1
|
Hu Q, Li Y, Lin Z, Zhang H, Chen H, Chao C, Zhao C. The Molecular Biological Mechanism of Hydrogen Therapy and Its Application in Spinal Cord Injury. Drug Des Devel Ther 2024; 18:1399-1414. [PMID: 38707612 PMCID: PMC11068043 DOI: 10.2147/dddt.s463177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024] Open
Abstract
Hydrogen, which is a novel biomedical molecule, is currently the subject of extensive research involving animal experiments and in vitro cell experiments, and it is gradually being applied in clinical settings. Hydrogen has been proven to possess anti-inflammatory, selective antioxidant, and antiapoptotic effects, thus exhibiting considerable protective effects in various diseases. In recent years, several studies have provided preliminary evidence for the protective effects of hydrogen on spinal cord injury (SCI). This paper provides a comprehensive review of the potential molecular biology mechanisms of hydrogen therapy and its application in treating SCI, with an aim to better explore the medical value of hydrogen and provide new avenues for the adjuvant treatment of SCI.
Collapse
Affiliation(s)
- Quan Hu
- Department of Neurosurgery, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Yingxiao Li
- Department of Gynecology, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Zhaochen Lin
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Hao Zhang
- Department of Rehabilitation Medical Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Haoyue Chen
- Department of Rehabilitation Medical Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Cui Chao
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Chuanliang Zhao
- Department of Orthopedics, the Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| |
Collapse
|
2
|
Li TT, Zhao DM, Wei YT, Li JB, Li XF, Wan Q, Zhang X, Liu XN, Yang WC, Li WZ. Effect and Mechanism of Sodium Butyrate on Neuronal Recovery and Prognosis in Diabetic Stroke. J Neuroimmune Pharmacol 2023; 18:366-382. [PMID: 37318680 DOI: 10.1007/s11481-023-10071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 05/18/2023] [Indexed: 06/16/2023]
Abstract
Ischemic stroke is a cerebrovascular lesion caused by local ischemia and hypoxia. Diabetes mellitus (DM) is a chronic inflammatory disease that disturbs immune homeostasis and predisposes patients to ischemic stroke. The mechanism by which DM exacerbates stroke remains unclear, although it may involve disturbances in immune homeostasis. Regulatory T cells (Tregs) play a regulatory role in many diseases, but the mechanism of Tregs in diabetes complicated by stroke remains unclear. Sodium butyrate is a short-chain fatty acid that increases Treg levels. This study examined the role of sodium butyrate in the prognosis of neurological function in diabetic stroke and the mechanism by which Tregs are amplified in the bilateral cerebral hemispheres. We evaluated the brain infarct volume, observed 48-h neuronal injury and 28-day behavioral changes, and calculated the 28-day survival rate in mice. We also measured Treg levels in peripheral blood and brain tissue, recorded changes in the blood‒brain barrier and water channel proteins and neurotrophic changes in mice, measured cytokine levels and peripheral B-cell distribution in bilateral hemispheres and peripheral blood, and examined the polarization of microglia and the distribution of peripheral T-cell subpopulations in bilateral hemispheres. Diabetes significantly exacerbated the poor prognosis and neurological deficits in mice with stroke, and sodium butyrate significantly improved infarct volume, prognosis, and neurological function and showed different mechanisms in brain tissue and peripheral blood. The potential regulatory mechanism in brain tissue involved modulating Tregs/TGF-β/microglia to suppress neuroinflammation, while that in peripheral blood involved improving the systemic inflammatory response through Tregs/TGF-β/T cells.
Collapse
Affiliation(s)
- Ting-Ting Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Deng-Ming Zhao
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Yu-Ting Wei
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Jing-Bo Li
- The Heilongjiang Key Laboratory of Anesthesia and Intensive Care Research, Harbin Medical University, 150081, Heilongjiang Province, Harbin, China
| | - Xue-Fei Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Qiang Wan
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, 650000, Yunnan Province, Kunming, China
| | - Xin Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Xiang-Nan Liu
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Wan-Chao Yang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Wen-Zhi Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China.
- The Heilongjiang Key Laboratory of Anesthesia and Intensive Care Research, Harbin Medical University, 150081, Heilongjiang Province, Harbin, China.
| |
Collapse
|
3
|
Zhao Q, Li H, Li H, Zhang J. Research progress on pleiotropic neuroprotective drugs for traumatic brain injury. Front Pharmacol 2023; 14:1185533. [PMID: 37475717 PMCID: PMC10354289 DOI: 10.3389/fphar.2023.1185533] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/26/2023] [Indexed: 07/22/2023] Open
Abstract
Traumatic brain injury (TBI) has become one of the most important causes of death and disability worldwide. A series of neuroinflammatory responses induced after TBI are key factors for persistent neuronal damage, but at the same time, such inflammatory responses can also promote debris removal and tissue repair after TBI. The concept of pleiotropic neuroprotection delves beyond the single-target treatment approach, considering the multifaceted impacts following TBI. This notion embarks deeper into the research-oriented treatment paradigm, focusing on multi-target interventions that inhibit post-TBI neuroinflammation with enhanced therapeutic efficacy. With an enriched comprehension of TBI's physiological mechanisms, this review dissects the advancements in developing pleiotropic neuroprotective pharmaceuticals to mitigate TBI. The aim is to provide insights that may contribute to the early clinical management of the condition.
Collapse
Affiliation(s)
- Qinghui Zhao
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Huige Li
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| | - Hongru Li
- Zhumadian Central Hospital, Zhumadian, China
| | - Jianhua Zhang
- Institute of Physical Culture, Huanghuai University, Zhumadian, China
| |
Collapse
|
4
|
Yang WC, Li TT, Wan Q, Zhang X, Sun LY, Zhang YR, Lai PC, Li WZ. Molecular Hydrogen Mediates Neurorestorative Effects After Stroke in Diabetic Rats: the TLR4/NF-κB Inflammatory Pathway. J Neuroimmune Pharmacol 2023; 18:90-99. [PMID: 35895245 PMCID: PMC10485112 DOI: 10.1007/s11481-022-10051-w] [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: 01/28/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022]
Abstract
Diabetes is an independent risk factor for stroke and amplifies inflammation. Diabetic stroke is associated with a higher risk of death and worse neural function. The identification of effective anti-inflammatory molecules with translational advantages is particularly important to promote perioperative neurorestorative effects. Applying molecular hydrogen, we measured blood glucose levels before and after middle cerebral artery occlusion (MCAO), 48-h cerebral oedema and infarct volumes, as well as 28-day weight, survival and neurological function. We also measured the levels of TLR4, NF-κB p65, phosphorylated NF-κB p65, catecholamines, acetylcholine and inflammatory factors. All measurements comprehensively showed the positive effect and translational advantage of molecular hydrogen on diabetic stroke. Molecular hydrogen improved the weight, survival and long-term neurological function of rats with diabetic stroke and alleviated changes in blood glucose levels before and after middle cerebral artery occlusion (MCAO), but no difference in circadian rhythm was observed. Molecular hydrogen inhibited the phosphorylation of NF-κB and significantly reduced inflammation. Molecular hydrogen mediates neurorestorative effects after stroke in diabetic rats. The effect is independent of circadian rhythms, indicating translational advantages. The molecular mechanism is related to the TLR4/NF-κB pathway and inflammation. Molecular hydrogen (H2) affects outcomes of ischemic stroke with diabetes mellitus (DM).
Collapse
Affiliation(s)
- Wan-Chao Yang
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Ting-ting Li
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Qiang Wan
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Xin Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Li-Ying Sun
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Yu-Rong Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Pei-Chen Lai
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, People’s Republic of China
- Asclepius Meditec Co., Ltd, Shanghai, China
| | - Wen-zhi Li
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
5
|
Mu G, Li Q, Lu B, Yu X. Amelioration of nerve demyelination by hydrogen-producing silicon-based agent in neuropathic pain rats. Int Immunopharmacol 2023; 117:110033. [PMID: 36933448 DOI: 10.1016/j.intimp.2023.110033] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Trigeminal neuralgia (TN) is a complex orofacial neuropathic pain. The crippling condition's underlying mechanism is still not completely understood. The main cause of lightning-like pain in patients with TN may be chronic inflammation that causes nerve demyelination. Nano-silicon (Si) can safely and continuously produce hydrogen in the alkaline environment of the intestine to exert systemic anti-inflammatory effects. Hydrogen has a promising anti-neuroinflammatory impact. The study aimed to determine how intra-intestinal application of a hydrogen-producing Si-based agent affected the demyelination of the trigeminal ganglion in TN rats. We discovered that increased expression of the NLRP3 inflammasome and inflammatory cell infiltration occurred concurrently with demyelination of the trigeminal ganglion in TN rats. We could determine that the neural effect of the hydrogen-producing Si-based agent was connected to the inhibition of microglial pyroptosis by using transmission electron microscopy. The results demonstrated that the Si-based agent reduced the infiltration of inflammatory cells and the degree of neural demyelination. In a subsequent study, it was discovered that hydrogen produced by a Si-based agent regulates the pyroptosis of microglia may through the NLRP3-caspase-1-GSDMD pathway, preventing the development of chronic neuroinflammation and consequently lowering the incidence of nerve demyelination. This study offers a novel strategy for elucidating the pathogenesis of TN and developing potential therapeutic drugs.
Collapse
Affiliation(s)
- Guo Mu
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, Sichuan Province 643000, PR China; Laboratory of Anesthesiology, Southwest Medical University, Luzhou, Sichuan Province 646000, PR China
| | - Qiang Li
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, Sichuan Province 643000, PR China
| | - Bin Lu
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, Sichuan Province 643000, PR China.
| | - Xuan Yu
- Department of Anesthesiology, Zigong Fourth People's Hospital, Zigong, Sichuan Province 643000, PR China.
| |
Collapse
|
6
|
Li TT, Wan Q, Zhang X, Xiao Y, Sun LY, Zhang YR, Liu XN, Yang WC. Stellate ganglion block reduces inflammation and improves neurological function in diabetic rats during ischemic stroke. Neural Regen Res 2022; 17:1991-1997. [PMID: 35142688 PMCID: PMC8848600 DOI: 10.4103/1673-5374.335162] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Diabetes mellitus is an independent risk factor for ischemic stroke. Both diabetes mellitus and stroke are linked to systemic inflammation that aggravates patient outcomes. Stellate ganglion block can effectively regulate the inflammatory response. Therefore, it is hypothesized that stellate ganglion block could be a potential therapy for ischemic stroke in diabetic subjects. In this study, we induced diabetes mellitus in rats by feeding them a high-fat diet for 4 successive weeks. The left middle cerebral artery was occluded to establish models of ischemic stroke in diabetic rats. Subsequently, we performed left stellate ganglion block with 1% lidocaine using the percutaneous posterior approach 15 minutes before reperfusion and again 20 and 44 hours after reperfusion. Our results showed that stellate ganglion block did not decrease the blood glucose level in diabetic rats with diabetes mellitus but did reduce the cerebral infarct volume and the cerebral water content. It also improved the recovery of neurological function, increased 28-day survival rate, inhibited Toll like receptor 4/nuclear factor kappa B signaling pathway and reduced inflammatory response in the plasma of rats. However, injection of Toll like receptor 4 agonist lipopolysaccharide 5 minutes before stellate ganglion block inhibited the effect of stellate ganglion block, whereas injection of Toll like receptor 4 inhibitor TAK242 had no such effect. We also found that stellate ganglion block performed at night had no positive effect on diabetic ischemic stroke. These findings suggest that stellate ganglion block is a potential therapy for diabetic ischemic stroke and that it may be mediated through the Toll like receptor 4/nuclear factor kappa B signaling pathway. We also found that the therapeutic effect of stellate ganglion block is affected by circadian rhythm.
Collapse
Affiliation(s)
- Ting-Ting Li
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Qiang Wan
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xin Zhang
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yuan Xiao
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Li-Ying Sun
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yu-Rong Zhang
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiang-Nan Liu
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wan-Chao Yang
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
7
|
Molecular hydrogen alleviates lung injury after traumatic brain injury: Pyroptosis and apoptosis. Eur J Pharmacol 2022; 914:174664. [PMID: 34883075 DOI: 10.1016/j.ejphar.2021.174664] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/19/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Traumatic brain injury (TBI)-induced acute lung injury (ALI) is a critical condition, and inflammation and apoptosis play essential roles. Molecular hydrogen (H2) exerts anti-inflammatory and anti-apoptotic effects. Our previous work has shown that 42% H2 can improve TBI. In the current study, we tested the hypothesis that inhalation of hydrogen (42% H2, 21% O2, balanced nitrogen) for 1 h per day can improve TBI-induced ALI. METHODS Sprague-Dawley male rats were randomly divided into 3 groups. Except for the sham group (group S), rats were subjected to a fluid percussion injury (FPI) and the H2 treatment group were given inhaled hydrogen for 1 h per day. We evaluated the lung function, pyroptosis and apoptosis at 24 h, 48 h and 72 h. RESULTS Compared with group S, the rats in the TBI group (group T) showed obvious pulmonary edema after a TBI. Inhalation of high-concentration hydrogen significantly improved the rats. During this process, rats had some tendency to heal on their own, and H2 also accelerated the self-healing process. Lung injury scores, oxygenation index and pulmonary edema were consistent. Compared with group S, the pyroptosis-related proteins Caspase-1, apoptosis-associated speck-like protein containing CARD (ASC) and Gasdermin-D (GSDM-D) in the lung tissues of the rats in group T were significantly increased after a TBI. In the H2 treatment group (group H), these proteins were significantly decreased. The levels of IL-1β and IL-18 were significantly increased after TBI while in group H were significantly decreased. At the same time, cleaved caspase-3 and BCL-2/Bax were also changed after H2 treatment. These demonstrates the powerful ameliorating effect of H2 on pyroptosis, apoptosis and systemic inflammation. However, rats also had tendency to heal on their own, and H2 also accelerated the self-healing process at the same time. CONCLUSIONS H2 improves TBI-ALI, and the mechanism may be due to the decrease of both pyroptosis and apoptosis and the alleviation of inflammation. These findings provide a reference and evidence for the use of H2 in TBI-ALI patients in the intensive care unit (ICU).
Collapse
|
8
|
Abstract
Traumatic brain injury (TBI) is a serious global public health problem. Survivors of TBI often suffer from long-term disability, which puts a heavy burden on society and families. Unfortunately, up to now, there is no efficacious treatment for TBI patients in clinical practice. As a reducing gas, hydrogen has been shown to be neuroprotective in multiple cerebral disease models; however, its efficacy in TBI remains controversial. In this review, we will focus on the results of hydrogen in experimental TBI, elaborate the potential mechanisms, and put forward for future researches based on our current understanding and views.
Collapse
Affiliation(s)
- Hong-Wei Hu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhi-Guo Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jian-Gang Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
9
|
Tatara Y, Shimada R, Kibayashi K. Effects of Preexisting Diabetes Mellitus on the Severity of Traumatic Brain Injury. J Neurotrauma 2020; 38:886-902. [PMID: 32998635 DOI: 10.1089/neu.2020.7118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Falls and traffic accidents can cause traumatic brain injury (TBI). Assessment of the injury severity is essential to determine the prognosis or the cause of death. Diabetes mellitus (DM) is a common preexisting disease in elderly adults. We hypothesized that preexisting DM exacerbates TBI secondary to prolonged inflammation. In this study, we investigated TBI-induced changes in nerve function and inflammatory cell migration to the injury site, and the extent of brain contusion in KK-Ay (DM) and C57BL/6J (non-DM) mice. A controlled cortical impact device was used to induce TBI in each mouse. The brain contusion volume was measured using magnetic resonance imaging. Nerve function changes were assessed using the following animal behavior tasks: neurological severity score (NSS), Morris water maze, forced swim test, and beam walking. Immunohistochemical examinations of brain sections were performed to assess the infiltration of neutrophils, astrocytes, microglia, and macrophages, and to detect apoptosis. These experiments were performed on post-injury days 1-90 (over five experiments/time-points in each group). Compared with non-DM mice, DM mice showed significantly greater brain contusion volume, greater deterioration in the NSS, and a higher number of neutrophils, macrophages, and apoptotic cells in the brain tissue specimens. This study indicates that the prognosis of normal mice and DM mice differs, even if they acquire a TBI of the same severity. Therefore, it is important to evaluate patients with TBI for DM and other preexisting diseases in order to provide adequate treatment or to determine the correct cause of death.
Collapse
Affiliation(s)
- Yuki Tatara
- Department of Legal Medicine, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Ryo Shimada
- Department of Legal Medicine, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Kazuhiko Kibayashi
- Department of Legal Medicine, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
10
|
Zhai X, Li J, Li L, Sun Y, Zhang X, Xue Y, Lv J, Gao Y, Li S, Yan W, Yin S, Xiao Z. L-lactate preconditioning promotes plasticity-related proteins expression and reduces neurological deficits by potentiating GPR81 signaling in rat traumatic brain injury model. Brain Res 2020; 1746:146945. [PMID: 32531223 DOI: 10.1016/j.brainres.2020.146945] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 01/16/2023]
Abstract
Currently, there is no efficacious pharmacological treatment for traumatic brain injury (TBI). Previous studies revealed that L-lactate preconditioning has shown rich neuroprotective effects against cerebral ischemia, and therefore has the potential to improve neurological outcomes after TBI. L-lactate played a neuroprotective role by activating GPR81 in diseases of the central nervous system (CNS) such as TBI and cerebral ischemia. In this study we investigated the effects of L-lactate preconditioning on TBI and explored the underlying mechanisms. In this study, the mNSS test revealed that L-lactate preconditioning alleviates the neurological deficit caused by TBI in rats. L-lactate preconditioning significantly increased the expression of GPR81, PSD95, GAP43, BDNF, and MCT2 24 h after TBI in the cortex and hippocampus compared with the sham group. Taken together, these data suggested that L-lactate preconditioning is an effective method with which to recover neurological function after TBI. This reveals the mechanism of L-lactate preconditioning on TBI and provides a potential therapeutic method for TBI in humans.
Collapse
Affiliation(s)
- Xiuli Zhai
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Jinying Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Liya Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Ye Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Xiaonan Zhang
- Department of Physiology, Dalian Medical University, Dalian 116044, China
| | - Ying Xue
- Department of Physiology, Dalian Medical University, Dalian 116044, China
| | - Jiaxin Lv
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Ye Gao
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Shouxin Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Wei Yan
- Department of Physiology, Dalian Medical University, Dalian 116044, China
| | - Shengming Yin
- Department of Physiology, Dalian Medical University, Dalian 116044, China.
| | - Zhaoyang Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China.
| |
Collapse
|
11
|
Zhang ZW, Liang J, Yan JX, Ye YC, Wang JJ, Chen C, Sun HT, Chen F, Tu Y, Li XH. TBHQ improved neurological recovery after traumatic brain injury by inhibiting the overactivation of astrocytes. Brain Res 2020; 1739:146818. [PMID: 32275911 DOI: 10.1016/j.brainres.2020.146818] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 01/01/2023]
Abstract
Traumatic brain injury (TBI) is a major leading cause of death and long-term disability. Although astrocytes play a key role in neuroprotection after TBI in the early stage, the overactivation of astrocytes can lead to long-term functional deficits, and the underlying pathophysiological mechanisms remain unclear. In addition, it is unknown whether the nuclear factor erythroid 2-related factor2/haem oxygenase-1 (Nrf-2/HO-1) pathway could elicit a neuroprotective effect by decreasing astrocyte overactivation after TBI. We aimed to study the effects of tert-butylhydroquinone (TBHQ) in reducing astrocyte overactivation after TBI and explored the underlying mechanisms. We first established a controlled cortical impact (CCI) model in rats and performed Haematoxylin and eosin (H&E) staining to observe brain tissue damage. The cognitive function of rats was assessed by modified neurological severity scoring (mNSS) and Morris water maze (MWM) test. Astrocyte and microglia activation was detected by immunofluorescence staining. Oxidative stress conditions were investigated using Western blotting. An enzyme-linked immunosorbent assay (ELISA) was designed to assess the level of the proinflammatory factor tumour necrosis factor-alpha (TNF-α). Dihydroethidium (DHE) staining was used to detect reactive oxygen species (ROS). Apoptosis was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The results showed that the administration of TBHQ ameliorated motor function and cognitive deficits and decreased the lesion volume. In addition, TBHQ significantly decreased astrocyte overactivation, diminished the pro-inflammatory phenotype M1 and inflammatory cytokines production after TBI, increased Nrf-2 nuclear accumulation, and enhanced the levels of the Nrf-2 downstream antioxidative genes HO-1 and NADPH-quinone oxidoreductase-1 (NQO-1). Furthermore, TBHQ treatment alleviated apoptosis and neuronal death in the cerebral cortex. Overall, our data indicated that the upregulation of Nrf-2 expression could enhance neuroprotection and decrease astrocyte overactivation and might represent a new theoretical basis for treating TBI.
Collapse
Affiliation(s)
- Zhen-Wen Zhang
- College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jun Liang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jing-Xing Yan
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Yi-Chao Ye
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Jing-Jing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Chong Chen
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Hong-Tao Sun
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China
| | - Feng Chen
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Yue Tu
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of PAPF, Tianjin 300162, China.
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China.
| |
Collapse
|