1
|
Zhu Z, Jiang T, Jia X, Wang X, Ren M. Whole-brain long-range connectivity of glutamatergic, GABAergic, parvalbumin-expressing and somatostatin-expressing neurons in mouse somatosensory cortex. Neurosci Res 2025; 217:104912. [PMID: 40436113 DOI: 10.1016/j.neures.2025.104912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 05/17/2025] [Accepted: 05/23/2025] [Indexed: 06/02/2025]
Abstract
Understanding the composition of cortical circuits at the whole-brain scale is crucial. However, the specific ways in which particular neuronal types in the primary somatosensory cortex (SSp) establish connections with upstream and downstream brain regions remain unclear. In this study, we used whole-brain imaging technology with submicron resolution to systematically reveal the long-range connectivity patterns of glutamatergic, GABAergic, parvalbumin-expressing (PV+), and somatostatin-expressing (SOM+) neurons in the SSp. Our results show that while glutamatergic, GABAergic, PV+ , and SOM+ neurons receive similar upstream afferent, specific thalamic subregions showed numerically stronger afferent to GABAergic, PV+ , and SOM+ neurons compared to glutamatergic neurons. Additionally, glutamatergic neurons exhibit a more complex collateral projection pattern in subcortical axonal pathways compared to PV+ neurons. These findings elucidate the long-range connectivity patterns of specific neuronal types in the SSp, offering new insights into the cell-type-specific mechanisms of sensory information processing.
Collapse
Affiliation(s)
- Zhaoxin Zhu
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya, Hainan, 572025, China
| | - Tao Jiang
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215125, China
| | - Xueyan Jia
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215125, China
| | - Xiaojun Wang
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya, Hainan, 572025, China
| | - Miao Ren
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya, Hainan, 572025, China.
| |
Collapse
|
2
|
Wang F, Tian ZC, Ding H, Yang XJ, Wang FD, Ji RX, Xu L, Cao ZX, Ma SB, Zhang M, Cui YT, Cong XY, Chu WG, Li ZZ, Han WJ, Gao YH, Yu YW, Zhao XH, Wang WT, Xie RG, Wu SX, Luo C. A sensory-motor-sensory circuit underlies antinociception ignited by primary motor cortex in mice. Neuron 2025:S0896-6273(25)00246-6. [PMID: 40239652 DOI: 10.1016/j.neuron.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/05/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025]
Abstract
Sensory-motor integration is crucial in the processing of chronic pain. The primary motor cortex (M1) is emerging as a promising target for chronic pain treatment. However, it remains elusive how nociceptive sensory inputs influence M1 activity and how rectifying M1 defects, in turn, regulates pain processing at cellular and network levels. We show that injury/inflammation leads to hypoactivity of M1Glu pyramidal neurons by excitation-inhibition imbalance between the primary somatosensory cortex (S1) and the M1. The impaired M1 output further weakens inputs to excitatory parvalbumin neurons of the lateral hypothalamus (LHPV) and impairs the descending inhibitory system, hence exacerbating spinal nociceptive sensitivity. When rectifying M1 defects with repetitive transcranial magnetic stimulation (rTMS), the imbalance of the S1-M1 microcircuitry can be effectively reversed, which aids in restoring the ability of the M1 to trigger the descending inhibitory system, thereby alleviating nociceptive hypersensitivity. Thus, a sensory-motor-sensory loop is identified for pain-related interactions between the sensory and motor systems and can be potentially exploited for treating chronic pain.
Collapse
Affiliation(s)
- Fei Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China; Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang 712046, China; Shaanxi Province Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Zhi-Cheng Tian
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Hui Ding
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xin-Jiang Yang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China; Department of Rehabilitation and Physical Therapy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Fu-Dong Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ruo-Xin Ji
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Lei Xu
- The Sixteenth Squadron of Fourth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Zi-Xuan Cao
- The Twenty-Second Squadron of Sixth Regiment, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Sui-Bin Ma
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ming Zhang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ya-Ting Cui
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xiang-Yu Cong
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wen-Guang Chu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Zhen-Zhen Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wen-Juan Han
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yong-Heng Gao
- Department of Respiration, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yuan-Wang Yu
- Shaanxi Province Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Xiang-Hui Zhao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wen-Ting Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Rou-Gang Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Sheng-Xi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China; Innovation Research Institute, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
3
|
Park J, Kim YG, Kim T, Baek M. Electrical Stimulation of the M1 Activates Somatostatin Interneurons in the S1: Potential Mechanisms Underlying Pain Suppression. eNeuro 2025; 12:ENEURO.0541-24.2025. [PMID: 40228867 PMCID: PMC12043047 DOI: 10.1523/eneuro.0541-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025] Open
Abstract
Chronic pain affects millions globally, yet no universally effective treatment exists. The primary motor cortex (M1) has been a key target for chronic pain therapies, with electrical stimulation of the M1 (eMCS) showing promise. However, the mechanisms underlying M1-mediated analgesic effects are not fully understood. We investigated the role of the primary somatosensory cortex (S1) in M1-mediated analgesia using a neuropathic pain mouse model. In this model, neuropathic pain is associated with increased spontaneous activity of layer V pyramidal neurons (LV-PNs) in the S1, partly attributed to the reduced activity of somatostatin-expressing inhibitory neurons (SST+ INs), which normally suppress LV-PNs. While manipulation of either LV-PNs or SST+ INs has been shown to alleviate pain, the role of S1 in M1-mediated analgesia has not been identified. Using multichannel silicon probes, we applied eMCS to neuropathic mice and observed significant analgesia. Histological analyses revealed that eMCS activated SST+ INs and suppressed hyperactivity of LV-PNs in the S1, suggesting that eMCS suppresses pain by modulating S1 neuronal circuits, alongside other pain-related regions. Notably, eMCS induced long-lasting analgesia, persisting for at least 2 d poststimulation. These findings implicate S1 as a critical mediator of eMCS-induced analgesia and suggest eMCS as a potential durable therapeutic strategy for chronic pain.
Collapse
Affiliation(s)
- Junhee Park
- Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea
| | - Yong Geon Kim
- Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea
| | - Taehyeon Kim
- Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea
| | - Myungin Baek
- Department of Brain Sciences, DGIST, Daegu 42988, Republic of Korea
| |
Collapse
|
4
|
Gu J, Wang J, Fan H, Wei Y, Li Y, Ma C, Xing K, Wang P, Wu Z, Wu T, Li X, Zhang L, Han Y, Chen T, Qu J, Yan X. Decoding the mechanism of proanthocyanidins in central analgesia: redox regulation and KCNK3 blockade. Exp Mol Med 2025; 57:567-583. [PMID: 40025170 PMCID: PMC11958645 DOI: 10.1038/s12276-025-01412-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 03/04/2025] Open
Abstract
Neuropathic pain causes enduring physical discomfort and emotional distress. Conventional pharmacological treatments often provide restricted relief and may result in undesirable side effects, posing a substantial clinical challenge. Peripheral and spinal redox homeostasis plays an important role in pain processing and perception. However, the roles of oxidative stress and antioxidants in pain and analgesia on the cortical region during chronic pain remains obscure. Here we focus on the ventrolateral orbital cortex (VLO), a brain region associated with pain severity and involved in pain inhibition. Using a spared nerve injury mouse model, we observed the notable reactive oxygen species (ROS)-mediated suppression of the excitability of pyramidal cells (PYRVLO) in the VLO. Nasal application or microinjection of the natural antioxidants proanthocyanidins (PACs) to the VLO specifically increased the activity of PYRVLO and induced a significant analgesic effect. Mechanistically, PACs activate PYRVLO by inhibiting distinct potassium channels in different ways: (1) by scavenging ROS to reduce ROS-sensitive voltage-gated potassium currents and (2) by acting as a channel blocker through direct binding to the cap structure of KCNK3 to inhibit the leak potassium current (Ileak). These results reveal the role of cortical oxidative stress in central hyperalgesia and elucidate the mechanism and potential translational significance of PACs in central analgesia. These findings suggest that the effects of PACs extend beyond their commonly assumed antioxidant or anti-inflammatory effects.
Collapse
Affiliation(s)
- Junxiang Gu
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Jian Wang
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongwei Fan
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Wei
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
- School of Medicine, Northwest University, Xi'an, China
| | - Yan Li
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Chengwen Ma
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Keke Xing
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Pan Wang
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Zhenyu Wu
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China
| | - Teng Wu
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyi Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yunyun Han
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Chen
- Department of Human Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, China.
| | - Jianqiang Qu
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Xianxia Yan
- Department of Neurosurgery, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
5
|
Katagiri T, Nakamura S, Tachibana Y, Nakayama K, Mochizuki A, Dantsuji M, Baba K, Inoue T. Tooth loss-associated neuroplasticity of mastication-related motor cortical neurons. J Oral Biosci 2025; 67:100606. [PMID: 39736390 DOI: 10.1016/j.job.2024.100606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/01/2025]
Abstract
OBJECTIVES The cerebral cortex contains neurons that play a pivotal role in controlling rhythmic masticatory jaw movements. However, the population characteristics of individual cortical neuronal activity during mastication and the impact of tooth loss on these characteristics remain unclear. Thus, in this study, we aimed to determine the activity patterns of mastication-related motor cortical neurons elicited during mastication and examine the effects of tooth extraction on neuronal activity using two-photon Ca2+ imaging in head-restrained awake mice. METHODS GCaMP6f-expressing adeno-associated virus serotype 1 was injected into the left motor cortex (centered 2 mm anterior and 2 mm lateral to the bregma) and electromyography (EMG) electrodes were implanted into the right masseter and digastric muscles of 6-8-week-old C57BL/6j mice. Three weeks after surgery, in vivo two-photon Ca2+ imaging of layer (L) 2/3 neurons and simultaneous EMG recordings were performed during the masticatory sequence. RESULTS Mastication induced a remarkable increase in the power and frequency of Ca2+ responses and correlated with majority of the mastication-related motor cortical L2/3 neuronal activity. These mastication-related changes correlated with the activity of neurons with low baseline activity that occurred before mastication. Extraction of the right upper three molars caused clear neuroplastic changes in the mastication-induced Ca2+ activity of L2/3 neurons. CONCLUSIONS Our in vivo imaging study provides new insights into the neuronal basis of tooth loss-induced cortical neuroplasticity, and suggests a possible therapeutic approach for oral sensorimotor dysfunction.
Collapse
Affiliation(s)
- Takafumi Katagiri
- Department of Prosthodontics, Showa University Graduate School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan; Department of Oral Physiology, Showa University Graduate School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Shiro Nakamura
- Department of Oral Physiology, Showa University Graduate School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan.
| | - Yoshihisa Tachibana
- Division of Physiology and Cell Biology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Hyogo, 650-0017, Japan
| | - Kiyomi Nakayama
- Department of Oral Physiology, Showa University Graduate School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Ayako Mochizuki
- Department of Oral Physiology, Showa University Graduate School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Masanori Dantsuji
- Department of Oral Physiology, Showa University Graduate School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Kazuyoshi Baba
- Department of Prosthodontics, Showa University Graduate School of Dentistry, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Tomio Inoue
- Department of Oral Physiology, Showa University Graduate School of Dentistry, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan; Department of Dental Hygiene, Kyoto Koka Women's College, 38 Nishikyogoku Kadono-cho, Ukyo-ku, Kyoto, 615-0882, Japan
| |
Collapse
|
6
|
Huang JY, Jin YX, Dong WY, Zhao W, Cheng PK, Miao JH, Liu A, Wang D, Li J, Zhang Z, Tao W, Zhu X. Intra-somatosensory cortical circuits mediating pain-induced analgesia. Nat Commun 2025; 16:1859. [PMID: 39984470 PMCID: PMC11845469 DOI: 10.1038/s41467-025-57050-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 02/05/2025] [Indexed: 02/23/2025] Open
Abstract
Pain in one part of the body profoundly diminishes the sensation of pain in other parts of the body in humans. Here, we found that pain-related behaviors in hindpaw are inhibited by noxious stimuli from diverse body regions in mice. Using activity-dependent cell labeling in male FosTRAP2 mice, we captured a neuronal ensemble in the layers 2-4 of secondary somatosensory cortex (S2) that was activated during pain at diverse body regions induced analgesia. Single-cell projection analysis showed that these S2 neurons receive projections from the contralateral S2 and specifically innervate the layer 4 of primary somatosensory cortex (S1). Microendoscopic calcium imaging and chemogenetic manipulation in freely moving mice showed that this S2 → S1 feedforward inhibitory circuit mediates ipsilateral pain-induced analgesia, whereas contralateral S2 innervation of the S2 → S1 circuit mediates contralateral pain-induced analgesia. Our study defines the intra-somatosensory cortical circuits underlying "pain inhibiting pain", expanding the scope of known circuit mechanisms involved in pain relief.
Collapse
Affiliation(s)
- Ji-Ye Huang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Yu-Xin Jin
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Wan-Ying Dong
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Wan Zhao
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of University of Science and Technique of China, Hefei, PR China
| | - Ping-Kai Cheng
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Jun-Hao Miao
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - An Liu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, PR China
| | - Di Wang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Juan Li
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China
| | - Zhi Zhang
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China.
- Department of Biophysics and Neurobiology, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, PR China.
| | - Wenjuan Tao
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, PR China.
- College & Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, PR China.
| | - Xia Zhu
- Department of Anesthesiology and Pain Medicine, The First Affiliated Hospital of USTC, Center for advance interdisciplinary science and biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, PR China.
| |
Collapse
|
7
|
Wei XY, Wang X, Shi GX, Tu JF, Yang JW, Ren MM, Liu JL, Lee CK, Zhou H, Wang ZY, Liu CZ. Acupuncture Modulation of Chronic Neuropathic Pain and Its Association With Brain Functional Properties. THE JOURNAL OF PAIN 2024; 25:104645. [PMID: 39089662 DOI: 10.1016/j.jpain.2024.104645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/22/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Chronic neuropathic pain has been one of the prominent causes of disability, and acupuncture has shown promise in treatment. The present study aimed to characterize acupuncture modulation of chronic neuropathic pain and explore the related functional brain changes. Sixty chronic sciatica patients were divided into acupuncture- or sham acupuncture groups and received 10 sessions of treatment during 4 weeks. The visual analog scale for leg pain, oswestry disability index (ODI), and resting-state functional magnetic resonance images were assessed at baseline and after treatment. Then, fractional amplitudes of low-frequency fluctuations (fALFF) and support vector regression analyses were performed. Compared with sham acupuncture, acupuncture significantly improved symptoms, including visual analog scale for leg pain and ODI. In addition, acupuncture exhibited increased fALFF of the right superior parietal lobule (SPL) and right postcentral gyrus. Furthermore, the actual 4-week ODI values were positively correlated with the support vector regression-predicted values based on the right SPL fALFF and baseline clinical measurements. These results indicate that the spontaneous neural activity of the right SPL and right postcentral gyrus may be involved in the modulation of acupuncture in chronic neuropathic pain. In addition, the spontaneous neural activity of the right SPL might be used as the predictor of response to acupuncture therapy. PERSPECTIVE: This clinical neuroimaging study elucidated the neural basis of acupuncture in chronic sciatica. Neurological indicators and clinical measurements could be used as potential predictors of acupuncture response. This study combines neuroimaging and artificial intelligence techniques to highlight the potential of acupuncture for the treatment of chronic neuropathic pain. TRIAL REGISTRATION NUMBER: Chinese Clinical Trial Registry, ChiCTR2100044585, http://www.chictr.org.cn.
Collapse
Affiliation(s)
- Xiao-Ya Wei
- School of Acupuncture-Moxibustion and Tuina, International Acupuncture and Moxibustion Innovation Institute, Beijing University of Chinese Medicine, Beijing, China
| | - Xu Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Guang-Xia Shi
- School of Acupuncture-Moxibustion and Tuina, International Acupuncture and Moxibustion Innovation Institute, Beijing University of Chinese Medicine, Beijing, China
| | - Jian-Feng Tu
- School of Acupuncture-Moxibustion and Tuina, International Acupuncture and Moxibustion Innovation Institute, Beijing University of Chinese Medicine, Beijing, China
| | - Jing-Wen Yang
- School of Acupuncture-Moxibustion and Tuina, International Acupuncture and Moxibustion Innovation Institute, Beijing University of Chinese Medicine, Beijing, China
| | - Meng-Meng Ren
- Department of Radiology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China
| | - Jun-Lian Liu
- Department of Radiology, Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University, Beijing, China
| | - Chih-Kai Lee
- School of Acupuncture-Moxibustion and Tuina, International Acupuncture and Moxibustion Innovation Institute, Beijing University of Chinese Medicine, Beijing, China
| | - Hang Zhou
- School of Acupuncture-Moxibustion and Tuina, International Acupuncture and Moxibustion Innovation Institute, Beijing University of Chinese Medicine, Beijing, China
| | - Ze-Yi Wang
- School of Acupuncture-Moxibustion and Tuina, International Acupuncture and Moxibustion Innovation Institute, Beijing University of Chinese Medicine, Beijing, China
| | - Cun-Zhi Liu
- School of Acupuncture-Moxibustion and Tuina, International Acupuncture and Moxibustion Innovation Institute, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
8
|
Yang L, Gomm A, Bai P, Ding W, Tanzi RE, Wang C, Shen S, Zhang C. The Effect of Pexidartinib on Neuropathic Pain via Influences on Microglia and Neuroinflammation in Mice. Anesth Analg 2024:00000539-990000000-01022. [PMID: 39475839 PMCID: PMC12041303 DOI: 10.1213/ane.0000000000007239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
BACKGROUND Chronic pain is a debilitating medical condition that lacks effective treatments. Increasing evidence suggests that microglia and neuroinflammation underlie pain pathophysiology, which therefore supports a potential strategy for developing pain therapeutics. Here, our study is testing the hypothesis that the promise of pain amelioration can be achieved using the small-molecule pexidartinib (PLX-3397), a previously food and drug administration (FDA)-approved cancer medicine and a colony-stimulating factor-1 receptor (CSF-1R) inhibitor that display microglia-depleting properties. METHOD We used the previously reported chronic constriction injury (CCI) mouse model, in which PLX-3397 or vehicle was orally administrated to mice daily for 21 days, then applied to the CCI model, followed by PLX-3397 or vehicle administration for an additional 28 days. Additionally, we examined microglia-related neuroinflammation markers using positron emission tomography (PET) neuroimaging and immunofluorescence (IF). RESULTS We showed that PLX-3397 significantly ameliorated pain-related behavioral changes throughout the entire experimental period after CCI (vehicle versus PLX-3397 at day 14, effect size: 2.57, P = .002). Microglia changes were first analyzed by live-animal PET neuroimaging, revealing PLX-3397-associated reduction of microglia by probing receptor-interacting serine/threonine-protein kinase 1 (RIPK1), a protein primarily expressed in microglia, which were further corroborated by postmortem immunohistochemistry (IHC) analysis using antibodies for microglia, including ionized Ca2+ binding adaptor molecule 1 (Iba-1) (somatosensory cortex, hindlimb area; vehicle versus PLX-3397, effect size 3.6, P = .011) and RIPK1 (somatosensory cortex, hindlimb area; vehicle versus PLX-3397, effect size 2.9, P = .023. The expression of both markers decreased in the PLX-3397 group. Furthermore, we found that PLX-3397 led to significant reductions in various proteins, including inducible nitric oxide synthase (iNOS) (somatosensory cortex, hindlimb area; vehicle versus PLX-3397, effect size: 2.3, P = .048), involved in neuroinflammation through IHC. CONCLUSIONS Collectively, our study showed PLX-3397-related efficacy in ameliorating pain linked to the reduction of microglia and neuroinflammation in mice. Furthermore, our research provided new proof-of-concept data supporting the promise of testing PLX-3397 as an analgesic.
Collapse
Affiliation(s)
- Liuyue Yang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129 United States
| | - Ashley Gomm
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, MA 02129
| | - Ping Bai
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, United States
| | - Weihua Ding
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129 United States
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, MA 02129
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, United States
| | - Shiqian Shen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129 United States
| | - Can Zhang
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, 114 16th Street, Charlestown, MA 02129
| |
Collapse
|
9
|
Zhang S, Liu W, Li J, Zhou D. Structural brain characteristics of epilepsy patients with comorbid migraine without aura. Sci Rep 2024; 14:21167. [PMID: 39256409 PMCID: PMC11387786 DOI: 10.1038/s41598-024-71000-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024] Open
Abstract
Migraine is a common bi-directional comorbidity of epilepsy, indicating potential complex interactions between the two conditions. However, no previous studies have used brain morphology analysis to assess possible interactions between epilepsy and migraine. Voxel-based morphometry (VBM), surface-based morphometry (SBM), and structural covariance networks (SCNs) can be used to detect morphological changes with high accuracy. We recruited 30 individuals with epilepsy and comorbid migraine without aura (EM), along with 20 healthy controls (HC) and 30 epilepsy controls (EC) without migraine. We used VBM, SBM, and SCN analysis to compare differences in gray matter volume, cortical thickness, and global level and local level graph theory indexes between the EM, EC, and HC groups to investigate structural brain changes in the EM patients. VBM analysis showed that the EM group had gray matter atrophy in the right temporal pole compared with the HC group (p < 0.001, false discovery rate correction [FDR]). Furthermore, the headache duration in the EM group was negatively correlated with the gray matter volume of the right temporal pole (p < 0.05). SBM analysis showed cortical atrophy in the left insula, left posterior cingulate gyrus, left postcentral gyrus, left middle temporal gyrus, and left fusiform gyrus in the EM compared with the HC group (p < 0.001, family wise error correction). We found a positive correlation between headache frequency and the cortical thickness of the left middle temporal gyrus (p < 0.05). SCN analysis revealed no differences in global parameters between the three groups. The area under the curve (AUC) of the nodal betweenness centrality in the right postcentral gyrus was lower in the EM group compared with the HC group (p < 0.001, FDR correction), and the AUC of the nodal degree in the right fusiform gyrus was lower in the EM group compared with the EC group (p < 0.001, FDR correction). We found clear differences in brain structure in the EM patients compared with the HC group. Accordingly, migraine episodes may influence brain structure in epilepsy patients. Conversely, abnormal brain structure may be an important factor in the development of epilepsy with comorbid migraine without aura. Further studies are needed to investigate the role of brain structure in individuals with epilepsy and comorbid migraine without aura.
Collapse
Affiliation(s)
- Shujiang Zhang
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenyu Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinmei Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China.
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
10
|
Wu X, Yang L, Li Z, Gu C, Jin K, Luo A, Rasheed NF, Fiutak I, Chao K, Chen A, Mao J, Chen Q, Ding W, Shen S. Aging-associated decrease of PGC-1α promotes pain chronification. Aging Cell 2024; 23:e14177. [PMID: 38760908 PMCID: PMC11320346 DOI: 10.1111/acel.14177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 05/20/2024] Open
Abstract
Aging is generally associated with declining somatosensory function, which seems at odds with the high prevalence of chronic pain in older people. This discrepancy is partly related to the high prevalence of degenerative diseases such as osteoarthritis in older people. However, whether aging alters pain processing in the primary somatosensory cortex (S1), and if so, whether it promotes pain chronification is largely unknown. Herein, we report that older mice displayed prolonged nociceptive behavior following nerve injury when compared with mature adult mice. The expression of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) in S1 was decreased in older mice, whereas PGC-1α haploinsufficiency promoted prolonged nociceptive behavior after nerve injury. Both aging and PGC-1α haploinsufficiency led to abnormal S1 neural dynamics, revealed by intravital two-photon calcium imaging. Manipulating S1 neural dynamics affected nociceptive behavior after nerve injury: chemogenetic inhibition of S1 interneurons aggravated nociceptive behavior in naive mice; chemogenetic activation of S1 interneurons alleviated nociceptive behavior in older mice. More interestingly, adeno-associated virus-mediated expression of PGC-1α in S1 interneurons ameliorated aging-associated chronification of nociceptive behavior as well as aging-related S1 neural dynamic changes. Taken together, our results showed that aging-associated decrease of PGC-1α promotes pain chronification, which might be harnessed to alleviate the burden of chronic pain in older individuals.
Collapse
Affiliation(s)
- Xinbo Wu
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Present address:
Shanghai 10th HospitalTongji University School of MedicineShanghaiChina
| | - Liuyue Yang
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Zihua Li
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Chenzheng Gu
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Kaiyan Jin
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Andrew Luo
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | | | | | - Kristina Chao
- Summer Intern ProgramMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Amy Chen
- Summer Intern ProgramMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Jianren Mao
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Qian Chen
- Chinese Academy of SciencesZhongshan Institute for Drug Discovery, Shanghai Institute of Materia MedicaShanghaiChina
| | - Weihua Ding
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Shiqian Shen
- Department of Anesthesia, Critical Care and Pain MedicineMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
11
|
Lou Q, Wei HR, Chen D, Zhang Y, Dong WY, Qun S, Wang D, Luo Y, Zhang Z, Jin Y. A noradrenergic pathway for the induction of pain by sleep loss. Curr Biol 2024; 34:2644-2656.e7. [PMID: 38810638 DOI: 10.1016/j.cub.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/31/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024]
Abstract
An epidemic of sleep loss currently affects modern societies worldwide and is implicated in numerous physiological disorders, including pain sensitization, although few studies have explored the brain pathways affected by active sleep deprivation (ASD; e.g., due to recreation). Here, we describe a neural circuit responsible for pain sensitization in mice treated with 9-h non-stress ASD. Using a combination of advanced neuroscience methods, we found that ASD stimulates noradrenergic inputs from locus coeruleus (LCNA) to glutamatergic neurons of the hindlimb primary somatosensory cortex (S1HLGlu). Moreover, artificial inhibition of this LCNA→S1HLGlu pathway alleviates ASD-induced pain sensitization in mice, while chemogenetic activation of this pathway recapitulates the pain sensitization observed following ASD. Our study thus implicates activation of the LCNA→S1HLGlu pathway in ASD-induced pain sensitization, expanding our fundamental understanding of the multisystem interplay involved in pain processing.
Collapse
Affiliation(s)
- Qianqian Lou
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Hong-Rui Wei
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Danyang Chen
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yuzhuo Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230022, China
| | - Wan-Ying Dong
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Sen Qun
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Di Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China.
| | - Yanli Luo
- Department of Psychological Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| | - Zhi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; The Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Department of Biophysics and Neurobiology, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei 230026, China.
| | - Yan Jin
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Department of Biophysics and Neurobiology, CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
12
|
Ye Q, Li J, Ren WJ, Zhang Y, Wang T, Rubini P, Yin HY, Illes P, Tang Y. Astrocyte activation in hindlimb somatosensory cortex contributes to electroacupuncture analgesia in acid-induced pain. Front Neurol 2024; 15:1348038. [PMID: 38633538 PMCID: PMC11021577 DOI: 10.3389/fneur.2024.1348038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/11/2024] [Indexed: 04/19/2024] Open
Abstract
Background Several studies have confirmed the direct relationship between extracellular acidification and the occurrence of pain. As an effective pain management approach, the mechanism of electroacupuncture (EA) treatment of acidification-induced pain is not fully understood. The purpose of this study was to assess the analgesic effect of EA in this type of pain and to explore the underlying mechanism(s). Methods We used plantar injection of the acidified phosphate-buffered saline (PBS; pH 6.0) to trigger thermal hyperalgesia in male Sprague-Dawley (SD) rats aged 6-8 weeks. The value of thermal withdrawal latency (TWL) was quantified after applying EA stimulation to the ST36 acupoint and/or chemogenetic control of astrocytes in the hindlimb somatosensory cortex. Results Both EA and chemogenetic astrocyte activation suppressed the acid-induced thermal hyperalgesia in the rat paw, whereas inhibition of astrocyte activation did not influence the hyperalgesia. At the same time, EA-induced analgesia was blocked by chemogenetic inhibition of astrocytes. Conclusion The present results suggest that EA-activated astrocytes in the hindlimb somatosensory cortex exert an analgesic effect on acid-induced pain, although these astrocytes might only moderately regulate acid-induced pain in the absence of EA. Our results imply a novel mode of action of astrocytes involved in EA analgesia.
Collapse
Affiliation(s)
- Qing Ye
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen-Jing Ren
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Zhang
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Wang
- Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Patrizia Rubini
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hai-Yan Yin
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peter Illes
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Rudolf Boehm Institute of Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
13
|
Di Cesare Mannelli L, Ghelardini C. Commentary on "Synchronized activity of sensory neurons initiates cortical synchrony in a model of neuropathic pain". Neural Regen Res 2024; 19:728. [PMID: 37843205 PMCID: PMC10664135 DOI: 10.4103/1673-5374.382219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 10/17/2023] Open
Affiliation(s)
- Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health – Neurofarba – Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health – Neurofarba – Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| |
Collapse
|
14
|
Wei HR, Tang L, Yang XL, Chen CM, Li LX, Mao Y, Wang D, Kang F, Qun S, Zhang Z, Cao P, Jin Y. A microglial activation cascade across cortical regions underlies secondary mechanical hypersensitivity to amputation. Cell Rep 2024; 43:113804. [PMID: 38368612 DOI: 10.1016/j.celrep.2024.113804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 08/03/2023] [Accepted: 01/31/2024] [Indexed: 02/20/2024] Open
Abstract
Neural mechanisms underlying amputation-related secondary pain are unclear. Using in vivo two-photon imaging, three-dimensional reconstruction, and fiber photometry recording, we show that a microglial activation cascade from the primary somatosensory cortex of forelimb (S1FL) to the primary somatosensory cortex of hindlimb (S1HL) mediates the disinhibition and subsequent hyperexcitation of glutamatergic neurons in the S1HL (S1HLGlu), which then drives secondary mechanical hypersensitivity development in ipsilateral hindpaws of mice with forepaw amputation. Forepaw amputation induces rapid S1FL microglial activation that further activates S1HL microglia via the CCL2-CCR2 signaling pathway. Increased engulfment of GABAergic presynapses by activated microglia stimulates S1HLGlu neuronal activity, ultimately leading to secondary mechanical hypersensitivity of hindpaws. It is widely believed direct neuronal projection drives interactions between distinct brain regions to prime specific behaviors. Our study reveals microglial interactions spanning different subregions of the somatosensory cortex to drive a maladaptive neuronal response underlying secondary mechanical hypersensitivity at non-injured sites.
Collapse
Affiliation(s)
- Hong-Rui Wei
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Lan Tang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Xin-Lu Yang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Department of Anesthesiology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, China
| | - Chang-Mao Chen
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Le-Xian Li
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yu Mao
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Di Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Fang Kang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Sen Qun
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Zhi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China.
| | - Peng Cao
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China.
| | - Yan Jin
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
15
|
Bak MS, Park H, Yoon H, Chung G, Shin H, Shin S, Kim TW, Lee K, Nägerl UV, Kim SJ, Kim SK. Machine learning-based evaluation of spontaneous pain and analgesics from cellular calcium signals in the mouse primary somatosensory cortex using explainable features. Front Mol Neurosci 2024; 17:1356453. [PMID: 38450042 PMCID: PMC10915002 DOI: 10.3389/fnmol.2024.1356453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction Pain that arises spontaneously is considered more clinically relevant than pain evoked by external stimuli. However, measuring spontaneous pain in animal models in preclinical studies is challenging due to methodological limitations. To address this issue, recently we developed a deep learning (DL) model to assess spontaneous pain using cellular calcium signals of the primary somatosensory cortex (S1) in awake head-fixed mice. However, DL operate like a "black box", where their decision-making process is not transparent and is difficult to understand, which is especially evident when our DL model classifies different states of pain based on cellular calcium signals. In this study, we introduce a novel machine learning (ML) model that utilizes features that were manually extracted from S1 calcium signals, including the dynamic changes in calcium levels and the cell-to-cell activity correlations. Method We focused on observing neural activity patterns in the primary somatosensory cortex (S1) of mice using two-photon calcium imaging after injecting a calcium indicator (GCaMP6s) into the S1 cortex neurons. We extracted features related to the ratio of up and down-regulated cells in calcium activity and the correlation level of activity between cells as input data for the ML model. The ML model was validated using a Leave-One-Subject-Out Cross-Validation approach to distinguish between non-pain, pain, and drug-induced analgesic states. Results and discussion The ML model was designed to classify data into three distinct categories: non-pain, pain, and drug-induced analgesic states. Its versatility was demonstrated by successfully classifying different states across various pain models, including inflammatory and neuropathic pain, as well as confirming its utility in identifying the analgesic effects of drugs like ketoprofen, morphine, and the efficacy of magnolin, a candidate analgesic compound. In conclusion, our ML model surpasses the limitations of previous DL approaches by leveraging manually extracted features. This not only clarifies the decision-making process of the ML model but also yields insights into neuronal activity patterns associated with pain, facilitating preclinical studies of analgesics with higher potential for clinical translation.
Collapse
Affiliation(s)
- Myeong Seong Bak
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Division of AI and Data Analysis, Neurogrin Inc., Seoul, Republic of Korea
| | - Haney Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Division of Preclinical R&D, Neurogrin Inc., Seoul, Republic of Korea
| | - Heera Yoon
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Division of Preclinical R&D, Neurogrin Inc., Seoul, Republic of Korea
| | - Geehoon Chung
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyunjin Shin
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Soonho Shin
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tai Wan Kim
- Department of Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Kyungjoon Lee
- Department of East-West Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - U. Valentin Nägerl
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297 and University of Bordeaux, Bordeaux, France
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sun Kwang Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Department of East-West Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
16
|
Yao D, Chen Y, Chen G. The role of pain modulation pathway and related brain regions in pain. Rev Neurosci 2023; 34:899-914. [PMID: 37288945 DOI: 10.1515/revneuro-2023-0037] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/18/2023] [Indexed: 06/09/2023]
Abstract
Pain is a multifaceted process that encompasses unpleasant sensory and emotional experiences. The essence of the pain process is aversion, or perceived negative emotion. Central sensitization plays a significant role in initiating and perpetuating of chronic pain. Melzack proposed the concept of the "pain matrix", in which brain regions associated with pain form an interconnected network, rather than being controlled by a singular brain region. This review aims to investigate distinct brain regions involved in pain and their interconnections. In addition, it also sheds light on the reciprocal connectivity between the ascending and descending pathways that participate in pain modulation. We review the involvement of various brain areas during pain and focus on understanding the connections among them, which can contribute to a better understanding of pain mechanisms and provide opportunities for further research on therapies for improved pain management.
Collapse
Affiliation(s)
- Dandan Yao
- Department of Anesthesiology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Gang Chen
- Department of Anesthesiology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| |
Collapse
|
17
|
Li J, Tian C, Yuan S, Yin Z, Wei L, Chen F, Dong X, Liu A, Wang Z, Wu T, Tian C, Niu L, Wang L, Wang P, Xie W, Cao F, Shen H. Neuropathic pain following spinal cord hemisection induced by the reorganization in primary somatosensory cortex and regulated by neuronal activity of lateral parabrachial nucleus. CNS Neurosci Ther 2023; 29:3269-3289. [PMID: 37170721 PMCID: PMC10580357 DOI: 10.1111/cns.14258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/04/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023] Open
Abstract
AIMS Neuropathic pain after spinal cord injury (SCI) remains a common and thorny problem, influencing the life quality severely. This study aimed to elucidate the reorganization of the primary sensory cortex (S1) and the regulatory mechanism of the lateral parabrachial nucleus (lPBN) in the presence of allodynia or hyperalgesia after left spinal cord hemisection injury (LHS). METHODS Through behavioral tests, we first identified mechanical allodynia and thermal hyperalgesia following LHS. We then applied two-photon microscopy to observe calcium activity in S1 during mechanical or thermal stimulation and long-term spontaneous calcium activity after LHS. By slice patch clamp recording, the electrophysiological characteristics of neurons in lPBN were explored. Finally, exploiting chemogenetic activation or inhibition of the neurons in lPBN, allodynia or hyperalgesia was regulated. RESULTS The calcium activity in left S1 was increased during mechanical stimulation of right hind limb and thermal stimulation of tail, whereas in right S1 it was increased only with thermal stimulation of tail. The spontaneous calcium activity in right S1 changed more dramatically than that in left S1 after LHS. The lPBN was also activated after LHS, and exploiting chemogenetic activation or inhibition of the neurons in lPBN could induce or alleviate allodynia and hyperalgesia in central neuropathic pain. CONCLUSION The neuronal activity changes in S1 are closely related to limb pain, which has accurate anatomical correspondence. After LHS, the spontaneously increased functional connectivity of calcium transient in left S1 is likely causing the mechanical allodynia in right hind limb and increased neuronal activity in bilateral S1 may induce thermal hyperalgesia in tail. This state of allodynia and hyperalgesia can be regulated by lPBN.
Collapse
Affiliation(s)
- Jing Li
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Chao Tian
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Shiyang Yuan
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Zhenyu Yin
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Liangpeng Wei
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Feng Chen
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Xi Dong
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Aili Liu
- Department of Cellular Biology, School of Basic ScienceTianjin Medical UniversityTianjinChina
| | - Zhenhuan Wang
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Tongrui Wu
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Chunxiao Tian
- School of Biomedical EngineeringTianjin Medical UniversityTianjinChina
| | - Lin Niu
- Department of Cellular Biology, School of Basic ScienceTianjin Medical UniversityTianjinChina
| | - Lei Wang
- Department of PhysiologyZhuhai Campus of Zunyi Medical UniversityZhuhaiChina
| | - Pu Wang
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Wanyu Xie
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Fujiang Cao
- Department of OrthopedicsTianjin Medical University General HospitalTianjinChina
| | - Hui Shen
- Department of Cellular Biology, School of Basic ScienceTianjin Medical UniversityTianjinChina
- Innovation Research Institute of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
18
|
Tian A, Gao H, Wang Z, Li N, Ma J, Guo L, Ma X. Brain structural correlates of postoperative axial pain in degenerative cervical myelopathy patients following posterior cervical decompression surgery: a voxel-based morphometry study. BMC Med Imaging 2023; 23:136. [PMID: 37726693 PMCID: PMC10507911 DOI: 10.1186/s12880-023-01057-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/18/2023] [Indexed: 09/21/2023] Open
Abstract
OBJECTIVE To investigate the brain structural correlates of postoperative axial pain (PAP) in degenerative cervical myelopathy (DCM) following posterior cervical decompression surgery. METHODS Structural images with high-resolution T1 weighting were collected from 62 patients with DCM and analyzed, in addition to 42 age/gender matched subjects who were healthy. Voxel-based morphometry (VBM) was analyzed, grey matter volume (GMV) was computed. One-way ANOVA was performed to reveal the GMV differences among DCM patients with PAP, patients without PAP and healthy controls (HC). Post-hoc analyses were conducted to identify the pair-wise GMV differences among these three groups. Analyses of correlations were conducted to uncover the link between clinical measurements and GMV variations. Last, support vector machine (SVM) was conducted to test the utility of GMV for classifying PAP and nPAP DCM patients. RESULTS Three main findings were observed: [1] Compared to healthy controls, DCM patients showed a significantly lower GMV in the precuneus preoperatively. DCM patients with PAP also exhibited a lower GMV within precuneus than those without; [2] In DCM patients with PAP, the precuneus GMV was inversely related to the postoperative pain intensity; [3] Moreover, successful classification between PAP and nPAP were observed via SVM based on precuneus GMV as features. CONCLUSION In summary, our results indicate that precuneus GMV may be linked to PAP in DCM, and could be employed to forecast the emergence of PAP in DCM patients.
Collapse
Affiliation(s)
- Aixian Tian
- Orthopedic Research Institute, Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, P. R. China
| | - Hongzhi Gao
- Radiology Department, Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, P. R. China
| | - Zhan Wang
- Orthopedic Research Institute, Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, P. R. China
| | - Na Li
- Orthopedic Research Institute, Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, P. R. China
| | - Jianxiong Ma
- Orthopedic Research Institute, Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, P. R. China
| | - Lin Guo
- Radiology Department, Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, P. R. China.
| | - Xinlong Ma
- Orthopedic Research Institute, Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, P. R. China.
| |
Collapse
|
19
|
Guo L, Wu C, Chen C, Zhang B, Wu J, Xie Y, Gui P. Anodal transcranial direct current stimulation over the right primary somatosensory cortex increases cough reflex sensitivity: a pilot randomised controlled crossover trial. ERJ Open Res 2023; 9:00238-2023. [PMID: 37850211 PMCID: PMC10577598 DOI: 10.1183/23120541.00238-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/15/2023] [Indexed: 10/19/2023] Open
Abstract
Background The cough reflex is a protective reflex of the human body. Increases or decreases in cough reflex sensitivity may be related to chronic cough, aspiration pneumonia and other diseases. The right primary somatosensory cortex (RS1) is the main activation centre for the urge to cough. Here, we discuss the effects of transcranial direct current stimulation (tDCS) of RS1 on the cough reflex and urge to cough. In addition, we explored the role of the left dorsolateral prefrontal cortex (lDLPFC) in cough using tDCS. Methods 24 healthy young adults completed this pilot randomised controlled crossover experiment. Each person was tested three times, receiving, in random order, anodal tDCS of RS1 or lDLPFC or sham stimulation. The current intensity was set to 2 mA, the stimulation time was 30 min and the interval between any two stimuli was ≥1 week. After each intervention, the citric acid cough challenge test was used immediately to assess the urge to cough and cough reflex sensitivity. Results The cough reflex thresholds, expressed as LogC2 and LogC5, were significantly reduced after RS1 anodal stimulation compared to sham stimulation, accompanied by increased urge-to-cough sensitivity (urge-to-cough log-log slope 1.19±0.40 point·L·g-1 versus 0.92±0.33 point·L·g-1, p=0.001), but the threshold for the urge to cough did not change significantly. There were no significant changes in the urge to cough and cough reflex sensitivity after tDCS anodal lDLPFC stimulation. Conclusion Anodal tDCS stimulation of the RS1 can increase urge-to-cough sensitivity and reduce cough reflex threshold. The effects of tDCS on cough reflex, as well as the underlying mechanisms driving those effects, should be explored further.
Collapse
Affiliation(s)
- Liya Guo
- Department of Rehabilitation Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chunwei Wu
- Department of Rehabilitation Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chen Chen
- Department of Rehabilitation Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bo Zhang
- Department of Rehabilitation Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jian Wu
- Department of Rehabilitation Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ying Xie
- Department of Rehabilitation Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Y. Xie and P. Gui contributed equally to this article as lead authors and supervised the work
| | - Peijun Gui
- Department of Rehabilitation Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Y. Xie and P. Gui contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
20
|
Bollmann Y, Modol L, Tressard T, Vorobyev A, Dard R, Brustlein S, Sims R, Bendifallah I, Leprince E, de Sars V, Ronzitti E, Baude A, Adesnik H, Picardo MA, Platel JC, Emiliani V, Angulo-Garcia D, Cossart R. Prominent in vivo influence of single interneurons in the developing barrel cortex. Nat Neurosci 2023; 26:1555-1565. [PMID: 37653166 DOI: 10.1038/s41593-023-01405-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 07/13/2023] [Indexed: 09/02/2023]
Abstract
Spontaneous synchronous activity is a hallmark of developing brain circuits and promotes their formation. Ex vivo, synchronous activity was shown to be orchestrated by a sparse population of highly connected GABAergic 'hub' neurons. The recent development of all-optical methods to record and manipulate neuronal activity in vivo now offers the unprecedented opportunity to probe the existence and function of hub cells in vivo. Using calcium imaging, connectivity analysis and holographic optical stimulation, we show that single GABAergic, but not glutamatergic, neurons influence population dynamics in the barrel cortex of non-anaesthetized mouse pups. Single GABAergic cells mainly exert an inhibitory influence on both spontaneous and sensory-evoked population bursts. Their network influence scales with their functional connectivity, with highly connected hub neurons displaying the strongest impact. We propose that hub neurons function in tailoring intrinsic cortical dynamics to external sensory inputs.
Collapse
Affiliation(s)
- Yannick Bollmann
- Aix Marseille Univ, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Laura Modol
- Aix Marseille Univ, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Thomas Tressard
- Aix Marseille Univ, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Artem Vorobyev
- Aix Marseille Univ, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Robin Dard
- Aix Marseille Univ, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Sophie Brustlein
- Aix Marseille Univ, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Ruth Sims
- Wavefront-Engineering Microscopy Group, Photonics Department, Vision Institute, Sorbonne University, INSERM, CNRS, Paris, France
| | - Imane Bendifallah
- Wavefront-Engineering Microscopy Group, Photonics Department, Vision Institute, Sorbonne University, INSERM, CNRS, Paris, France
| | - Erwan Leprince
- Aix Marseille Univ, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Vincent de Sars
- Wavefront-Engineering Microscopy Group, Photonics Department, Vision Institute, Sorbonne University, INSERM, CNRS, Paris, France
| | - Emiliano Ronzitti
- Wavefront-Engineering Microscopy Group, Photonics Department, Vision Institute, Sorbonne University, INSERM, CNRS, Paris, France
| | - Agnès Baude
- Aix Marseille Univ, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Hillel Adesnik
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Michel Aimé Picardo
- Aix Marseille Univ, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Jean-Claude Platel
- Aix Marseille Univ, Inserm, INMED, Turing Center for Living Systems, Marseille, France
| | - Valentina Emiliani
- Wavefront-Engineering Microscopy Group, Photonics Department, Vision Institute, Sorbonne University, INSERM, CNRS, Paris, France
| | - David Angulo-Garcia
- Departamento de Matemáticas y Estadística, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Colombia, Manizales, Colombia
| | - Rosa Cossart
- Aix Marseille Univ, Inserm, INMED, Turing Center for Living Systems, Marseille, France.
| |
Collapse
|
21
|
Pegado R, Silva-Filho E, Micussi MTABC. Letter to the Editor Regarding 'Neuromodulation for Management of Chronic Pelvic Pain: A Comprehensive Review'. Pain Ther 2023; 12:1095-1097. [PMID: 37148446 PMCID: PMC10289954 DOI: 10.1007/s40122-023-00517-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/06/2023] [Indexed: 05/08/2023] Open
Affiliation(s)
- Rodrigo Pegado
- Graduate Program in Physical Therapy, Federal University of Rio Grande Do Norte, Natal, Brazil
- Graduate Program in Health Science, Federal University of Rio Grande Do Norte, Natal, Brazil
| | - Edson Silva-Filho
- Graduate Program in Health Science, Federal University of Rio Grande Do Norte, Natal, Brazil.
| | | |
Collapse
|
22
|
Ding W, Yang L, Chen Q, Hu K, Liu Y, Bao E, Wang C, Mao J, Shen S. Foramen lacerum impingement of trigeminal nerve root as a rodent model for trigeminal neuralgia. JCI Insight 2023; 8:e168046. [PMID: 37159265 PMCID: PMC10393239 DOI: 10.1172/jci.insight.168046] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/03/2023] [Indexed: 05/10/2023] Open
Abstract
Trigeminal neuralgia (TN) is a classic neuralgic pain condition with distinct clinical characteristics. Modeling TN in rodents is challenging. Recently, we found that a foramen in the rodent skull base, the foramen lacerum, provides direct access to the trigeminal nerve root. Using this access, we developed a foramen lacerum impingement of trigeminal nerve root (FLIT) model and observed distinct pain-like behaviors in rodents, including paroxysmal asymmetric facial grimaces, head tilt when eating, avoidance of solid chow, and lack of wood chewing. The FLIT model recapitulated key clinical features of TN, including lancinating pain-like behavior and dental pain-like behavior. Importantly, when compared with a trigeminal neuropathic pain model (infraorbital nerve chronic constriction injury [IoN-CCI]), the FLIT model was associated with significantly higher numbers of c-Fos-positive cells in the primary somatosensory cortex (S1), unraveling robust cortical activation in the FLIT model. On intravital 2-photon calcium imaging, synchronized S1 neural dynamics were present in the FLIT but not the IoN-CCI model, revealing differential implication of cortical activation in different pain models. Taken together, our results indicate that FLIT is a clinically relevant rodent model of TN that could facilitate pain research and therapeutics development.
Collapse
Affiliation(s)
- Weihua Ding
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Liuyue Yang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Qian Chen
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Kun Hu
- Department of Pathology, Tuft University School of Medicine, Boston, Massachusetts, USA
| | - Yan Liu
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eric Bao
- Brooks School, North Andover, Massachusetts, USA
| | - Changning Wang
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jianren Mao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shiqian Shen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Hashimoto A, Kawamura N, Tarusawa E, Takeda I, Aoyama Y, Ohno N, Inoue M, Kagamiuchi M, Kato D, Matsumoto M, Hasegawa Y, Nabekura J, Schaefer A, Moorhouse AJ, Yagi T, Wake H. Microglia enable cross-modal plasticity by removing inhibitory synapses. Cell Rep 2023; 42:112383. [PMID: 37086724 DOI: 10.1016/j.celrep.2023.112383] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/13/2023] [Accepted: 03/28/2023] [Indexed: 04/24/2023] Open
Abstract
Cross-modal plasticity is the repurposing of brain regions associated with deprived sensory inputs to improve the capacity of other sensory modalities. The functional mechanisms of cross-modal plasticity can indicate how the brain recovers from various forms of injury and how different sensory modalities are integrated. Here, we demonstrate that rewiring of the microglia-mediated local circuit synapse is crucial for cross-modal plasticity induced by visual deprivation (monocular deprivation [MD]). MD relieves the usual inhibition of functional connectivity between the somatosensory cortex and secondary lateral visual cortex (V2L). This results in enhanced excitatory responses in V2L neurons during whisker stimulation and a greater capacity for vibrissae sensory discrimination. The enhanced cross-modal response is mediated by selective removal of inhibitory synapse terminals on pyramidal neurons by the microglia in the V2L via matrix metalloproteinase 9 signaling. Our results provide insights into how cortical circuits integrate different inputs to functionally compensate for neuronal damage.
Collapse
Affiliation(s)
- Akari Hashimoto
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Nanami Kawamura
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Etsuko Tarusawa
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Ikuko Takeda
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yuki Aoyama
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, Jichi Medical University, Shimotsuke 329-0498, Japan; Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Mio Inoue
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Mai Kagamiuchi
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Daisuke Kato
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Mami Matsumoto
- Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Yoshihiro Hasegawa
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; Department of Physiological Sciences, Graduate University for Advanced Studies, SOKENDAI, Hayama 240-0193, Japan
| | - Anne Schaefer
- Center for Glial Biology, Department of Neuroscience and Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029-6574, USA; Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
| | - Andrew J Moorhouse
- School of Biomedical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Takeshi Yagi
- KOKORO-Biology Group, Laboratories for Integrated Biology, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Division of Multicellular Circuit Dynamics, National Institute for Physiological Sciences, Okazaki 444-8585, Japan; Center for Optical Scattering Image Science, Kobe University, Kobe 657-8501, Japan; Department of Physiological Sciences, Graduate University for Advanced Studies, SOKENDAI, Hayama 240-0193, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan.
| |
Collapse
|
24
|
Ziegler K, Folkard R, Gonzalez AJ, Burghardt J, Antharvedi-Goda S, Martin-Cortecero J, Isaías-Camacho E, Kaushalya S, Tan LL, Kuner T, Acuna C, Kuner R, Mease RA, Groh A. Primary somatosensory cortex bidirectionally modulates sensory gain and nociceptive behavior in a layer-specific manner. Nat Commun 2023; 14:2999. [PMID: 37225702 DOI: 10.1038/s41467-023-38798-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 05/16/2023] [Indexed: 05/26/2023] Open
Abstract
The primary somatosensory cortex (S1) is a hub for body sensation of both innocuous and noxious signals, yet its role in somatosensation versus pain is debated. Despite known contributions of S1 to sensory gain modulation, its causal involvement in subjective sensory experiences remains elusive. Here, in mouse S1, we reveal the involvement of cortical output neurons in layers 5 (L5) and 6 (L6) in the perception of innocuous and noxious somatosensory signals. We find that L6 activation can drive aversive hypersensitivity and spontaneous nocifensive behavior. Linking behavior to neuronal mechanisms, we find that L6 enhances thalamic somatosensory responses, and in parallel, strongly suppresses L5 neurons. Directly suppressing L5 reproduced the pronociceptive phenotype induced by L6 activation, suggesting an anti-nociceptive function for L5 output. Indeed, L5 activation reduced sensory sensitivity and reversed inflammatory allodynia. Together, these findings reveal a layer-specific and bidirectional role for S1 in modulating subjective sensory experiences.
Collapse
Affiliation(s)
- Katharina Ziegler
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Ross Folkard
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Antonio J Gonzalez
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Jan Burghardt
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Sailaja Antharvedi-Goda
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Jesus Martin-Cortecero
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Emilio Isaías-Camacho
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Sanjeev Kaushalya
- Department of Molecular Pharmacology, Institute for Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Linette Liqi Tan
- Department of Molecular Pharmacology, Institute for Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Thomas Kuner
- Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Claudio Acuna
- Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Rohini Kuner
- Department of Molecular Pharmacology, Institute for Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Rebecca Audrey Mease
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany.
| | - Alexander Groh
- Medical Biophysics, Institute for Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
25
|
Chen P, Huang NY, Pang B, Ye ZJ, Luo RX, Liu C, Gong Q, Wang C, Wang L. Proteomic and metabolomic approaches elucidate the molecular mechanism of emodin against neuropathic pain through modulating the gamma-aminobutyric acid (GABA)-ergic pathway and PI3K/AKT/NF-κB pathway. Phytother Res 2023; 37:1883-1899. [PMID: 36723382 DOI: 10.1002/ptr.7704] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 10/12/2022] [Accepted: 11/20/2022] [Indexed: 02/02/2023]
Abstract
Neuropathic pain (NeP) is a major health concern. Due to the complex pathological mechanisms, management of NeP is challenging. Emodin, a natural anthraquinone derivative, exerts excellent analgesic effects. However, its mechanisms of action are still poorly understood. In this study, we investigated the mechanisms underlying pain-relief effects of emodin in the cerebral cortex using proteomic and metabolomic approaches. After 15 days of emodin administration, the mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) values in the emodin groups were significantly higher than those in the chronic constriction injury (CCI) group (p < .05), suggesting emodin treatment could reverse CCI-induced hyperalgesia. Emodin treatment evoked the expression alteration of 402 proteins (153 up-regulated and 249 down-regulated) in the CCI models, which were primarily involved in PI3K/AKT signaling pathway, gamma-aminobutyric acid (GABA) receptor signaling, complement and coagulation cascades, cGMP/PKG signaling pathway, MAPK signaling pathway, and calcium signaling pathway. In parallel, emodin intervention regulated the abundance alteration of 27 brain metabolites (20 up-regulated and 7 down-regulated) in the CCI rats, which were primarily implicated in carbon metabolism, biosynthesis of amino acids, pentose phosphate pathway, and glucagon signaling pathway. After a comprehensive analysis and western blot validation, we demonstrated that emodin alleviated NeP mainly through regulating GABAergic pathway and PI3K/AKT/NF-κB pathway.
Collapse
Affiliation(s)
- Peng Chen
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ning-Yu Huang
- Accreditation Center of Traditional Chinese Medicine Physician, National Administration of Traditional Chinese Medicine, Beijing, China
| | - Bo Pang
- Medical Ward, Traditional Chinese Medicine Hospital of Huangpu District, Guangzhou, China
| | - Zeng-Jie Ye
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rui-Xi Luo
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Chang Liu
- Basic Medical School, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qian Gong
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chen Wang
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Traditional Chinese Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
26
|
Ding W, Fischer L, Chen Q, Li Z, Yang L, You Z, Hu K, Wu X, Zhou X, Chao W, Hu P, Dagnew TM, Dubreuil DM, Wang S, Xia S, Bao C, Zhu S, Chen L, Wang C, Wainger B, Jin P, Mao J, Feng G, Harnett MT, Shen S. Highly synchronized cortical circuit dynamics mediate spontaneous pain in mice. J Clin Invest 2023; 133:e166408. [PMID: 36602876 PMCID: PMC9974100 DOI: 10.1172/jci166408] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023] Open
Abstract
Cortical neural dynamics mediate information processing for the cerebral cortex, which is implicated in fundamental biological processes such as vision and olfaction, in addition to neurological and psychiatric diseases. Spontaneous pain is a key feature of human neuropathic pain. Whether spontaneous pain pushes the cortical network into an aberrant state and, if so, whether it can be brought back to a "normal" operating range to ameliorate pain are unknown. Using a clinically relevant mouse model of neuropathic pain with spontaneous pain-like behavior, we report that orofacial spontaneous pain activated a specific area within the primary somatosensory cortex (S1), displaying synchronized neural dynamics revealed by intravital two-photon calcium imaging. This synchronization was underpinned by local GABAergic interneuron hypoactivity. Pain-induced cortical synchronization could be attenuated by manipulating local S1 networks or clinically effective pain therapies. Specifically, both chemogenetic inhibition of pain-related c-Fos-expressing neurons and selective activation of GABAergic interneurons significantly attenuated S1 synchronization. Clinically effective pain therapies including carbamazepine and nerve root decompression could also dampen S1 synchronization. More important, restoring a "normal" range of neural dynamics through attenuation of pain-induced S1 synchronization alleviated pain-like behavior. These results suggest that spontaneous pain pushed the S1 regional network into a synchronized state, whereas reversal of this synchronization alleviated pain.
Collapse
Affiliation(s)
- Weihua Ding
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Lukas Fischer
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Qian Chen
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ziyi Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Liuyue Yang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Zerong You
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Kun Hu
- Department of Pathology, Tufts University School of Medicine, Medford, Massachusetts, USA
| | - Xinbo Wu
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Xue Zhou
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Wei Chao
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Peter Hu
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tewodros Mulugeta Dagnew
- MGH/HST Martinos Center for Biomedical Imaging, Department of Radiology, MGH, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel M. Dubreuil
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Shiyu Wang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Suyun Xia
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Caroline Bao
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Shengmei Zhu
- Department of Anesthesiology, the First Affiliate Hospital of Zhejiang University, Hangzhou, China
| | - Lucy Chen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Changning Wang
- MGH/HST Martinos Center for Biomedical Imaging, Department of Radiology, MGH, Harvard Medical School, Boston, Massachusetts, USA
| | - Brian Wainger
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Peng Jin
- Department of Human Genetics, Emory University, Atlanta, Georgia, USA
| | - Jianren Mao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| | - Guoping Feng
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mark T. Harnett
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Shiqian Shen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital (MGH), Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Zhou H, Li M, Zhao R, Sun L, Yang G. A sleep-active basalocortical pathway crucial for generation and maintenance of chronic pain. Nat Neurosci 2023; 26:458-469. [PMID: 36690899 PMCID: PMC10010379 DOI: 10.1038/s41593-022-01250-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 12/12/2022] [Indexed: 01/24/2023]
Abstract
Poor sleep is associated with the risk of developing chronic pain, but how sleep contributes to pain chronicity remains unclear. Here we show that following peripheral nerve injury, cholinergic neurons in the anterior nucleus basalis (aNB) of the basal forebrain are increasingly active during nonrapid eye movement (NREM) sleep in a mouse model of neuropathic pain. These neurons directly activate vasoactive intestinal polypeptide-expressing interneurons in the primary somatosensory cortex (S1), causing disinhibition of pyramidal neurons and allodynia. The hyperactivity of aNB neurons is caused by the increased inputs from the parabrachial nucleus (PB) driven by the injured peripheral afferents. Inhibition of this pathway during NREM sleep, but not wakefulness, corrects neuronal hyperactivation and alleviates pain. Our results reveal that the PB-aNB-S1 pathway during sleep is critical for the generation and maintenance of chronic pain. Inhibiting this pathway during the sleep phase could be important for treating neuropathic pain.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Miao Li
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Ruohe Zhao
- Department of Neuroscience and Physiology, Skirball Institute, New York University School of Medicine, New York, NY, USA
| | - Linlin Sun
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Guang Yang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
28
|
Chen C, Sun L, Adler A, Zhou H, Zhang L, Zhang L, Deng J, Bai Y, Zhang J, Yang G, Gan WB, Tang P. Synchronized activity of sensory neurons initiates cortical synchrony in a model of neuropathic pain. Nat Commun 2023; 14:689. [PMID: 36755026 PMCID: PMC9908980 DOI: 10.1038/s41467-023-36093-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023] Open
Abstract
Increased low frequency cortical oscillations are observed in people with neuropathic pain, but the cause of such elevated cortical oscillations and their impact on pain development remain unclear. By imaging neuronal activity in a spared nerve injury (SNI) mouse model of neuropathic pain, we show that neurons in dorsal root ganglia (DRG) and somatosensory cortex (S1) exhibit synchronized activity after peripheral nerve injury. Notably, synchronized activity of DRG neurons occurs within hours after injury and 1-2 days before increased cortical oscillations. This DRG synchrony is initiated by axotomized neurons and mediated by local purinergic signaling at the site of nerve injury. We further show that synchronized DRG activity after SNI is responsible for increasing low frequency cortical oscillations and synaptic remodeling in S1, as well as for inducing animals' pain-like behaviors. In naive mice, enhancing the synchrony, not the level, of DRG neuronal activity causes synaptic changes in S1 and pain-like behaviors similar to SNI mice. Taken together, these results reveal the critical role of synchronized DRG neuronal activity in increasing cortical plasticity and oscillations in a neuropathic pain model. These findings also suggest the potential importance of detection and suppression of elevated cortical oscillations in neuropathic pain states.
Collapse
Affiliation(s)
- Chao Chen
- Department of Orthopaedics, Peking 301 Hospital, Beijing, China
- Department of Hand Surgery, Shenzhen People's Hospital, Second Clinical Medicine College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Linlin Sun
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, Neuroscience Research Institute, Peking University, Beijing, China
| | - Avital Adler
- Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Hang Zhou
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA
| | - Licheng Zhang
- Department of Orthopaedics, Peking 301 Hospital, Beijing, China
| | - Lihai Zhang
- Department of Orthopaedics, Peking 301 Hospital, Beijing, China
| | - Junhao Deng
- Department of Orthopaedics, Peking 301 Hospital, Beijing, China
| | - Yang Bai
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Jinhui Zhang
- Department of Orthopaedics, the Affiliated Southeast Hospital of Xiamen University, Zhangzhou 175 Hospital, Zhangzhou, Fujian, China
| | - Guang Yang
- Department of Anesthesiology, Columbia University Medical Center, New York, NY, USA.
| | - Wen-Biao Gan
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China.
| | - Peifu Tang
- Department of Orthopaedics, Peking 301 Hospital, Beijing, China.
| |
Collapse
|
29
|
Li T, Gao Y, He M, Gui Z, Zhao B, Cao Y, Chen T, Zhu J, Wang J, Zhong Q, Zhang Z. P2X7 receptor-activated microglia in cortex is critical for sleep disorder under neuropathic pain. Front Neurosci 2023; 17:1095718. [PMID: 36816134 PMCID: PMC9936193 DOI: 10.3389/fnins.2023.1095718] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Neuropathic pain (NP) is associated with sleep disturbances, which may substantially influence the quality of life. Clinical and animal studies demonstrated that neurotransmitter is one of the main contributors to cause sleep disturbances induced by NP. Recently, it was reported that P2X7 receptors (P2X7R) are widely expressed in microglia, which serves crucial role in neuronal activity in the pain and sleep-awake cycle. In this study, we adopted the chronic constriction injury (CCI) model to establish the progress of chronic pain and investigated whether P2X7R of microglia in cortex played a critical role in sleep disturbance induced by NP. At electroencephalogram (EEG) level, sleep disturbance was observed in mice treated with CCI as they exhibited mechanical and thermal hypersensitivity, and inhibition of P2X7R ameliorated these changes. We showed a dramatic high level of P2X7R and Iba-1 co-expression in the cortical region, and the inhibition of P2X7R also adversely affected it. Furthermore, the power of LFPs in ventral posterior nucleus (VP) and primary somatosensory cortex (S1) which changed in the CCI group was adverse after the inhibition of P2X7R. Furthermore, inhibition of P2X7R also decreased the VP-S1 coherence which increased in CCI group. Nuclear magnetic resonance demonstrated inhibition of P2X7R decreased glutamate (Glu) levels in thalamic and cortical regions which were significantly increased in the CCI mice. Our findings provide evidence that NP has a critical effect on neuronal activity linked to sleep and may built up a new target for the development of sleep disturbances under chronic pain conditions.
Collapse
Affiliation(s)
- Tingting Li
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Yunling Gao
- Xiangyang Central Hospital, Institute of Neuroscience and Brain Diseases, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Mengying He
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China
| | - Zhu Gui
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China,University of Chinese Academy of Sciences, Beijing, China
| | - Bingchu Zhao
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China,School of Computer Science, Wuhan University, Wuhan, Hubei, China
| | - Yue Cao
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Ting Chen
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Jinpiao Zhu
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China
| | - Jie Wang
- Xiangyang Central Hospital, Institute of Neuroscience and Brain Diseases, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, China,University of Chinese Academy of Sciences, Beijing, China
| | - Qi Zhong
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,*Correspondence: Qi Zhong,
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China,Zongze Zhang,
| |
Collapse
|
30
|
Kobayashi S, O'Hashi K, Kobayashi M. Repetitive nociceptive stimulation increases spontaneous neural activation similar to nociception-induced activity in mouse insular cortex. Sci Rep 2022; 12:15190. [PMID: 36071208 PMCID: PMC9452502 DOI: 10.1038/s41598-022-19562-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/31/2022] [Indexed: 11/09/2022] Open
Abstract
Recent noninvasive neuroimaging technology has revealed that spatiotemporal patterns of cortical spontaneous activity observed in chronic pain patients are different from those in healthy subjects, suggesting that the spontaneous cortical activity plays a key role in the induction and/or maintenance of chronic pain. However, the mechanisms of the spontaneously emerging activities supposed to be induced by nociceptive inputs remain to be established. In the present study, we investigated spontaneous cortical activities in sessions before and after electrical stimulation of the periodontal ligament (PDL) by applying wide-field and two-photon calcium imaging to anesthetized GCaMP6s transgenic mice. First, we identified the sequential cortical activation patterns from the primary somatosensory and secondary somatosensory cortices to the insular cortex (IC) by PDL stimulation. We, then found that spontaneous IC activities that exhibited a similar spatiotemporal cortical pattern to evoked activities by PDL stimulation increased in the session after repetitive PDL stimulation. At the single-cell level, repetitive PDL stimulation augmented the synchronous neuronal activity. These results suggest that cortical plasticity induced by the repetitive stimulation leads to the frequent PDL stimulation-evoked-like spontaneous IC activation. This nociception-induced spontaneous activity in IC may be a part of mechanisms that induces chronic pain.
Collapse
Affiliation(s)
- Shutaro Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.,Department of Oral Surgery, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kazunori O'Hashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan. .,Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan. .,Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan.
| | - Masayuki Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan. .,Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan. .,Molecular Imaging Research Center, RIKEN, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
31
|
Chung G, Yun YC, Kim CY, Kim SK, Kim SJ. Metabotropic Glutamate Receptor 5 in the Dysgranular Zone of Primary Somatosensory Cortex Mediates Neuropathic Pain in Rats. Biomedicines 2022; 10:1633. [PMID: 35884938 PMCID: PMC9313034 DOI: 10.3390/biomedicines10071633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022] Open
Abstract
The primary somatosensory cortex (S1) plays a key role in the discrimination of somatic sensations. Among subdivisions in S1, the dysgranular zone of rodent S1 (S1DZ) is homologous to Brodmann's area 3a of primate S1, which is involved in the processing of noxious signals from the body. However, molecular changes in this region and their role in the pathological pain state have never been studied. In this study, we identified molecular alteration of the S1DZ in a rat model of neuropathic pain induced by right L5 spinal nerve ligation (SNL) surgery and investigated its functional role in pain symptoms. Brain images acquired from SNL group and control group in our previous study were analyzed, and behaviors were measured using the von Frey test, acetone test, and conditioned place preference test. We found that metabotropic glutamate receptor 5 (mGluR5) levels were significantly upregulated in the S1DZ contralateral to the nerve injury in the SNL group compared to the sham group. Pharmacological deactivation of mGluR5 in S1DZ ameliorated symptoms of neuropathic allodynia, which was shown by a significant increase in the mechanical paw withdrawal threshold and a decrease in the behavioral response to cold stimuli. We further confirmed that this treatment induced relief from the tonic-aversive state of chronic neuropathic pain, as a place preference memory associated with the treatment-paired chamber was formed in rats with neuropathic pain. Our data provide evidence that mGluR5 in the S1DZ is involved in the manifestation of abnormal pain sensations in the neuropathic pain state.
Collapse
Affiliation(s)
- Geehoon Chung
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (G.C.); (S.K.K.)
- Department of Physiology, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea; (Y.-C.Y.); (C.Y.K.)
| | - Yeong-Chan Yun
- Department of Physiology, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea; (Y.-C.Y.); (C.Y.K.)
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Chae Young Kim
- Department of Physiology, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea; (Y.-C.Y.); (C.Y.K.)
- Department of Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea
- Institut du Cerveau—Paris Brain Institute—ICM, INSERM, Sorbonne Université, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, F-75013 Paris, France
| | - Sun Kwang Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (G.C.); (S.K.K.)
| | - Sang Jeong Kim
- Department of Physiology, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea; (Y.-C.Y.); (C.Y.K.)
- Department of Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| |
Collapse
|
32
|
Osaki H, Kanaya M, Ueta Y, Miyata M. Distinct nociception processing in the dysgranular and barrel regions of the mouse somatosensory cortex. Nat Commun 2022; 13:3622. [PMID: 35768422 PMCID: PMC9243138 DOI: 10.1038/s41467-022-31272-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/07/2022] [Indexed: 11/23/2022] Open
Abstract
Nociception, a somatic discriminative aspect of pain, is, like touch, represented in the primary somatosensory cortex (S1), but the separation and interaction of the two modalities within S1 remain unclear. Here, we show spatially distinct tactile and nociceptive processing in the granular barrel field (BF) and adjacent dysgranular region (Dys) in mouse S1. Simultaneous recordings of the multiunit activity across subregions revealed that Dys neurons are more responsive to noxious input, whereas BF neurons prefer tactile input. At the single neuron level, nociceptive information is represented separately from the tactile information in Dys layer 2/3. In contrast, both modalities seem to converge on individual layer 5 neurons of each region, but to a different extent. Overall, these findings show layer-specific processing of nociceptive and tactile information between Dys and BF. We further demonstrated that Dys activity, but not BF activity, is critically involved in pain-like behavior. These findings provide new insights into the role of pain processing in S1. The processing of nociception in the somatosensory cortex (S1) has yet to be fully understood. Here, the authors demonstrate that the dysgranular region in S1 has an affinity for nociception and is critically involved in pain-like behavior.
Collapse
Affiliation(s)
- Hironobu Osaki
- Division of Neurophysiology, Department of Physiology, Graduate School of Medicine, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan. .,Laboratory of Functional Brain Circuit Construction, Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto, Japan.
| | - Moeko Kanaya
- Division of Neurophysiology, Department of Physiology, Graduate School of Medicine, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Yoshifumi Ueta
- Division of Neurophysiology, Department of Physiology, Graduate School of Medicine, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | - Mariko Miyata
- Division of Neurophysiology, Department of Physiology, Graduate School of Medicine, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan.
| |
Collapse
|
33
|
Hsiao IH, Liao HY, Lin YW. Optogenetic modulation of electroacupuncture analgesia in a mouse inflammatory pain model. Sci Rep 2022; 12:9067. [PMID: 35641558 PMCID: PMC9156770 DOI: 10.1038/s41598-022-12771-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/22/2022] [Indexed: 11/09/2022] Open
Abstract
Peripheral tissue damage and associated inflammation can trigger neuroplastic changes in somatic pain pathways, such as reduced neuronal firing thresholds and synaptic potentiation, that ultimately lead to peripheral sensitization and chronic pain. Electroacupuncture (EA) can relieve chronic inflammatory pain, but the underlying mechanisms are unknown, including the contributions of higher pain centers such as somatosensory cortex (SSC). We investigated these mechanisms using optogenetic modulation of SSC activity in a mouse inflammatory pain model. Injection of Complete Freund's Adjuvant into the hind paw reliably induced inflammation accompanied by reduced mechanical and thermal pain thresholds (hyperalgesia) within three days (mechanical: 1.54 ± 0.13 g; thermal: 3.94 ± 0.43 s). Application of EA produced significant thermal and mechanical analgesia, but these responses were reversed by optogenetic activation of SSC neurons, suggesting that EA-induced analgesia involves modulation of central pain pathways. Western blot and immunostaining revealed that EA also attenuated CaMKIIα signaling in the dorsal root ganglion, central spinal cord, SSC, and anterior cingulate cortex (ACC). In contrast, optogenetic activation of the SSC induced CaMKIIα signaling in SSC and ACC. These findings suggest that AE can relieve inflammatory pain by suppressing CaMKIIα-dependent plasticity in cortical pain pathways. The SSC and ACC CaMKIIα signaling pathways may be valuable therapeutic targets for chronic inflammatory pain treatment.
Collapse
Affiliation(s)
- I-Han Hsiao
- College of Chinese Medicine, Graduate Institute of Acupuncture Science, China Medical University, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan
- Department of Neurosurgery, China Medical University Hospital, Taichung, 404332, Taiwan
| | - Hsien-Yin Liao
- College of Chinese Medicine, School of Post-Baccalaureate Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan.
| | - Yi-Wen Lin
- College of Chinese Medicine, Graduate Institute of Acupuncture Science, China Medical University, 91 Hsueh-Shih Road, Taichung, 40402, Taiwan.
- Chinese Medicine Research Center, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
34
|
Hiraga SI, Itokazu T, Nishibe M, Yamashita T. Neuroplasticity related to chronic pain and its modulation by microglia. Inflamm Regen 2022; 42:15. [PMID: 35501933 PMCID: PMC9063368 DOI: 10.1186/s41232-022-00199-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/19/2022] [Indexed: 01/03/2023] Open
Abstract
Neuropathic pain is often chronic and can persist after overt tissue damage heals, suggesting that its underlying mechanism involves the alteration of neuronal function. Such an alteration can be a direct consequence of nerve damage or a result of neuroplasticity secondary to the damage to tissues or to neurons. Recent studies have shown that neuroplasticity is linked to causing neuropathic pain in response to nerve damage, which may occur adjacent to or remotely from the site of injury. Furthermore, studies have revealed that neuroplasticity relevant to chronic pain is modulated by microglia, resident immune cells of the central nervous system (CNS). Microglia may directly contribute to synaptic remodeling and altering pain circuits, or indirectly contribute to neuroplasticity through property changes, including the secretion of growth factors. We herein highlight the mechanisms underlying neuroplasticity that occur in the somatosensory circuit of the spinal dorsal horn, thalamus, and cortex associated with chronic pain following injury to the peripheral nervous system (PNS) or CNS. We also discuss the dynamic functions of microglia in shaping neuroplasticity related to chronic pain. We suggest further understanding of post-injury ectopic plasticity in the somatosensory circuits may shed light on the differential mechanisms underlying nociceptive, neuropathic, and nociplastic-type pain. While one of the prominent roles played by microglia appears to be the modulation of post-injury neuroplasticity. Therefore, future molecular- or genetics-based studies that address microglia-mediated post-injury neuroplasticity may contribute to the development of novel therapies for chronic pain.
Collapse
Affiliation(s)
- Shin-Ichiro Hiraga
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takahide Itokazu
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan. .,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Mariko Nishibe
- Center for Strategic Innovative Dentistry, Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | - Toshihide Yamashita
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan. .,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan. .,WPI Immunology Frontier Research Center, Osaka, Japan. .,Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| |
Collapse
|
35
|
Hirai A, Sugio S, Nimako C, Nakayama SMM, Kato K, Takahashi K, Arizono K, Hirano T, Hoshi N, Fujioka K, Taira K, Ishizuka M, Wake H, Ikenaka Y. Ca 2+ imaging with two-photon microscopy to detect the disruption of brain function in mice administered neonicotinoid insecticides. Sci Rep 2022; 12:5114. [PMID: 35332220 PMCID: PMC8948258 DOI: 10.1038/s41598-022-09038-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 03/14/2022] [Indexed: 12/02/2022] Open
Abstract
Neonicotinoid pesticides are a class of insecticides that reportedly have harmful effects on bees and dragonflies, causing a reduction in their numbers. Neonicotinoids act as neuroreceptor modulators, and some studies have reported their association with neurodevelopmental disorders. However, the precise effect of neonicotinoids on the central nervous system has not yet been identified. Herein, we conducted in vivo Ca2+ imaging using a two-photon microscope to detect the abnormal activity of neuronal circuits in the brain after neonicotinoid application. The oral administration of acetamiprid (ACE) (20 mg/kg body weight (BW) in mature mice with a quantity less than the no-observed-adverse-effect level (NOAEL) and a tenth or half of the median lethal dose (LD50) of nicotine (0.33 or 1.65 mg/kg BW, respectively), as a typical nicotinic acetylcholine receptor (nAChR) agonist, increased anxiety-like behavior associated with altered activities of the neuronal population in the somatosensory cortex. Furthermore, we detected ACE and its metabolites in the brain, 1 h after ACE administration. The results suggested that in vivo Ca2+ imaging using a two-photon microscope enabled the highly sensitive detection of neurotoxicant-mediated brain disturbance of nerves.
Collapse
Affiliation(s)
- Anri Hirai
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan
| | - Shouta Sugio
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumi-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Collins Nimako
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan
| | - Shouta M M Nakayama
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan
| | - Keisuke Kato
- Faculty of Pharmaceutical Sciences, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan
| | - Keisuke Takahashi
- Faculty of Pharmaceutical Sciences, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan
| | - Koji Arizono
- Faculty of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, 3-1-100 Tsukide, Higashi-ku, Kumamoto, 862-8502, Japan
| | - Tetsushi Hirano
- Life Science Research Center, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Nobuhiko Hoshi
- Student Affairs Section, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
| | - Kazutoshi Fujioka
- Albany College of Pharmacy and Health Sciences, 106 New Scotland Ave, Albany, NY, USA
| | - Kumiko Taira
- Department of Anesthesiology, Medical Center East, Tokyo Women's Medical University, Tokyo, Japan
| | - Mayumi Ishizuka
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumi-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yoshinori Ikenaka
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan. .,Water Research Group, Unit for Environmental Sciences and Management, North-West University, 11 Hoffman Street, Potchefstroom, 2531, South Africa. .,One Health Research Center, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan. .,Translational Research Unit, Faculty of Veterinary Medicine, Veterinary Teaching Hospital, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, 060-0818, Japan.
| |
Collapse
|
36
|
Hirata Y, Nomura K, Kato D, Tachibana Y, Niikura T, Uchiyama K, Hosooka T, Fukui T, Oe K, Kuroda R, Hara Y, Adachi T, Shibasaki K, Wake H, Ogawa W. A Piezo1/KLF15/IL-6 axis mediates immobilization-induced muscle atrophy. J Clin Invest 2022; 132:1-13. [PMID: 35290243 PMCID: PMC9159676 DOI: 10.1172/jci154611] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 03/01/2022] [Indexed: 11/17/2022] Open
Abstract
Although immobility is a common cause of muscle atrophy, the mechanism underlying this causality is unclear. We here show that Krüppel-like factor 15 (KLF15) and IL-6 are upregulated in skeletal muscle of limb-immobilized mice and that mice with KLF15 deficiency in skeletal muscle or with systemic IL-6 deficiency are protected from immobility-induced muscle atrophy. A newly developed Ca2+ bioimaging revealed that the cytosolic Ca2+ concentration ([Ca2+]i) of skeletal muscle is reduced to below the basal level by immobilization, which is associated with the downregulation of Piezo1. Acute disruption of Piezo1 in skeletal muscle induced Klf15 and Il6 expression as well as muscle atrophy, which was prevented by antibodies against IL-6. A role for the Piezo1/KLF15/IL-6 axis in immobility-induced muscle atrophy was validated in human samples. Our results thus uncover a paradigm for Ca2+ signaling in that a decrease in [Ca2+]i from the basal level triggers a defined biological event.
Collapse
Affiliation(s)
- Yu Hirata
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazuhiro Nomura
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Daisuke Kato
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihisa Tachibana
- Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takahiro Niikura
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kana Uchiyama
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tetsuya Hosooka
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoaki Fukui
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Keisuke Oe
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuji Hara
- Department of Integrative Physiology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Takahiro Adachi
- Department of Precision Health, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koji Shibasaki
- Laboratory of Neurochemistry, Graduate School of Human Health Science, University of Nagasaki, Nagasaki, Japan
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
37
|
Nishi M, Sugio S, Hirano T, Kato D, Wake H, Shoda A, Murata M, Ikenaka Y, Tabuchi Y, Mantani Y, Yokoyama T, Hoshi N. Elucidation of the neurological effects of clothianidin exposure at the no-observed-adverse-effect level (NOAEL) using two-photon microscopy in vivo imaging. J Vet Med Sci 2022; 84:585-592. [PMID: 35264496 PMCID: PMC9096047 DOI: 10.1292/jvms.22-0013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Neonicotinoid pesticides (NNs) cause behavioral abnormalities in mammals, raising
concerns about their effects on neural circuit activity. We herein examined the
neurological effects of the NN clothianidin (CLO) by in vivo
Ca2+ imaging using two-photon microscopy. Mice were fed the
no-observed-adverse-effect-level (NOAEL) dose of CLO for 2 weeks and their neuronal
activity in the primary somatosensory cortex (S1) was observed weekly for 2 weeks. CLO
exposure caused a sustained influx of Ca2+ in neurons in the S1 2/3 layers,
indicating hyperactivation of neurons. In addition, microarray gene expression analysis
suggested the induction of neuroinflammation and changes in synaptic activity. These
results demonstrate that exposure to the NOAEL dose of CLO can overactivate neurons and
disrupt neuronal homeostasis.
Collapse
Affiliation(s)
- Misaki Nishi
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University
| | - Shouta Sugio
- Department of Anatomy and Molecular Cell Biology, Graduate School of Medicine, Nagoya University
| | | | - Daisuke Kato
- Department of Anatomy and Molecular Cell Biology, Graduate School of Medicine, Nagoya University
| | - Hiroaki Wake
- Department of Anatomy and Molecular Cell Biology, Graduate School of Medicine, Nagoya University
| | - Asuka Shoda
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University
| | - Midori Murata
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University
| | - Yoshinori Ikenaka
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University.,Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University.,One Health Research Center, Hokkaido University.,Water Research Group, Unit for Environmental Sciences and Management, North-West University
| | | | - Youhei Mantani
- Laboratory of Histophysiology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University
| | - Toshifumi Yokoyama
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University
| | - Nobuhiko Hoshi
- Laboratory of Animal Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University
| |
Collapse
|
38
|
Kim YR, Kim SJ. Altered synaptic connections and inhibitory network of the primary somatosensory cortex in chronic pain. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:69-75. [PMID: 35203057 PMCID: PMC8890942 DOI: 10.4196/kjpp.2022.26.2.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
Chronic pain is induced by tissue or nerve damage and is accompanied by pain hypersensitivity (i.e., allodynia and hyperalgesia). Previous studies using in vivo two-photon microscopy have shown functional and structural changes in the primary somatosensory (S1) cortex at the cellular and synaptic levels in inflammatory and neuropathic chronic pain. Furthermore, alterations in local cortical circuits were revealed during the development of chronic pain. In this review, we summarize recent findings regarding functional and structural plastic changes of the S1 cortex and alteration of the S1 inhibitory network in chronic pain. Finally, we discuss potential neuromodulators driving modified cortical circuits and suggest further studies to understand the cortical mechanisms that induce pain hypersensitivity.
Collapse
Affiliation(s)
- Yoo Rim Kim
- Departments of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sang Jeong Kim
- Departments of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Departments of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
39
|
Wei HL, Xu CH, Wang JJ, Zhou GP, Guo X, Chen YC, Yu YS, He ZZ, Yin X, Li J, Zhang H. Disrupted Functional Connectivity of the Amygdala Predicts the Efficacy of Non-steroidal Anti-inflammatory Drugs in Migraineurs Without Aura. Front Mol Neurosci 2022; 15:819507. [PMID: 35283727 PMCID: PMC8908446 DOI: 10.3389/fnmol.2022.819507] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Machine learning (ML) has been largely applied for predicting migraine classification. However, the prediction of efficacy of non-steroidal anti-inflammatory drugs (NSAIDs) in migraine is still in the early stages. This study aims to evaluate whether the combination of machine learning and amygdala-related functional features could help predict the efficacy of NSAIDs in patients with migraine without aura (MwoA). A total of 70 MwoA patients were enrolled for the study, including patients with an effective response to NSAIDs (M-eNSAIDs, n = 35) and MwoA patients with ineffective response to NSAIDs (M-ieNSAIDs, n = 35). Furthermore, 33 healthy controls (HCs) were matched for age, sex, and education level. The study participants were subjected to resting-state functional magnetic resonance imaging (fMRI) scanning. Disrupted functional connectivity (FC) patterns from amygdala-based FC analysis and clinical characteristics were considered features that could promote classification through multivariable logistic regression (MLR) and support vector machine (SVM) for predicting the efficacy of NSAIDs. Further, receiver operating characteristic (ROC) curves were drawn to evaluate the predictive ability of the models. The M-eNSAIDs group exhibited enhanced FC with ipsilateral calcarine sulcus (CAL), superior parietal gyrus (SPG), paracentral lobule (PCL), and contralateral superior frontal gyrus (SFG) in the left amygdala. However, the M-eNSAIDs group showed decreased FC with ipsilateral caudate nucleus (CAU), compared to the M-ieNSAIDs group. Moreover, the M-eNSAIDs group showed higher FC with left pre-central gyrus (PreCG) and post-central gyrus (PoCG) compared to HCs. In contrast, the M-ieNSAIDs group showed lower FC with the left anterior cingulate cortex (ACC) and right SFG. Furthermore, the MwoA patients showed increased FC with the left middle frontal gyrus (MFG) in the right amygdala compared to HCs. The disrupted left amygdala-related FC patterns exhibited significant correlations with migraine characteristics in the M-ieNSAIDs group. The MLR and SVM models discriminated clinical efficacy of NSAIDs with an area under the curve (AUC) of 0.891 and 0.896, sensitivity of 0.971 and 0.833, and specificity of 0.629 and 0.875, respectively. These findings suggest that the efficacy of NSAIDs in migraine could be predicted using ML algorithm. Furthermore, this study highlights the role of amygdala-related neural function in revealing underlying migraine-related neuroimaging mechanisms.
Collapse
Affiliation(s)
- Heng-Le Wei
- Department of Radiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Chen-Hui Xu
- Department of Neurology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Jin-Jin Wang
- Department of Radiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Gang-Ping Zhou
- Department of Radiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Xi Guo
- Department of Radiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Yu-Chen Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Sheng Yu
- Department of Radiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Zhen-Zhen He
- Department of Radiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Xindao Yin
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Junrong Li
- Department of Neurology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Junrong Li,
| | - Hong Zhang
- Department of Radiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
- Hong Zhang,
| |
Collapse
|
40
|
Guinto MC, Haruta M, Kurauchi Y, Saigo T, Kurasawa K, Ryu S, Ohta Y, Kawahara M, Takehara H, Tashiro H, Sasagawa K, Katsuki H, Ohta J. Modular head-mounted cortical imaging device for chronic monitoring of intrinsic signals in mice. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:026501. [PMID: 35166087 PMCID: PMC8843356 DOI: 10.1117/1.jbo.27.2.026501] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/24/2022] [Indexed: 06/14/2023]
Abstract
SIGNIFICANCE Intrinsic optical signals (IOS) generated in the cortical tissue as a result of various interacting metabolic processes are used extensively to elucidate the underlying mechanisms that govern neurovascular coupling. However, current IOS measurements still often rely on bulky, tabletop imaging systems, and there remains a dearth of studies in freely moving subjects. Lightweight, miniature head-mounted imaging devices provide unique opportunities for investigating cortical dynamics in small animals under a variety of naturalistic behavioral settings. AIM The aim of this work was to monitor IOS in the somatosensory cortex of wild-type mice by developing a lightweight, biocompatible imaging device that readily lends itself to animal experiments in freely moving conditions. APPROACH Herein we describe a method for realizing long-term IOS imaging in mice using a 0.54-g, compact, CMOS-based, head-mounted imager. The two-part module, consisting of a tethered sensor plate and a base plate, allows facile assembly prior to imaging sessions and disassembly when the sensor is not in use. LEDs integrated into the device were chosen to illuminate the cortical mantle at two different wavelengths in the visible regime (λcenter: 535 and 625 nm) for monitoring volume- and oxygenation state-dependent changes in the IOS, respectively. To test whether the system can detect robust cortical responses, we recorded sensory-evoked IOS from mechanical stimulation of the hindlimbs (HL) of anesthetized mice in both acute and long-term implantation conditions. RESULTS Cortical IOS recordings in the primary somatosensory cortex hindlimb receptive field (S1HL) of anesthetized mice under green and red LED illumination revealed robust, multiphasic profiles that were time-locked to the mechanical stimulation of the contralateral plantar hindpaw. Similar intrinsic signal profiles observed in S1HL at 40 days postimplantation demonstrated the viability of the approach for long-term imaging. Immunohistochemical analysis showed that the brain tissue did not exhibit appreciable immune response due to the device implantation and operation. A proof-of-principle imaging session in a freely behaving mouse showed minimal locomotor impediment for the animal and also enabled estimation of blood flow speed. CONCLUSIONS We demonstrate the utility of a miniature cortical imaging device for monitoring IOS and related hemodynamic processes in both anesthetized and freely moving mice, cueing potential for applications to some neuroscientific studies of sensation and naturalistic behavior.
Collapse
Affiliation(s)
- Mark Christian Guinto
- Nara Institute of Science and Technology, Graduate School of Science and Technology, Division of Materials Science, Ikoma, Japan
| | - Makito Haruta
- Nara Institute of Science and Technology, Graduate School of Science and Technology, Division of Materials Science, Ikoma, Japan
| | - Yuki Kurauchi
- Kumamoto University, Graduate School of Pharmaceutical Sciences, Department of Chemico-Pharmacological Sciences, Kumamoto, Japan
| | - Taisuke Saigo
- Nara Institute of Science and Technology, Graduate School of Science and Technology, Division of Materials Science, Ikoma, Japan
| | - Kazuki Kurasawa
- Nara Institute of Science and Technology, Graduate School of Science and Technology, Division of Materials Science, Ikoma, Japan
| | - Sumika Ryu
- Kumamoto University, Graduate School of Pharmaceutical Sciences, Department of Chemico-Pharmacological Sciences, Kumamoto, Japan
| | - Yasumi Ohta
- Nara Institute of Science and Technology, Graduate School of Science and Technology, Division of Materials Science, Ikoma, Japan
| | - Mamiko Kawahara
- Nara Institute of Science and Technology, Graduate School of Science and Technology, Division of Materials Science, Ikoma, Japan
| | - Hironari Takehara
- Nara Institute of Science and Technology, Graduate School of Science and Technology, Division of Materials Science, Ikoma, Japan
| | - Hiroyuki Tashiro
- Nara Institute of Science and Technology, Graduate School of Science and Technology, Division of Materials Science, Ikoma, Japan
- Kyushu University, Division of Medical Technology, Department of Health Sciences, Faculty of Medical Sciences, Fukuoka, Japan
| | - Kiyotaka Sasagawa
- Nara Institute of Science and Technology, Graduate School of Science and Technology, Division of Materials Science, Ikoma, Japan
| | - Hiroshi Katsuki
- Kumamoto University, Graduate School of Pharmaceutical Sciences, Department of Chemico-Pharmacological Sciences, Kumamoto, Japan
| | - Jun Ohta
- Nara Institute of Science and Technology, Graduate School of Science and Technology, Division of Materials Science, Ikoma, Japan
| |
Collapse
|