1
|
Wang W, Li K, Ma W, Li Y, Liu F, Kong Y, Wang L, Yi F, Sang Y, Li G, Liu H, Qiu J. Ultrasound-activated piezoelectric nanostickers for neural stem cell therapy of traumatic brain injury. NATURE MATERIALS 2025:10.1038/s41563-025-02214-w. [PMID: 40329084 DOI: 10.1038/s41563-025-02214-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 03/17/2025] [Indexed: 05/08/2025]
Abstract
Traumatic brain injury (TBI) is associated with life-threatening and permanent disabilities. Given the limited capacity of neurons to regenerate, effective treatments for TBI are lacking. Neural stem cells (NSCs) can differentiate into fully functioning neurons and thus hold promise for TBI treatment. Nonetheless, NSC differentiation and proliferation are slow and inefficient. Studies have shown that piezoelectric stimulation is capable of promoting the differentiation and proliferation of NSCs. Here, we describe barium titanate-reduced graphene oxide (BTO/rGO) hybrid piezoelectric nanostickers that promote NSC proliferation and differentiation. These hybrid nanostickers attach to NSC membranes, serving as long-term generators of piezoelectric potentials upon ultrasound stimulation. BTO/rGO nanostickers promote rapid neuronal differentiation and maturation by activating the voltage-gated calcium channel/Ca2+/calmodulin-dependent protein kinase II/cAMP response element-binding protein pathways. Transplantation of NSCs with BTO/rGO nanostickers into the injured brain region of rats with TBI substantially repairs brain tissue and effectively restores physiological functions after 28 d following 5-min ultrasound irradiation every 2 d. These results demonstrate the potential of the combination of NSCs and BTO/rGO nanostickers for TBI treatment.
Collapse
Affiliation(s)
- Wenhan Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Keyi Li
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Wenjun Ma
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Yiwei Li
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Feng Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Ying Kong
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Liang Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| | - Yuanhua Sang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China.
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China.
- Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, China.
| | - Jichuan Qiu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, China.
| |
Collapse
|
2
|
Zhao N, Qin R, Liu B, Zhang D. Sevoflurane versus propofol on immediate postoperative cognitive dysfunction in patients undergoing cardiac surgery under cardiopulmonary bypass: a comparative analysis. J Cardiothorac Surg 2025; 20:54. [PMID: 39794827 PMCID: PMC11724596 DOI: 10.1186/s13019-024-03327-0] [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: 11/12/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
OBJECTIVE This study aims to compare the effects of sevoflurane (SEV) and propofol (PRO) on postoperative cognitive dysfunction (POCD) in patients undergoing cardiac surgery (CS) under cardiopulmonary bypass (CPB), with a focus on evaluating the efficacy of these anesthetic agents in preventing POCD. METHODS A total of 113 patients undergoing CS with CPB were grouped into two: PRO group (n = 58) and SEV group (n = 55). Baseline data, anesthesia effects (CPB duration, anesthesia time, respiratory recovery time, and anesthesia recovery time), Montreal Cognitive Assessment (MoCA) scores, POCD incidence, neurological function markers (NSE, S-100β, MMP9), and serum inflammatory markers (IL-6, IL-8, TNF-α) were analyzed. The study was conducted between March 2018 and May 2021. RESULTS The PRO group showed significantly shorter anesthesia time (P < 0.05), respiratory recovery time (P < 0.05), and anesthesia recovery time (P < 0.05) compared to the SEV group. The postoperative MoCA score in the PRO group reduced markedly compared with the baseline, but still higher than that in the SEV group (P < 0.05). The incidence of POCD was significantly lower in the PRO group (5.17% vs. 27.27%, P = 0.001). The levels of NSE, S-100β, MMP9, IL-6, IL-8, and TNF-α were significantly elevated compared to baseline values, but still lower than those in the SEV group (P < 0.05 for all comparisons). CONCLUSION PRO is more effective than SEV in preventing POCD in patients undergoing CS with CPB. It provides superior anesthetic effects and offers better protection against neuronal damage and serum inflammation compared to SEV. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Na Zhao
- Department of Anesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750001, China
| | - Rui Qin
- Department of Anesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750001, China
| | - Bin Liu
- Department of Anesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750001, China
| | - Dongmei Zhang
- Department of Anesthesia and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750001, China.
| |
Collapse
|
3
|
Hou Y, Ye W, Tang Z, Li F. Anesthetics in pathological cerebrovascular conditions. J Cereb Blood Flow Metab 2025; 45:32-47. [PMID: 39450477 PMCID: PMC11563546 DOI: 10.1177/0271678x241295857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/21/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
The increasing prevalence of pathological cerebrovascular conditions, including stroke, hypertensive encephalopathy, and chronic disorders, underscores the importance of anesthetic considerations for affected patients. Preserving cerebral oxygenation and blood flow during anesthesia is paramount to prevent neurological deterioration. Furthermore, protecting vulnerable neurons from damage is crucial for optimal outcomes. Recent research suggests that anesthetic agents may provide a potentially therapeutic approach for managing pathological cerebrovascular conditions. Anesthetics target neural mechanisms underlying cerebrovascular dysfunction, thereby modulating neuroinflammation, protecting neurons against ischemic injury, and improving cerebral hemodynamics. However, optimal strategies regarding mechanisms, dosage, and indications remain uncertain. This review aims to clarify the physiological effects, mechanisms of action, and reported neuroprotective benefits of anesthetics in patients with various pathological cerebrovascular conditions. Investigating anesthetic effects in cerebrovascular disease holds promise for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuhui Hou
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Wei Ye
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Ziyuan Tang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Fengxian Li
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Zhang H, Tian Y, Ma S, Ji Y, Wang Z, Xiao P, Xu Y. Chaperone-Mediated Autophagy in Brain Injury: A Double-Edged Sword with Therapeutic Potentials. Mol Neurobiol 2024; 61:10671-10683. [PMID: 38775879 DOI: 10.1007/s12035-024-04230-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 05/10/2024] [Indexed: 11/24/2024]
Abstract
Autophagy is an intracellular recycling process that maintains cellular homeostasis by degrading excess or defective macromolecules and organelles. Chaperone-mediated autophagy (CMA) is a highly selective form of autophagy in which a substrate containing a KFERQ-like motif is recognized by a chaperone protein, delivered to the lysosomal membrane, and then translocated to the lysosome for degradation with the assistance of lysosomal membrane protein 2A. Normal CMA activity is involved in the regulation of cellular proteostasis, metabolism, differentiation, and survival. CMA dysfunction disturbs cellular homeostasis and directly participates in the pathogenesis of human diseases. Previous investigations on CMA in the central nervous system have primarily focus on neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. Recently, mounting evidence suggested that brain injuries involve a wider range of types and severities, making the involvement of CMA in the bidirectional processes of damage and repair even more crucial. In this review, we summarize the basic processes of CMA and its associated regulatory mechanisms and highlight the critical role of CMA in brain injury such as cerebral ischemia, traumatic brain injury, and other specific brain injuries. We also discuss the potential of CMA as a therapeutic target to treat brain injury and provide valuable insights into clinical strategies.
Collapse
Affiliation(s)
- Huiyi Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ye Tian
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuai Ma
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yichen Ji
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhihang Wang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peilun Xiao
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Xu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
5
|
Antonova VV, Silachev DN, Plotnikov EY, Pevzner IB, Ivanov ME, Boeva EA, Kalabushev SN, Yadgarov MY, Cherpakov RA, Grebenchikov OA, Kuzovlev AN. Positive Effects of Argon Inhalation After Traumatic Brain Injury in Rats. Int J Mol Sci 2024; 25:12673. [PMID: 39684384 DOI: 10.3390/ijms252312673] [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: 10/28/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
The noble gas argon is one of the most promising neuroprotective agents for hypoxic-reperfusion injuries of the brain. However, its effect on traumatic injuries has been insufficiently studied. The aim of this study was to analyze the effect of the triple inhalation of the argon-oxygen mixture Ar 70%/O2 30% on physical and neurological recovery and the degree of brain damage after traumatic brain injury and to investigate the possible molecular mechanisms of the neuroprotective effect. The experiments were performed in male Wistar rats. A controlled brain injury model was used to investigate the effects of argon treatment and the underlying molecular mechanisms. The results of the study showed that animals with craniocerebral injuries that were treated with argon inhalation exhibited better physical recovery rates, better neurological status, and less brain damage. Argon treatment significantly reduced the expression of the proinflammatory markers TNFα and CD68 caused by TBI, increased the expression of phosphorylated protein kinase B (pAKT), and promoted the expression of the transcription factor Nrf2 in intact animals. Treatment with an argon-oxygen breathing mixture after traumatic brain injury has a neuroprotective effect by suppressing the inflammatory response and activating the antioxidant and anti-ischemic system.
Collapse
Affiliation(s)
- Viktoriya V Antonova
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Denis N Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Egor Y Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Irina B Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Mikhail E Ivanov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Ekaterina A Boeva
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Sergey N Kalabushev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Mikhail Ya Yadgarov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Rostislav A Cherpakov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Oleg A Grebenchikov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Artem N Kuzovlev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| |
Collapse
|
6
|
Chu JMT, Chiu SPW, Wang J, Chang RCC, Wong GTC. Adiponectin deficiency is a critical factor contributing to cognitive dysfunction in obese mice after sevoflurane exposure. Mol Med 2024; 30:177. [PMID: 39415089 PMCID: PMC11481458 DOI: 10.1186/s10020-024-00954-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND The number of major operations performed in obese patients is expected to increase given the growing prevalence of obesity. Obesity is a risk factor for a range of postoperative complications including perioperative neurocognitive disorders. However, the mechanisms underlying this vulnerability are not well defined. We hypothesize that obese subjects are more vulnerable to general anaesthesia induced neurotoxicity due to reduced levels of adiponectin. This hypothesis was tested using a murine surgical model in obese and adiponectin knockout mice exposed to the volatile anaesthetic agent sevoflurane. METHODS Obese mice were bred by subjecting C57BL/6 mice to a high fat diet. Cognitive function, neuroinflammatory responses and neuronal degeneration were assessed in both obese and lean mice following exposure to 2 h of sevoflurane to confirm sevoflurane-induced neurotoxicity. Thereafter, to confirm the role of adiponectin deficiency in, adiponectin knockout mice were established and exposed to the sevoflurane. Finally, the neuroprotective effects of adiponectin receptor agonist (AdipoRon) were examined. RESULTS Sevoflurane triggered significant cognitive dysfunction, neuroinflammatory responses and neuronal degeneration in the obese mice while no significant impact was observed in the lean mice. Similar cognitive dysfunction and neuronal degeneration were also observed in the adiponectin knockout mice after sevoflurane exposure. Administration of AdipoRon partially prevented the deleterious effects of sevoflurane in both obese and adiponectin knockout mice. CONCLUSIONS Our findings demonstrate that obese mice are more susceptible to sevoflurane-induced neurotoxicity and cognitive impairment in which adiponectin deficiency is one of the underlying mechanisms. Treatment with adiponectin receptor agonist ameliorates this vulnerability. These findings may have therapeutic implications in reducing the incidence of anaesthesia related neurotoxicity in obese subjects.
Collapse
Affiliation(s)
- John Man Tak Chu
- Department of Anaesthesiology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, HKSAR, China
- Laboratory of Neurodegenerative Disease, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, L4-49, Laboratory Block, 21 Sassoon Road, Hong Kong, HKSAR, China
| | - Suki Pak Wing Chiu
- Department of Anaesthesiology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, HKSAR, China
| | - Jiaqi Wang
- Department of Anaesthesiology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, HKSAR, China
| | - Raymond Chuen Chung Chang
- Laboratory of Neurodegenerative Disease, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, L4-49, Laboratory Block, 21 Sassoon Road, Hong Kong, HKSAR, China.
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, HKSAR, China.
| | - Gordon Tin Chun Wong
- Department of Anaesthesiology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Room K424, Queen Mary Hospital, Pokfulam, Hong Kong, HKSAR, China.
| |
Collapse
|
7
|
Xie A, Zhang X, Ju F, Zhou Y, Wu D, Han J. Sevoflurane impedes neuropathic pain by maintaining endoplasmic reticulum stress and oxidative stress homeostasis through inhibiting the activation of the PLCγ/CaMKII/IP3R signaling pathway. Aging (Albany NY) 2024; 16:11062-11071. [PMID: 38975935 PMCID: PMC11272110 DOI: 10.18632/aging.206001] [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: 06/13/2023] [Accepted: 05/29/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVE To investigate the effect of sevoflurane on neuropathic pain induced by chronic constriction injury (CCI) of sciatic nerve in mice, and to elucidate its mechanism by animal experiments. METHODS AND RESULTS Thirty-two C57BL/6 mice were randomly divided into four groups: Sham group, Model group, Control group and Sevoflurane group. First, a mouse model of neuropathic pain was established. Then, the mice in each group were killed on Day 14 after operation to harvest the enlarged lumbosacral spinal cord. In contrast with the Model group, the Sevoflurane group displayed a significantly increased paw withdrawal mechanical threshold (PWMT) and significantly prolonged paw withdrawal thermal latency (PWTL) from Day 5 after operation. The morphological changes of lumbosacral spinal cord were observed by hematoxylin-eosin (HE) staining and transmission electron microscopy. Pathological results showed that sevoflurane reduced nuclear pyknosis in lumbosacral spinal cord tissue, with a large number of mitochondrial crista disappearance and mitochondrial swelling. The results of Western blotting showed that sevoflurane significantly decreased the protein expressions of phosphorylated phospholipase Cγ (p-PLCγ), phosphorylated calcium/calmodulin-dependent protein kinase II (p-CaMKII) and phosphorylated inositol 1,4,5-triphosphate receptor (p-IP3R), and reduced the protein expressions of endoplasmic reticulum (ER) stress proteins glucose-regulated protein 78 (GRP78) and GRP94, oxidative stress-related proteins P22 and P47 and inflammatory factors nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), interleukin-1 β (IL-1β), and tumor necrosis factor-α (TNF-α). CONCLUSIONS Sevoflurane inhibits neuropathic pain by maintaining ER stress and oxidative stress homeostasis through inhibiting the activation of the PLCγ/CaMKII/IP3R signaling pathway.
Collapse
Affiliation(s)
- An Xie
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| | - Xianjie Zhang
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| | - Feng Ju
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| | - Yukai Zhou
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| | - Dan Wu
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| | - Jia Han
- Department of Anesthesiology, People’s Hospital of Deyang, Deyang, Sichuan, China
| |
Collapse
|
8
|
Hao S, Yuan S, Liu Z, Hou B, Feng S, Zhang D. Neuroprotective effects of takinib on an experimental traumatic brain injury rat model via inhibition of transforming growth factor beta-activated kinase 1. Heliyon 2024; 10:e29484. [PMID: 38644820 PMCID: PMC11033159 DOI: 10.1016/j.heliyon.2024.e29484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/23/2024] Open
Abstract
Transforming growth factor β-activated kinase 1 (TAK1) plays a significant role in controlling several signaling pathways involved with regulating inflammation and apoptosis. As such, it represents an important potential target for developing treatments for traumatic brain injury (TBI). Takinib, a small molecule and selective TAK1 inhibitor, has potent anti-inflammatory activity and has shown promising activity in preclinical studies using rat models to evaluate the potential neuroprotective impact on TBI. The current study used a modified Feeney's weight-drop model to cause TBI in mature Sprague-Dawley male rats. At 30 min post-induction of TBI in the rats, they received an intracerebroventricular (ICV) injection of Takinib followed by assessment of their histopathology and behavior. The results of this study demonstrated how Takinib suppressed TBI progression in the rats by decreasing TAK1, p-TAK1, and nuclear p65 levels while upregulating IκB-α expression. Takinib was also shown to significantly inhibit the production of two pro-inflammatory factors, namely tumor necrosis factor-α and interleukin-1β. Furthermore, Takinib greatly upregulated the expression of tight junction proteins zonula occludens-1 and claudin-5, reducing cerebral edema. Additionally, Takinib effectively suppressed apoptosis via downregulation of cleaved caspase 3 and Bax and reduction of TUNEL-positive stained cell count. As a result, an enhancement of neuronal function and survival was observed post-TBI. These findings highlight the medicinal value of Takinib in the management of TBI and offer an experimental justification for further investigation of TAK1 as a potential pharmacological target.
Collapse
Affiliation(s)
- Shuangying Hao
- School of Medicine, Henan Polytechnic University, Jiaozuo, Henan, PR China
| | - Shuai Yuan
- School of Medicine, Henan Polytechnic University, Jiaozuo, Henan, PR China
| | - Zhiqiang Liu
- School of Medicine, Henan Polytechnic University, Jiaozuo, Henan, PR China
| | - Baohua Hou
- School of Medicine, Henan Polytechnic University, Jiaozuo, Henan, PR China
| | - Sijie Feng
- School of Medicine, Henan Polytechnic University, Jiaozuo, Henan, PR China
| | - Dingding Zhang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, PR China
| |
Collapse
|
9
|
Jiang W, Luo H, Zhao M, Fan Q, Ye C, Li X, He J, Lai J, He S, Chen W, Xian W, Chen S, Chen Z, Li D, Chen R, Wang B. Evaluation of canine adipose-derived mesenchymal stem cells for neurological functional recovery in a rat model of traumatic brain injury. BMC Vet Res 2024; 20:110. [PMID: 38500105 PMCID: PMC10946090 DOI: 10.1186/s12917-024-03912-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/04/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a common condition in veterinary medicine that is difficult to manage.Veterinary regenerative therapy based on adipose mesenchymal stem cells seem to be an effective strategy for the treatment of traumatic brain injury. In this study, we evaluated therapeutic efficacy of canine Adipose-derived mesenchymal stem cells (AD-MSCs)in a rat TBI model, in terms of improved nerve function and anti-neuroinflammation. RESULTS Canine AD-MSCs promoted neural functional recovery, reduced neuronal apoptosis, and inhibited the activation of microglia and astrocytes in TBI rats. According to the results in vivo, we further investigated the regulatory mechanism of AD-MSCs on activated microglia by co-culture in vitro. Finally, we found that canine AD-MSCs promoted their polarization to the M2 phenotype, and inhibited their polarization to the M1 phenotype. What's more, AD-MSCs could reduce the migration, proliferation and Inflammatory cytokines of activated microglia, which is able to inhibit inflammation in the central system. CONCLUSIONS Collectively, the present study demonstrates that transplantation of canine AD-MSCs can promote functional recovery in TBI rats via inhibition of neuronal apoptosis, glial cell activation and central system inflammation, thus providing a theoretical basis for canine AD-MSCs therapy for TBI in veterinary clinic.
Collapse
Affiliation(s)
- Wenkang Jiang
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, 526000, China
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Huina Luo
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Mingming Zhao
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Quanbao Fan
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Cailing Ye
- Deja Lab, VetCell Biotechnology Company Limited, Foshan, 528225, China
| | - Xingying Li
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Jing He
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Jianyi Lai
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Shi He
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Wojun Chen
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Weihang Xian
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Shengfeng Chen
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Zhisheng Chen
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China
| | - Dongsheng Li
- Deja Lab, VetCell Biotechnology Company Limited, Foshan, 528225, China.
| | - Ruiai Chen
- Zhaoqing Branch Center of Guangdong Laboratory for Lingnan Modern Agricultural Science and Technology, Zhaoqing, 526000, China.
| | - Bingyun Wang
- School of Life Science and Engineering, Foshan University, Foshan, 528225, China.
| |
Collapse
|
10
|
Xie X, Zhang X, Li S, Du W. Involvement of Fgf2-mediated tau protein phosphorylation in cognitive deficits induced by sevoflurane in aged rats. Mol Med 2024; 30:39. [PMID: 38493090 PMCID: PMC10943822 DOI: 10.1186/s10020-024-00784-0] [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: 10/30/2023] [Accepted: 01/11/2024] [Indexed: 03/18/2024] Open
Abstract
OBJECTIVE Anesthetics have been linked to cognitive alterations, particularly in the elderly. The current research delineates how Fibroblast Growth Factor 2 (Fgf2) modulates tau protein phosphorylation, contributing to cognitive impairments in aged rats upon sevoflurane administration. METHODS Rats aged 3, 12, and 18 months were subjected to a 2.5% sevoflurane exposure to form a neurotoxicity model. Cognitive performance was gauged, and the GEO database was employed to identify differentially expressed genes (DEGs) in the 18-month-old cohort post sevoflurane exposure. Bioinformatics tools, inclusive of STRING and GeneCards, facilitated detailed analysis. Experimental validations, both in vivo and in vitro, examined Fgf2's effect on tau phosphorylation. RESULTS Sevoflurane notably altered cognitive behavior in older rats. Out of 128 DEGs discerned, Fgf2 stood out as instrumental in regulating tau protein phosphorylation. Sevoflurane exposure spiked Fgf2 expression in cortical neurons, intensifying tau phosphorylation via the PI3K/AKT/Gsk3b trajectory. Diminishing Fgf2 expression correspondingly curtailed tau phosphorylation, neurofibrillary tangles, and enhanced cognitive capacities in aged rats. CONCLUSION Sevoflurane elicits a surge in Fgf2 expression in aging rats, directing tau protein phosphorylation through the PI3K/AKT/Gsk3b route, instigating cognitive aberrations.
Collapse
Affiliation(s)
- Xin Xie
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dandong District, Liaoning Province, Shenyang, 110042, P. R. China
| | - Xiaomin Zhang
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dandong District, Liaoning Province, Shenyang, 110042, P. R. China
| | - Songze Li
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dandong District, Liaoning Province, Shenyang, 110042, P. R. China
| | - Wei Du
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan Road, Dandong District, Liaoning Province, Shenyang, 110042, P. R. China.
| |
Collapse
|
11
|
Ni H, Kan X, Rui Q, Zhang Y, Zhai W, Zhang B, Yu Z. RACK1 promotes autophagy via the PERK signaling pathway to protect against traumatic brain injury in rats. CNS Neurosci Ther 2024; 30:e14691. [PMID: 38532543 PMCID: PMC10966134 DOI: 10.1111/cns.14691] [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: 10/29/2023] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/28/2024] Open
Abstract
AIMS Neuronal cell death is a primary factor that determines the outcome after traumatic brain injury (TBI). We previously revealed the importance of receptor for activated C kinase (RACK1), a multifunctional scaffold protein, in maintaining neuronal survival after TBI, but the specific mechanism remains unclear. The aim of this study was to explore the mechanism underlying RACK1-mediated neuroprotection in TBI. METHODS TBI model was established using controlled cortical impact injury in Sprague-Dawley rats. Genetic intervention and pharmacological inhibition of RACK1 and PERK-autophagy signaling were administrated by intracerebroventricular injection. Western blotting, coimmunoprecipitation, transmission electron microscopy, real-time PCR, immunofluorescence, TUNEL staining, Nissl staining, neurobehavioral tests, and contusion volume assessment were performed. RESULTS Endogenous RACK1 was upregulated and correlated with autophagy induction after TBI. RACK1 knockdown markedly inhibited TBI-induced autophagy, whereas RACK1 overexpression exerted the opposite effects. Moreover, RACK1 overexpression ameliorated neuronal apoptosis, neurological deficits, and cortical tissue loss after TBI, and these effects were abrogated by the autophagy inhibitor 3-methyladenine or siRNAs targeting Beclin1 and Atg5. Mechanistically, RACK1 interacted with PERK and activated PERK signaling. Pharmacological and genetic inhibition of the PERK pathway abolished RACK1-induced autophagy after TBI. CONCLUSION Our findings indicate that RACK1 protected against TBI-induced neuronal damage partly through autophagy induction by regulating the PERK signaling pathway.
Collapse
Affiliation(s)
- Haibo Ni
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
- Department of NeurosurgeryThe Fourth Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xugang Kan
- Department of Neurobiology and Cell Biology, Xuzhou Key Laboratory of NeurobiologyXuzhou Medical UniversityXuzhouChina
| | - Qin Rui
- Department of Center of Clinical LaboratoryThe Fourth Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yang Zhang
- Department of Neurobiology and Cell Biology, Xuzhou Key Laboratory of NeurobiologyXuzhou Medical UniversityXuzhouChina
| | - Weiwei Zhai
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Baole Zhang
- Department of Neurobiology and Cell Biology, Xuzhou Key Laboratory of NeurobiologyXuzhou Medical UniversityXuzhouChina
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research LaboratoryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
12
|
Liu Y, Zhao Z, Guo J, Ma Y, Li J, Ji H, Chen Z, Zheng J. Anacardic acid improves neurological deficits in traumatic brain injury by anti-ferroptosis and anti-inflammation. Exp Neurol 2023; 370:114568. [PMID: 37820939 DOI: 10.1016/j.expneurol.2023.114568] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/23/2023] [Accepted: 10/07/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) is an important cause of disability and death. TBI leads to multiple forms of nerve cell death including ferroptosis due to iron-dependent lipid peroxidation. Anacardic acid (AA) is a natural component extracted from cashew nut shells, which has been reported to have neuroprotective effects in traumatic brain injury. We investigated whether AA has an anti-ferroptosis effect in TBI. METHODS We used the Feeney free-fall impact method to construct a TBI model to investigate the effect of AA on ferroptosis caused by TBI, in which Ferrostatin-1 (Fer-1), a ferroptosis inhibitor, served as a positive control group. We first identified the therapeutic effect of AA on TBI through modified neurological severity score (mNSS) and determined the appropriate concentration. Secondly, we investigated the effect of AA on the expression level of the key protein of ferroptosis by Western blotting and immunohistochemistry. Then the effect of AA on nerve tissue injury and nerve function improvement was verified. Finally, enzym-linked immunosorbent assay (ELISA) was used to verify that AA could reduce inflammation after TBI. RESULTS We found the intensely inhibitory effect of AA on ferroptosis, which is in parallel with the results obtained after Fer-1 treatment. In addition, AA and Fer-1 mitigated TBI-mediated tissue defects, destruction of the blood-brain barrier, and neurodegeneration. Novel object recognition (NOR), mNSS and water maze test showed that AA could significantly reduce the impairment of neural function and behavioral cognitive ability caused by TBI. Finally, we also demonstrated that AA has not only an anti-ferroptosis effect, but also an anti-inflammation effect. CONCLUSIONS AA can reduce the neurological impairment and behavioral cognitive impairment caused by TBI through the dual effect of anti-ferroptosis and anti-inflammation.
Collapse
Affiliation(s)
- Yu Liu
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China; Xuzhou Medical University, Xuzhou 221000, China
| | - Zongren Zhao
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China
| | - Jianqiang Guo
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China; Xuzhou Medical University, Xuzhou 221000, China
| | - Yuanhao Ma
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China; Xuzhou Medical University, Xuzhou 221000, China
| | - Jing Li
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China
| | - Huanhuan Ji
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China
| | - Zhongjun Chen
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China
| | - Jinyu Zheng
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223022, China.
| |
Collapse
|
13
|
Zhang L, Xu L. Fgf2 and Ptpn11 play a role in cerebral injury caused by sevoflurane anesthesia. Medicine (Baltimore) 2023; 102:e36108. [PMID: 37960778 PMCID: PMC10637467 DOI: 10.1097/md.0000000000036108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Sevoflurane is a new inhaled anesthetic, which has better physical properties than the existing inhalational anesthetics, rapid induction, less tissue uptake, and faster recovery. Sevoflurane can directly dilators cerebral blood vessels and increase cerebral blood flow, but it also reduces cerebral oxygen metabolism rate, thereby reducing cerebral blood flow. However, the role of Fgf2 and Ptpn11 in cerebral injury caused by sevoflurane anesthesia remains unclear. The sevoflurane anesthesia brain tissue datasets GSE139220 and GSE141242 were downloaded from gene expression omnibus (GEO). Differentially expressed genes (DEGs) were screened and weighted gene co-expression network analysis (WGCNA) was performed. Construction and analysis of protein-protein interaction (PPI) Network. Gene Ontology (GO) and Kyoto Encyclopedia of Gene and Genome (KEGG), comparative toxicogenomics database (CTD) were performed. A heat map of gene expression was drawn. TargetScan was used to screen miRNAs regulating DEGs. 500 DEGs were identified. According to GO, in Biological Process analysis, they were mainly enriched in response to hypoxia, blood vessel development, inner ear development, neural tube closure, and aging. In Cellular Component (CC), they were mainly enriched in plasma membrane, integral component of membrane, and basal lamina. In Molecular Function (MF), they were mainly associated with protein binding, Wnt-activated receptor activity, and organic anion transmembrane transporter activity. In the KEGG analysis, they were mainly enriched in proteoglycans in cancer, pathways in cancer, transcriptional misregulation in cancer, basal cell carcinoma, thyroid hormone signaling pathway. In the Metascape enrichment analysis, the GO enrichment items revealed upregulated regulation of vascular endothelial cell proliferation, platelet-derived growth factor receptor signaling pathway, inner ear development, and response to hypoxia. A total of 20 modules were generated. Gene Expression Heatmap showed that the core genes (Fgf2, Pdgfra, Ptpn11, Slc2a1) were highly expressed in sevoflurane anesthesia brain tissue samples. CTD Analysis showed that the 4 core genes (Fgf2, Pdgfra, Ptpn11, Slc2a1) were associated with neurodegenerative diseases, brain injuries, memory disorders, cognitive disorders, neurotoxicity, drug-induced abnormalities, neurological disorders, developmental disorders, and intellectual disabilities. Fgf2 and Ptpn11 are highly expressed in brain tissue after sevoflurane anesthesia, higher the expression level of Fgf2 and Ptpn11, worse the prognosis.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Lingyan Xu
- Department of Disease Control and Prevention, The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Diaz MD, Kandell RM, Wu JR, Chen A, Christman KL, Kwon EJ. Infusible Extracellular Matrix Biomaterial Promotes Vascular Integrity and Modulates the Inflammatory Response in Acute Traumatic Brain Injury. Adv Healthc Mater 2023; 12:e2300782. [PMID: 37390094 PMCID: PMC10592293 DOI: 10.1002/adhm.202300782] [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: 03/13/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
Traumatic brain injury (TBI) affects millions of people each year and, in many cases, results in long-term disabilities. Once a TBI has occurred, there is a significant breakdown of the blood-brain barrier resulting in increased vascular permeability and progression of the injury. In this study, the use of an infusible extracellular matrix-derived biomaterial (iECM) for its ability to reduce vascular permeability and modulate gene expression in the injured brain is investigated. First, the pharmacokinetics of iECM administration in a mouse model of TBI is characterized, and the robust accumulation of iECM at the site of injury is demonstrated. Next, it is shown that iECM administration after injury can reduce the extravasation of molecules into the brain, and in vitro, iECM increases trans-endothelial electrical resistance across a monolayer of TNFα-stimulated endothelial cells. In gene expression analysis of brain tissue, iECM induces changes that are indicative of downregulation of the proinflammatory response 1-day post-injury/treatment and neuroprotection at 5 days post-injury/treatment. Therefore, iECM shows potential as a treatment for TBI.
Collapse
Affiliation(s)
- Miranda D. Diaz
- Shu‐Chien Gene Lay Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
- Sanford Consortium for Regenerative MedicineLa JollaCA92037USA
| | - Rebecca M. Kandell
- Shu‐Chien Gene Lay Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
- Sanford Consortium for Regenerative MedicineLa JollaCA92037USA
| | - Jason R. Wu
- Shu‐Chien Gene Lay Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
- Sanford Consortium for Regenerative MedicineLa JollaCA92037USA
| | - Alexander Chen
- Shu‐Chien Gene Lay Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
- Sanford Consortium for Regenerative MedicineLa JollaCA92037USA
| | - Karen L. Christman
- Shu‐Chien Gene Lay Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
- Sanford Consortium for Regenerative MedicineLa JollaCA92037USA
| | - Ester J. Kwon
- Shu‐Chien Gene Lay Department of BioengineeringUniversity of California San DiegoLa JollaCA92093USA
- Sanford Consortium for Regenerative MedicineLa JollaCA92037USA
| |
Collapse
|
15
|
Wang Y, Wu D, Li D, Zhou X, Fan D, Pan J. The role of PERK-eIF2α-ATF4-CHOP pathway in sevoflurane induced neuroapoptosis and cognitive dysfunction in aged mice. Cell Signal 2023; 110:110841. [PMID: 37549858 DOI: 10.1016/j.cellsig.2023.110841] [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: 05/28/2023] [Revised: 07/24/2023] [Accepted: 08/02/2023] [Indexed: 08/09/2023]
Abstract
Postoperative cognitive dysfunction (POCD) is a common surgical complication that causes additional pain in patients and affects their quality of life. To address this problem, emerging studies have focused on the POCD. Recent studies have shown that aging and anesthetic exposure are the two major risk factors for developing POCD. However, few reports described the exact molecular mechanisms underlying POCD in elderly patients. In the previous studies, the endoplasmic reticulum (ER) stress and neuroapoptosis in the hippocampus were associated with inducing POCD; however, no further information on the related signaling pathways could be disclosed. The PERK-eIF2α-ATF4-CHOP pathway is identified as the main regulatory pathway involved in ER stress and cell apoptosis. Therefore, we assume that the occurrence of POCD induced by sevoflurane inhalation may potentially result from ER stress and neuroapoptosis in the hippocampus of aged mice mediated by the PERK-eIF2α-ATF4-CHOP pathway. In our study, we found a relationship between sevoflurane inhalation concentration and memory decline in aged mice, with a 'ceiling effect'. We have confirmed that POCD induced by sevoflurane results from ER stress and neuroapoptosis in the hippocampus of aged mice, which is regulated by the over-expression of PERK-eIF2α-ATF4-CHOP pathway. Furthermore, we also showed that the dephosphorylation inhibitor of eIF2α (salubrinal) could down-regulate PERK-eIF2α-ATF4-CHOP pathway expression to inhibit ER stress and enhance the cognitive function of aged mice. In general, our study has elucidated one of the molecular mechanisms of sevoflurane-related cognitive dysfunction in aged groups and provided new strategies for treating sevoflurane-induced POCD.
Collapse
Affiliation(s)
- Yuhao Wang
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China; Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Di Wu
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Danni Li
- Department of Anesthesiology, Sichuan Academy of Medical Science, Sichuan Provincial People's Hospital, Chengdu 610072, PR China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, PR China
| | - Xueer Zhou
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China; Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Dan Fan
- Department of Anesthesiology, Sichuan Academy of Medical Science, Sichuan Provincial People's Hospital, Chengdu 610072, PR China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, PR China.
| | - Jian Pan
- State Key Laboratory of Oral Disease & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China; Frontier Innovation Center for Dental Medicine Plus, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, PR China.
| |
Collapse
|
16
|
Gu T, Xu C, Meng X, Gao D, Jiang G, Yin A, Liu Q, Zhang L. Sevoflurane Preconditioning Alleviates Posttraumatic Stress Disorder-Induced Apoptosis in the Hippocampus via the EZH2-Regulated Akt/mTOR Axis and Improves Synaptic Plasticity. J Mol Neurosci 2023; 73:225-236. [PMID: 36930428 DOI: 10.1007/s12031-023-02114-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
Posttraumatic stress disorder (PTSD) is a persistent and severe psychological and mental disorder resulting from experiences of serious trauma or stress and is suffered by many individuals. Previous studies have shown that pretreatment with sevoflurane is efficient in reducing the incidence of PTSD. However, we require a more comprehensive understanding of the specific mechanisms by which sevoflurane works. Enhancer of zeste homolog 2 (EZH2) has been reported to be regulated by sevoflurane, and to improve patient cognition. In this study, we aimed to explore the mechanisms of sevoflurane and the role of EZH2 in PTSD cases. We explored the effects of sevoflurane and EPZ-6438 (inhibitor of EZH2) on rat behavior, followed by an investigation of EZH2 mRNA and protein expression. The effects of sevoflurane and EZH2 on neuronal survival were assessed by western blotting and TUNEL staining, while western blotting was used to examine the expression of PSD95 and the AKT/mTOR proteins. Sevoflurane preconditioning restored EZH2 expression and significantly inhibited apoptosis by regulating phosphorylation of the AKT/mTOR pathway. Synaptic plasticity was also significantly improved. These results suggest that pretreatment with sevoflurane could play an important role in PTSD prevention by regulating EZH2 expression.
Collapse
Affiliation(s)
- Tingting Gu
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chang Xu
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaozhou Meng
- Department of Anesthesiology, Jinling Hospital, Medical College of Nanjing Medical University, Nanjing, China
| | - Dapeng Gao
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guanghao Jiang
- Department of Anesthesiology, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, China
| | - Anqi Yin
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qingzhen Liu
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Lidong Zhang
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China. .,Department of Anesthesiology, Jinling Hospital, Medical College of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
17
|
Du Y, Xu M, Su Y, Liu Y, Zhou Y, Gu X, Xia T. Long-term sevoflurane exposure relieves stress-enhanced fear learning and anxiety in PTSD mice. Transl Neurosci 2023; 14:20220313. [PMID: 37901139 PMCID: PMC10612489 DOI: 10.1515/tnsci-2022-0313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 10/31/2023] Open
Abstract
Objectives Post-traumatic stress disorder (PTSD) is characterized by recurrent episodes of severe anxiety after exposure to traumatic events. It is believed that these episodes are triggered at least in part by environmental stimuli associated with the precipitating trauma through classical conditioning, termed conditioned fear. However, traditional methods of conditioned fear memory extinction are frequently ineffective for PTSD treatment due to the contribution of non-associative sensitization caused by trauma. Anesthetics have shown promise for treating various psychiatric diseases such as depression. Methods In this study, we examined if the inhaled anesthetic sevoflurane can suppress stress-enhanced fear learning (SEFL) in PTSD model mice. Model mice exposed to 2.4% sevoflurane for 6 h exhibited reduced freezing time and behavioral anxiety compared to sham-treated model mice. To explore the underlying mechanisms, we evaluated the regional expression levels of glucocorticoid receptors (GRs), cannabinoid CB1 receptors (CB1Rs), D1 dopamine receptors (D1Rs), and D2 dopamine receptors (D2Rs). Results We verified that both GR and CB1R were significantly upregulated in the hippocampus, amygdaloid nucleus, and prefrontal cortex (PFC) of model mice, while D1R and D2R were downregulated. All of these expression changes were partially normalized in the PFC by 6 h but not with 2 h sevoflurane exposure. Conclusions These results showed that sevoflurane exposure following traumatic events may be an effective treatment for PTSD.
Collapse
Affiliation(s)
- Ying Du
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Minhui Xu
- Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yan Su
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yujia Liu
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yiming Zhou
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Xiaoping Gu
- Department of Anesthesiology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Tianjiao Xia
- Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| |
Collapse
|