pain remains unclear. Pirt is a membrane protein and binds to TRPV1 to enhance its activity. It was suggested that Pirt should also be involved in neuropathic pain. In this study, we investigated the role of Pirt in neuropathic pain (CCI model); the results show that mechanical allodynia and thermal hyperalgesia were alleviated in Pirt−/− mice in CCI models. TRPV1 expression was increased by immunofluorescence and real-time PCR experiments. The increase in TRPV1 expression was less in Pirt knockout mice in CCI models. Moreover, the number of capsaicin-responding neurons and the magnitude of evoked calcium response were attenuated in DRG neurons from Pirt−/− mice in CCI models. Finally, we found that the pain behavior attenuated in dysfunction of both Pirt and TRPV1 was much stronger than in dysfunction of Pirt or TRPV1 only in a CCI model in vitro study. Taken together, Pirt together with TRPV1 is involved in CCI-induced neuropathic pain.
Collapse
|
188
|
Skaper SD, Facci L, Zusso M, Giusti P. An Inflammation-Centric View of Neurological Disease: Beyond the Neuron. Front Cell Neurosci 2018; 12:72. [PMID: 29618972 PMCID: PMC5871676 DOI: 10.3389/fncel.2018.00072] [Citation(s) in RCA: 301] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 02/27/2018] [Indexed: 12/13/2022] Open
Abstract
Inflammation is a complex biological response fundamental to how the body deals with injury and infection to eliminate the initial cause of cell injury and effect repair. Unlike a normally beneficial acute inflammatory response, chronic inflammation can lead to tissue damage and ultimately its destruction, and often results from an inappropriate immune response. Inflammation in the nervous system (“neuroinflammation”), especially when prolonged, can be particularly injurious. While inflammation per se may not cause disease, it contributes importantly to disease pathogenesis across both the peripheral (neuropathic pain, fibromyalgia) and central [e.g., Alzheimer disease, Parkinson disease, multiple sclerosis, motor neuron disease, ischemia and traumatic brain injury, depression, and autism spectrum disorder] nervous systems. The existence of extensive lines of communication between the nervous system and immune system represents a fundamental principle underlying neuroinflammation. Immune cell-derived inflammatory molecules are critical for regulation of host responses to inflammation. Although these mediators can originate from various non-neuronal cells, important sources in the above neuropathologies appear to be microglia and mast cells, together with astrocytes and possibly also oligodendrocytes. Understanding neuroinflammation also requires an appreciation that non-neuronal cell—cell interactions, between both glia and mast cells and glia themselves, are an integral part of the inflammation process. Within this context the mast cell occupies a key niche in orchestrating the inflammatory process, from initiation to prolongation. This review will describe the current state of knowledge concerning the biology of neuroinflammation, emphasizing mast cell-glia and glia-glia interactions, then conclude with a consideration of how a cell's endogenous mechanisms might be leveraged to provide a therapeutic strategy to target neuroinflammation.
Collapse
Affiliation(s)
- Stephen D Skaper
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Laura Facci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Pietro Giusti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| |
Collapse
|
189
|
A Longitudinal Mapping Study on Cortical Plasticity of Peripheral Nerve Injury Treated by Direct Anastomosis and Electroacupuncture in Rats. World Neurosurg 2018. [PMID: 29524702 DOI: 10.1016/j.wneu.2018.02.173] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE We used functional magnetic resonance imaging to provide a longitudinal description of cortical plasticity caused by electroacupuncture (EA) of sciatic nerve transection and direct anastomosis in rats. METHODS Sixteen rats in a sciatic nerve transection and direct anastomosis model were randomly divided into intervention and control groups. EA intervention in the position of ST-36, GB-30 was conducted continuously for 4 months in the intervention group. Functional magnetic resonance imaging and gait assessment were performed every month after intervention. RESULTS The somatosensory area was more activated in the first 2 months and then deactivated in the rest 2 months when EA was applied. The pain-related areas had the same activation pattern as the somatosensory area. The limbic/paralimbic areas fluctuated more during the EA intervention, which was not constantly activated or deactivated as previous studies reported. We attributed such changes in somatosensory and pain-related areas to the gradual reduction of sensory afferentation. The alterations in limbic/paralimbic system might be associated with the confrontation between the upregulating effect of paresthesia or pain and the downregulating effect of EA intervention through the autonomic nerve system. The gait analysis showed significantly higher maximum contact mean intensity in the intervention group. CONCLUSIONS The alterations in the brain brought about by the long-term therapeutic effect of EA could be described as a synchronized activation pattern in the somatosensory and pain-related areas and a fluctuating pattern in the limbic/paralimbic system.
Collapse
|
190
|
Dual extra-retinal origins of microglia in the model of retinal microglia repopulation. Cell Discov 2018; 4:9. [PMID: 29507754 PMCID: PMC5827656 DOI: 10.1038/s41421-018-0011-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 01/03/2018] [Indexed: 12/14/2022] Open
Abstract
Elucidating the origin of microglia is crucial for understanding their functions and homeostasis. Previous study has indicated that Nestin-positive progenitor cells differentiate into microglia and replenish the brain after depleting most brain microglia. Microglia have also shown the capacity to repopulate the retina after eliminating all retinal microglia. However, the origin(s) of repopulated retinal microglia is/are unknown. In this study, we aim to investigate the origins of repopulated microglia in the retina. Interestingly, we find that repopulated retinal microglia are not derived from Nestin-positive progenitor cells. Instead, they have two origins: the center-emerging microglia are derived from residual microglia in the optic nerve and the periphery-emerging microglia are derived from macrophages in the ciliary body/iris. Therefore, we have for the first time identified the extra-retinal origins of microglia in the adult mammalian retina by using a model of microglial repopulation, which may shed light on the target exploration of therapeutic interventions for retinal degenerative disorders.
Collapse
|
191
|
Microglia in neuropathic pain: cellular and molecular mechanisms and therapeutic potential. Nat Rev Neurosci 2018; 19:138-152. [DOI: 10.1038/nrn.2018.2] [Citation(s) in RCA: 365] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
192
|
Meacham K, Shepherd A, Mohapatra DP, Haroutounian S. Neuropathic Pain: Central vs. Peripheral Mechanisms. Curr Pain Headache Rep 2018; 21:28. [PMID: 28432601 DOI: 10.1007/s11916-017-0629-5] [Citation(s) in RCA: 259] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW Our goal is to examine the processes-both central and peripheral-that underlie the development of peripherally-induced neuropathic pain (pNP) and to highlight recent evidence for mechanisms contributing to its maintenance. While many pNP conditions are initiated by damage to the peripheral nervous system (PNS), their persistence appears to rely on maladaptive processes within the central nervous system (CNS). The potential existence of an autonomous pain-generating mechanism in the CNS creates significant implications for the development of new neuropathic pain treatments; thus, work towards its resolution is crucial. Here, we seek to identify evidence for PNS and CNS independently generating neuropathic pain signals. RECENT FINDINGS Recent preclinical studies in pNP support and provide key details concerning the role of multiple mechanisms leading to fiber hyperexcitability and sustained electrical discharge to the CNS. In studies regarding central mechanisms, new preclinical evidence includes the mapping of novel inhibitory circuitry and identification of the molecular basis of microglia-neuron crosstalk. Recent clinical evidence demonstrates the essential role of peripheral mechanisms, mostly via studies that block the initially damaged peripheral circuitry. Clinical central mechanism studies use imaging to identify potentially self-sustaining infra-slow CNS oscillatory activity that may be unique to pNP patients. While new preclinical evidence supports and expands upon the key role of central mechanisms in neuropathic pain, clinical evidence for an autonomous central mechanism remains relatively limited. Recent findings from both preclinical and clinical studies recapitulate the critical contribution of peripheral input to maintenance of neuropathic pain. Further clinical investigations on the possibility of standalone central contributions to pNP may be assisted by a reconsideration of the agreed terms or criteria for diagnosing the presence of central sensitization in humans.
Collapse
Affiliation(s)
- Kathleen Meacham
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew Shepherd
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Durga P Mohapatra
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Simon Haroutounian
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
193
|
Abstract
The Cre/loxP system is a widely applied technology for site-specific genetic manipulation in mice. This system allows for deletion of the genes of interest in specific cells, tissues, and whole organism to generate a diversity of conditional knockout mouse strains. Additionally, the Cre/loxP system is useful for development of cell- and tissue-specific reporter mice for lineage tracing, and cell-specific conditional depletion models in mice. Recently, the Cre/loxP technique was extensively adopted to characterize the monocyte/macrophage biology in mouse models. Compared to other relatively homogenous immune cell types such as neutrophils, mast cells, and basophils, monocytes/macrophages represent a highly heterogeneous population which lack specific markers or transcriptional factors. Though great efforts have been made toward establishing macrophage-specific Cre driver mice in the past decade, all of the current available strains are not perfect with regard to their depletion efficiency and targeting specificity for endogenous macrophages. Here we overview the commonly used Cre driver mouse strains targeting macrophages and discuss their major applications and limitations.
Collapse
|
194
|
Macrophage depletion by liposome-encapsulated clodronate suppresses seizures but not hippocampal damage after acute viral encephalitis. Neurobiol Dis 2017; 110:192-205. [PMID: 29208406 DOI: 10.1016/j.nbd.2017.12.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/09/2017] [Accepted: 12/01/2017] [Indexed: 12/12/2022] Open
Abstract
Viral encephalitis is a major risk factor for the development of seizures and epilepsy, but the underlying mechanisms are only poorly understood. Mouse models such as viral encephalitis induced by intracerebral infection with Theiler's virus in C57BL/6 (B6) mice allow advancing our understanding of the immunological and virological aspects of infection-induced seizures and their treatment. Previous studies using the Theiler's virus model in B6 mice have indicated that brain-infiltrating inflammatory macrophages and the cytokines released by these cells are key to the development of acute seizures and hippocampal damage in this model. However, approaches used to prevent or reduce macrophage infiltration were not specific, so contribution of other mechanisms could not be excluded. In the present study, we used a more selective and widely used approach for macrophage depletion, i.e., systemic administration of clodronate liposomes, to study the contribution of macrophage infiltration to development of seizures and hippocampal damage. By this approach, almost complete depletion of monocytic cells was achieved in spleen and blood of Theiler's virus infected B6 mice, which was associated with a 70% decrease in the number of brain infiltrating macrophages as assessed by flow cytometry. Significantly less clodronate liposome-treated mice exhibited seizures than liposome controls (P<0.01), but the development of hippocampal damage was not prevented or reduced. Clodronate liposome treatment did not reduce the increased Iba1 and Mac3 labeling in the hippocampus of infected mice, indicating that activated microglia may contribute to hippocampal damage. The unexpected mismatch between occurrence of seizures and hippocampal damage is thought-provoking and suggests that the mechanisms involved in degeneration of specific populations of hippocampal neurons in encephalitis-induced epilepsy are more complex than previously thought.
Collapse
|
195
|
Kiguchi N, Kobayashi D, Saika F, Matsuzaki S, Kishioka S. Pharmacological Regulation of Neuropathic Pain Driven by Inflammatory Macrophages. Int J Mol Sci 2017; 18:ijms18112296. [PMID: 29104252 PMCID: PMC5713266 DOI: 10.3390/ijms18112296] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 10/27/2017] [Accepted: 10/31/2017] [Indexed: 12/16/2022] Open
Abstract
Neuropathic pain can have a major effect on quality of life but current therapies are often inadequate. Growing evidence suggests that neuropathic pain induced by nerve damage is caused by chronic inflammation. Upon nerve injury, damaged cells secrete pro-inflammatory molecules that activate cells in the surrounding tissue and recruit circulating leukocytes to the site of injury. Among these, the most abundant cell type is macrophages, which produce several key molecules involved in pain enhancement, including cytokines and chemokines. Given their central role in the regulation of peripheral sensitization, macrophage-derived cytokines and chemokines could be useful targets for the development of novel therapeutics. Inhibition of key pro-inflammatory cytokines and chemokines prevents neuroinflammation and neuropathic pain; moreover, recent studies have demonstrated the effectiveness of pharmacological inhibition of inflammatory (M1) macrophages. Nicotinic acetylcholine receptor ligands and T helper type 2 cytokines that reduce M1 macrophages are able to relieve neuropathic pain. Future translational studies in non-human primates will be crucial for determining the regulatory mechanisms underlying neuroinflammation-associated neuropathic pain. In turn, this knowledge will assist in the development of novel pharmacotherapies targeting macrophage-driven neuroinflammation for the treatment of intractable neuropathic pain.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Pharmacology, Wakayama Medical University, Wakayama 641-0012, Japan.
| | - Daichi Kobayashi
- Department of Pharmacology, Wakayama Medical University, Wakayama 641-0012, Japan.
| | - Fumihiro Saika
- Department of Pharmacology, Wakayama Medical University, Wakayama 641-0012, Japan.
| | - Shinsuke Matsuzaki
- Department of Pharmacology, Wakayama Medical University, Wakayama 641-0012, Japan.
| | - Shiroh Kishioka
- Department of Pharmacology, Wakayama Medical University, Wakayama 641-0012, Japan.
| |
Collapse
|
196
|
Attenuation of mechanical pain hypersensitivity by treatment with Peptide5, a connexin-43 mimetic peptide, involves inhibition of NLRP3 inflammasome in nerve-injured mice. Exp Neurol 2017; 300:1-12. [PMID: 29055716 DOI: 10.1016/j.expneurol.2017.10.016] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/22/2017] [Accepted: 10/17/2017] [Indexed: 02/07/2023]
Abstract
Connexin43 (Cx43) hemichannels in spinal cord astrocytes are implicated in the maintenance of neuropathic pain following peripheral nerve injury. Peptide5 is a Cx43 mimetic peptide that blocks hemichannels. In this study, we investigated the effects of spinal delivery of Peptide5 on mechanical pain hypersensitivity in two mouse models of neuropathic pain, peripheral nerve injury and chemotherapy-induced peripheral neuropathy (CIPN). We demonstrated that 10days following a chronic constriction injury (CCI) of the sciatic nerve, Cx43 expression, co-localised predominantly with astrocytes, was increased in the ipsilateral L3-L5 lumbar spinal cord. An intrathecal injection of Peptide5 into nerve-injured mice, on day 10 when pain was well-established, caused significant improvement in mechanical pain hypersensitivity 8h after injection. Peptide5 treatment resulted in significantly reduced Cx43, and microglial and astrocyte activity in the dorsal horn of the spinal cord, as compared to control saline-treated CCI mice. Further in vitro investigations on primary astrocyte cultures showed that 1h pre-treatment with Peptide5 significantly reduced adenosine triphosphate (ATP) release in response to extracellular calcium depletion. Since ATP is a known activator of the NOD-like receptor protein 3 (NLRP3) inflammasome complex, a key mediator of neuroinflammation, we examined the effects of Peptide5 treatment on NLRP3 inflammasome expression. We found that NLRP3, its adaptor apoptosis-associated spec-like protein (ASC) and caspase-1 protein were increased in the ipsilateral spinal cord of CCI mice and reduced to naïve levels following Peptide5 treatment. In the models of oxaliplatin- and paclitaxel-induced peripheral neuropathy, treatment with Peptide5 had no effect on mechanical pain hypersensitivity. Interestingly, in these CIPN models, although spinal Cx43 expression was significantly increased at day 13 following chemotherapy, NLRP3 expression was not altered. These results suggest that the analgesic effect of Peptide5 is specifically achieved by reducing NLRP3 expression. Together, our findings demonstrate that blocking Cx43 hemichannels with Peptide5 after nerve injury attenuates mechanical pain hypersensitivity by specifically targeting the NLRP3 inflammasome in the spinal cord.
Collapse
|
197
|
T-Cell Mediation of Pregnancy Analgesia Affecting Chronic Pain in Mice. J Neurosci 2017; 37:9819-9827. [PMID: 28877966 DOI: 10.1523/jneurosci.2053-17.2017] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 01/02/2023] Open
Abstract
It has been reported consistently that many female chronic pain sufferers have an attenuation of symptoms during pregnancy. Rats display increased pain tolerance during pregnancy due to an increase in opioid receptors in the spinal cord. Past studies did not consider the role of non-neuronal cells, which are now known to play an important role in chronic pain processing. Using an inflammatory (complete Freund's adjuvant) or neuropathic (spared nerve injury) model of persistent pain, we observed that young adult female mice in early pregnancy switch from a microglia-independent to a microglia-dependent pain hypersensitivity mechanism. During late pregnancy, female mice show no evidence of chronic pain whatsoever. This pregnancy-related analgesia is reversible by intrathecal administration of naloxone, suggesting an opioid-mediated mechanism; pharmacological and genetic data suggest the importance of δ-opioid receptors. We also observe that T-cell-deficient (nude and Rag1-null mutant) pregnant mice do not exhibit pregnancy analgesia, which can be rescued with the adoptive transfer of CD4+ or CD8+ T cells from late-pregnant wild-type mice. These results suggest that T cells are a mediator of the opioid analgesia exhibited during pregnancy.SIGNIFICANCE STATEMENT Chronic pain symptoms often subside during pregnancy. This pregnancy-related analgesia has been demonstrated for acute pain in rats. Here, we show that pregnancy analgesia can produce a complete cessation of chronic pain behaviors in mice. We show that the phenomenon is dependent on pregnancy hormones (estrogen and progesterone), δ-opioid receptors, and T cells of the adaptive immune system. These findings add to the recent but growing evidence of sex-specific T-cell involvement in chronic pain processing.
Collapse
|
198
|
Richner M, Vaegter CB. Glucocorticoids - Efficient analgesics against postherpetic neuralgia? Scand J Pain 2017; 16:61-63. [PMID: 28850413 DOI: 10.1016/j.sjpain.2017.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Mette Richner
- Department of Biomedicine, Aarhus University, Denmark.
| | | |
Collapse
|
199
|
Hone AJ, Servent D, McIntosh JM. α9-containing nicotinic acetylcholine receptors and the modulation of pain. Br J Pharmacol 2017; 175:1915-1927. [PMID: 28662295 DOI: 10.1111/bph.13931] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/09/2017] [Accepted: 06/13/2017] [Indexed: 01/01/2023] Open
Abstract
Neuropathic pain is a complex and debilitating syndrome for which there are few effective pharmacological treatments. Opioid-based medications are initially effective for acute pain, but tolerance to their analgesic effects quickly develops, and long-term use often leads to physical dependence and addiction. Furthermore, neuropathic pain is generally resistant to non-steroidal anti-inflammatory drugs. Other classes of medications including antidepressants, antiepileptics and voltage-gated calcium channel inhibitors are only partially effective in most patients, may be associated with significant side effects and have few disease-modifying effects on the underlying pathology. Medications that act through new mechanisms of action, and particularly ones that have disease-modifying properties, would be highly desirable. In the last decade, a potential new target for the treatment of neuropathic pain has emerged: the α9-containing nicotinic acetylcholine receptor (nAChR). Recent studies indicate that antagonists of α9-containing nAChRs are analgesic in animal models of neuropathic pain. These nerve injury models include chronic constriction injury, partial sciatic nerve ligation, streptozotocin-induced diabetic neuropathy and chemotherapeutic-induced neuropathy. This review details the history and state of the field regarding the role that α9-containing nAChRs may play in neuropathic pain. An alternative hypothesis that α-conotoxins exert their therapeutic effect through blocking N-type calcium channels via activation of GABAB receptors is also reviewed. Understanding how antagonists of α9-containing nAChRs exert their therapeutic effects may ultimately result in the development of medications that not only treat but also prevent the development of neuropathic pain states. LINKED ARTICLES This article is part of a themed section on Nicotinic Acetylcholine Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.11/issuetoc.
Collapse
Affiliation(s)
- Arik J Hone
- Department of Biology, University of Utah, Salt Lake City, UT, USA
| | - Denis Servent
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - J Michael McIntosh
- Department of Biology, University of Utah, Salt Lake City, UT, USA.,George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA.,Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
200
|
Lund H, Pieber M, Harris RA. Lessons Learned about Neurodegeneration from Microglia and Monocyte Depletion Studies. Front Aging Neurosci 2017; 9:234. [PMID: 28804456 PMCID: PMC5532389 DOI: 10.3389/fnagi.2017.00234] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/05/2017] [Indexed: 12/20/2022] Open
Abstract
While bone marrow-derived Ly6Chi monocytes can infiltrate the central nervous system (CNS) they are developmentally and functionally distinct from resident microglia. Our understanding of the relative importance of these two populations in the distinct processes of pathogenesis and resolution of inflammation during neurodegenerative disorders was limited by a lack of tools to specifically manipulate each cell type. During recent years, the development of experimental cell-specific depletion models has enabled this issue to be addressed. Herein we compare and contrast the different depletion approaches that have been used, focusing on the respective functionalities of microglia and monocyte-derived macrophages in a range of neurodegenerative disease states, and discuss their prospects for immunotherapy.
Collapse
Affiliation(s)
- Harald Lund
- Department of Clinical Neuroscience, Karolinska Institutet, Centre for Molecular Medicine, Karolinska Hospital at SolnaSolna, Sweden
| | - Melanie Pieber
- Department of Clinical Neuroscience, Karolinska Institutet, Centre for Molecular Medicine, Karolinska Hospital at SolnaSolna, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Karolinska Institutet, Centre for Molecular Medicine, Karolinska Hospital at SolnaSolna, Sweden
| |
Collapse
|