1
|
Zou Y, He X, Ye Z, Li Z, Guo Q, Zou W, Peng Q. Inhibition of the glutamatergic PVT-NAc projections attenuates local anesthetic-induced neurotoxic behaviors. Reg Anesth Pain Med 2025; 50:243-251. [PMID: 38233353 DOI: 10.1136/rapm-2023-104964] [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: 08/29/2023] [Accepted: 12/28/2023] [Indexed: 01/19/2024]
Abstract
INTRODUCTION Local anesthetic-induced neurotoxicity contributes to perioperative nerve damage; however, the underlying mechanisms remain unclear. Here, we investigated the role of the paraventricular thalamus (PVT)-nucleus accumbens (NAc) projections in neurotoxicity induced by ropivacaine, a local anesthetic agent. METHODS Ropivacaine (58 mg/kg, intraperitoneal administration) was used to construct the local anesthetic systemic toxicity (LAST) mice model. We first identified neural projections from the PVT to the NAc through the expression of a retrograde tracer and virus. The inhibitory viruses (rAAV-EF1α-DIO-hm4D(Gi)-mCherry-WPREs: AAV2/retro and rAAV-CaMKII-CRE-WPRE-hGh: AAV2/9) were injected into the mice model to assess the effects of the specific inhibition of the PVT-NAc pathway on neurological behaviors in the presence of clozapine-N-oxide. The inhibition of the PVT-NAc pathway was evaluated by immunofluorescence staining of c-Fos-positive neurons and Ca2+ signals in CaMKIIa neurons. RESULTS We successfully identified a circuit connecting the PVT and NAc in C57BL/6 mice. Ropivacaine administration induced the activation of the PVT-NAc pathway and seizures. Specific inhibition of NAc-projecting CaMKII neurons in the PVT was sufficient to inhibit the neuronal activity in the NAc, which subsequently decreased ropivacaine-induced neurotoxicity. CONCLUSION These results reveal the presence of a dedicated PVT-NAc circuit that regulates local anesthetic-induced neurotoxicity and provide a potential mechanistic explanation for the treatment and prevention of LAST.
Collapse
Affiliation(s)
- Yu Zou
- Department of Anesthesia, Xiangya Hospital Central South University, Changsha, Hunan Province, China
| | - Xin He
- Department of Anesthesia, Xiangya Hospital Central South University, Changsha, Hunan Province, China
| | - Zhiwen Ye
- Department of Critical Care Medicine, Xiangya Hospital Central South University, Changsha, Hunan Province, China
- Xiangya Hospital Central South University National Clinical Research Center for Geriatric Disorders, Changsha, Hunan Province, China
| | - Zhengyiqi Li
- Department of Anesthesia, Xiangya Hospital Central South University, Changsha, Hunan Province, China
| | - Qulian Guo
- Department of Anesthesia, Xiangya Hospital Central South University, Changsha, Hunan Province, China
| | - Wangyuan Zou
- Department of Anesthesia, Xiangya Hospital Central South University, Changsha, Hunan Province, China
| | - Qianyi Peng
- Department of Critical Care Medicine, Xiangya Hospital Central South University, Changsha, Hunan Province, China
- Xiangya Hospital Central South University National Clinical Research Center for Geriatric Disorders, Changsha, Hunan Province, China
| |
Collapse
|
2
|
Qiu G, Wang P, Rao J, Qing X, Cao C, Wang D, Mei B, Zhang J, Liu H, Yang Z, Liu X. Dexmedetomidine Inhibits Paraventricular Corticotropin-releasing Hormone Neurons that Attenuate Acute Stress-induced Anxiety-like Behavior in Mice. Anesthesiology 2024; 140:1134-1152. [PMID: 38498811 DOI: 10.1097/aln.0000000000004982] [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: 03/20/2024]
Abstract
BACKGROUND Dexmedetomidine has repeatedly shown to improve anxiety, but the precise neural mechanisms underlying this effect remain incompletely understood. This study aims to explore the role of corticotropin-releasing hormone-producing hypothalamic paraventricular nucleus (CRHPVN) neurons in mediating the anxiolytic effects of dexmedetomidine. METHODS A social defeat stress mouse model was used to evaluate the anxiolytic effects induced by dexmedetomidine through the elevated plus maze, open-field test, and measurement of serum stress hormone levels. In vivo Ca2+ signal fiber photometry and ex vivo patch-clamp recordings were used to determine the excitability of CRHPVN neurons and investigate the specific mechanism involved. CRHPVN neuron modulation was achieved through chemogenetic activation or inhibition. RESULTS Compared with saline, dexmedetomidine (40 µg/kg) alleviated anxiety-like behaviors. Additionally, dexmedetomidine reduced CRHPVN neuronal excitability. Chemogenetic activation of CRHPVN neurons decreased the time spent in the open arms of the elevated plus maze and in the central area of the open-field test. Conversely, chemogenetic inhibition of CRHPVN neurons had the opposite effect. Moreover, the suppressive impact of dexmedetomidine on CRHPVN neurons was attenuated by the α2-receptor antagonist yohimbine. CONCLUSIONS The results indicate that the anxiety-like effects of dexmedetomidine are mediated via α2-adrenergic receptor-triggered inhibition of CRHPVN neuronal excitability in the hypothalamus. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Gaolin Qiu
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Peng Wang
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Jin Rao
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Xin Qing
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Chenchen Cao
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Dijia Wang
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Bin Mei
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Jiqian Zhang
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Hu Liu
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Zhilai Yang
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Xuesheng Liu
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Zhou Q, Jiang N, Dong Y, Tian K. Dexmedetomidine alleviates anxiety-like behaviors in female mice with musculoskeletal pain through SIRT1/p53 axis. Brain Res Bull 2023; 201:110698. [PMID: 37406884 DOI: 10.1016/j.brainresbull.2023.110698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/25/2023] [Accepted: 07/02/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Musculoskeletal pain is the most common form of chronic pain. Anxiety increases pain intensity and appears to have a major impact on the prevalence and also disability of musculoskeletal pain in women. We examined the effect of dexmedetomidine (DEX) on anxiety-like behaviors associated with musculoskeletal pain and the underlying molecular mechanism in female mice. METHODS Musculoskeletal pain was induced by injection of acidified saline into the gastrocnemius muscle in adult female mice, and the von Frey filament test is used to measure mechanical sensitivity. DEX and EX527 (SIRT1 inhibitor) were administered after modelling. Behavioral tests were used for anxiety and motor activity tests. SIRT1, p53 and acetyl-p53 were quantified by Western blot. RESULTS Adult female mice with musculoskeletal pain exhibit increased fear-like behavior by reducing SIRT1 expression in the medial prefrontal cortex (mPFC). While administration of DEX was able to alleviate mechanical hypersensitivity and anxiety-like behaviors by blocking SIRT1 decline and acetyl-p53 upregulation in mPFC, EX527 inhibited acetyl-p53 rise and reversed the antinociceptive and anxiolytic effects of DEX. CONCLUSION DEX may alleviate anxiety-like behaviors in mice with musculoskeletal pain via the SIRT1/p53 axis. These results suggest that DEX may have a potential therapeutic role in musculoskeletal pain-induced anxiety.
Collapse
Affiliation(s)
- Quan Zhou
- Department of Anesthesiology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China; School of Psychology, Shenzhen University, Shenzhen, 518060, China
| | - Ningbin Jiang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital,Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Yinv Dong
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ke Tian
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
4
|
Yin JB, Liu HX, Shi W, Ding T, Hu HQ, Guo HW, Jin S, Wang XL, Zhang T, Lu YC, Cao BZ. Various BDNF administrations attenuate SPS-induced anxiety-like behaviors. Neurosci Lett 2022; 788:136851. [PMID: 36007708 DOI: 10.1016/j.neulet.2022.136851] [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: 04/03/2022] [Revised: 08/02/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022]
Abstract
Post-traumatic stress disorder (PTSD) has become epidemic following severely stressful incidents. Previous studies have shown that brain-derived neurotrophic factor (BDNF) has anxiolytic effects on various anxiety or depression disorders including PTSD. However, the detailed mechanisms of BDNF for treating PTSD were rarely investigated. In the current study, single-prolonged stress (SPS) was used as an animal model recapitulating specific aspects for a PTSD-like phenotype. The effects of BDNF on SPS-induced anxiety-like behaviors were investigated. We showed that the levels of BDNF in the cerebro-spinal fluid (CSF) were significantly reduced after the rats experienced SPS. The SPS-induced reductions of percentages of time spent in the central area to total time in the open field test, were dose-dependently mitigated after BDNF intracerebroventricular (i.c.v.) injections. BDNF i.c.v. administration also dose-dependently increased the preference of the light box in the light-dark box test. Both expressions of tyrosine kinase receptor B (TrkB) protein and mRNA in the prefrontal cortex (PFC) and amygdala were significantly increased after SPS challenges. BDNF i.c.v. administration attenuated these compensatory increases of TrkB. At last, the anxiolytic effects of BDNF on SPS model were also observed by using other two injection methods. These results inspired us to study that different administrations of BDNF were used in patients with PTSD in the future, in-depthly.
Collapse
Affiliation(s)
- Jun-Bin Yin
- Department of Neurology, The 960(th) Hospital of Joint Logistics Support, PLA, Jinan 250031, PR China; Department of Human Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Hai-Xia Liu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Jinan 250021, PR China
| | - Wei Shi
- Department of Neurosurgery, The 960th Hospital of Joint Logistics Force, PLA, Jinan 250031, PR China
| | - Tan Ding
- Department of Human Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, PR China; Institute of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Huai-Qiang Hu
- Department of Neurology, The 960(th) Hospital of Joint Logistics Support, PLA, Jinan 250031, PR China
| | - Hong-Wei Guo
- Department of Neurology, The 960(th) Hospital of Joint Logistics Support, PLA, Jinan 250031, PR China
| | - Shan Jin
- Department of Neurology, The 960(th) Hospital of Joint Logistics Support, PLA, Jinan 250031, PR China
| | - Xiao-Ling Wang
- Department of Neurology, The 960(th) Hospital of Joint Logistics Support, PLA, Jinan 250031, PR China
| | - Ting Zhang
- Department of Human Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Ya-Cheng Lu
- Department of Human Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Bing-Zhen Cao
- Department of Neurology, The 960(th) Hospital of Joint Logistics Support, PLA, Jinan 250031, PR China.
| |
Collapse
|
5
|
Unchiti K, Leurcharusmee P, Samerchua A, Pipanmekaporn T, Chattipakorn N, Chattipakorn SC. The potential role of dexmedetomidine on neuroprotection and its possible mechanisms: Evidence from in vitro and in vivo studies. Eur J Neurosci 2021; 54:7006-7047. [PMID: 34561931 DOI: 10.1111/ejn.15474] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022]
Abstract
Neurological disorders following brain injuries and neurodegeneration are on the rise worldwide and cause disability and suffering in patients. It is crucial to explore novel neuroprotectants. Dexmedetomidine, a selective α2-adrenoceptor agonist, is commonly used for anxiolysis, sedation and analgesia in clinical anaesthesia and critical care. Recent studies have shown that dexmedetomidine exerts protective effects on multiple organs. This review summarized and discussed the current neuroprotective effects of dexmedetomidine, as well as the underlying mechanisms. In preclinical studies, dexmedetomidine reduced neuronal injury and improved functional outcomes in several models, including hypoxia-induced neuronal injury, ischaemic-reperfusion injury, intracerebral haemorrhage, post-traumatic brain injury, anaesthetic-induced neuronal injury, substance-induced neuronal injury, neuroinflammation, epilepsy and neurodegeneration. Several mechanisms are associated with the neuroprotective function of dexmedetomidine, including neurotransmitter regulation, inflammatory response, oxidative stress, apoptotic pathway, autophagy, mitochondrial function and other cell signalling pathways. In summary, dexmedetomidine has the potential to be a novel neuroprotective agent for a wide range of neurological disorders.
Collapse
Affiliation(s)
- Kantarakorn Unchiti
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Prangmalee Leurcharusmee
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Artid Samerchua
- Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Tanyong Pipanmekaporn
- Department of Anesthesiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
6
|
The Anticonvulsant Effects of Alpha-2 Adrenoceptor Agonist Dexmedetomidine on Pentylenetetrazole-Induced Seizures in Rats. Neurochem Res 2021; 47:305-314. [PMID: 34491515 DOI: 10.1007/s11064-021-03445-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/12/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
Alpha2-adrenoreceptor (α2-AR) is a noradrenergic receptor that is frequently studied for modulation of seizure activity. However, the precise role of this receptor agonists in regulating seizure activity is still unclear. Our aim in this study was to investigate the effects of α2-AR agonist dexmedetomidine (DEX) and atipamezole (α2-AR antagonist, ATI) on seziures in rats. In the study, 32 adult male Wistar Albino rats (weighing 220-260 g) were used. To induce seizures in rats, pentylenetetrazole (PTZ, 35 mg/kg) was injected intraperitoneally (i.p.) and seizure stages were determined according to the Racine scale. After induction of seizures, DEX (0.1 mg/kg, i.p.) and ATI (1 mg/kg, i.p.) were administered to rats and their effects determined on seizures. GABA levels of the brain hippocampal tissue sample were measured using an ELISA kit and c-Fos positive cells of the dentate gyrus and hippocampal regions were quantitatively analyzed with Image J software. The results showed that DEX decreased the seizure stages according to the Racine scale, significantly prolonged the onset time of first myoclonic jerk (FMJ) and reduced the number of spikes and percentage seizure duration (p < 0.05). In contrast, ATI increased the seizure stage, the number of spikes and percentage seizure duration. The hippocampal GABA level was significantly decreased in rats with only PTZ injection (p < 0.05). In addition, DEX reduced the number of c-Fos positive cells in dentate gyrus and the hippocampal CA1 and CA3 regions. In conclusion, our findings showed that α2-AR agonist DEX had a reducing activity on PTZ-induced seizure, while α2-AR antagonist ATI facilitated seizure formation.
Collapse
|
7
|
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
|
8
|
TRPV1 Contributes to the Neuroprotective Effect of Dexmedetomidine in Pilocarpine-Induced Status Epilepticus Juvenile Rats. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7623635. [PMID: 32337274 PMCID: PMC7168755 DOI: 10.1155/2020/7623635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/08/2020] [Accepted: 02/20/2020] [Indexed: 11/25/2022]
Abstract
To investigate the antiepileptic and neuroprotective effects of dexmedetomidine (Dex) in pilocarpine- (Pilo-) induced status epilepticus (SE) juvenile rats, rats were randomly assigned to the following six groups (n = 20): normal, normal+Dex, SE, SE+Cap, SE+Dex, and SE+Dex+Cap. The rats were treated with either diazepam (i.p., an antiepileptic drug) or Dex after the onset of SE. The Morris water maze was used to assess rat cognitive behavior. Flow cytometry was used to detect the concentrations of Ca2+, mitochondrial membrane potential, and reactive oxygen species. Transmission electron microscopy was performed to evaluate specimens of brain tissue. The levels of caspase 3 and TRPV1 were examined by western blot and immunohistochemistry (IHC). Treatment with Dex significantly decreased the escape latency of the SE rats (P < 0.05). Capsaicin, a TRPV1 agonist, delivery aggravated the performance of SE rats. Pathological changes in SE rat were attenuated by Dex and deteriorated by capsaicin. Swollen mitochondria and abnormal endoplasmic reticulum were found in SE rats and were then aggravated by capsaicin and reversed by Dex. Moreover, our data showed that Dex significantly restrained calcium overload, ROS production, and mitochondrial membrane potential loss, all of which were induced by Pilo and capsaicin (P < 0.05). Dex decreased the apoptotic rate in the Model SE group (P < 0.05) and TRPV1 and caspase 3 expression in the Dex treatment group (P < 0.05). Interestingly, all these effects of Dex were partially counteracted by the TRPV1 agonist, capsaicin (P < 0.05). Our study showed that Dex exerted a neuroprotective effect in Pilo-induced SE rats by inhibiting TRPV1 expression and provided information for therapy to SE patients.
Collapse
|
9
|
Lv K, Yang C, Xiao R, Yang L, Liu T, Zhang R, Fan X. Dexmedetomidine attenuates ethanol-induced inhibition of hippocampal neurogenesis in neonatal mice. Toxicol Appl Pharmacol 2020; 390:114881. [PMID: 31954762 DOI: 10.1016/j.taap.2020.114881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND/AIMS Ethanol (EtOH) exposure during a period comparable to the third trimester in human results in obvious neurotoxicity in the developing hippocampus and persistent deficits in hippocampal neurogenesis. Dexmedetomidine (DEX), a highly selective α-2-adrenergic agonist has been demonstrated to restore the impaired neurogenesis and neuronal plasticity in the dentate gyrus (DG) that follows neurological insult. However, the protective roles of DEX in the EtOH-induced deficits of postnatal neurogenesis in the hippocampus are still unknown. METHODS Mice were pretreated with DEX prior to EtOH exposure to determine its protective effects on impaired postnatal hippocampal neurogenesis. Six-day-old neonatal mice were treated with DEX (125 μg/kg) or saline, followed by EtOH at a total of 5 g/kg or an equivalent volume of saline on P7. Immunohistochemistry and immunofluorescence were used to evaluate the neurogenesis and activated microglia in the DG. Quantitative real time PCR (qRT-PCR) was utilized to assess the expression of inflammatory factors in the hippocampus. RESULTS DEX pretreatment attenuated the inhibition of EtOH-mediated hippocampal neurogenesis and the reduction of hippocampal neural precursor cells (NPCs). We further confirmed that DEX pretreatment reversed the EtOH-induced microglia activation in the DG as well as the upregulation of the hippocampal TNFα, MCP-1, IL-6, and IL-1β mRNA levels. CONCLUSION Our findings indicate that DEX pretreatment protects against EtOH-mediated inhibition of hippocampal neurogenesis in postnatal mice and reverses EtOH-induced neuroinflammation via repressing microglia activation and the expression of inflammatory cytokines.
Collapse
Affiliation(s)
- Keyi Lv
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China
| | - Congwen Yang
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Rui Xiao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China
| | - Ling Yang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China
| | - Tianyao Liu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China
| | - Ruiyu Zhang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
10
|
Qiu Z, Lu P, Wang K, Zhao X, Li Q, Wen J, Zhang H, Li R, Wei H, Lv Y, Zhang S, Zhang P. Dexmedetomidine Inhibits Neuroinflammation by Altering Microglial M1/M2 Polarization Through MAPK/ERK Pathway. Neurochem Res 2019; 45:345-353. [PMID: 31823113 DOI: 10.1007/s11064-019-02922-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/18/2019] [Accepted: 11/25/2019] [Indexed: 12/31/2022]
Abstract
Neuroinflammation is critical in the pathogenesis of neurological diseases. Microglial pro-inflammatory (M1) and anti-inflammatory (M2) status determines the outcome of neuroinflammation. Dexmedetomidine exerts anti-inflammatory effects in many neurological conditions. Whether dexmedetomidine functions via modulation of microglia M1/M2 polarization remains to be fully elucidated. In the present study, we investigated the anti-inflammatory effects of dexmedetomidine on the neuroinflammatory cell model and explored the potential mechanism. BV2 cells were stimulated with LPS to establish a neuroinflammatory model. The cell viability was determined with MTT assay. NO levels were assessed using a NO detection kit. The protein levels of IL-10, TNF-α, iNOS, CD206, ERK1/2, and pERK1/2 were quantified using Western blotting. LPS significantly increased pro-inflammatory factors TNF-α and NO, and M1 phenotypic marker iNOS, and decreased anti-inflammatory factor IL-10 and M2 phenotypic marker CD206 in BV2 cells. Furthermore, exposure of BV2 cells to LPS significantly raised pERK1/2 expression. Pretreatment with dexmedetomidine attenuated LPS-elicited changes in p-ERK, iNOS, TNF-α, NO, CD206 and IL-10 levels in BV2 cells. However, co-treatment with dexmedetomidine and LM22B-10, an agonist of ERK, reversed dexmedetomidine-elicited changes in p-ERK, iNOS, TNF-α, NO, CD206 and IL-10 levels in LPS-exposed BV2 cells. We, for the first time, showed that dexmedetomidine increases microglial M2 polarization by inhibiting phosphorylation of ERK1/2, by which it exerts anti-inflammatory effects in BV2 cells.
Collapse
Affiliation(s)
- Zhengguo Qiu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China.,Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Medical University, Shaanxi, 710038, China
| | - Pan Lu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Kui Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Xijuan Zhao
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Qianqian Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Jieqiong Wen
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Hong Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Rong Li
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Yuying Lv
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China
| | - Shuyue Zhang
- Institute of Neurobiology, National Key Academic Subject of Physiology of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157# West 5 Road, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
11
|
Wang S, Lin Q, Wang Z, Pan X. Ropivacaine induces neurotoxicity by activating MAPK/p38 signal to upregulate Fas expression in neurogliocyte. Neurosci Lett 2019; 706:7-11. [DOI: 10.1016/j.neulet.2019.04.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/19/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023]
|
12
|
Tan X, Tu Z, Han W, Song X, Cheng L, Chen H, Tu S, Li P, Liu W, Jiang L. Anticonvulsant and Neuroprotective Effects of Dexmedetomidine on Pilocarpine-Induced Status Epilepticus in Rats Using a Metabolomics Approach. Med Sci Monit 2019; 25:2066-2078. [PMID: 30892279 PMCID: PMC6437718 DOI: 10.12659/msm.912283] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Status epilepticus (SE) is the most extreme form of seizure. It is a medical and neurological emergency that requires prompt and appropriate treatment and early neuroprotection. Dexmedetomidine (DEX) is mainly used for its sedative, analgesic, anxiolytic, and neuroprotective effects with light respiratory depression. The purpose of this study was to comprehensively analyze the metabolic events associated with anticonvulsion and neuroprotection of DEX on pilocarpine-induced status epilepticus rats by LC-MS/MS-based on metabolomics methods combined with histopathology. Material/Methods In this research, rats were divided into 3 groups: a normal group, an SE group, and an SE+DEX group. Hippocampus of rats from each group were collected for further LC-MS/MS-based metabolomic analysis. We collected brains for HE staining and Nissl staining. Multivariate analysis and KEGG enrichment analysis were performed. Results Results of metabolic profiles of the hippocampus tissues of rats proved that dexmedetomidine relieved rats suffering from the status epilepticus by restoring the damaged neuromodulatory metabolism and neurotransmitters, reducing the disturbance in energy, improving oxidative stress, and alleviating nucleic acid metabolism and amino acid in pilocarpine-induced status epilepticus rats. Conclusions This integral metabolomics research provides an extremely effective method to access the therapeutic effects of DEX. This research will further development of new treats for status epilepticus and provide new insights into the anticonvulsive and neuroprotective effects of DEX on status epilepticus.
Collapse
Affiliation(s)
- Xingqin Tan
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China (mainland).,Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Zhenzhen Tu
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China (mainland).,Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Wei Han
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China (mainland)
| | - Xiaojie Song
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China (mainland)
| | - Li Cheng
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China (mainland)
| | - Hengsheng Chen
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China (mainland)
| | - Shengfen Tu
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China (mainland).,Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Pan Li
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China (mainland).,Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Wei Liu
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China (mainland).,Department of Anesthesiology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| | - Li Jiang
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China (mainland).,Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China (mainland)
| |
Collapse
|
13
|
Zou Y, He X, Peng QY, Guo QL. Inhibition of CD38/Cyclic ADP-ribose Pathway Protects Rats against Ropivacaine-induced Convulsion. Chin Med J (Engl) 2018; 130:2354-2360. [PMID: 28937043 PMCID: PMC5634088 DOI: 10.4103/0366-6999.215333] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The CD38/cyclic ADP-ribose (cADPR) pathway plays a role in various central nervous system diseases and in morphine tolerance, but its role in local anesthetic intoxication is unknown. The aim of this study was to determine the role of the CD38/cADPR pathway in ropivacaine-induced convulsion. METHODS Forty male Sprague-Dawley rats were randomly divided into five groups (n = 8 per group): sham group, ropivacaine group, ropivacaine+8-Br-cADPR (5 nmol) group, ropivacaine+8-Br-cADPR (10 nmol) group, and ropivacaine+8-Br-cADPR (20 nmol) group (no rats died). Rats were intracerebroventricularly injected with normal saline or 8-Br-cADPR 30 min before receiving an intraperitoneal injection of ropivacaine. Electroencephalography and convulsion behavior scores were recorded. The hippocampus was harvested from each group and subjected to nicotinamide adenine dinucleotide and cADPR assays, Western blotting analysis, and malondialdehyde (MDA) and superoxide dismutase (SOD) assays. RESULTS Intraperitoneal injection of ropivacaine (33.8 mg/kg) induced convulsions in rats. CD38 and cADPR levels increased significantly following ropivacaine-induced convulsion (P = 0.031 and 0.020, respectively, compared with the sham group). Intraventricular injection of 8-Br-cADPR (5, 10, and 20 nmol) significantly prolonged convulsion latency (P = 0.037, 0.034, and 0.000, respectively), reduced convulsion duration (P = 0.005, 0.005, and 0.005, respectively), and reduced convulsion behavior scores (P = 0.015, 0.015, and 0.000, respectively). Intraventricular injection of 8-Br-cADPR (10 nmol) also increased the B-cell lymphoma-2 (Bcl-2)/Bcl-2-associated X protein ratio (P = 0.044) and reduced cleaved Caspase 3/Caspase 3 ratio, inducible nitric oxide synthase, MDA and SOD levels (P = 0.014, 0.044, 0.001, and 0.010, respectively) compared with the ropivacaine group. CONCLUSIONS The CD38/cADPR pathway is activated in ropivacaine-induced convulsion. Inhibiting this pathway alleviates ropivacaine-induced convulsion and protects the brain from apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Yu Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xin He
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Qian-Yi Peng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Qu-Lian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
14
|
Zhang L, Yin JB, Hu W, Zhao WJ, Fan QR, Qiu ZC, He MJ, Ding T, Sun Y, Kaye AD, Wang ER. Analgesic Effects of Duloxetine on Formalin-Induced Hyperalgesia and Its Underlying Mechanisms in the CeA. Front Pharmacol 2018; 9:317. [PMID: 29692727 PMCID: PMC5902556 DOI: 10.3389/fphar.2018.00317] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/20/2018] [Indexed: 12/17/2022] Open
Abstract
In rodents, the amygdala has been proposed to serve as a key center for the nociceptive perception. Previous studies have shown that extracellular signal-regulated kinase (ERK) signaling cascade in the central nucleus of amygdala (CeA) played a functional role in inflammation-induced peripheral hypersensitivity. Duloxetine (DUL), a serotonin and noradrenaline reuptake inhibitor, produced analgesia on formalin-induced spontaneous pain behaviors. However, it is still unclear whether single DUL pretreatment influences formalin-induced hypersensitivity and what is the underlying mechanism. In the current study, we revealed that systemic pretreatment with DUL not only dose-dependently suppressed the spontaneous pain behaviors, but also relieved mechanical and thermal hypersensitivity induced by formalin hindpaw injection. Consistent with the analgesic effects of DUL on the pain behaviors, the expressions of Fos and pERK that were used to check the neuronal activities in the spinal cord and CeA were also dose-dependently reduced following DUL pretreatment. Meanwhile, no emotional aversive behaviors were observed at 24 h after formalin injection. The concentration of 5-HT in the CeA was correlated with the dose of DUL in a positive manner at 24 h after formalin injection. Direct injecting 5-HT into the CeA suppressed both the spontaneous pain behaviors and hyperalgesia induced by formalin injection. However, DUL did not have protective effects on the formalin-induced edema of hindpaw. In sum, the activation of CeA neurons may account for the transition from acute pain to long-term hyperalgesia after formalin injection. DUL may produce potent analgesic effects on the hyperalgesia and decrease the expressions of p-ERK through increasing the concentration of serotonin in the CeA.
Collapse
Affiliation(s)
- Lie Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jun-Bin Yin
- Department of Neurosurgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China.,Department of Neurology, The 456th Hospital of PLA, Jinan, China.,Department of Human Anatomy, The Fourth Military Medical University, Xi'an, China
| | - Wei Hu
- Department of Neurosurgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China.,Department of Human Anatomy, The Fourth Military Medical University, Xi'an, China
| | - Wen-Jun Zhao
- Department of Human Anatomy, The Fourth Military Medical University, Xi'an, China
| | - Qing-Rong Fan
- Department of Neurosurgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Zhi-Chun Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Ming-Jie He
- Department of Neurosurgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Tan Ding
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yan Sun
- Cadet Bridge, The Fourth Military Medical University, Xi'an, China
| | - Alan D Kaye
- Departments of Anesthesiology and Pharmacology, Louisiana State University School of Medicine, New Orleans, LA, United States
| | - En-Ren Wang
- Department of Neurosurgery, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
15
|
McCarren HS, Arbutus JA, Ardinger C, Dunn EN, Jackson CE, McDonough JH. Dexmedetomidine stops benzodiazepine-refractory nerve agent-induced status epilepticus. Epilepsy Res 2018; 141:1-12. [PMID: 29414381 DOI: 10.1016/j.eplepsyres.2018.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/21/2017] [Accepted: 01/07/2018] [Indexed: 12/15/2022]
Abstract
Nerve agents are highly toxic chemicals that pose an imminent threat to soldiers and civilians alike. Nerve agent exposure leads to an increase in acetylcholine within the central nervous system, resulting in development of protracted seizures known as status epilepticus (SE). Currently, benzodiazepines are the standard of care for nerve agent-induced SE, but their efficacy quickly wanes as the time to treatment increases. Here, we examine the role of the α2-adrenoceptor in termination of nerve agent-induced SE using the highly specific agonist dexmedetomidine (DEX). Adult male rats were exposed to soman and entered SE as confirmed by electroencephalograph (EEG). We observed that administration of DEX in combination with the benzodiazepine midazolam (MDZ) 20 or 40 min after the onset of SE stopped seizures and returned processed EEG measurements to baseline levels. The protective effect of DEX was blocked by the α2-adrenoceptor antagonist atipamezole (ATI), but ATI failed to restore seizure activity after it was already halted by DEX in most cases, suggesting that α2-adrenoceptors may be involved in initiating SE cessation rather than merely suppressing seizure activity. Histologically, treatment with DEX + MDZ significantly reduced the number of dying neurons as measured by FluoroJade B in the amygdala, thalamus, and piriform cortex, but did not protect the hippocampus or parietal cortex even when SE was successfully halted. We conclude that DEX serves not just as a valuable potential addition to the anticonvulsant regimen for nerve agent exposure, but also as a tool for dissecting the neural circuitry that drives SE.
Collapse
Affiliation(s)
- Hilary S McCarren
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States.
| | - Julia A Arbutus
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - Cherish Ardinger
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - Emily N Dunn
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - Cecelia E Jackson
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| | - John H McDonough
- USAMRICD, Medical Toxicology Research Division, Neuroscience Branch, 2900 Ricketts Point Rd, Aberdeen Proving Ground, MD 21010, United States
| |
Collapse
|
16
|
Neuroprotection and neurotoxicity in the developing brain: an update on the effects of dexmedetomidine and xenon. Neurotoxicol Teratol 2017; 60:102-116. [PMID: 28065636 DOI: 10.1016/j.ntt.2017.01.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 12/30/2016] [Accepted: 01/04/2017] [Indexed: 12/13/2022]
Abstract
Growing and consistent preclinical evidence, combined with early clinical epidemiological observations, suggest potentially neurotoxic effects of commonly used anesthetic agents in the developing brain. This has prompted the FDA to issue a safety warning for all sedatives and anesthetics approved for use in children under three years of age. Recent studies have identified dexmedetomidine, the potent α2-adrenoceptor agonist, and xenon, the noble gas, as effective anesthetic adjuvants that are both less neurotoxic to the developing brain, and also possess neuroprotective properties in neonatal and other settings of acute ongoing neurologic injury. Dexmedetomidine and xenon are effective anesthetic adjuvants that appear to be less neurotoxic than other existing agents and have the potential to be neuroprotective in the neonatal and pediatric settings. Although results from recent clinical trials and case reports have indicated the neuroprotective potential of xenon and dexmedetomidine, additional randomized clinical trials corroborating these studies are necessary. By reviewing both the existing preclinical and clinical evidence on the neuroprotective effects of dexmedetomidine and xenon, we hope to provide insight into the potential clinical efficacy of these agents in the management of pediatric surgical patients.
Collapse
|
17
|
Pu-erh Tea Protects the Nervous System by Inhibiting the Expression of Metabotropic Glutamate Receptor 5. Mol Neurobiol 2016; 54:5286-5299. [PMID: 27578019 PMCID: PMC5533841 DOI: 10.1007/s12035-016-0064-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/17/2016] [Indexed: 12/31/2022]
Abstract
Glutamate is one of the major excitatory neurotransmitters of the CNS and is essential for numerous key neuronal functions. However, excess glutamate causes massive neuronal death and brain damage owing to excitotoxicity via the glutamate receptors. Metabotropic glutamate receptor 5 (mGluR5) is one of the glutamate receptors and represents a promising target for studying neuroprotective agents of potential application in neurodegenerative diseases. Pu-erh tea, a fermented tea, mainly produced in Yunnan province, China, has beneficial effects, including the accommodation of the CNS. In this study, pu-erh tea markedly decreased the transcription and translation of mGluR5 compared to those by black and green teas. Pu-erh tea also inhibited the expression of Homer, one of the synaptic scaffolding proteins binding to mGluR5. Pu-erh tea protected neural cells from necrosis via blocked Ca2+ influx and inhibited protein kinase C (PKC) activation induced by excess glutamate. Pu-erh tea relieved rat epilepsy induced by LiCl-pilocarpine in behavioural and physiological assays. Pu-erh tea also decreased the expression of mGluR5 in the hippocampus. These results show that the inhibition of mGluR5 plays a role in protecting neural cells from glutamate. The results also indicate that pu-erh tea contains biological compounds binding transcription factors and inhibiting the expression of mGluR5 and identify pu-erh tea as a novel natural neuroprotective agent.
Collapse
|
18
|
Wang J, Li ZH, Feng B, Zhang T, Zhang H, Li H, Chen T, Cui J, Zang WD, Li YQ. Corticotrigeminal Projections from the Insular Cortex to the Trigeminal Caudal Subnucleus Regulate Orofacial Pain after Nerve Injury via Extracellular Signal-Regulated Kinase Activation in Insular Cortex Neurons. Front Cell Neurosci 2015; 9:493. [PMID: 26733817 PMCID: PMC4689789 DOI: 10.3389/fncel.2015.00493] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 12/08/2015] [Indexed: 11/16/2022] Open
Abstract
Cortical neuroplasticity alterations are implicated in the pathophysiology of chronic orofacial pain. However, the relationship between critical cortex excitability and orofacial pain maintenance has not been fully elucidated. We recently demonstrated a top-down corticospinal descending pain modulation pathway from the anterior cingulate cortex (ACC) to the spinal dorsal horn that could directly regulate nociceptive transmission. Thus, we aimed to investigate possible corticotrigeminal connections that directly influence orofacial nociception in rats. Infraorbital nerve chronic constriction injury (IoN-CCI) induced significant orofacial nociceptive behaviors as well as pain-related negative emotions such as anxiety/depression in rats. By combining retrograde and anterograde tract tracing, we found powerful evidence that the trigeminal caudal subnucleus (Vc), especially the superficial laminae (I/II), received direct descending projections from granular and dysgranular parts of the insular cortex (IC). Extracellular signal-regulated kinase (ERK), an important signaling molecule involved in neuroplasticity, was significantly activated in the IC following IoN-CCI. Moreover, in IC slices from IoN-CCI rats, U0126, an inhibitor of ERK activation, decreased both the amplitude and the frequency of spontaneous excitatory postsynaptic currents (sEPSCs) and reduced the paired-pulse ratio (PPR) of Vc-projecting neurons. Additionally, U0126 also reduced the number of action potentials in the Vc-projecting neurons. Finally, intra-IC infusion of U0126 obviously decreased Fos expression in the Vc, accompanied by the alleviation of both nociceptive behavior and negative emotions. Thus, the corticotrigeminal descending pathway from the IC to the Vc could directly regulate orofacial pain, and ERK deactivation in the IC could effectively alleviate neuropathic pain as well as pain-related negative emotions in IoN-CCI rats, probably through this top–down pathway. These findings may help researchers and clinicians to better understand the underlying modulation mechanisms of orofacial neuropathic pain and indicate a novel mechanism of ERK inhibitor-induced analgesia.
Collapse
Affiliation(s)
- Jian Wang
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Zhi-Hua Li
- Basic Medical College, Zhengzhou University Zhengzhou, China
| | - Ban Feng
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Ting Zhang
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Han Zhang
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Hui Li
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Tao Chen
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical University Xi'an, China
| | - Jing Cui
- Basic Medical College, Zhengzhou University Zhengzhou, China
| | - Wei-Dong Zang
- Basic Medical College, Zhengzhou University Zhengzhou, China
| | - Yun-Qing Li
- Department of Anatomy and K. K. Leung Brain Research Centre, Fourth Military Medical UniversityXi'an, China; Collaborative Innovation Center for Brain Science, Fudan UniversityShanghai, China
| |
Collapse
|