1
|
Cao Z, Yung WH, Ke Y. Repeated activation of preoptic area recipient neurons in posterior paraventricular nucleus mediates chronic heat-induced negative emotional valence and hyperarousal states. eLife 2025; 13:RP101302. [PMID: 40202515 PMCID: PMC11981607 DOI: 10.7554/elife.101302] [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] [Indexed: 04/10/2025] Open
Abstract
Mental and behavioral disorders are associated with extended period of hot weather as found in heatwaves, but the underlying neural circuit mechanism remains poorly known. The posterior paraventricular thalamus (pPVT) is a hub for emotional processing and receives inputs from the hypothalamic preoptic area (POA), the well-recognized thermoregulation center. The present study was designed to explore whether chronic heat exposure leads to aberrant activities in POA recipient pPVT neurons and subsequent changes in emotional states. By devising an air heating paradigm mimicking the condition of heatwaves and utilizing emotion-related behavioral tests, viral tract tracing, in vivo calcium recordings, optogenetic manipulations, and electrophysiological recordings, we found that chronic heat exposure for 3 weeks led to negative emotional valence and hyperarousal states in mice. The pPVT neurons receive monosynaptic excitatory and inhibitory innervations from the POA. These neurons exhibited a persistent increase in neural activity following chronic heat exposure, which was essential for chronic heat-induced emotional changes. Notably, these neurons were also prone to display stronger neuronal activities associated with anxiety responses to stressful situations. Furthermore, we observed saturated neuroplasticity in the POA-pPVT excitatory pathway after chronic heat exposure that occluded further potentiation. Taken together, long-term aberration in the POA to pPVT pathway offers a neurobiological mechanism of emotional and behavioral changes seen in extended periods of hot weather like heatwaves.
Collapse
Affiliation(s)
- Zhiping Cao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong KongChina
| | - Wing-Ho Yung
- Department of Neuroscience, College of Biomedicine, City University of Hong KongHong KongChina
| | - Ya Ke
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong KongHong KongChina
| |
Collapse
|
2
|
Wang W, Liu W, Liu S, Duan D, Ma Y, Zhang Z, Li C, Tang Y, Wang Z, Xing Y. Specific Activation of Dopamine Receptor D1 Expressing Neurons in the PrL Alleviates CSDS-Induced Anxiety-Like Behavior Comorbidity with Postoperative Hyperalgesia in Male Mice. Mol Neurobiol 2025; 62:2817-2834. [PMID: 39177734 DOI: 10.1007/s12035-024-04444-6] [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: 02/22/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
Postoperative pain is a type of pain that occurs in clinical patients after surgery. Among the factors influencing the transition from acute postoperative pain to chronic postoperative pain, chronic stress has received much attention in recent years. Here, we investigated the role of dopamine receptor D1/D2 expressing pyramidal neurons in the prelimbic cortex (PrL) in modulating chronic social defeat stress (CSDS)-induced anxiety-like behavior comorbidity with postoperative hyperalgesia in male mice. Our results showed that preoperative CSDS induced anxiety-like behavior and significantly prolonged postoperative pain caused by plantar incision, but did not affect plantar wound recovery and inflammation. Reduced activation of dopamine receptor D1 or D2 expressing neurons in the PrL is a remarkable feature of male mice after CSDS, and chronic inhibition of dopamine receptor D1 or D2 expressing neurons in the PrL induced anxiety-like behavior and persistent postoperative pain. Further studies found that activation of D1 expressing but not D2 expressing neurons in the PrL ameliorated CSDS-induced anxiety-like behavior and postoperative hyperalgesia. Our results suggest that dopamine receptor D1 expressing neurons in the PrL play a crucial role in CSDS-induced anxiety-like behavior comorbidity with postoperative hyperalgesia in male mice.
Collapse
Affiliation(s)
- Wang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, No.100 Science Road, Gao-Xin District, Henan, 450001, Zhengzhou, China
- The Academy of Medical Sciences of Zhengzhou University, Henan, 450001, Zhengzhou, China
| | - Weizhen Liu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, No.100 Science Road, Gao-Xin District, Henan, 450001, Zhengzhou, China
- The Academy of Medical Sciences of Zhengzhou University, Henan, 450001, Zhengzhou, China
| | - Sufang Liu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX75246, USA
| | - Dongxiao Duan
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, No.100 Science Road, Gao-Xin District, Henan, 450001, Zhengzhou, China
| | - Yajing Ma
- College of Biology and Food, Shangqiu Normal University, Shangqiu, 476000, China
| | - Zijuan Zhang
- School of Basic Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Changsheng Li
- Department of Anesthesiology, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yuanyuan Tang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zhiju Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, No.100 Science Road, Gao-Xin District, Henan, 450001, Zhengzhou, China.
| | - Ying Xing
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, No.100 Science Road, Gao-Xin District, Henan, 450001, Zhengzhou, China.
| |
Collapse
|
3
|
Duan L, Wang Q, Chen J, Fan Z, Zhang W, Yan J. Unraveling the PVT Glu-mPFC Glu circuit: A new frontier in chronic pain management for bone cancer pain. Brain Res Bull 2025; 222:111235. [PMID: 39922506 DOI: 10.1016/j.brainresbull.2025.111235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025]
Abstract
Bone cancer pain (BCP) is a type of ongoing or breakthrough pain caused by a primary bone tumor or bone metastasis. BCP impairs patients' quality of life. Depending upon clinical observations, the administration of centrally acting analgesic has been associated with the alleviation of pain symptoms BCP patients. Central nervous system sensitization performs a crucial role in pain-regulating perception in BCP. Nevertheless, the precise neural circuitry and mechanism of action remain enigmatic. In the present study, we observed the activation of glutamatergic neurons in the Prelimbic cortex (mPFC) and paraventricular thalamus (PVT) in BCP mice. Experimental validation using viral tracers confirmed the existence of a projection pathway between the PVT and mPFC. Inhibition of the input from PVT glutamatergic neurons to mPFC glutamatergic neurons alleviates chronic pain in BCP, whereas activation of the PVTGlu-mPFCGlu projection induces chronic pain in mice. These findings imply a pivotal role for the PVTGlu-mPFCGlu circuit in the regulation of chronic pain in BCP.
Collapse
Affiliation(s)
- Liqun Duan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China; Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qianliang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Jianpeng Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Zelin Fan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Wenzhi Zhang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Jun Yan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China.
| |
Collapse
|
4
|
Ma L, Sun D, Wen S, Yuan J, Li J, Tan X, Cao S. PSD-95 Protein: A Promising Therapeutic Target in Chronic Pain. Mol Neurobiol 2025; 62:3361-3375. [PMID: 39285025 DOI: 10.1007/s12035-024-04485-x] [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: 10/04/2023] [Accepted: 09/04/2024] [Indexed: 02/04/2025]
Abstract
Chronic pain, as a social public health problem, has a serious impact on the quality of patients' life. Currently, the main drugs used to treat chronic pain are opioids, antipyretic, and nonsteroidal anti-inflammatory drugs (NSAIDs). But the obvious side effects limit their use, so it is urgent to find new therapeutic targets. Postsynaptic density (PSD)-95 protein plays an important role in the occurrence and development of chronic pain. The over-expression of the PSD-95 protein and its interaction with other proteins are closely related to the chronic pain. Besides, the PSD-95-related drugs that inhibit the expression of PSD-95 as well as the interaction with other protein have been proved to treat chronic pain significantly. In conclusion, although more deep studies are needed in the future, these studies indicate that PSD-95 and the related proteins, such as NMDA receptor (NMDAR) subunit 2B (GluN2B), AMPA receptor (AMPAR), calmodulin-dependent protein kinase II (CaMKII), 5-hydroxytryptamine 2A receptor (5-HT2AR), and neuronal nitric oxide synthase (nNOS), are the promising therapeutic targets for chronic pain.
Collapse
Affiliation(s)
- Lulin Ma
- Department of Pain Medicine, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, China
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Dongdong Sun
- Hengyang Medical School, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Song Wen
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Jie Yuan
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Jing Li
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Xinran Tan
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
- Department of Pain Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, Guizhou, China
| | - Song Cao
- Department of Pain Medicine, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong, China.
| |
Collapse
|
5
|
Chen J, Yang L, Shen J, Lu J, Mo X, Huang L, Chen L, Yu C. Distinct Roles of Astrocytes and GABAergic Neurons in the Paraventricular Thalamic Nucleus in Modulating Diabetic Neuropathic Pain. J Neurosci 2025; 45:e1013242024. [PMID: 39622642 PMCID: PMC11841761 DOI: 10.1523/jneurosci.1013-24.2024] [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: 05/30/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 02/21/2025] Open
Abstract
Diabetic neuropathic pain (DNP) is a common chronic complication of diabetes mellitus and a clinically common form of neuropathic pain. The thalamus is an important center for the conduction and modulation of nociceptive signals. The paraventricular thalamic nucleus (PVT) is an important midline nucleus of the thalamus involved in sensory processing, but the specific role of PVT astrocytes and GABAergic neurons in DNP remains unclear. Here, we examined the activity of PVT astrocytes and neurons at various time points during the development of DNP by fluorescence immunohistochemistry and found that the activity of PVT astrocytes was significantly increased while that of PVT neurons was significantly decreased 14 d after streptozotocin injection in male rats. The inhibition of PVT astrocytes by chemogenetic manipulation relieved mechanical allodynia in male DNP model rats, whereas the activation of PVT astrocytes induced mechanical allodynia in normal male rats. Interestingly, chemogenetic activation of GABAergic neurons in the PVT alleviated mechanical allodynia in male DNP model rats, whereas chemogenetic inhibition of GABAergic neurons in the PVT induced mechanical allodynia in normal male rats. These data demonstrate the distinct roles of PVT astrocytes and GABAergic neurons in modulating DNP, revealing the mechanism of DNP pathogenesis and the role of the PVT in pain modulation.
Collapse
Affiliation(s)
- Jian Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Lan Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Jinhuang Shen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Jingshan Lu
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Fujian Center for Safety Evaluation of New Drug, Fujian Medical University, Fuzhou 350122, China
| | - Xiaona Mo
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Linyi Huang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Li Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Changxi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| |
Collapse
|
6
|
Zhao M, Chen L, Cheng Z, Wang X, Zhang S, Li M, Hao Z, Sun X, Zhang J, Yu Y, Ren J, Jia X. Altered brain functional connectivity in patients with tension-type headache. Headache 2025; 65:216-229. [PMID: 39801497 DOI: 10.1111/head.14900] [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: 11/30/2023] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 02/06/2025]
Abstract
OBJECTIVE To evaluate whether patients with tension-type headache (TTH) exhibit abnormal brain functional connectivity compared to healthy controls. BACKGROUND TTH is one of the most prevalent headache disorders throughout the world. The present study delves into brain functional connectivity in patients with TTH to enhance the understanding of its underlying pathophysiology. METHODS A cross-sectional study was conducted, enrolling patients with TTH diagnosed in line with the International Classification of Headache Disorders, 3rd edition beta criteria and a cohort of healthy controls (HCs). We used four metrics-global brain functional connectivity, functional connectivity, Granger causality analysis, and dynamic functional connectivity-to evaluate alterations of functional connectivity patterns in patients with TTH from both static and dynamic perspectives. Furthermore, correlational analyses were performed to explore the relationships between abnormal brain activities and clinical characteristics. RESULTS A total of 33 patients with TTH (mean age = 42.3; 13 males/20 females) and 30 HCs (mean age = 37.1; 13 males/17 females) were included in the current study. Compared to HCs, patients with TTH showed altered global brain functional connectivity in the right dorsolateral superior frontal gyrus (SFGdor, t = 4.60). Abnormal functional connectivity was also detected between the right SFGdor and the right superior temporal gyrus (t = 4.56). Furthermore, the right SFGdor exhibited altered information flow with several brain regions, including the left precuneus (t = 5.16), right middle temporal gyrus (MTG, t = 4.72/-4.41), right inferior temporal gyrus (t = 4.64), right caudate nucleus (t = 4.09), and right thalamus (THA, t = -4.04). In terms of dynamic functional connectivity, disconnection was observed between the right SFGdor and the right MTG (t = -3.10), right Rolandic operculum (ROL, t = 3.60), left opercular inferior frontal gyrus (t = -3.48), and left medial superior frontal gyrus (t = -3.00). In addition, the correlation analyses revealed that activities in the MTG (r = 0.48), THA (r = -0.38), and ROL (r = 0.36) were significantly correlated with disease duration, while THA activity was associated with Visual Analogue Scale scores (r = 0.50). CONCLUSIONS This study revealed alterations in both static and dynamic brain functional connectivity in patients with TTH within regions implicated in sensory perception, emotional processing, cognition, and pain regulation. These results may promote the understanding of the neural networks involved in TTH and potentially inform future therapeutic approaches for the condition.
Collapse
Affiliation(s)
- Mengqi Zhao
- School of Psychology, Zhejiang Normal University, Jinhua, China
- School of Medical Imaging, Shandong Second Medical University, Weifang, China
| | - Lanfen Chen
- School of Medical Imaging, Shandong Second Medical University, Weifang, China
| | - Zhixiang Cheng
- Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Xizhen Wang
- Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Shuxian Zhang
- Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Mengting Li
- School of Psychology, Zhejiang Normal University, Jinhua, China
| | - Zeqi Hao
- School of Psychology, Zhejiang Normal University, Jinhua, China
| | - Xihe Sun
- School of Medical Imaging, Shandong Second Medical University, Weifang, China
| | - Jianxin Zhang
- School of Foreign Studies, China University of Petroleum (East China), Qingdao, China
| | - Yang Yu
- Department of Psychiatry, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Ren
- School of Psychology, Zhejiang Normal University, Jinhua, China
| | - Xize Jia
- School of Psychology, Zhejiang Normal University, Jinhua, China
| |
Collapse
|
7
|
He Z, Zhang J, Xu J, Wang Y, Zheng X, Wang W. Differential Neuronal Activation of Nociceptive Pathways in Neuropathic Pain After Spinal Cord Injury. Cell Mol Neurobiol 2025; 45:18. [PMID: 39883258 PMCID: PMC11782389 DOI: 10.1007/s10571-025-01532-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/12/2025] [Indexed: 01/31/2025]
Abstract
Neuropathic pain, a prevalent complication following spinal cord injury (SCI), severely impairs the life quality of patients. No ideal treatment exists due to incomplete knowledge on underlying neural processes. To explore the SCI-induced effect on nociceptive circuits, the protein expression of c-Fos was analyzed as an indicator of neuronal activation in a rat contusion model exhibiting below-level pain. Additional stimuli were delivered to mimic the different peripheral sensory inputs in daily life. Following noxious rather than innocuous or no stimulation, a greater number of spinal dorsal horn (DH) neurons were activated after SCI, mainly in the deep DH. SCI facilitated the activation of excitatory but not inhibitory DH neurons. Moreover, excitatory interneurons expressing protein kinase C gamma (PKCγ) in laminae II-III, which are known to play a role in mechanical allodynia after peripheral nerve injury, responded in larger amounts to both innocuous and noxious stimulation following SCI. Accordingly, more spinal projection neurons in lamina I were activated. Within supraspinal nuclei processing pain, differentially enhanced activation in response to noxious stimulation was detected after SCI, with a significant increase in the locus coeruleus and medial thalamus, a slight increase in the periaqueductal gray and dorsal raphe, and no change in the lateral parabrachial nucleus or primary sensory cortex. These findings indicated differential hyperexcitability along the sensory neuroaxis following SCI, with a particular emphasis on the involvement of specific neuron subtypes, such as spinal PKCγ interneurons and locus coeruleus noradrenergic neurons, which may serve as crucial targets for potential therapies.
Collapse
Affiliation(s)
- Ziyu He
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Zhang
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Neurosurgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia Xu
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Rehabilitation, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
- Stem Cell Research Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Wang
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Neurosurgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaolong Zheng
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wei Wang
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Neurological Diseases of Chinese Ministry of Education, the School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
8
|
Liu X, Zhang X, Wang D, Cao Y, Zhang L, Li Z, Zhang Q, Shen Y, Lu X, Fan K, Liu M, Wei J, Hu S, Liu H. A Neural Circuit From Paraventricular Nucleus of the Thalamus to the Nucleus Accumbens Mediates Inflammatory Pain in Mice. Brain Behav 2025; 15:e70218. [PMID: 39740781 DOI: 10.1002/brb3.70218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 11/22/2024] [Accepted: 12/01/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Pain is a prevalent comorbidity in numerous clinical conditions and causes suffering; however, the mechanism of pain is intricate, and the neural circuitry underlying pain in the brain remains incompletely elucidated. More research into the perception and modulation of pain within the central nervous system is essential. The nucleus accumbens (NAc) plays a pivotal role in the regulation of animal behavior, and extensive research has unequivocally demonstrated its significant involvement in the occurrence and development of pain. NAc receives projections from various other neural nuclei within the brain, including the paraventricular nucleus of the thalamus (PVT). In this experiment, we demonstrate that the specific glutamatergic neural circuit projection from PVT to NAc (PVTGlut→NAc) is implicated in the modulation of inflammatory pain in mice. METHODS We compared the difference in pain thresholds between complete Freund's adjuvant (CFA)-induced inflammatory pain models and controls. Then in a well-established mouse model of CFA-induced inflammatory pain, immunofluorescence staining was utilized to evaluate changes in c-Fos protein expression within PVT neurons. To investigate the role of PVTGlut→NAc in the modulation of pain, we used optogenetics to modulate this neural circuit, and nociceptive behavioral tests were employed to investigate the functional role of the PVTGlut→NAc circuit in the modulation of inflammatory pain. RESULTS In the mice with the inflammatory pain group, both the paw withdrawal latencies (PWLs) and paw withdrawal thresholds (PWTs) of the right hind paw were decreased compared to the control group. In addition, compared to the control group, CFA-induced inflammatory pain led to increased c-Fos protein expression in PVT, which means that some of the neurons in this area of the brain region have been activated. Following the injection of retrograde transport fluorescent-labeled virus into NAc, glutamatergic neurons projecting from the PVT to NAc were observed, confirming the projection relationship between PVT and NAc. In the experiments in optogenetic regulation, normal mice exhibited pain behavior when the PVTGlut→NAc circuit was stimulated by a 473 nm blue laser, resulting in decreased PWLs and PWTs compared to the control group, which means activating this neural circuit can lead to painful behaviors. In the CFA-induced pain group, inhibition of the PVTGlut→NAc circuit by a 589 nm yellow laser alleviated pain behavior, leading to increased PWLs and PWTs compared to the control group, representing the fact that inhibition of this neural circuit relieves pain behaviors. CONCLUSIONS The findings unveil a pivotal role of the PVTGlut→NAc circuit in modulating inflammatory pain induced by CFA in mice.
Collapse
Affiliation(s)
- Xi Liu
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
- Department of Anesthesiology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Xi Zhang
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
- School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongxu Wang
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Ya Cao
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Ling Zhang
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Zhonghua Li
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Qin Zhang
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Yu Shen
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Xian Lu
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Keyu Fan
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Mingxia Liu
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Jingqiu Wei
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
- Department of Education & Training, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Hangzhou, China
| | - Siping Hu
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - He Liu
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine & Key Laboratory of Anesthesia and Analgesia Application Technology, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| |
Collapse
|
9
|
Cao Y, Wu Z, Zhang M, Ji R, Zhang H, Song L. Microglial adenosine A 2A receptor in the paraventricular thalamic nucleus regulates pain sensation and analgesic effects independent of opioid and cannabinoid receptors. Front Pharmacol 2024; 15:1467305. [PMID: 39749202 PMCID: PMC11693661 DOI: 10.3389/fphar.2024.1467305] [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: 07/19/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction The paraventricular thalamic nucleus (PVT) is recognized for its critical role in pain regulation, yet the precise molecular mechanisms involved remain poorly understood. Here, we demonstrated an essential role of the microglial adenosine A2A receptor (A2AR) in the PVT in regulating pain sensation and non-opioid analgesia. Method and results Specifically, A2AR was predominantly expressed in ionized calcium binding adapter molecule 1 (Iba1)-positive microglia cells within the PVT, with expression levels remaining unchanged in mice experiencing persistent inflammatory pain induced by complete Freund's adjuvant (CFA). Pharmacological activation of local PVT A2AR with its agonist CGS21680 induced significantly decreased 50% paw withdrawal threshold (50%PWTs) and paw withdrawal latency (PWLs), as measured by the Von Frey test and Hargreaves test in adult mice. Conversely, intra-PVT infusion of A2AR antagonist SCH58261 increased 50%PWTs and PWLs in mice; a robust analgesic effect was also observed in CFA mice with inflammatory pain. Importantly, these analgesic effects of A2AR antagonist SCH58261 were not affected by adjunctive intraperitoneal administration of naloxone or rimonabant, inhibitors of opioid receptor and cannabinoid CB1 receptor (CB1R), respectively. Discussion Overall, these pharmacological experiments underscore an essential role of microglia-expressed A2AR with in PVT in pain sensation while revealing a novel analgesic action independent of opioid and cannabinoids receptors. Thus, these findings highlight PVT microglial adenosine A2A receptor as a promising target for novel approaches to pain modulation and future analgesic development.
Collapse
Affiliation(s)
- Yiping Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhou Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Moruo Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ran Ji
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lingzhen Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
10
|
Wu Y, Zhang D, Liu J, Jiang J, Xie K, Wu L, Leng Y, Liang P, Zhu T, Zhou C. Activity of the Sodium Leak Channel Maintains the Excitability of Paraventricular Thalamus Glutamatergic Neurons to Resist Anesthetic Effects of Sevoflurane in Mice. Anesthesiology 2024; 141:56-74. [PMID: 38625708 DOI: 10.1097/aln.0000000000005015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
BACKGROUND Stimulation of the paraventricular thalamus has been found to enhance anesthesia recovery; however, the underlying molecular mechanism by which general anesthetics modulate paraventricular thalamus is unclear. This study aimed to test the hypothesis that the sodium leak channel (NALCN) maintains neuronal activity in the paraventricular thalamus to resist anesthetic effects of sevoflurane in mice. METHODS Chemogenetic and optogenetic manipulations, in vivo multiple-channel recordings, and electroencephalogram recordings were used to investigate the role of paraventricular thalamus neuronal activity in sevoflurane anesthesia. Virus-mediated knockdown and/or overexpression was applied to determine how NALCN influenced excitability of paraventricular thalamus glutamatergic neurons under sevoflurane. Viral tracers and local field potentials were used to explore the downstream pathway. RESULTS Single neuronal spikes in the paraventricular thalamus were suppressed by sevoflurane anesthesia and recovered during emergence. Optogenetic activation of paraventricular thalamus glutamatergic neurons shortened the emergence period from sevoflurane anesthesia, while chemogenetic inhibition had the opposite effect. Knockdown of the NALCN in the paraventricular thalamus delayed the emergence from sevoflurane anesthesia (recovery time: from 24 ± 14 to 64 ± 19 s, P < 0.001; concentration for recovery of the righting reflex: from 1.13% ± 0.10% to 0.97% ± 0.13%, P < 0.01). As expected, the overexpression of the NALCN in the paraventricular thalamus produced the opposite effects. At the circuit level, knockdown of the NALCN in the paraventricular thalamus decreased the neuronal activity of the nucleus accumbens, as indicated by the local field potential and decreased single neuronal spikes in the nucleus accumbens. Additionally, the effects of NALCN knockdown in the paraventricular thalamus on sevoflurane actions were reversed by optical stimulation of the nucleus accumbens. CONCLUSIONS Activity of the NALCN maintains the excitability of paraventricular thalamus glutamatergic neurons to resist the anesthetic effects of sevoflurane in mice.
Collapse
Affiliation(s)
- Yujie Wu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Donghang Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jingyao Jiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Keyu Xie
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Wu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Leng
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Liang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Zhang F, Wei Y, Weng R, Xu Q, Li R, Yu Y, Xu G. Paraventricular thalamus-insular cortex circuit mediates colorectal visceral pain induced by neonatal colonic inflammation in mice. CNS Neurosci Ther 2024; 30:e14534. [PMID: 37994678 PMCID: PMC11017444 DOI: 10.1111/cns.14534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
AIMS Irritable bowel syndrome (IBS) is a common functional gastrointestinal disorder, but its pathogenesis remains incompletely understood, particularly the involvements of central nervous system sensitization in colorectal visceral pain. Our study was to investigate whether the paraventricular thalamus (PVT) projected to the insular cortex (IC) to regulate colorectal visceral pain in neonatal colonic inflammation (NCI) mice and underlying mechanisms. METHODS We applied optogenetic, chemogenetic, or pharmacological approaches to manipulate the glutamatergicPVT-IC pathway. Fiber photometry was used to assess neuronal activity. Electromyography activities in response to colorectal distension (CRD) were measured to evaluate the colorectal visceral pain. RESULTS NCI enhanced c-Fos expression and calcium activity upon CRD in the ICGlu, and optogenetic manipulation of them altered colorectal visceral pain responses accordingly. Viral tracing indicated that the PVTGlu projected to the ICGlu. Optogenetic manipulation of PVTGlu changed colorectal visceral pain responses. Furthermore, selective optogenetic modulation of PVT projections in the IC influenced colorectal visceral pain, which was reversed by chemogenetic manipulation of downstream ICGlu. CONCLUSIONS This study identified a novel PVT-IC neural circuit playing a critical role in colorectal visceral pain in a mouse model of IBS.
Collapse
Affiliation(s)
- Fu‐Chao Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of NeuroscienceSoochow UniversitySuzhouJiangsuP. R. China
| | - Ying‐Xue Wei
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of NeuroscienceSoochow UniversitySuzhouJiangsuP. R. China
| | - Rui‐Xia Weng
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuP. R. China
| | - Qi‐Ya Xu
- Department of AnesthesiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuP. R. China
| | - Rui Li
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuP. R. China
| | - Yang Yu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of NeuroscienceSoochow UniversitySuzhouJiangsuP. R. China
| | - Guang‐Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of NeuroscienceSoochow UniversitySuzhouJiangsuP. R. China
| |
Collapse
|
12
|
Li YC, Zhang FC, Xu TW, Weng RX, Zhang HH, Chen QQ, Hu S, Gao R, Li R, Xu GY. Advances in the pathological mechanisms and clinical treatments of chronic visceral pain. Mol Pain 2024; 20:17448069241305942. [PMID: 39673493 PMCID: PMC11645724 DOI: 10.1177/17448069241305942] [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: 10/09/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 12/16/2024] Open
Abstract
Chronic visceral pain stems from internal organs and is frequently associated with functional gastrointestinal disorders, like irritable bowel syndrome (IBS). Since the underlying mechanisms of visceral pain remain largely unclear, clinical management is often limited and ineffective. Comprehensive research into the pathogenesis of visceral pain, along with the development of personalized therapeutic strategies, is crucial for advancing treatment options. Studies suggest that imbalances in purinergic receptors and neural circuit function are closely linked to the onset of visceral pain. In this review, we will explore the etiology and pathological mechanisms underlying visceral pain, with a focus on ion channels, epigenetic factors, and neural circuits, using functional gastrointestinal disorders as case studies. Finally, we will summarize and evaluate emerging treatments and potential initiatives aimed at managing visceral pain.
Collapse
Affiliation(s)
- Yong-Chang Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fu-Chao Zhang
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Timothy W Xu
- Department of Earth Sciences, University College London, London, UK
| | - Rui-Xia Weng
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong-Hong Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qian-Qian Chen
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Shufen Hu
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Rong Gao
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rui Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guang-Yin Xu
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
13
|
Tang QQ, Wu Y, Tao Q, Shen Y, An X, Liu D, Xu Z. Direct paraventricular thalamus-basolateral amygdala circuit modulates neuropathic pain and emotional anxiety. Neuropsychopharmacology 2024; 49:455-466. [PMID: 37848732 PMCID: PMC10724280 DOI: 10.1038/s41386-023-01748-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/05/2023] [Accepted: 09/27/2023] [Indexed: 10/19/2023]
Abstract
The comorbidity of chronic pain and mental dysfunctions such as anxiety disorders has long been recognized, but the underlying mechanisms remained poorly understood. Here, using a mouse model of neuropathic pain, we demonstrated that the thalamic paraventricular nucleus (PVT) played a critical role in chronic pain-induced anxiety-like behavioral abnormalities. Fiber photometry and electrophysiology demonstrated that chronic pain increased the activities in PVT glutamatergic neurons. Chemogenetic manipulation revealed that suppression of PVT glutamatergic neurons relieved pain-like behavior and anxiety-like behaviors. Conversely, selective activation of PVT glutamatergic neurons showed algesic and anxiogenic effects. Furthermore, the elevated excitability of PVT glutamatergic neurons resulted in increased excitatory inputs to the basolateral complex (BLA) neurons. Optogenetic manipulation of the PVT-BLA pathway bilaterally modulates both the pain-like behavior and anxiety-like phenotypes. These findings shed light on how the PVT-BLA pathway contributed to the processing of pain-like behavior and maladaptive anxiety, and targeting this pathway might be a straightforward therapeutic strategy to both alleviate nociceptive hypersensitivity and rescue anxiety behaviors in chronic pain conditions.
Collapse
Affiliation(s)
- Qian-Qian Tang
- Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, 910 Hengshan Road, Shanghai, China
| | - Yuanyuan Wu
- Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, 910 Hengshan Road, Shanghai, China
| | - Qiang Tao
- Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, 910 Hengshan Road, Shanghai, China
| | - Yanan Shen
- Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, 910 Hengshan Road, Shanghai, China
| | - Xiaohu An
- Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, 910 Hengshan Road, Shanghai, China
| | - Di Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zifeng Xu
- Department of Anesthesiology, the International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, 910 Hengshan Road, Shanghai, China.
| |
Collapse
|
14
|
Li JN, Wu XM, Zhao LJ, Sun HX, Hong J, Wu FL, Chen SH, Chen T, Li H, Dong YL, Li YQ. Central medial thalamic nucleus dynamically participates in acute itch sensation and chronic itch-induced anxiety-like behavior in male mice. Nat Commun 2023; 14:2539. [PMID: 37137899 PMCID: PMC10156671 DOI: 10.1038/s41467-023-38264-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/20/2023] [Indexed: 05/05/2023] Open
Abstract
Itch is an annoying sensation consisting of both sensory and emotional components. It is known to involve the parabrachial nucleus (PBN), but the following transmission nodes remain elusive. The present study identified that the PBN-central medial thalamic nucleus (CM)-medial prefrontal cortex (mPFC) pathway is essential for itch signal transmission at the supraspinal level in male mice. Chemogenetic inhibition of the CM-mPFC pathway attenuates scratching behavior or chronic itch-related affective responses. CM input to mPFC pyramidal neurons is enhanced in acute and chronic itch models. Specifically chronic itch stimuli also alter mPFC interneuron involvement, resulting in enhanced feedforward inhibition and a distorted excitatory/inhibitory balance in mPFC pyramidal neurons. The present work underscores CM as a transmit node of the itch signal in the thalamus, which is dynamically engaged in both the sensory and affective dimensions of itch with different stimulus salience.
Collapse
Affiliation(s)
- Jia-Ni Li
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xue-Mei Wu
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Human Anatomy, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Liu-Jie Zhao
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China
| | - Han-Xue Sun
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Human Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Jie Hong
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Human Anatomy, Baotou Medical College Inner Mongolia University of Science and Technology, Baotou, 014040, China
| | - Feng-Ling Wu
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China
| | - Si-Hai Chen
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China
| | - Tao Chen
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Li
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Lin Dong
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology, K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China.
- Department of Human Anatomy, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China.
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China.
- Department of Human Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China.
- Department of Human Anatomy, Baotou Medical College Inner Mongolia University of Science and Technology, Baotou, 014040, China.
| |
Collapse
|
15
|
Wu FL, Chen SH, Li JN, Zhao LJ, Wu XM, Hong J, Zhu KH, Sun HX, Shi SJ, Mao E, Zang WD, Cao J, Kou ZZ, Li YQ. Projections from the Rostral Zona Incerta to the Thalamic Paraventricular Nucleus Mediate Nociceptive Neurotransmission in Mice. Metabolites 2023; 13:metabo13020226. [PMID: 36837844 PMCID: PMC9966812 DOI: 10.3390/metabo13020226] [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: 01/12/2023] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Zona incerta (ZI) is an integrative subthalamic region in nociceptive neurotransmission. Previous studies demonstrated that the rostral ZI (ZIR) is an important gamma-aminobutyric acid-ergic (GABAergic) source to the thalamic paraventricular nucleus (PVT), but whether the ZIR-PVT pathway participates in nociceptive modulation is still unclear. Therefore, our investigation utilized anatomical tracing, fiber photometry, chemogenetic, optogenetic and local pharmacological approaches to investigate the roles of the ZIRGABA+-PVT pathway in nociceptive neurotransmission in mice. We found that projections from the GABAergic neurons in ZIR to PVT were involved in nociceptive neurotransmission. Furthermore, chemogenetic and optogenetic activation of the ZIRGABA+-PVT pathway alleviates pain, whereas inhibiting the activities of the ZIRGABA+-PVT circuit induces mechanical hypersensitivity and partial heat hyperalgesia. Importantly, in vivo pharmacology combined with optogenetics revealed that the GABA-A receptor (GABAAR) is crucial for GABAergic inhibition from ZIR to PVT. Our data suggest that the ZIRGABA+-PVT pathway acts through GABAAR-expressing glutamatergic neurons in PVT mediates nociceptive neurotransmission.
Collapse
Affiliation(s)
- Feng-Ling Wu
- Department of Human Anatomy, College of Preclinical Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
| | - Si-Hai Chen
- Department of Human Anatomy, College of Preclinical Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
| | - Jia-Ni Li
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
| | - Liu-Jie Zhao
- Department of Human Anatomy, College of Preclinical Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
| | - Xue-Mei Wu
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
- Department of Human Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China
| | - Jie Hong
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
- Department of Human Anatomy, Baotou Medical College Inner Mongolia University of Science and Technology, Baotou 014040, China
| | - Ke-Hua Zhu
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
| | - Han-Xue Sun
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
- Department of Human Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China
| | - Su-Juan Shi
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
| | - E Mao
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
| | - Wei-Dong Zang
- Department of Human Anatomy, College of Preclinical Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jing Cao
- Department of Human Anatomy, College of Preclinical Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhen-Zhen Kou
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
- Correspondence: (Z.-Z.K.); (Y.-Q.L.); Tel.: +86-29-8477-2706; Fax: +86-29-8328-3229 (Y.-Q.L.)
| | - Yun-Qing Li
- Department of Human Anatomy, College of Preclinical Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi’an 710032, China
- Department of Geriatrics, Tangdu Hospital, The Fourth Military Medical University, Xi’an 710038, China
- Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou 571199, China
- Department of Anatomy, College of Basic Medicine, Dali University, Dali 671000, China
- Correspondence: (Z.-Z.K.); (Y.-Q.L.); Tel.: +86-29-8477-2706; Fax: +86-29-8328-3229 (Y.-Q.L.)
| |
Collapse
|
16
|
Li L, Zhang H, Zheng Z, Ma N, Zhang Y, Liu Y, Zhang J, Su S, Zang W, Shao J, Cao J. Perioperative sleep deprivation activates the paraventricular thalamic nucleus resulting in persistent postoperative incisional pain in mice. Front Neuroanat 2022; 16:1074310. [PMID: 36620195 PMCID: PMC9813598 DOI: 10.3389/fnana.2022.1074310] [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: 10/19/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Background The duration of postsurgical pain is closely correlated with perioperative stress. Most patients suffer short-term sleep disorder/deprivation before and/or after surgery, which leads to extended postsurgical pain by an undetermined mechanism. The paraventricular thalamus (PVT) is a critical area that contributes to the regulation of feeding, awakening, and emotional states. However, whether the middle PVT is involved in postoperative pain or the extension of postoperative pain caused by perioperative sleep deprivation has not yet been investigated. Methods We established a model of postoperative pain by plantar incision with perioperative rapid eye movement sleep deprivation (REMSD) 6 h/day for 3 consecutive days in mice. The excitability of the CaMKIIα+ neurons in the middle PVT (mPVTCaMKIIα) was detected by immunofluorescence and fiber photometry. The activation/inhibition of mPVTCaMKIIα neurons was conducted by chemogenetics. Results REMSD prolonged the duration of postsurgical pain and increased the excitability of mPVTCaMKIIα neurons. In addition, mPVTCaMKIIα neurons showed increased excitability in response to nociceptive stimuli or painful conditions. However, REMSD did not delay postsurgical pain recovery following the ablation of CaMKIIα neurons in the mPVT. The activation of mPVTCaMKIIα neurons prolonged the duration of postsurgical pain and elicited anxiety-like behaviors. In contrast, inhibition of mPVTCaMKIIα neurons reduced the postsurgical pain after REMSD. Conclusion Our data revealed that the CaMKIIα neurons in the mPVT are involved in the extension of the postsurgical pain duration induced by REMSD, and represented a novel potential target to treat postoperative pain induced by REMSD.
Collapse
Affiliation(s)
- Lei Li
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Huijie Zhang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenli Zheng
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China,Department of Medical Record Management, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Nan Ma
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yidan Zhang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yaping Liu
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China,Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jingjing Zhang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Songxue Su
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Weidong Zang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China,Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, Henan, China
| | - Jinping Shao
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China,Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, Henan, China,*Correspondence: Jinping Shao,
| | - Jing Cao
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China,Neuroscience Research Institute, Zhengzhou University Academy of Medical Sciences, Zhengzhou, Henan, China,Jing Cao,
| |
Collapse
|
17
|
Plasticity of neuronal excitability and synaptic balance in the anterior nucleus of paraventricular thalamus after nerve injury. Brain Res Bull 2022; 188:1-10. [PMID: 35850188 DOI: 10.1016/j.brainresbull.2022.07.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/20/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022]
Abstract
The anterior nucleus of the paraventricular thalamus (aPVT) integrates various synaptic inputs and conveys information to the downstream brain regions for arousal and pain regulation. Recent studies have indicated that the PVT plays a crucial role in the regulation of chronic pain, but the plasticity mechanism of neuronal excitability and synaptic inputs for aPVT neurons in neuropathic pain remains unclear. Here, we report that spinal nerve ligation (SNL) significantly increased the neuronal excitability and reset the excitatory/inhibitory (E/I) synaptic inputs ratio of aPVT neurons in mice. SNL significantly increased the membrane input resistance, firing frequency, and the half-width of action potential. Additionally, SNL enlarged the area of afterdepolarization and prolonged the rebound low-threshold spike following a hyperpolarized current injection. Further results indicate that an inwardly rectifying current density was decreased in SNL animals. SNL also decreased the amplitude, but not the frequency of spontaneous excitatory postsynaptic currents (sEPSCs), nor the amplitude or frequency of spontaneous inhibitory postsynaptic currents (sIPSCs) of aPVT neurons. Moreover, SNL disrupted the E/I synaptic ratio, caused a decrease in weighted tau and half-width of averaged sIPSCs, but did not change these physiological properties of averaged sEPSCs. Finally, pharmacological activation of the GABAA receptor at aPVT could effective relieve SNL-induced mechanical allodynia in mice. These results reveal the plasticity of intrinsic neuronal excitability and E/I synaptic balance in the aPVT neurons after nerve injury and it may play an important role in the development of pain sensitization.
Collapse
|
18
|
Neyama H, Nishiyori M, Cui Y, Watanabe Y, Ueda H. Lysophosphatidic acid receptor type-1 mediates brain activation in micro-Positron Emission Tomography analysis in a fibromyalgia-like mouse model. Eur J Neurosci 2022; 56:4224-4233. [PMID: 35666711 DOI: 10.1111/ejn.15729] [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: 11/15/2021] [Accepted: 05/30/2022] [Indexed: 11/26/2022]
Abstract
The intermittent cold stress-induced generalized pain response mimics the pathophysiological and pharmacotherapeutic features reported for fibromyalgia patients, including the presence of chronic generalized pain and female dominance. In addition, the intermittent cold stress-induced generalized pain is abolished in lysophosphatidic acid receptor type-1 knockout mice, as reported in many cases of neuropathic pain models. This study aimed to identify the brain loci involved in the intermittent cold stress generalized pain response and test their dependence on the lysophosphatidic acid receptor type-1. Positron emission tomography analyses using 2-deoxy-2-[18 F]fluoro-D-glucose in the presence of a pain stimulus showed that intermittent cold stress causes a significant increase in uptake in the ipsilateral regions, including the salience networking-related anterior cingulate cortex and insular cortex and the cognition-related hippocampus. A significant decrease was observed in the default mode network-related posterior cingulate cortex. Almost these intermittent cold stress-induced changes were abolished in lysophosphatidic acid receptor type-1 knockout mice. There results suggest that the intermittent cold stress-induced generalized pain response is mediated by the lysophosphatidic acid receptor type-1 in specific brain loci related to salience networking and cognition, which may lead to further developments in the treatment of fibromyalgia.
Collapse
Affiliation(s)
- Hiroyuki Neyama
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan.,Laboratory for Biofunction Dynamics Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Michiko Nishiyori
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Yilong Cui
- Laboratory for Biofunction Dynamics Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan.,Laboratory for the Study of Pain, Research Institute for Production Development, Kyoto, Japan
| |
Collapse
|
19
|
Chemogenetics as a neuromodulatory approach to treating neuropsychiatric diseases and disorders. Mol Ther 2022; 30:990-1005. [PMID: 34861415 PMCID: PMC8899595 DOI: 10.1016/j.ymthe.2021.11.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/12/2021] [Accepted: 11/29/2021] [Indexed: 01/01/2023] Open
Abstract
Chemogenetics enables precise, non-invasive, and reversible modulation of neural activity via the activation of engineered receptors that are pharmacologically selective to endogenous or exogenous ligands. With recent advances in therapeutic gene delivery, chemogenetics is poised to support novel interventions against neuropsychiatric diseases and disorders. To evaluate its translational potential, we performed a scoping review of applications of chemogenetics that led to the reversal of molecular and behavioral deficits in studies relevant to neuropsychiatric diseases and disorders. In this review, we present these findings and discuss the potential and challenges for using chemogenetics as a precision medicine-based neuromodulation strategy.
Collapse
|
20
|
Isagulyan ED, Mikhailova VA, Aslakhanova KS, Slavin KV. Prospects of neuromodulation for chronic pain. BRAIN DISORDERS 2022. [DOI: 10.1016/j.dscb.2021.100027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
21
|
Jefferson T, Kelly CJ, Martina M. Differential Rearrangement of Excitatory Inputs to the Medial Prefrontal Cortex in Chronic Pain Models. Front Neural Circuits 2022; 15:791043. [PMID: 35002635 PMCID: PMC8738091 DOI: 10.3389/fncir.2021.791043] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/02/2021] [Indexed: 11/16/2022] Open
Abstract
Chronic pain patients suffer a disrupted quality of life not only from the experience of pain itself, but also from comorbid symptoms such as depression, anxiety, cognitive impairment, and sleep disturbances. The heterogeneity of these symptoms support the idea of a major involvement of the cerebral cortex in the chronic pain condition. Accordingly, abundant evidence shows that in chronic pain the activity of the medial prefrontal cortex (mPFC), a brain region that is critical for executive function and working memory, is severely impaired. Excitability of the mPFC depends on the integrated effects of intrinsic excitability and excitatory and inhibitory inputs. The main extracortical sources of excitatory input to the mPFC originate in the thalamus, hippocampus, and amygdala, which allow the mPFC to integrate multiple information streams necessary for cognitive control of pain including sensory information, context, and emotional salience. Recent techniques, such as optogenetic methods of circuit dissection, have made it possible to tease apart the contributions of individual circuit components. Here we review the synaptic properties of these main glutamatergic inputs to the rodent mPFC, how each is altered in animal models of chronic pain, and how these alterations contribute to pain-associated mPFC deactivation. By understanding the contributions of these individual circuit components, we strive to understand the broad spectrum of chronic pain and comorbid pathologies, how they are generated, and how they might be alleviated.
Collapse
Affiliation(s)
- Taylor Jefferson
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | | | - Marco Martina
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
22
|
Kimmey BA, McCall NM, Wooldridge LM, Satterthwaite T, Corder G. Engaging endogenous opioid circuits in pain affective processes. J Neurosci Res 2022; 100:66-98. [PMID: 33314372 PMCID: PMC8197770 DOI: 10.1002/jnr.24762] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 01/03/2023]
Abstract
The pervasive use of opioid compounds for pain relief is rooted in their utility as one of the most effective therapeutic strategies for providing analgesia. While the detrimental side effects of these compounds have significantly contributed to the current opioid epidemic, opioids still provide millions of patients with reprieve from the relentless and agonizing experience of pain. The human experience of pain has long recognized the perceived unpleasantness entangled with a unique sensation that is immediate and identifiable from the first-person subjective vantage point as "painful." From this phenomenological perspective, how is it that opioids interfere with pain perception? Evidence from human lesion, neuroimaging, and preclinical functional neuroanatomy approaches is sculpting the view that opioids predominately alleviate the affective or inferential appraisal of nociceptive neural information. Thus, opioids weaken pain-associated unpleasantness rather than modulate perceived sensory qualities. Here, we discuss the historical theories of pain to demonstrate how modern neuroscience is revisiting these ideas to deconstruct the brain mechanisms driving the emergence of aversive pain perceptions. We further detail how targeting opioidergic signaling within affective or emotional brain circuits remains a strong avenue for developing targeted pharmacological and gene-therapy analgesic treatments that might reduce the dependence on current clinical opioid options.
Collapse
Affiliation(s)
- Blake A. Kimmey
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Equal contributions
| | - Nora M. McCall
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Equal contributions
| | - Lisa M. Wooldridge
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Theodore Satterthwaite
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Informatics and Neuroimaging Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gregory Corder
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
23
|
Miyashiro S, Yamada Y, Nagaoka M, Shima R, Muta T, Ishikawa H, Abe T, Hori M, Oka K, Koshikawa F, Ito E. Pain relief associated with decreased oxyhemoglobin level in left dorsolateral prefrontal cortex. PLoS One 2021; 16:e0256626. [PMID: 34424921 PMCID: PMC8382195 DOI: 10.1371/journal.pone.0256626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/09/2021] [Indexed: 01/10/2023] Open
Abstract
Pain in the elbow, shoulder, knee, lower back, and various other joints is relieved by adhesion of pyramidal thorn patches. To elucidate the pain relief mechanism induced by the patches, we established a quantitative method for estimating the pain reduction and investigated the brain regions that change in association with pain relief. We first attempted to quantify the pain relief using transcutaneous electric stimulation (TCES) and a visual analog scale (VAS), and then applied near-infrared spectroscopy (NIRS) to the prefrontal cortex, including the dorsolateral prefrontal cortex (DLPFC) and the orbitofrontal cortex (OFC). We also examined the salivary oxytocin levels, which are thought to reflect oxytocin secretion levels from the posterior pituitary in the brain. Application of pyramidal thorn patches to pain regions decreased the pain degree estimated using TCES and VAS. Oxyhemoglobin levels were likely to be decreased in the left DLPFC on the basis of NIRS measurements during patch treatment, suggesting that the left DLPFC is involved in pain relief. On the other hand, the salivary oxytocin levels varied widely. A potential reason for the varying salivary oxytocin levels is its utilization in the pain region as an analgesic agent. Our results suggest that the left DLPFC will become a target brain region for pain therapy.
Collapse
Affiliation(s)
| | - Yurika Yamada
- Department of Biology, Waseda University, Tokyo, Japan
| | | | - Rei Shima
- Department of Biology, Waseda University, Tokyo, Japan
| | - Toshizumi Muta
- Department of Psychology, Waseda University, Tokyo, Japan
| | - Haruyuki Ishikawa
- Department of Culture, Media and Society, Waseda University, Tokyo, Japan
| | - Tetsuri Abe
- Department of Psychology, Waseda University, Tokyo, Japan
| | - Masashi Hori
- Department of Educational Psychology, Waseda University, Tokyo, Japan
| | - Kotaro Oka
- Department of Bioscience and Informatics, Keio University, Yokohama, Japan
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | - Etsuro Ito
- Department of Biology, Waseda University, Tokyo, Japan
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
24
|
The paraventricular nucleus of the thalamus: an integrative node underlying homeostatic behavior. Trends Neurosci 2021; 44:538-549. [PMID: 33775435 DOI: 10.1016/j.tins.2021.03.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/20/2021] [Accepted: 03/03/2021] [Indexed: 12/15/2022]
Abstract
Early anatomical evidence suggested that the paraventricular nucleus of the thalamus (PVT) regulates arousal, as well as emotional and motivated behaviors. We discuss recent studies using modern techniques which now confirm and expand the involvement of the rodent PVT in these functions. Despite the emerging notion that the PVT is implicated in various behavioral processes, a recurrent theme is that activity in this brain region depends on internal state information arriving from the hypothalamus and brainstem, and is influenced by prior experience. We propose that the primary function of the PVT is to detect homeostatic challenges by integrating information about prior experiences, competing needs, and internal state to guide adaptive behavioral responses aimed at restoring homeostasis.
Collapse
|
25
|
Zhou K, Zhu L, Hou G, Chen X, Chen B, Yang C, Zhu Y. The Contribution of Thalamic Nuclei in Salience Processing. Front Behav Neurosci 2021; 15:634618. [PMID: 33664657 PMCID: PMC7920982 DOI: 10.3389/fnbeh.2021.634618] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
The brain continuously receives diverse information about the external environment and changes in the homeostatic state. The attribution of salience determines which stimuli capture attention and, therefore, plays an essential role in regulating emotions and guiding behaviors. Although the thalamus is included in the salience network, the neural mechanism of how the thalamus contributes to salience processing remains elusive. In this mini-review, we will focus on recent advances in understanding the specific roles of distinct thalamic nuclei in salience processing. We will summarize the functional connections between thalamus nuclei and other key nodes in the salience network. We will highlight the convergence of neural circuits involved in reward and pain processing, arousal, and attention control in thalamic structures. We will discuss how thalamic activities represent salience information in associative learning and how thalamic neurons modulate adaptive behaviors. Lastly, we will review recent studies which investigate the contribution of thalamic dysfunction to aberrant salience processing in neuropsychiatric disorders, such as drug addiction, posttraumatic stress disorder (PTSD), and schizophrenia. Based on emerging evidence from both human and rodent research, we propose that the thalamus, different from previous studies that as an information relay, has a broader role in coordinating the cognitive process and regulating emotions.
Collapse
Affiliation(s)
- Kuikui Zhou
- Shenzhen Key Laboratory of Drug Addiction, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Lin Zhu
- Department of Neonatology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Guoqiang Hou
- Shenzhen Key Laboratory of Drug Addiction, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Xueyu Chen
- Department of Neonatology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Bo Chen
- Shenzhen Key Laboratory of Drug Addiction, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Chuanzhong Yang
- Department of Neonatology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Yingjie Zhu
- Shenzhen Key Laboratory of Drug Addiction, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| |
Collapse
|
26
|
Yin JB, Liang SH, Li F, Zhao WJ, Bai Y, Sun Y, Wu ZY, Ding T, Sun Y, Liu HX, Lu YC, Zhang T, Huang J, Chen T, Li H, Chen ZF, Cao J, Ren R, Peng YN, Yang J, Zang WD, Li X, Dong YL, Li YQ. dmPFC-vlPAG projection neurons contribute to pain threshold maintenance and antianxiety behaviors. J Clin Invest 2021; 130:6555-6570. [PMID: 32841213 DOI: 10.1172/jci127607] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 08/20/2020] [Indexed: 12/21/2022] Open
Abstract
The dorsal medial prefrontal cortex (dmPFC) has been recognized as a key cortical area for nociceptive modulation. However, the underlying neural pathway and the function of specific cell types remain largely unclear. Here, we show that lesions in the dmPFC induced an algesic and anxious state. Using multiple tracing methods including a rabies-based transsynaptic tracing method, we outlined an excitatory descending neural pathway from the dmPFC to the ventrolateral periaqueductal gray (vlPAG). Specific activation of the dmPFC/vlPAG neural pathway by optogenetic manipulation produced analgesic and antianxiety effects in a mouse model of chronic pain. Inhibitory neurons in the dmPFC were specifically activated using a chemogenetic approach, which logically produced an algesic and anxious state under both normal and chronic pain conditions. Antagonists of the GABAA receptor (GABAAR) or mGluR1 were applied to the dmPFC, which produced analgesic and antianxiety effects. In summary, the results of our study suggest that the dmPFC/vlPAG neural pathway might participate in the maintenance of pain thresholds and antianxiety behaviors under normal conditions, while silencing or suppressing the dmPFC/vlPAG pathway might be involved in the initial stages and maintenance of chronic pain and the emergence of anxiety-like behaviors.
Collapse
Affiliation(s)
- Jun-Bin Yin
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Department of Neurology, the 960th Hospital of PLA, Jinan, China.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA.,Key Laboratory of Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Haikou, China
| | - Shao-Hua Liang
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Department of Human Anatomy, Binzhou Medical College, Yantai, China
| | - Fei Li
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Cadet Brigade, and
| | - Wen-Jun Zhao
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Cadet Brigade, and
| | - Yang Bai
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yi Sun
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Human Anatomy, Binzhou Medical College, Yantai, China
| | - Zhen-Yu Wu
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tan Ding
- Department of Orthopedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | | | - Hai-Xia Liu
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Ya-Cheng Lu
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Ting Zhang
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Jing Huang
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Tao Chen
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Hui Li
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zhou-Feng Chen
- Center for the Study of Itch, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jing Cao
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Rui Ren
- Key Laboratory of Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Haikou, China
| | - Ya-Nan Peng
- Key Laboratory of Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Haikou, China
| | - Juan Yang
- Key Laboratory of Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Haikou, China
| | - Wei-Dong Zang
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Xiang Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu-Lin Dong
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, China.,Key Laboratory of Brain Science Research and Transformation in the Tropical Environment of Hainan Province, Haikou, China.,Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
27
|
Li JN, Ren JH, Zhao LJ, Wu XM, Li H, Dong YL, Li YQ. Projecting neurons in spinal dorsal horn send collateral projections to dorsal midline/intralaminar thalamic complex and parabrachial nucleus. Brain Res Bull 2021; 169:184-195. [PMID: 33508400 DOI: 10.1016/j.brainresbull.2021.01.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 10/22/2022]
Abstract
Itch is an annoying sensation that always triggers scratching behavior, yet little is known about its transmission pathway in the central nervous system. Parabrachial nucleus (PBN), an essential transmission nucleus in the brainstem, has been proved to be the first relay station in itch sensation. Meanwhile, dorsal midline/intralaminar thalamic complex (dMITC) is proved to be activated with nociceptive stimuli. However, whether the PBN-projecting neurons in spinal dorsal horn (SDH) send collateral projections to dMITC, and whether these projections involve in itch remain unknown. In the present study, a double retrograde tracing method was applied when the tetramethylrhodamine-dextran (TMR) was injected into the dMITC and Fluoro-gold (FG) was injected into the PBN, respectively. Immunofluorescent staining for NeuN, substance P receptor (SPR), substance P (SP), or FOS induced by itch or pain stimulations with TMR and FG were conducted to provide morphological evidence. The results revealed that TMR/FG double-labeled neurons could be predominately observed in superficial laminae and lateral spinal nucleus (LSN) of SDH; Meanwhile, most of the collateral projection neurons expressed SPR and some of them expressed FOS in acute itch model induced by histamine. The present results implicated that some of the SPR-expressing neurons in SDH send collateral projections to the dMITC and PBN in itch transmission, which might be involved in itch related complex affective/emotional processing to the higher brain centers.
Collapse
Affiliation(s)
- Jia-Ni Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jia-Hao Ren
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Liu-Jie Zhao
- Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China
| | - Xue-Mei Wu
- Department of Human Anatomy, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Hui Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Lin Dong
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China; Department of Anatomy, Basic Medical College, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Haikou, China.
| |
Collapse
|
28
|
Cortical Modulation of Nociception. Neuroscience 2021; 458:256-270. [PMID: 33465410 DOI: 10.1016/j.neuroscience.2021.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/28/2020] [Accepted: 01/03/2021] [Indexed: 02/06/2023]
Abstract
Nociception is the neuronal process of encoding noxious stimuli and could be modulated at peripheral, spinal, brainstem, and cortical levels. At cortical levels, several areas including the anterior cingulate cortex (ACC), prefrontal cortex (PFC), ventrolateral orbital cortex (VLO), insular cortex (IC), motor cortex (MC), and somatosensory cortices are involved in nociception modulation through two main mechanisms: (i) a descending modulatory effect at spinal level by direct corticospinal projections or mostly by activation of brainstem structures (i.e. periaqueductal grey matter (PAG), locus coeruleus (LC), the nucleus of raphe (RM) and rostroventral medulla (RVM)); and by (ii) cortico-cortical or cortico-subcortical interactions. This review summarizes evidence related to the participation of the aforementioned cortical areas in nociception modulation and different neurotransmitters or neuromodulators that have been studied in each area. Besides, we point out the importance of considering intracortical neuronal populations and receptors expression, as well as, nociception-induced cortical changes, both functional and connectional, to better understand this modulatory effect. Finally, we discuss the possible mechanisms that could potentiate the use of cortical stimulation as a promising procedure in pain alleviation.
Collapse
|
29
|
A Neural Circuit from Thalamic Paraventricular Nucleus to Central Amygdala for the Facilitation of Neuropathic Pain. J Neurosci 2020; 40:7837-7854. [PMID: 32958568 DOI: 10.1523/jneurosci.2487-19.2020] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 09/04/2020] [Accepted: 09/09/2020] [Indexed: 12/23/2022] Open
Abstract
As one of the thalamic midline nuclei, the thalamic paraventricular nucleus (PVT) is considered to be an important signal integration site for many descending and ascending pathways that modulate a variety of behaviors, including feeding, emotions, and drug-seeking. A recent study has demonstrated that the PVT is implicated in the acute visceral pain response, but it is unclear whether the PVT plays a critical role in the central processing of chronic pain. Here, we report that the neurons in the posterior portion of the PVT (pPVT) and their downstream pathway are involved in descending nociceptive facilitation regarding the development of neuropathic pain conditions in male rats. Lesions or inhibition of pPVT neurons alleviated mechanical allodynia induced by spared nerve injury (SNI). The excitability of pPVT-central amygdala (CeA) projection neurons was significantly increased in SNI rats. Importantly, selective optogenetic activation of the pPVT-CeA pathway induced obvious mechanical hypersensitivity in naive rats. In addition, we used rabies virus (RV)-based and cell-type-specific retrograde transsynaptic tracing techniques to define a novel neuronal circuit in which glutamatergic neurons in the vlPAG were the target of the pPVT-CeA descending facilitation pathway. Our data suggest that this pPVTGlu+-CeA-vlPAGGlu+ circuit mediates central mechanisms of descending pain facilitation underlying persistent pain conditions.SIGNIFICANCE STATEMENT Studies have shown that the interactions between the posterior portion of the thalamic paraventricular nucleus (pPVT) and central amygdala (CeA) play a critical role in pain-related emotional regulation. However, most reports have associated this circuit with fear and anxiety behaviors. Here, an integrative approach of behavioral tests, electrophysiology, and immunohistochemistry was used to advance the novel concept that the pPVT-CeA pathway activation facilitates neuropathic pain processing. Using rabies virus (RV)-based and cell-type-specific retrograde transsynaptic tracing techniques, we found that glutamatergic neurons in the vlPAG were the target of the pPVT-CeA pathway. Thus, this study indicates the involvement of a pPVTGlu+-CeA-vlPAGGlu+ pathway in a descending facilitatory mechanism underlying neuropathic pain.
Collapse
|
30
|
nNOS-expressing neurons in the vmPFC transform pPVT-derived chronic pain signals into anxiety behaviors. Nat Commun 2020; 11:2501. [PMID: 32427844 PMCID: PMC7237711 DOI: 10.1038/s41467-020-16198-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 04/21/2020] [Indexed: 01/30/2023] Open
Abstract
Anxiety is common in patients suffering from chronic pain. Here, we report anxiety-like behaviors in mouse models of chronic pain and reveal that nNOS-expressing neurons in ventromedial prefrontal cortex (vmPFC) are essential for pain-induced anxiety but not algesia, using optogenetic and chemogenetic strategies. Additionally, we determined that excitatory projections from the posterior subregion of paraventricular thalamic nucleus (pPVT) provide a neuronal input that drives the activation of vmPFC nNOS-expressing neurons in our chronic pain models. Our results suggest that the pain signal becomes an anxiety signal after activation of vmPFC nNOS-expressing neurons, which causes subsequent release of nitric oxide (NO). Finally, we show that the downstream molecular mechanisms of NO likely involve enhanced glutamate transmission in vmPFC CaMKIIα-expressing neurons through S-nitrosylation-induced AMPAR trafficking. Overall, our data suggest that pPVT excitatory neurons drive chronic pain-induced anxiety through activation of vmPFC nNOS-expressing neurons, resulting in NO-mediated AMPAR trafficking in vmPFC pyramidal neurons. Chronic pain usually induces anxiety. Here, the authors report that vmPFC nNOS-expressing neurons are activated by excitatory inputs from pPVT during chronic pain and subsequently induce anxiety-like behaviors in mice through promoting AMPAR trafficking.
Collapse
|
31
|
Kummer KK, Mitrić M, Kalpachidou T, Kress M. The Medial Prefrontal Cortex as a Central Hub for Mental Comorbidities Associated with Chronic Pain. Int J Mol Sci 2020; 21:E3440. [PMID: 32414089 PMCID: PMC7279227 DOI: 10.3390/ijms21103440] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic pain patients frequently develop and suffer from mental comorbidities such as depressive mood, impaired cognition, and other significant constraints of daily life, which can only insufficiently be overcome by medication. The emotional and cognitive components of pain are processed by the medial prefrontal cortex, which comprises the anterior cingulate cortex, the prelimbic, and the infralimbic cortex. All three subregions are significantly affected by chronic pain: magnetic resonance imaging has revealed gray matter loss in all these areas in chronic pain conditions. While the anterior cingulate cortex appears hyperactive, prelimbic, and infralimbic regions show reduced activity. The medial prefrontal cortex receives ascending, nociceptive input, but also exerts important top-down control of pain sensation: its projections are the main cortical input of the periaqueductal gray, which is part of the descending inhibitory pain control system at the spinal level. A multitude of neurotransmitter systems contributes to the fine-tuning of the local circuitry, of which cholinergic and GABAergic signaling are particularly emerging as relevant components of affective pain processing within the prefrontal cortex. Accordingly, factors such as distraction, positive mood, and anticipation of pain relief such as placebo can ameliorate pain by affecting mPFC function, making this cortical area a promising target region for medical as well as psychosocial interventions for pain therapy.
Collapse
Affiliation(s)
| | | | | | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.K.K.); (M.M.); (T.K.)
| |
Collapse
|
32
|
Bai Y, Chen YB, Qiu XT, Chen YB, Ma LT, Li YQ, Sun HK, Zhang MM, Zhang T, Chen T, Fan BY, Li H, Li YQ. Nucleus tractus solitarius mediates hyperalgesia induced by chronic pancreatitis in rats. World J Gastroenterol 2019; 25:6077-6093. [PMID: 31686764 PMCID: PMC6824279 DOI: 10.3748/wjg.v25.i40.6077] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/06/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Central sensitization plays a pivotal role in the maintenance of chronic pain induced by chronic pancreatitis (CP). We hypothesized that the nucleus tractus solitarius (NTS), a primary central site that integrates pancreatic afferents apart from the thoracic spinal dorsal horn, plays a key role in the pathogenesis of visceral hypersensitivity in a rat model of CP.
AIM To investigate the role of the NTS in the visceral hypersensitivity induced by chronic pancreatitis.
METHODS CP was induced by the intraductal injection of trinitrobenzene sulfonic acid (TNBS) in rats. Pancreatic hyperalgesia was assessed by referred somatic pain via von Frey filament assay. Neural activation of the NTS was indicated by immunohistochemical staining for Fos. Basic synaptic transmission within the NTS was assessed by electrophysiological recordings. Expression of vesicular glutamate transporters (VGluTs), N-methyl-D-aspartate receptor subtype 2B (NR2B), and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subtype 1 (GluR1) was analyzed by immunoblotting. Membrane insertion of NR2B and GluR1 was evaluated by electron microscopy. The regulatory role of the NTS in visceral hypersensitivity was detected via pharmacological approach and chemogenetics in CP rats.
RESULTS TNBS treatment significantly increased the number of Fos-expressing neurons within the caudal NTS. The excitatory synaptic transmission was substantially potentiated within the caudal NTS in CP rats (frequency: 5.87 ± 1.12 Hz in CP rats vs 2.55 ± 0.44 Hz in sham rats, P < 0.01; amplitude: 19.60 ± 1.39 pA in CP rats vs 14.71 ± 1.07 pA in sham rats; P < 0.01). CP rats showed upregulated expression of VGluT2, and increased phosphorylation and postsynaptic trafficking of NR2B and GluR1 within the caudal NTS. Blocking excitatory synaptic transmission via the AMPAR antagonist CNQX and the NMDAR antagonist AP-5 microinjection reversed visceral hypersensitivity in CP rats (abdominal withdraw threshold: 7.00 ± 1.02 g in CNQX group, 8.00 ± 0.81 g in AP-5 group and 1.10 ± 0.27 g in saline group, P < 0.001). Inhibiting the excitability of NTS neurons via chemogenetics also significantly attenuated pancreatic hyperalgesia (abdominal withdraw threshold: 13.67 ± 2.55 g in Gi group, 2.00 ± 1.37 g in Gq group, and 2.36 ± 0.67 g in mCherry group, P < 0.01).
CONCLUSION Our findings suggest that enhanced excitatory transmission within the caudal NTS contributes to pancreatic pain and emphasize the NTS as a pivotal hub for the processing of pancreatic afferents, which provide novel insights into the central sensitization of painful CP.
Collapse
Affiliation(s)
- Yang Bai
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Ying-Biao Chen
- Department of Anatomy, Fujian Health College, Fuzhou 350101, Fujian Province, China
| | - Xin-Tong Qiu
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Yan-Bing Chen
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Li-Tian Ma
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Ying-Qi Li
- Department of Cardiology, The Second Affiliated Hospital of Xian Jiaotong University, Xian Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Hong-Ke Sun
- Department of Cardiology, The Second Affiliated Hospital of Xian Jiaotong University, Xian Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Ming-Ming Zhang
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Ting Zhang
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Tao Chen
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Bo-Yuan Fan
- Department of Cardiology, The Second Affiliated Hospital of Xian Jiaotong University, Xian Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Hui Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an 710032, Shaanxi Province, China
- Joint Laboratory of Neuroscience at Hainan Medical University and Fourth Military Medical University, Hainan Medical University, Haikou 571199, Hainan Province, China
| |
Collapse
|
33
|
Vierck C. Mechanisms of Below-Level Pain Following Spinal Cord Injury (SCI). THE JOURNAL OF PAIN 2019; 21:262-280. [PMID: 31493490 DOI: 10.1016/j.jpain.2019.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/05/2019] [Accepted: 08/07/2019] [Indexed: 12/18/2022]
Abstract
Mechanisms of below-level pain are discoverable as neural adaptations rostral to spinal injury. Accordingly, the strategy of investigations summarized here has been to characterize behavioral and neural responses to below-level stimulation over time following selective lesions of spinal gray and/or white matter. Assessments of human pain and the pain sensitivity of humans and laboratory animals following spinal injury have revealed common disruptions of pain processing. Interruption of the spinothalamic pathway partially deafferents nocireceptive cerebral neurons, rendering them spontaneously active and hypersensitive to remaining inputs. The spontaneous activity among these neurons is disorganized and unlikely to generate pain. However, activation of these neurons by their remaining inputs can result in pain. Also, injury to spinal gray matter results in a cascade of secondary events, including excitotoxicity, with rostral propagation of excitatory influences that contribute to chronic pain. Establishment and maintenance of below-level pain results from combined influences of injured and spared axons in the spinal white matter and injured neurons in spinal gray matter on processing of nociception by hyperexcitable cerebral neurons that are partially deafferented. A model of spinal stenosis suggests that ischemic injury to the core spinal region can generate below-level pain. Additional questions are raised about demyelination, epileptic discharge, autonomic activation, prolonged activity of C nocireceptive neurons, and thalamocortical plasticity in the generation of below-level pain. PERSPECTIVE: An understanding of mechanisms can direct therapeutic approaches to prevent development of below-level pain or arrest it following spinal cord injury. Among the possibilities covered here are surgical and other means of attenuating gray matter excitotoxicity and ascending propagation of excitatory influences from spinal lesions to thalamocortical systems involved in pain encoding and arousal.
Collapse
Affiliation(s)
- Chuck Vierck
- Department of Neuroscience, University of Florida College of Medicine and McKnight Brain Institute, Gainesville, Florida.
| |
Collapse
|
34
|
Li JN, Sun Y, Ji SL, Chen YB, Ren JH, He CB, Wu ZY, Li H, Dong YL, Li YQ. Collateral Projections from the Medullary Dorsal Horn to the Ventral Posteromedial Thalamic Nucleus and the Parafascicular Thalamic Nucleus in the Rat. Neuroscience 2019; 410:293-304. [PMID: 31075313 DOI: 10.1016/j.neuroscience.2019.04.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 04/24/2019] [Accepted: 04/26/2019] [Indexed: 10/26/2022]
Abstract
Medullary dorsal horn (MDH), the homolog of spinal dorsal horn, plays essential roles in processing of nociceptive signals from orofacial region toward higher centers, such as the ventral posteromedial thalamic nucleus (VPM) and parafascicular thalamic nucleus (Pf), which belong to the sensory-discriminative and affective aspects of pain transmission systems at the thalamic level, respectively. In the present study, in order to provide morphological evidence for whether neurons in the MDH send collateral projections to the VPM and Pf, a retrograde double tracing method combined with immunofluorescence staining for substance P (SP), SP receptor (SPR) and Fos protein was used. Fluoro-gold (FG) was injected into the VPM and the tetramethylrhodamine-dextran (TMR) was injected into the Pf. The result revealed that both FG- and TMR-labeled projection neurons were observed throughout the entire extent of the MDH, while the FG/TMR double-labeled neurons were mainly located in laminae I and III. It was also found that some of the FG/TMR double-labeled neurons within lamina I expressed SPR and were in close contact with SP-immunoreactive (SP-ir) terminals. After formalin injection into the orofacial region, 41.4% and 34.3% of the FG/TMR double-labeled neurons expressed Fos protein in laminae I and III, respectively. The present results provided morphological evidence for that some SPR-expressing neurons within the MDH send collateral projections to both VPM and Pf and might be involved in sensory-discriminative and affective aspects of acute orofacial nociceptive information transmission.
Collapse
Affiliation(s)
- Jia-Ni Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China
| | - Yi Sun
- Department of Human Anatomy, Histology and Embryology, Fujian Medical University, Fuzhou 350108, China
| | - Song-Ling Ji
- Department of Anatomy, The Zunyi Medical Collage, Zunyi 563000, China
| | - Yan-Bing Chen
- Department of Human Anatomy, Histology and Embryology, Fujian Medical University, Fuzhou 350108, China
| | - Jia-Hao Ren
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China
| | - Cheng-Bo He
- Department of Anatomy, The Zunyi Medical Collage, Zunyi 563000, China
| | - Zhen-Yu Wu
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China
| | - Hui Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China
| | - Yu-Lin Dong
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China.
| | - Yun-Qing Li
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Joint Laboratory of Neuroscience at Hainan Medical University and The Fourth Military Medical University, Hainan Medical University, Haikou 571199, China.
| |
Collapse
|
35
|
Jacobson GM, Voss LJ, Klockars A, Bird S, Dimitrov I, Denny WA, Olszewski PK, Sleigh JW, Harvey MG. Transcriptional changes in response to ketamine ester-analogs SN 35210 and SN 35563 in the rat brain. BMC Genomics 2019; 20:281. [PMID: 30971208 PMCID: PMC6458767 DOI: 10.1186/s12864-019-5649-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/27/2019] [Indexed: 12/29/2022] Open
Abstract
Background Ketamine ester analogs, SN 35210 and SN 35563, demonstrate different pharmacological profiles to ketamine in animal models. Both confer hypnosis with predictably rapid offset yet, paradoxically, SN35563 induces a prolonged anti-nociceptive state. To explore underlying mechanisms, broad transcriptome changes were measured and compared across four relevant target regions of the rat brain. Results SN 35563 produced large-scale alteration of gene expression in the Basolateral Amygdala (BLA) and Paraventricular Nucleus of the Thalamus (PVT), in excess of 10x that induced by ketamine and SN 35210. A smaller and quantitatively similar number of gene changes were observed in the Insula (INS) and Nucleus Accumbens (ACB) for all three agents. In the BLA and PVT, SN 35563 caused enrichment for gene pathways related to the function and structure of glutamatergic synapses in respect to: release of neurotransmitter, configuration of postsynaptic AMPA receptors, and the underlying cytoskeletal scaffolding and alignment. Conclusion The analgesic ketamine ester analog SN 35563 induces profound large-scale changes in gene expression in key pain-related brain regions reflecting its unique prolonged pharmacodynamic profile. Electronic supplementary material The online version of this article (10.1186/s12864-019-5649-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gregory M Jacobson
- Faculty of Science and Engineering, The University of Waikato, Hillcrest, Hamilton, 3216, New Zealand.
| | - Logan J Voss
- Waikato District Health Board, Pembroke Street, Hamilton, 3204, New Zealand
| | - Anica Klockars
- Faculty of Science and Engineering, The University of Waikato, Hillcrest, Hamilton, 3216, New Zealand
| | - Steve Bird
- Faculty of Science and Engineering, The University of Waikato, Hillcrest, Hamilton, 3216, New Zealand
| | - Ivo Dimitrov
- The University of Auckland, Grafton, Auckland, 1023, New Zealand
| | - William A Denny
- The University of Auckland, Grafton, Auckland, 1023, New Zealand
| | - Pawel K Olszewski
- Faculty of Science and Engineering, The University of Waikato, Hillcrest, Hamilton, 3216, New Zealand
| | - James W Sleigh
- Waikato District Health Board, Pembroke Street, Hamilton, 3204, New Zealand
| | - Martyn G Harvey
- Waikato District Health Board, Pembroke Street, Hamilton, 3204, New Zealand
| |
Collapse
|
36
|
Kinlein SA, Phillips DJ, Keller CR, Karatsoreos IN. Role of corticosterone in altered neurobehavioral responses to acute stress in a model of compromised hypothalamic-pituitary-adrenal axis function. Psychoneuroendocrinology 2019; 102:248-255. [PMID: 30594817 PMCID: PMC7649055 DOI: 10.1016/j.psyneuen.2018.12.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/01/2018] [Accepted: 12/10/2018] [Indexed: 01/08/2023]
Abstract
An organism's capacity to cope with stressful experiences is dependent on its ability to appropriately engage central and peripheral systems, such as the hypothalamic-pituitary-adrenal (HPA) axis, to adapt to changing environmental demands. The HPA axis is a primary neuroendocrine mediator of neural and behavioral responses to stress, and dysfunction of this system is linked to increased risk for developing mental health disorders such as depression, anxiety, and post-traumatic stress disorder. However, the mechanisms by which dysregulated HPA function results in abnormal behavioral responses to stress are poorly understood. Here, we tested how corticosterone (CORT)-induced HPA axis disruption affects behavioral responses to stress in male C57BL/6 N mice, and probed correlates of these behaviors in the brain. We show that chronic HPA disruption blunts acute stress-induced grooming and rearing behaviors in the open field test, effects which were accompanied by decreased FOS immunoreactivity in the paraventricular nucleus of the hypothalamus (PVH) and paraventricular nucleus of the thalamus (PVT). Blockade of CORT secretion with metyrapone injection prior to acute stress did not recapitulate the effects of chronic HPA disruption on open field behavior, and acute CORT replacement did not rescue normal behavioral stress responses following chronic HPA disruption. This suggests that under acute conditions, CORT is not necessary for these responses normally, nor sufficient to rescue the deficits of chronic HPA dysregulation. Together, these findings support the hypothesis that chronic HPA dysregulation causes adaptation in stress-related brain circuits and demonstrate that these changes can influence an organism's behavioral response to stress exposure.
Collapse
Affiliation(s)
- Scott A. Kinlein
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Derrick J. Phillips
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Chandler R. Keller
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA
| | - Ilia N. Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, USA,Corresponding author: Ilia N. Karatsoreos, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, ., T:509-335-4829
| |
Collapse
|
37
|
Aldrin-Kirk P, Björklund T. Practical Considerations for the Use of DREADD and Other Chemogenetic Receptors to Regulate Neuronal Activity in the Mammalian Brain. Methods Mol Biol 2019; 1937:59-87. [PMID: 30706390 DOI: 10.1007/978-1-4939-9065-8_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Chemogenetics is the process of genetically expressing a macromolecule receptor capable of modulating the activity of the cell in response to selective chemical ligand. This chapter will cover the chemogenetic technologies that are available to date, focusing on the commonly available engineered or otherwise modified ligand-gated ion channels and G-protein-coupled receptors in the context of neuromodulation. First, we will give a brief overview of each chemogenetic approach as well as in vitro/in vivo applications, then we will list their strengths and weaknesses. Finally, we will provide tips for ligand application in each case.Each technology has specific limitations that make them more or less suitable for different applications in neuroscience although we will focus mainly on the most commonly used and versatile family named designer receptors exclusively activated by designer drugs or DREADDs. We here describe the most common cases where these can be implemented and provide tips on how and where these technologies can be applied in the field of neuroscience.
Collapse
Affiliation(s)
- Patrick Aldrin-Kirk
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Tomas Björklund
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.
| |
Collapse
|
38
|
Li J. Chronic myocardial infarction changed the excitatory-inhibitory synaptic balance in the medial prefrontal cortex of rat. Mol Pain 2018; 14:1744806918809586. [PMID: 30303032 PMCID: PMC6243403 DOI: 10.1177/1744806918809586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The medial prefrontal cortex is a key area for the regulation of pain and emotion. However, the functional involvement of the medial prefrontal cortex for visceral nociception, at the neuronal or synaptic level, is obscure yet. In the present study, the properties of excitatory and inhibitory synaptic transmission within the layer II/III of rat medial prefrontal cortex after chronic myocardial infarction were studied. It is found that the excitation–inhibition ratio of the medial prefrontal cortex was greatly changed, with enhanced excitation and decreased inhibition inputs to the pyramidal cells of the medial prefrontal cortex, which largely due to decreased spike firing in gamma-aminobutyric acid-ergic neurons. Behaviorally, inhibition of gamma-aminobutyric acid-ergic synaptic transmission alleviated the visceral pain and anxiety. It is thus for the first time showing that the excitation–inhibition ratio is increased in the medial prefrontal cortex after chronic myocardial infarction, which may come from the reduced intrinsic activity of gamma-aminobutyric acid-ergic neurons and is important for regulating the angina pectoris and anxiety induced by chronic myocardial infarction.
Collapse
Affiliation(s)
- Jing Li
- 1 Department of Psychology, Institute of Public Health, Xi'an Medical University, Xi'an, China.,2 School of Public Health, Institute for Research on Health Information and Technology, Xi'an Medical University, Xi'an, China
| |
Collapse
|
39
|
Abstract
Chemogenetic technologies enable selective pharmacological control of specific cell populations. An increasing number of approaches have been developed that modulate different signaling pathways. Selective pharmacological control over G protein-coupled receptor signaling, ion channel conductances, protein association, protein stability, and small molecule targeting allows modulation of cellular processes in distinct cell types. Here, we review these chemogenetic technologies and instances of their applications in complex tissues in vivo and ex vivo.
Collapse
Affiliation(s)
- Deniz Atasoy
- Department of Physiology, School of Medicine and Regenerative-Restorative Medicine Research Center (REMER), Istanbul Medipol University , Istanbul , Turkey ; and Janelia Research Campus, Howard Hughes Medical Institute , Ashburn, Virginia
| | - Scott M Sternson
- Department of Physiology, School of Medicine and Regenerative-Restorative Medicine Research Center (REMER), Istanbul Medipol University , Istanbul , Turkey ; and Janelia Research Campus, Howard Hughes Medical Institute , Ashburn, Virginia
| |
Collapse
|
40
|
Xie YF, Wang J, Bonin RP. Optogenetic exploration and modulation of pain processing. Exp Neurol 2018; 306:117-121. [PMID: 29729250 DOI: 10.1016/j.expneurol.2018.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/21/2018] [Accepted: 05/01/2018] [Indexed: 12/26/2022]
Abstract
Intractable pain is the single most common cause of disability, affecting more than 20% of the population world-wide. There is accordingly a global effort to decipher how changes in nociceptive processing in the peripheral and central nervous systems contribute to the onset and maintenance of chronic pain. The past several years have brought rapid progress in the adaptation of optogenetic approaches to study and manipulate the activity of sensory afferents and spinal cord neurons in freely behaving animals, and to investigate cortical processing and modulation of pain responses. This review discusses methodological advances that underlie this recent progress, and discusses practical considerations for the optogenetic modulation of nociceptive sensory processing.
Collapse
Affiliation(s)
- Yu-Feng Xie
- Leslie Dan Faculty of Pharmacy, University of Toronto, Canada.
| | - Jing Wang
- The Department of Osteoporosis, the People's Hospital of Baoan District, Shenzhen, Guangdong Province, China
| | - Robert P Bonin
- Leslie Dan Faculty of Pharmacy, University of Toronto, Canada.
| |
Collapse
|
41
|
Stankewitz A, Sorg C, von Kalckreuth A, Schulz E, Valet M, Neufang S, Zimmer C, Henningsen P, Gündel H, Wohlschläger AM, Tölle TR. Fronto-Insular Connectivity during Pain Distraction Is Impaired in Patients with Somatoform Pain. J Neuroimaging 2018; 28:621-628. [PMID: 30028554 DOI: 10.1111/jon.12547] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 07/04/2018] [Accepted: 07/05/2018] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Somatoform pain disorder is characterized by chronic pain and various psychological symptoms including increased attention to mental and physical processes. Given that the medial prefrontal cortex (mPFC) of the default mode network (DMN) and the anterior insula of the salience network are critically involved in intrinsic and attentional processes, we investigated the involvement of these networks during the distraction from physical pain in somatoform pain patients. METHODS During painful and nonpainful heat stimulation, attentional distraction from physical processes was modulated with a Stroop task. Thirteen patients were investigated with functional magnetic resonance imaging (fMRI) and compared to 13 controls. Main outcomes were spatial maps of coherent fMRI activity based on independent component analysis and functional connectivity (FC) resulting from psychophysiological interaction analysis. RESULTS Behavioral pain intensity ratings were reduced during the distraction task in both groups. At brain level, we found deviant network activities in the DMN (particularly in the mPFC) and in the salience network (bilaterally in the anterior insula) in patients. During pain stimulation, Stroop-induced distraction decreased the FC between the mPFC and anterior insula in controls but not in patients. CONCLUSIONS Modulating the FC between the mPFC and the insula may be highly relevant for shifting the attention away from external stimuli, including nociceptive input. The observed alterations in somatoform pain patients may foster new strategies in cognitive behavioral training tools for these patients.
Collapse
Affiliation(s)
- Anne Stankewitz
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christian Sorg
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,TUM Neuroimaging Center, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Alexander von Kalckreuth
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Enrico Schulz
- Department of Neurology, Klinikum der Universität München, Ludwig-Maximilians-Universität, München, Germany
| | - Michael Valet
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Benedictus Krankenhaus, Feldafing, Germany
| | - Susanne Neufang
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Wuerzburg, Germany
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Peter Henningsen
- Department of Psychosomatic Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Harald Gündel
- Department of Psychosomatic Medicine, University of Ulm, Ulm, Germany
| | - Afra M Wohlschläger
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,TUM Neuroimaging Center, Klinikum rechts der Isar, Technische Universität München, Germany
| | - Thomas R Tölle
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
42
|
Xiao X, Zhang YQ. A new perspective on the anterior cingulate cortex and affective pain. Neurosci Biobehav Rev 2018; 90:200-211. [DOI: 10.1016/j.neubiorev.2018.03.022] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/22/2018] [Accepted: 03/22/2018] [Indexed: 12/24/2022]
|
43
|
Bajic D, Monory K, Conrad A, Maul C, Schmid RM, Wotjak CT, Stein-Thoeringer CK. Cannabinoid Receptor Type 1 in the Brain Regulates the Affective Component of Visceral Pain in Mice. Neuroscience 2018; 384:397-405. [PMID: 29885522 DOI: 10.1016/j.neuroscience.2018.05.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 12/16/2022]
Abstract
Endocannabinoids acting through cannabinoid receptor type 1 (CB1) are major modulators of peripheral somatic and visceral nociception. Although only partially studied, some evidence suggests a particular role of CB1 within the brain in nociceptive processes. As the endocannabinoid system regulates affect and emotional behaviors, we hypothesized that cerebral CB1 influences affective processing of visceral pain-related behaviors in laboratory animals. To study nocifensive responses modulated by supraspinal CB1, we used conditional knock-out mice lacking CB1 either in cortical glutamatergic neurons (Glu-CB1-KO), or in forebrain GABAergic neurons (GABA-CB1-KO), or in principal neurons of the forebrain (CaMK-CB1-KO). These mutant mice and mice treated with the CB1 antagonist SR141716 were tested for different pain-related behaviors. In an acetic acid-induced abdominal constriction test, supraspinal CB1 deletions did not affect nocifensive responses. In the cerulein-model of acute pancreatitis, mechanical allodynia or hyperalgesia were not changed, but Glu-CB1- and CaMK-CB1-KO mice showed significantly increased facial grimacing scores indicating increased affective responses to this noxious visceral stimulus. Similarly, these brain-specific CB1 KO mice also showed significantly changed thermal nociception in a hot-plate test. These results reveal a novel, and important role of CB1 expressed by cortical glutamatergic neurons in the affective component of visceral nociception.
Collapse
Affiliation(s)
- Danica Bajic
- Klinik und Poliklinik fuer Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Krisztina Monory
- Institute of Physiological Chemistry, University Medical Center, Mainz, Germany
| | - Andrea Conrad
- Institute of Physiological Chemistry, University Medical Center, Mainz, Germany
| | - Christina Maul
- Institute of Physiological Chemistry, University Medical Center, Mainz, Germany
| | - Roland M Schmid
- Klinik und Poliklinik fuer Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Carsten T Wotjak
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, Munich, Germany
| | - Christoph K Stein-Thoeringer
- Klinik und Poliklinik fuer Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
| |
Collapse
|
44
|
Ong WY, Stohler CS, Herr DR. Role of the Prefrontal Cortex in Pain Processing. Mol Neurobiol 2018; 56:1137-1166. [PMID: 29876878 PMCID: PMC6400876 DOI: 10.1007/s12035-018-1130-9] [Citation(s) in RCA: 413] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/14/2018] [Indexed: 12/16/2022]
Abstract
The prefrontal cortex (PFC) is not only important in executive functions, but also pain processing. The latter is dependent on its connections to other areas of the cerebral neocortex, hippocampus, periaqueductal gray (PAG), thalamus, amygdala, and basal nuclei. Changes in neurotransmitters, gene expression, glial cells, and neuroinflammation occur in the PFC during acute and chronic pain, that result in alterations to its structure, activity, and connectivity. The medial PFC (mPFC) could serve dual, opposing roles in pain: (1) it mediates antinociceptive effects, due to its connections with other cortical areas, and as the main source of cortical afferents to the PAG for modulation of pain. This is a ‘loop’ where, on one side, a sensory stimulus is transformed into a perceptual signal through high brain processing activity, and perceptual activity is then utilized to control the flow of afferent sensory stimuli at their entrance (dorsal horn) to the CNS. (2) It could induce pain chronification via its corticostriatal projection, possibly depending on the level of dopamine receptor activation (or lack of) in the ventral tegmental area-nucleus accumbens reward pathway. The PFC is involved in biopsychosocial pain management. This includes repetitive transcranial magnetic stimulation, transcranial direct current stimulation, antidepressants, acupuncture, cognitive behavioral therapy, mindfulness, music, exercise, partner support, empathy, meditation, and prayer. Studies demonstrate the role of the PFC during placebo analgesia, and in establishing links between pain and depression, anxiety, and loss of cognition. In particular, losses in PFC grey matter are often reversible after successful treatment of chronic pain.
Collapse
Affiliation(s)
- Wei-Yi Ong
- Department of Anatomy, National University of Singapore, Singapore, 119260, Singapore.
- Neurobiology and Ageing Research Programme, National University of Singapore, Singapore, 119260, Singapore.
| | | | - Deron R Herr
- Department of Pharmacology, National University of Singapore, Singapore, 119260, Singapore.
| |
Collapse
|
45
|
Maldonado R, Baños JE, Cabañero D. Usefulness of knockout mice to clarify the role of the opioid system in chronic pain. Br J Pharmacol 2018; 175:2791-2808. [PMID: 29124744 DOI: 10.1111/bph.14088] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 12/29/2022] Open
Abstract
Several lines of knockout mice deficient in the genes encoding each component of the endogenous opioid system have been used for decades to clarify the specific role of the different opioid receptors and peptide precursors in many physiopathological conditions. The use of these genetically modified mice has improved our knowledge of the specific involvement of each endogenous opioid component in nociceptive transmission during acute and chronic pain conditions. The present review summarizes the recent advances obtained using these genetic tools in understanding the role of the opioid system in the pathophysiological mechanisms underlying chronic pain. Behavioural data obtained in these chronic pain models are discussed considering the peculiarities of the behavioural phenotype of each line of knockout mice. These studies have identified the crucial role of specific components of the opioid system in different manifestations of chronic pain and have also opened new possible therapeutic approaches, such as the development of opioid compounds simultaneously targeting several opioid receptors. However, several questions still remain open and require further experimental effort to be clarified. The novel genetic tools now available to manipulate specific neuronal populations and precise genome editing in mice will facilitate in a near future the elucidation of the role of each component of the endogenous opioid system in chronic pain. LINKED ARTICLES This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.
Collapse
Affiliation(s)
- Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Josep Eladi Baños
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
46
|
de la Puente B, Zamanillo D, Romero L, Vela JM, Merlos M, Portillo-Salido E. Pharmacological sensitivity of reflexive and nonreflexive outcomes as a correlate of the sensory and affective responses to visceral pain in mice. Sci Rep 2017; 7:13428. [PMID: 29044171 PMCID: PMC5647413 DOI: 10.1038/s41598-017-13987-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/04/2017] [Indexed: 12/19/2022] Open
Abstract
Pain encompasses both sensory and affective dimensions which can be differentially modulated by drugs. Here, we compare the pharmacological sensitivity of the sensory and affective responses using acetic acid-induced abdominal writhings (sensory-reflexive outcome) and acetic acid-induced depression of reward seeking behaviour (RSB, affective-nonreflexive outcome) to a highly palatable food in mice. We found that the expression of RSB critically depends on factors such as sex and previous knowledge and type of the food stimulus. Intraperitoneal administration of acetic acid (iAA) produced a long-lasting (beyond the resolution of writhing behaviour) and concentration-dependent decrease on both appetitive-approach and consummatory dimensions of RSB. Ibuprofen and diclofenac were much more potent in reversing AA-induced changes in RSB: latency to eat (ED50 = 2 and 0.005 mg/kg, intraperinoneally, respectively) and amount consumed (ED50 = 11 and 0.1 mg/kg) than in AA-induced writhing (ED50 = 123 and 60 mg/kg). Morphine and duloxetine inhibited the writhing response (ED50 = 0.8 and 6 mg/kg, respectively) but not the AA-induced changes in RSB. Caffeine was ineffective in both AA-induced writhing and RSB changes. Overall, this study characterized a preclinical mouse model of hedonic deficits induced by pain that can be used to assess affective responses as well as complementary classic reflexive approaches in the evaluation of candidate analgesics.
Collapse
Affiliation(s)
| | - Daniel Zamanillo
- Drug Discovery and Preclinical Development, ESTEVE, Barcelona, Spain
| | - Luz Romero
- Drug Discovery and Preclinical Development, ESTEVE, Barcelona, Spain
| | - José M Vela
- Drug Discovery and Preclinical Development, ESTEVE, Barcelona, Spain
| | - Manuel Merlos
- Drug Discovery and Preclinical Development, ESTEVE, Barcelona, Spain
| | | |
Collapse
|
47
|
Lyubashina O, Sivachenko I. The 5-HT 4 receptor-mediated inhibition of visceral nociceptive neurons in the rat caudal ventrolateral medulla. Neuroscience 2017; 359:277-288. [DOI: 10.1016/j.neuroscience.2017.07.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/26/2017] [Accepted: 07/17/2017] [Indexed: 12/23/2022]
|
48
|
Martins I, Tavares I. Reticular Formation and Pain: The Past and the Future. Front Neuroanat 2017; 11:51. [PMID: 28725185 PMCID: PMC5497058 DOI: 10.3389/fnana.2017.00051] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/19/2017] [Indexed: 01/10/2023] Open
Abstract
The involvement of the reticular formation (RF) in the transmission and modulation of nociceptive information has been extensively studied. The brainstem RF contains several areas which are targeted by spinal cord afferents conveying nociceptive input. The arrival of nociceptive input to the RF may trigger alert reactions which generate a protective/defense reaction to pain. RF neurons located at the medulla oblongata and targeted by ascending nociceptive information are also involved in the control of vital functions that can be affected by pain, namely cardiovascular control. The RF contains centers that belong to the pain modulatory system, namely areas involved in bidirectional balance (decrease or enhancement) of pain responses. It is currently accepted that the imbalance of pain modulation towards pain facilitation accounts for chronic pain. The medullary RF has the peculiarity of harboring areas involved in bidirectional pain control namely by the existence of specific neuronal populations involved in antinociceptive or pronociceptive behavioral responses, namely at the rostroventromedial medulla (RVM) and the caudal ventrolateral medulla (VLM). Furthermore the dorsal reticular nucleus (also known as subnucleus reticularis dorsalis; DRt) may enhance nociceptive responses, through a reverberative circuit established with spinal lamina I neurons and inhibit wide-dynamic range (WDR) neurons of the deep dorsal horn. The components of the triad RVM-VLM-DRt are reciprocally connected and represent a key gateway for top-down pain modulation. The RVM-VLM-DRt triad also represents the neurobiological substrate for the emotional and cognitive modulation of pain, through pathways that involve the periaqueductal gray (PAG)-RVM connection. Collectively, we propose that the RVM-VLM-DRt triad represents a key component of the “dynamic pain connectome” with special features to provide integrated and rapid responses in situations which are life-threatening and involve pain. The new available techniques in neurobiological studies both in animal and human studies are producing new and fascinating data which allow to understand the complex role of the RF in pain modulation and its integration with several body functions and also how the RF accounts for chronic pain.
Collapse
Affiliation(s)
- Isabel Martins
- Departamento de Biomedicina, Faculdade de Medicina do PortoPorto, Portugal.,Unidade de Biologia Experimental, Faculdade de Medicina do Porto, Universidade do PortoPorto, Portugal.,Instituto de Biologia Celular e Molecular (IBMC), Universidade do PortoPorto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S)Porto, Portugal
| | - Isaura Tavares
- Departamento de Biomedicina, Faculdade de Medicina do PortoPorto, Portugal.,Unidade de Biologia Experimental, Faculdade de Medicina do Porto, Universidade do PortoPorto, Portugal.,Instituto de Biologia Celular e Molecular (IBMC), Universidade do PortoPorto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto (I3S)Porto, Portugal
| |
Collapse
|
49
|
Geng KW, He T, Wang RR, Li CL, Luo WJ, Wu FF, Wang Y, Li Z, Lu YF, Guan SM, Chen J. Ethanol Increases Mechanical Pain Sensitivity in Rats via Activation of GABAA Receptors in Medial Prefrontal Cortex. Neurosci Bull 2016; 32:433-44. [PMID: 27628528 DOI: 10.1007/s12264-016-0063-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/14/2016] [Indexed: 12/23/2022] Open
Abstract
Ethanol is widely known for its ability to cause dramatic changes in emotion, social cognition, and behavior following systemic administration in humans. Human neuroimaging studies suggest that alcohol dependence and chronic pain may share common mechanisms through amygdala-medial prefrontal cortex (mPFC) interactions. However, whether acute administration of ethanol in the mPFC can modulate pain perception is unknown. Here we showed that bilateral microinjections of ethanol into the prelimbic and infralimbic areas of the mPFC lowered the bilateral mechanical pain threshold for 48 h without influencing thermal pain sensitivity in adult rats. However, bilateral microinjections of artificial cerebrospinal fluid into the mPFC or bilateral microinjections of ethanol into the dorsolateral PFC (also termed as motor cortex area 1 in Paxinos and Watson's atlas of The Rat Brain. Elsevier Academic Press, Amsterdam, 2005) failed to do so, suggesting regional selectivity of the effects of ethanol. Moreover, bilateral microinjections of ethanol did not change the expression of either pro-apoptotic (caspase-3 and Bax) or anti-apoptotic (Bcl-2) proteins, suggesting that the dose was safe and validating the method used in the current study. To determine whether γ-aminobutyric acid A (GABAA) receptors are involved in mediating the ethanol effects, muscimol, a selective GABAA receptor agonist, or bicuculline, a selective GABAA receptor antagonist, was administered alone or co-administered with ethanol through the same route into the bilateral mPFC. The results showed that muscimol mimicked the effects of ethanol while bicuculline completely reversed the effects of ethanol and muscimol. In conclusion, ethanol increases mechanical pain sensitivity through activation of GABAA receptors in the mPFC of rats.
Collapse
Affiliation(s)
- Kai-Wen Geng
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Ting He
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Rui-Rui Wang
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Chun-Li Li
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Wen-Jun Luo
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Fang-Fang Wu
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Yan Wang
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Zhen Li
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China
| | - Yun-Fei Lu
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Su-Min Guan
- School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Jun Chen
- Institute for Biomedical Sciences of Pain and Institute for Functional Brain Disorders, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.
- Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an, 710038, China.
- Beijing Institute for Brain Disorders, Beijing, 100069, China.
| |
Collapse
|
50
|
Liang SH, Yin JB, Sun Y, Bai Y, Zhou KX, Zhao WJ, Wang W, Dong YL, Li YQ. Collateral projections from the lateral parabrachial nucleus to the paraventricular thalamic nucleus and the central amygdaloid nucleus in the rat. Neurosci Lett 2016; 629:245-250. [PMID: 27423318 DOI: 10.1016/j.neulet.2016.07.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/12/2016] [Indexed: 10/21/2022]
Abstract
Combined the retrograde double tracing with immunofluorescence histochemical staining, we examined the neurons in the lateral parabrachial nucleus (LPB) sent collateral projections to the paraventricular thalamic nucleus (PVT) and central amygdaloid nucleus (CeA) and their roles in the nociceptive transmission in the rat. After the injection of Fluoro-gold (FG) into the PVT and tetramethylrhodamine-dextran (TMR) into the CeA, respectively, FG/TMR double-labeled neurons were observed in the LPB. The percentages of FG/TMR double-labeled neurons to the total number of FG- or TMR-labeled neurons were 6.18% and 9.09%, respectively. Almost all of the FG/TMR double-labeled neurons (95%) exhibited calcitonin gene-related peptide (CGRP) immunoreactivity. In the condition of neuropathic pain, 94% of these neurons showed FOS immunoreactivity. The present data indicates that some of CGRP-expressing neurons in the LPB may transmit nociceptive information toward the PVT and CeA by way of axon collaterals.
Collapse
Affiliation(s)
- Shao-Hua Liang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Jun-Bin Yin
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China
| | - Yi Sun
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Yang Bai
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China
| | - Kai-Xiang Zhou
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Student Brigade, The Fourth Military Medical University, Xi'an 710032, China
| | - Wen-Jun Zhao
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Student Brigade, The Fourth Military Medical University, Xi'an 710032, China
| | - Wei Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.
| | - Yu-Lin Dong
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China.
| | - Yun-Qing Li
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China; Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|