1
|
Ren B, Kang J, Wang Y, Meng X, Huang Y, Bai Y, Feng Z. Transcranial direct current stimulation promotes angiogenesis and improves neurological function via the OXA-TF-AKT/ERK signaling pathway in traumatic brain injury. Aging (Albany NY) 2024; 16:6566-6587. [PMID: 38604164 PMCID: PMC11042948 DOI: 10.18632/aging.205724] [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: 12/18/2023] [Accepted: 03/19/2024] [Indexed: 04/13/2024]
Abstract
Traumatic brain injury (TBI) and its resulting complications pose a major challenge to global public health, resulting in increased rates of disability and mortality. Cerebrovascular dysfunction is nearly universal in TBI cases and is closely associated with secondary injury after TBI. Transcranial direct current stimulation (tDCS) shows great potential in the treatment of TBI; however, the exact mechanism remains elusive. In this study, we performed in vivo and in vitro experiments to explore the effects and mechanisms of tDCS in a controlled cortical impact (CCI) rat model simulating TBI. In vivo experiments show that tDCS can effectively reduce brain tissue damage, cerebral edema and neurological deficits. The potential mechanism may be that tDCS improves the neurological function of rats by increasing orexin A (OXA) secretion, upregulating the TF-AKT/ERK signaling pathway, and promoting angiogenesis at the injury site. Cellular experiments showed that OXA promoted HUVEC migration and angiogenesis, and these effects were counteracted by the ERK1/2 inhibitor LY3214996. The results of Matrigel experiment in vivo showed that TNF-a significantly reduced the ability of HUVEC to form blood vessels, but OXA could rescue the effect of TNF-a on the ability of HUVEC to form blood vessels. However, LY3214996 could inhibit the therapeutic effect of OXA. In summary, our preliminary study demonstrates that tDCS can induce angiogenesis through the OXA-TF-AKT/ERK signaling pathway, thereby improving neurological function in rats with TBI.
Collapse
Affiliation(s)
- Bingkai Ren
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Junwei Kang
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Yan Wang
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Xiangqiang Meng
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Ying Huang
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Yang Bai
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| | - Zhen Feng
- The Affiliated Rehabilitation Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330003, Jiangxi, China
| |
Collapse
|
2
|
Yang Z, You Y, Liu X, Wan Q, Xu Z, Shuai Y, Wang J, Guo T, Hu J, Lv J, Zhang M, Yang M, Mao C, Yang S. Injectable Bombyx mori (B. mori) silk fibroin/MXene conductive hydrogel for electrically stimulating neural stem cells into neurons for treating brain damage. J Nanobiotechnology 2024; 22:111. [PMID: 38486273 PMCID: PMC10941401 DOI: 10.1186/s12951-024-02359-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/20/2024] [Indexed: 03/17/2024] Open
Abstract
Brain damage is a common tissue damage caused by trauma or diseases, which can be life-threatening. Stem cell implantation is an emerging strategy treating brain damage. The stem cell is commonly embedded in a matrix material for implantation, which protects stem cell and induces cell differentiation. Cell differentiation induction by this material is decisive in the effectiveness of this treatment strategy. In this work, we present an injectable fibroin/MXene conductive hydrogel as stem cell carrier, which further enables in-vivo electrical stimulation upon stem cells implanted into damaged brain tissue. Cell differentiation characterization of stem cell showed high effectiveness of electrical stimulation in this system, which is comparable to pure conductive membrane. Axon growth density of the newly differentiated neurons increased by 290% and axon length by 320%. In addition, unfavored astrocyte differentiation is minimized. The therapeutic effect of this system is proved through traumatic brain injury model on rats. Combined with in vivo electrical stimulation, cavities formation is reduced after traumatic brain injury, and rat motor function recovery is significantly promoted.
Collapse
Affiliation(s)
- Zhangze Yang
- Institute of Applied Bioresource Research, College of Animal Science, Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, 310058, Zhejiang, China
| | - Yuxin You
- Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Xiangyu Liu
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, Zhejiang, China
| | - Quan Wan
- Institute of Applied Bioresource Research, College of Animal Science, Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, 310058, Zhejiang, China
| | - Zongpu Xu
- Institute of Applied Bioresource Research, College of Animal Science, Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, 310058, Zhejiang, China
| | - Yajun Shuai
- Institute of Applied Bioresource Research, College of Animal Science, Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, 310058, Zhejiang, China
| | - Jie Wang
- Institute of Applied Bioresource Research, College of Animal Science, Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, 310058, Zhejiang, China
| | - Tingbiao Guo
- Centre for Optical and Electromagnetic Research National Engineering Research Center for Optical Instruments Zhejiang University, Hangzhou, 310058, China
| | - Jiaqi Hu
- Institute of Applied Bioresource Research, College of Animal Science, Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, 310058, Zhejiang, China
| | - Junhui Lv
- Department of Neurosurgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China
| | - Meng Zhang
- Institute of Applied Bioresource Research, College of Animal Science, Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, 310058, Zhejiang, China
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, 310058, Zhejiang, China.
| | - Chuanbin Mao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, Zhejiang, China.
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR.
| | - Shuxu Yang
- Department of Neurosurgery, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
3
|
Qian J, Liang T, Xu Y, Liu ZP, Jing LL, Luo HB. Effect of the Novel Free Radical Scavenger 4'-Hydroxyl-2-Substituted Phenylnitronyl Nitroxide on Oxidative Stress, Mitochondrial Dysfunction and Apoptosis Induced by Cerebral Ischemia-Reperfusion in Rats. Neuroscience 2024; 540:1-11. [PMID: 38242279 DOI: 10.1016/j.neuroscience.2023.12.016] [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: 09/28/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/21/2024]
Abstract
Mitochondrial dysfunction, which results in the overproduction of oxygen free radicals, is a crucial mechanism underlying cerebral ischemia-reperfusion injury. 4'-Hydroxyl-2-substituted phenylnitronyl nitroxide (HPN), which is an antioxidant and free radical scavenger, can effectively scavenge oxygen free radicals, suggesting its potential as a protective agent against cerebral ischemia-reperfusion injury. In this study, we investigated the effects of HPN on mitochondrial function and apoptosis following cerebral ischemia/reperfusion injury in rats. Healthy adult SD rats were chosen as the experimental subjects, and the rat ischemia/reperfusion injury model was generated using the modified Zea Longa method. The administration of HPN significantly enhanced the activity of endogenous antioxidant enzymes, such as superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase (CAT). Additionally, HPN effectively preserved the morphology and function of mitochondria, reduced the protein and gene expression of Caspase-3 and Bax, increased the protein and gene expression of Bcl-2, mitigated neuronal apoptosis, improved neurological deficits, and decreased the volume of cerebral infarction. Of interest, the protective effect on brain tissue was more evident with increasing doses of HPN. These findings indicate that HPN can serve as an effective protective agent against cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Jun Qian
- Department of Neurology, Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, PR China
| | - Tao Liang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, PR China
| | - Yu Xu
- Department of Respiratory Medicine, Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, PR China
| | - Zhi-Peng Liu
- Department of Neurology, Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, PR China
| | - Lin-Lin Jing
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Hong-Bo Luo
- Department of Neurology, Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, PR China.
| |
Collapse
|
4
|
Liu J, Xin X, Sun J, Fan Y, Zhou X, Gong W, Yang M, Li Z, Wang Y, Yang Y, Gao C. Dual-targeting AAV9P1-mediated neuronal reprogramming in a mouse model of traumatic brain injury. Neural Regen Res 2024; 19:629-635. [PMID: 37721294 PMCID: PMC10581548 DOI: 10.4103/1673-5374.380907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/09/2023] [Accepted: 06/06/2023] [Indexed: 09/19/2023] Open
Abstract
Traumatic brain injury results in neuronal loss and glial scar formation. Replenishing neurons and eliminating the consequences of glial scar formation are essential for treating traumatic brain injury. Neuronal reprogramming is a promising strategy to convert glial scars to neural tissue. However, previous studies have reported inconsistent results. In this study, an AAV9P1 vector incorporating an astrocyte-targeting P1 peptide and glial fibrillary acidic protein promoter was used to achieve dual-targeting of astrocytes and the glial scar while minimizing off-target effects. The results demonstrate that AAV9P1 provides high selectivity of astrocytes and reactive astrocytes. Moreover, neuronal reprogramming was induced by downregulating the polypyrimidine tract-binding protein 1 gene via systemic administration of AAV9P1 in a mouse model of traumatic brain injury. In summary, this approach provides an improved gene delivery vehicle to study neuronal programming and evidence of its applications for traumatic brain injury.
Collapse
Affiliation(s)
- Jingzhou Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xin Xin
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jiejie Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yueyue Fan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Xun Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
5
|
Wu H, Zhang Q, Xu P, Chen J, Duan L, Xu F, Zhang F. Nattokinase Promotes Post-stroke Neurogenesis and Cognition Recovery via Increasing Circulating Irisin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37466380 DOI: 10.1021/acs.jafc.2c08718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
The therapeutic potential of treatments for post-stroke cognitive impairment (PSCI) is severely limited by the autonomic regeneration capacity of the adult brain. Nattokinase (NK), a serine protease from the traditional food natto, has many beneficial effects on the cardiovascular system by modulating the blood system. While the role of blood factors in neurogenesis and cognition is well-established, it remains unclear whether NK can serve as an anti-PSCI agent through these factors. Our study demonstrates that NK protects against acute ischemic stroke and impressively promotes neurogenesis in rat models by increasing peripheral blood irisin, leading to improved cognitive functions. Our findings demonstrate NK to be a promising candidate for treating PSCI, and we also highlight irisin as a novel target of NK, suggesting its potential role in the peripheral blood-to-brain axis.
Collapse
Affiliation(s)
- Hao Wu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China
| | - Qian Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China
| | - Pu Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China
| | - Jiepeng Chen
- Sungen Biotech Company, Limited, Shantou, Guangdong 515000, People's Republic of China
| | - Lili Duan
- Sungen Biotech Company, Limited, Shantou, Guangdong 515000, People's Republic of China
| | - Feng Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China
| | - Fengjiao Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, People's Republic of China
| |
Collapse
|
6
|
Liu W, He X, Lin H, Yang M, Dai Y, Chen L, Li C, Liang S, Tao J, Chen L. Ischemic stroke rehabilitation through optogenetic modulation of parvalbumin neurons in the contralateral motor cortex. Exp Neurol 2023; 360:114289. [PMID: 36471512 DOI: 10.1016/j.expneurol.2022.114289] [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: 06/10/2022] [Revised: 10/09/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Based on the theory of interhemispheric inhibition and the bimodal balance-recovery model in stroke, we explored the effects of excitation/inhibition (E/I) of parvalbumin (PV) neurons in the contralateral primary motor cortex (cM1) connecting the ipsilateral M1 (iM1) via the corpus callosum (cM1-CC-iM1) of ischemic stroke rats by optogenetic stimulation. METHODS We tested this by injecting anterograde and retrograde virus in rats with middle cerebral artery occlusion (MCAO), and evaluated the neurological scores, motor behavior, volume of cerebral infarction and the E/I balance of the bilateral M1 two weeks after employing optogenetic treatment. RESULTS We found that concentrations of Glu and GABA decreased and increased, respectively, in the iM1 of MCAO rats, and that the former increased in the cM1, suggesting E/I imbalance in bilateral M1 after ischemic stroke. Interestingly, optogenetic stimulation improved M1 E/I imbalance, as illustrated by the increase of Glu in the iM1 and the decrease of GABA in both iM1 and cM1, which were accompanied by an improvement in neurological deficit and motor dysfunction. In addition, we observed a reduced infarct volume, an increase in the expression of the NMDAR and AMPAR, and a decrease in GAD67 in the iM1 after intervention. CONCLUSIONS Optogenetic modulation of PV neurons of the iM1-CC-cM1 improve E/I balance, leading to reduced neurological deficit and improved motor dysfunction following ischemic stroke in rats.
Collapse
Affiliation(s)
- Weilin Liu
- Rehabilitation Industry institute, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Xiaojun He
- Rehabilitation Industry institute, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Huawei Lin
- Rehabilitation Industry institute, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Minguang Yang
- Rehabilitation Industry institute, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Yaling Dai
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Lewen Chen
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Chaohui Li
- General surgery, Anxi General Hospital of Traditional Chinese Medicine, Quanzhou, Fujian 362400, China
| | - Shengxiang Liang
- Rehabilitation Industry institute, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Jing Tao
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Lidian Chen
- Rehabilitation Industry institute, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| |
Collapse
|
7
|
Wu H, Li Y, Zhang Q, Wang H, Xiu W, Xu P, Deng Y, Huang W, Wang DO. Crocetin antagonizes parthanatos in ischemic stroke via inhibiting NOX2 and preserving mitochondrial hexokinase-I. Cell Death Dis 2023; 14:50. [PMID: 36681688 PMCID: PMC9867762 DOI: 10.1038/s41419-023-05581-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/22/2023]
Abstract
Parthanatos is one of the major pathways of programmed cell death in ischemic stroke characterized by DNA damage, poly (ADP-ribose) polymerases (PARP) activation, and poly (ADP-ribose) (PAR) formation. Here we demonstrate that crocetin, a natural potent antioxidant compound from Crocus sativus, antagonizes parthanatos in ischemic stroke. We reveal that mechanistically, crocetin inhibits NADPH oxidase 2 (NOX2) activation to reduce reactive oxygen species (ROS) and PAR production at the early stage of parthanatos. Meanwhile we demonstrate that PARylated hexokinase-I (HK-I) is a novel substrate of E3 ligase RNF146 and that crocetin interacts with HK-I to suppress RNF146-mediated HK-I degradation at the later stage of parthanatos, preventing mitochondrial dysfunction and DNA damage that ultimately trigger the irreversible cell death. Our study supports further development of crocetin as a potential drug candidate for preventing and/or treating ischemic stroke.
Collapse
Affiliation(s)
- Hao Wu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Ying Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Qian Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hanxun Wang
- College of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wenyu Xiu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Pu Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yujie Deng
- Guangzhou National Laboratory, Guangzhou, Guangdong, 510530, China
| | - Wanxu Huang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China.
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510700, China.
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, Jiangsu, 222001, China.
| | - Dan Ohtan Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China.
- Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan.
- Graduate School of Biostudies, Kyoto University, Yoshida Hon-Machi, Kyoto, 606-8501, Japan.
| |
Collapse
|
8
|
Li MC, Tian Q, Liu S, Han SM, Zhang W, Qin XY, Chen JH, Liu CL, Guo YJ. The mechanism and relevant mediators associated with neuronal apoptosis and potential therapeutic targets in subarachnoid hemorrhage. Neural Regen Res 2023; 18:244-252. [PMID: 35900398 PMCID: PMC9396483 DOI: 10.4103/1673-5374.346542] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a dominant cause of death and disability worldwide. A sharp increase in intracranial pressure after SAH leads to a reduction in cerebral perfusion and insufficient blood supply for neurons, which subsequently promotes a series of pathophysiological responses leading to neuronal death. Many previous experimental studies have reported that excitotoxicity, mitochondrial death pathways, the release of free radicals, protein misfolding, apoptosis, necrosis, autophagy, and inflammation are involved solely or in combination in this disorder. Among them, irreversible neuronal apoptosis plays a key role in both short- and long-term prognoses after SAH. Neuronal apoptosis occurs through multiple pathways including extrinsic, mitochondrial, endoplasmic reticulum, p53 and oxidative stress. Meanwhile, a large number of blood contents enter the subarachnoid space after SAH, and the secondary metabolites, including oxygenated hemoglobin and heme, further aggravate the destruction of the blood-brain barrier and vasogenic and cytotoxic brain edema, causing early brain injury and delayed cerebral ischemia, and ultimately increasing neuronal apoptosis. Even there is no clear and effective therapeutic strategy for SAH thus far, but by understanding apoptosis, we might excavate new ideas and approaches, as targeting the upstream and downstream molecules of apoptosis-related pathways shows promise in the treatment of SAH. In this review, we summarize the existing evidence on molecules and related drugs or molecules involved in the apoptotic pathway after SAH, which provides a possible target or new strategy for the treatment of SAH.
Collapse
|
9
|
Prediction of adult post-hemorrhagic hydrocephalus: a risk score based on clinical data. Sci Rep 2022; 12:12213. [PMID: 35842469 PMCID: PMC9288433 DOI: 10.1038/s41598-022-16577-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/12/2022] [Indexed: 11/08/2022] Open
Abstract
There is lacking research on risk factors and prediction models associated with Post-hemorrhagic hydrocephalus (PHH). Thus, this present study aimed to analyze the risk factors of PHH and establish a risk-scoring system through a large-scale study. A retrospective study of 382 patients with intracranial hemorrhage assessed age, history and diagnosis, Glasgow coma score (GCS), and fever time. After univariate and logistic regression analysis, a risk scoring system was established according to independent risk factors and evaluated using the area under the curve (AUC). Of the 382 patients, 133 (34.8%) had PHH, 43 (11.3%) received surgical treatment. Factor classification showed that age > 60 years old [odds ratio (OR): 0.347, II = 5 points], GCS < 5 (OR: 0.09, IV = 10 points), GCS 6‒8 (OR = 0.232, III = 6 points), fever time > 9 (OR: 0.202, III = 7 points), fever time 5-9 (OR: 0.341, II = 5 points), CSF-TP x time > 14,4000 group (OR: 0.267, IV = 6 points), and CSF-TP x time 9,601‒14,400 group (OR: 0.502, III = 3 points) were independent risk factors. The result of the receiver operating characteristic (ROC) prediction showed that AUC = 0.790 (0.744‒0.836). Low-risk (IV-VII), moderate (VIII-X), and high-risk group (XI-XIII) incidence of PHH were 11.76%, 50.55%, and 70.00% (p < 0.001), respectively. The coincidence rates in the validation cohort were 26.00%, 74.07%, and 100.0% (p < 0.001), respectively. AUC value was 0.860 (0.780‒0.941). The predictive model was conducive to determining the occurrence of PHH and facilitating early intervention.
Collapse
|
10
|
Li Y, Zhang J, Wang H, Zhu L, Zhang H, Ma Q, Liu X, Dong L, Lu G. Does erythropoietin affect the outcome and complication rates of patient with traumatic brain injury? A pooled-analysis. Neurol Sci 2022; 43:3783-3793. [PMID: 35044560 DOI: 10.1007/s10072-022-05877-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 01/08/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The aim of this meta-analysis was to review the scientific literature published until April 18, 2021, to summarize existing knowledge on the efficacy and safety of erythropoietin (EPO) for traumatic brain injury (TBI). METHODS This systematic review followed PRISMA guidelines. Randomized controlled trials (RCTs) reporting on the efficacy and safety of EPO in the treatment of TBI were systematically searched in relevant electronic databases according to a pre-designed search strategy. The primary outcomes are the mortality; and secondary outcomes are the good functional outcome (GFO) and adverse events (AEs). RESULTS A total of 10 RCTs involving 2,402 participants fulfilled the inclusion criteria. The results showed that there is a significant difference in terms of the mortality (RR = 0.67, 95% CI = 0.54-0.84, P = 0.0003) and seizure rate (RR = 0.52, 95% CI = 0.29-0.96, P = 0.04) between the EPO groups compared to those in the control groups. However, compared with the control groups, the GFO in the EPO groups was not statistically significant (RR = 1.18, 95% CI = 0.93-1.48, P = 0.17). CONCLUSIONS Findings of the present meta-analysis suggest that the use of EPO could reduce mortality rate in patients with TBI, without increasing the incidence of AEs. EPO has potential research and application value in the treatment of TBI.
Collapse
Affiliation(s)
- Yuping Li
- Neuro Intensive Care Unit, Department of Neurosurgery, Clinical Medical College, Yangzhou University, Yangzhou, China.,Department of Neurosurgery, Yangzhou Clinical Medical College of Xuzhou Medical University, Xuzhou, China
| | - Jun Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Haili Wang
- Department of Clinical Medicine, Dalian Medical University, Dalian Liaoning, China
| | - Lei Zhu
- Neuro Intensive Care Unit, Department of Neurosurgery, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Hengzhu Zhang
- Department of Neurosurgery, Yangzhou Clinical Medical College of Xuzhou Medical University, Xuzhou, China
| | - Qiang Ma
- Neuro Intensive Care Unit, Department of Neurosurgery, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Xiaoguang Liu
- Neuro Intensive Care Unit, Department of Neurosurgery, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Lun Dong
- Neuro Intensive Care Unit, Department of Neurosurgery, Clinical Medical College, Yangzhou University, Yangzhou, China.,Department of Neurosurgery, Yangzhou Clinical Medical College of Xuzhou Medical University, Xuzhou, China
| | - Guangyu Lu
- Institute of Public Health, Medical College, Yangzhou University, Yangzhou, China.
| |
Collapse
|
11
|
Guo R, Wang X, Fang Y, Chen X, Chen K, Huang W, Chen J, Hu J, Liang F, Du J, Dordoe C, Tian X, Lin L. rhFGF20 promotes angiogenesis and vascular repair following traumatic brain injury by regulating Wnt/β-catenin pathway. Biomed Pharmacother 2021; 143:112200. [PMID: 34649342 DOI: 10.1016/j.biopha.2021.112200] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 11/29/2022] Open
Abstract
The pathology of cerebrovascular disorders takes an important role in traumatic brain injury (TBI) by increasing intracranial pressure. Fibroblast growth factor 20 (FGF20) is a brain-derived neurotrophic factor, that has been shown to play an important role in the survival of dopaminergic neurons and the treatment of Parkinson's disease (PD). However, little is known about the role of FGF20 in the treatment of TBI and its underlying mechanism. The purpose of this study was to evaluate the protective effect of recombinant human FGF20 (rhFGF20) on protecting cerebral blood vessels after TBI. In this study, we indicated that rhFGF20 could reduce brain edema, Evans blue penetration and upregulated the expression of blood-brain barrier (BBB)-related tight junction (TJ) proteins, exerting a protective effect on the BBB in vivo after TBI. In the TBI repair phase, rhFGF20 promoted angiogenesis, neurological and cognitive function recovery. In tumor necrosis factor-α (TNF-α)-induced human brain microvascular endothelial cells (hCMEC/D3), an in vitro BBB disruption model, rhFGF20 reversed the impairment in cell migration and tube formation induced by TNF-α. Moreover, in both the TBI mouse model and the in vitro model, rhFGF20 increased the expression of β-catenin and GSK3β, which are the two key regulators in the Wnt/β-catenin signaling pathway. In addition, the Wnt/β-catenin inhibitor IWR-1-endo significantly reversed the effects of rhFGF20. These results indicate that rhFGF20 may prevent vascular repair and angiogenesis through the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Ruili Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yani Fang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiongjian Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Kun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenting Huang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China
| | - Jun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Fei Liang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jingting Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Confidence Dordoe
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xianxi Tian
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China.
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China; Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Beijing 100730, China.
| |
Collapse
|
12
|
Animal models of traumatic brain injury: a review of pathophysiology to biomarkers and treatments. Exp Brain Res 2021; 239:2939-2950. [PMID: 34324019 DOI: 10.1007/s00221-021-06178-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
Traumatic brain injury (TBI) is one of the main causes of death and disability in both civilian and military population. TBI may occur via a variety of etiologies, all of which involve trauma to the head. However, the neuroprotective drugs which were found to be very effective in animal TBI models failed in phase II or phase III clinical trials, emphasizing a compelling need to review the current status of animal TBI models and therapeutic strategies. No single animal model can adequately mimic all aspects of human TBI owing to the heterogeneity of clinical TBI. However, due to the ethical limitations, it is difficult to precisely emulate the TBI mechanisms that occur in humans. Therefore, many animal models with varying severity and mechanisms of brain injury have been developed, and each model has its own pros and cons in its implementation for TBI research. These challenges pose a need for study of continued TBI mechanisms, brain injury severity, duration, treatment strategies, and optimization of animal models across the neurotrauma research community. The aim of this review is to discuss (1) causes of TBI, (2) its prevalence in military and civilian population, (3) classification and pathophysiology of TBI, (4) biomarkers and detection methods, (5) animal models of TBI, and (6) the advantages and disadvantages of each model and the species used, as well as possible treatments.
Collapse
|
13
|
Michinaga S, Koyama Y. Pathophysiological Responses and Roles of Astrocytes in Traumatic Brain Injury. Int J Mol Sci 2021; 22:ijms22126418. [PMID: 34203960 PMCID: PMC8232783 DOI: 10.3390/ijms22126418] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is immediate damage caused by a blow to the head resulting from traffic accidents, falls, and sporting activity, which causes death or serious disabilities in survivors. TBI induces multiple secondary injuries, including neuroinflammation, disruption of the blood–brain barrier (BBB), and brain edema. Despite these emergent conditions, current therapies for TBI are limited or insufficient in some cases. Although several candidate drugs exerted beneficial effects in TBI animal models, most of them failed to show significant effects in clinical trials. Multiple studies have suggested that astrocytes play a key role in the pathogenesis of TBI. Increased reactive astrocytes and astrocyte-derived factors are commonly observed in both TBI patients and experimental animal models. Astrocytes have beneficial and detrimental effects on TBI, including promotion and restriction of neurogenesis and synaptogenesis, acceleration and suppression of neuroinflammation, and disruption and repair of the BBB via multiple bioactive factors. Additionally, astrocytic aquaporin-4 is involved in the formation of cytotoxic edema. Thus, astrocytes are attractive targets for novel therapeutic drugs for TBI, although astrocyte-targeting drugs have not yet been developed. This article reviews recent observations of the roles of astrocytes and expected astrocyte-targeting drugs in TBI.
Collapse
Affiliation(s)
- Shotaro Michinaga
- Department of Pharmacodynamics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan;
| | - Yutaka Koyama
- Laboratory of Pharmacology, Kobe Pharmaceutical University, 4-19-1 Motoyama-Kita Higashinada, Kobe 668-8558, Japan
- Correspondence: ; Tel.: +81-78-441-7572
| |
Collapse
|
14
|
Shi YH, Zhang XL, Ying PJ, Wu ZQ, Lin LL, Chen W, Zheng GQ, Zhu WZ. Neuroprotective Effect of Astragaloside IV on Cerebral Ischemia/Reperfusion Injury Rats Through Sirt1/Mapt Pathway. Front Pharmacol 2021; 12:639898. [PMID: 33841157 PMCID: PMC8033022 DOI: 10.3389/fphar.2021.639898] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/17/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Ischemic stroke is a common disease with poor prognosis, which has become one of the leading causes of morbidity and mortality worldwide. Astragaloside IV (AS-IV) is the main bioactive ingredient of Astragali Radix (which has been used for ischemic stroke for thousands of years) and has been found to have multiple bioactivities in the nervous system. In the present study, we aimed to explore the neuroprotective effects of AS-IV in rats with cerebral ischemia/reperfusion (CIR) injury targeting the Sirt1/Mapt pathway. Methods: Sprague-Dawley rats (male, 250-280 g) were randomly divided into the Sham group, middle cerebral artery occlusion/reperfusion (MCAO/R) group, AS-IV group, MCAO/R + EX527 (SIRT1-specific inhibitor) group, and AS-IV + EX527 group. Each group was further assigned into several subgroups according to ischemic time (6 h, 1 d, 3 d, and 7 days). The CIR injury was induced in MCAO/R group, AS-IV group, MCAO/R + EX527 group, and AS-IV + EX527 group by MCAO surgery in accordance with the modified Zea Longa criteria. Modified Neurological Severity Scores (mNSS) were used to evaluate the neurological deficits; TTC (2,3,5-triphenyltetrazolium chloride) staining was used to detect cerebral infarction area; Western Blot was used to assess the protein levels of SIRT1, acetylated MAPT (ac-MAPT), phosphorylated MAPT (p-MAPT), and total MAPT (t-MAPT); Real-time Quantitative Polymerase Chain Reaction (qRT-PCR) was used in the detection of Sirt1 and Mapt transcriptions. Results: Compared with the MCAO/R group, AS-IV can significantly improve the neurological dysfunction (p < 0.05), reduce the infarction area (p < 0.05), raise the expression of SIRT1 (p < 0.05), and alleviate the abnormal hyperacetylation and hyperphosphorylation of MAPT (p < 0.05). While compared with the AS-IV group, AS-IV + EX527 group showed higher mNSS scores (p < 0.05), more severe cerebral infarction (p < 0.05), lower SIRT1 expression (p < 0.01), and higher ac-MAPT and p-MAPT levels (p < 0.05). Conclusion: AS-IV can improve the neurological deficit after CIR injury in rats and reduce the cerebral infarction area, which exerts neuroprotective effects probably through the Sirt1/Mapt pathway.
Collapse
Affiliation(s)
- Yi-Hua Shi
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xi-Le Zhang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peng-Jie Ying
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zi-Qian Wu
- Department of Neurology, Wenzhou Hospital of Traditional Chinese Medicine Affiliated toZhejiang Chinese Medical University, Wenzhou, China
| | - Le-Le Lin
- Department of Neurology, Wenzhou Hospital of Traditional Chinese Medicine Affiliated toZhejiang Chinese Medical University, Wenzhou, China
| | - Wei Chen
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guo-Qing Zheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wen-Zong Zhu
- Department of Neurology, Wenzhou Hospital of Traditional Chinese Medicine Affiliated toZhejiang Chinese Medical University, Wenzhou, China
| |
Collapse
|
15
|
Zeng W, Lei Q, Ma J, Gao S, Ju R. Endothelial Progenitor Cell-Derived Microvesicles Promote Angiogenesis in Rat Brain Microvascular Endothelial Cells In vitro. Front Cell Neurosci 2021; 15:638351. [PMID: 33679329 PMCID: PMC7930325 DOI: 10.3389/fncel.2021.638351] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 01/26/2021] [Indexed: 12/14/2022] Open
Abstract
Brain microvascular endothelial cells (BMECs) are a major component of the blood-brain barrier that maintains brain homeostasis. Preserving and restoring the normal biological functions of BMECs can reverse or reduce brain injury. Endothelial progenitor cells (EPCs) may promote brain vascular remodeling and restore normal endothelial function. As a novel vehicle for cell-cell communication, microvesicles (MVs) have varied biological functions. The present study investigated the biological effects of EPC-derived MVs (EPC-MVs) on BMECs in vitro. We isolated MVs from the supernatant of EPCs in a serum-depleted medium. BMECs were cultured alone or in the presence of EPC-MVs. BMEC viability and proliferation were evaluated with the Cell Counting Kit-8 and by flow cytometry, and the proangiogenic effect of EPC-MVs on BMECs was assessed with the transwell migration, wound healing, and tube formation assays. Our results showed that EPC-derived MVs labeled with DiI were internalized by cultured BMECs; this enhanced BMEC viability and promoted their proliferation. EPC-MVs also stimulated migration and tube formation in BMECs. These results demonstrate that EPC-derived MVs exert a proangiogenic effect on BMECs, which has potential applications in cell-free therapy for brain injury.
Collapse
Affiliation(s)
- Wen Zeng
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiaoling Lei
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiao Ma
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shuqiang Gao
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rong Ju
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
16
|
Effect of Tetramethylpyrazine on Neuroplasticity after Transient Focal Cerebral Ischemia Reperfusion in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:1587241. [PMID: 33531914 PMCID: PMC7834793 DOI: 10.1155/2021/1587241] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/23/2020] [Accepted: 01/07/2021] [Indexed: 11/22/2022]
Abstract
Tetramethylpyrazine (TMP) has been widely used in ischemic stroke in China. The regulation of neuroplasticity may underlie the recovery of some neurological functions in ischemic stroke. Middle cerebral artery occlusion (MCAO) model was established in this study. Rats were divided into three groups: sham group, model group, and TMP group. The neurological function was evaluated using modified neurological severity score (mNSS). Following the neurological function test, expression of synaptophysin (SYP) and growth-associated protein 43 (GAP-43) were analyzed through immunohistochemistry at 3 d, 7 d, 14 d, and 28 d after MCAO. Finally, the synaptic structural plasticity was investigated using transmission electron microscopy (TEM). The TMP group showed better neurological function comparing to the model group. SYP levels increased gradually in ischemic penumbra (IP) in the model group and could be enhanced by TMP treatment at 7 d, 14 d, and 28 d, whereas GAP-43 levels increased from 3 d to 7 d and thereafter decreased gradually from 14 d to 28 d in the model group, which showed no significant improvement in the TMP group. The results of TEM showed a flatter synaptic interface, a thinner postsynaptic density (PSD), and a wider synaptic cleft in the model group, and the first two alterations could be ameliorated by TMP. Then, a Pearson's correlation test revealed mNSS markedly correlated with SYP and synaptic ultrastructures. Taken together, TMP is capable of promoting functional outcome after ischemic stroke, and the mechanisms may be partially associated with regulation of neuroplasticity.
Collapse
|
17
|
Xue Y, Ding J, Liu Y, Pan Y, Zhao P, Ren Z, Xu J, Ye L, Xu Y. Preparation and Evaluation of Recombinant Human Erythropoietin Loaded Tween 80-Albumin Nanoparticle for Traumatic Brain Injury Treatment. Int J Nanomedicine 2020; 15:8495-8506. [PMID: 33154639 PMCID: PMC7608583 DOI: 10.2147/ijn.s264025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/25/2020] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Traumatic brain injury (TBI) is a serious health problem with few available treatment options. Rh-erythropoietin (rh-EPO) is a potential therapeutic drug for TBI, but it cannot cross the blood-brain barrier (BBB) directly. In this regard, a novel strategy to deliver rh-EPO for enhanced TBI treatment is via the development of Tween 80 modified albumin nanoparticles using electrostatic spray technology. METHODS The rh-EPO loaded Tween 80 modified albumin nanoparticles (rh-EPO-Tw-ABNPs) were prepared by electrostatic spray technology, while the process parameters were optimized via a single factor design. Investigation of physicochemical properties, bioactivity and stability of rh-EPO-Tw-ABNPs was carried out. The in vitro release and biocompatibility with nerve cells were also analyzed. The in vivo brain targeting efficiency, brain edema relieving effect and the expression of aquaporin 4 (AQP4) and glial fibrillary acidic protein (GFAP) in the brain were evaluated in TBI model rats. RESULTS The particle size of optimal rh-EPO-Tw-ABNPs was about 438 ± 45 nm, with a zeta potential of -25.42 ± 0.8 mv. The average drug loading ratio of rh-EPO-Tw-ABNPs was 21.3± 3.7 IU/mg with a relative bioactivity of 91.6 ± 4.1%. The in vitro release of rh-EPO from the nanoparticles was rather slow, while neither the blank Tw-ABNPs nor rh-EPO-Tw-ABNPs exhibited toxicity on the microglia cells. Furthermore, in vivo experiments indicated that the rh-EPO-Tw-ABNPs could enhance the distribution of EPO in the brain and relieve brain edema more effectively. Moreover, compared with an rh-EPO injection, the rh-EPO-Tw-ABNPs could increase the AQP4 level but reduced GFAP expression in the brain with more efficiency. CONCLUSION The rh-EPO-Tw-ABNPs could enhance the transport of rh-EPO into the brain with superior therapeutic effect for TBI.
Collapse
Affiliation(s)
- Yuanfeng Xue
- Department of Neurosurgery, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch Southeast University, Nanjing211200, People’s Republic of China
| | - Junhong Ding
- Department of Neurosurgery, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch Southeast University, Nanjing211200, People’s Republic of China
| | - Yulong Liu
- College of Pharmacy, Jiangsu University, Zhenjiang212013, People’s Republic of China
| | - Yuchun Pan
- Department of Neurosurgery, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch Southeast University, Nanjing211200, People’s Republic of China
| | - Penglai Zhao
- Department of Neurosurgery, Brain Hospital Affiliated to Nanjing Medical University, Nanjing210029, People’s Republic of China
| | - Zhiwen Ren
- Department of Neurosurgery, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch Southeast University, Nanjing211200, People’s Republic of China
| | - Jian Xu
- Department of Neurosurgery, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch Southeast University, Nanjing211200, People’s Republic of China
| | - Liangliang Ye
- Department of Neurosurgery, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch Southeast University, Nanjing211200, People’s Republic of China
| | - Ying Xu
- College of Pharmacy, Jiangsu University, Zhenjiang212013, People’s Republic of China
| |
Collapse
|
18
|
Zhou Y, Sun B, Guo J, Zhou G. Intranasal injection of recombinant human erythropoietin improves cognitive and visual impairments in chronic cerebral ischemia rats. Biomed Rep 2020; 13:40. [PMID: 32934813 DOI: 10.3892/br.2020.1347] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 07/13/2020] [Indexed: 01/02/2023] Open
Abstract
The present study aimed to study the protective effect of intranasally delivered recombinant human erythropoietin (rhEPO) on cognitive and visual impairments in a permanent bilateral common carotid artery occlusion (2VO)-induced chronic cerebral ischemia (CCI) rat model. Male Sprague-Dawley rats (age, 6 months) with 2VO-induced CCI were treated with intranasal rhEPO (50 U/100 g) once per week for 8 weeks. A Morris water maze was used to evaluate the spatial learning and memory of the rats. Flash visual evoked potentials were measured to assess retinal function. Hematoxylin and eosin staining was performed to visualize and evaluate histopathological changes in the cerebral cortex, the hippocampus CA1 region and the retina. CCI-induced learning, memory and visual impairments were significantly alleviated in rats treated with rhEPO compared with those treated with a saline vehicle control. This was evidenced by remarkably decreased escape latency, increased frequency of crossing the hidden platform and elevated amplitude of primary wave in the rats treated with rhEPO. In addition, the rats experienced CCI-induced histopathological alterations, demonstrated by thinning of the cerebral cortex and retina, and losses of neurons and retinal ganglion cells. These alterations were significantly reversed in response to rhEPO administration compared with the saline vehicle control group. rhEPO may exert a protective role against cognitive and visual impairments in rats with CCI at least partially through preventing the thinning of the cerebral cortex and retina, as well as by inhibiting the loss of neurons and retinal ganglion cells.
Collapse
Affiliation(s)
- Yanhui Zhou
- Department of Internal Medicine, Shanxi Eye Hospital, Taiyuan, Shanxi 030002, P.R. China
| | - Bin Sun
- Department of Orbitopathy, Shanxi Eye Hospital, Taiyuan, Shanxi 030002, P.R. China
| | - Junhong Guo
- Department of Neurology, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Guohong Zhou
- Department of Lacrimal Duct, Shanxi Eye Hospital, Taiyuan, Shanxi 030002, P.R. China
| |
Collapse
|
19
|
Effects and Mechanisms of Five Psoralea Prenylflavonoids on Aging-Related Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2128513. [PMID: 32655760 PMCID: PMC7320294 DOI: 10.1155/2020/2128513] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/12/2020] [Accepted: 05/28/2020] [Indexed: 01/09/2023]
Abstract
During the aging process, senescent cells gradually accumulate in the organs; they secrete proinflammatory cytokines and other factors, collectively known as the senescence-associated secretory phenotype (SASP). SASP secretions contribute to “inflammaging,” which is a state of chronic, systemic, sterility, low-grade inflammatory microenvironment and a key risk factor in the development of aging-related diseases. Fructus psoraleae is a traditional Chinese medical herb best known for delaying aging and treating osteoporosis. Prenylflavonoids from fructus psoraleae are the main bioactive compounds responsible for its pharmacological applications, such as beaching, bavachinin, bavachalcone, isobavachalcone, and neobavaisoflavone. In previous decades, there have been some promising studies on the pharmacology of fructus psoraleae. Here, we focus on the anti-inflammatory and antiaging diseases of five psoralea prenylflavonoids, such as cardiovascular protection, diabetes and obesity intervention, neuroprotection, and osteoporosis, and discuss the mechanism of these active ingredients for better understanding the material basis and drug application of fructus psoraleae in Chinese medicine.
Collapse
|
20
|
Xia M, Ruan Z, Chen B, Wang Y, Zhou Z, Ren S, Wu L, Tang N. Wuzang Wenyang Huayu decoction regulates differentially expressed transcripts in the rats' hippocampus after cerebral hypoperfusion. J Cell Mol Med 2020; 24:294-303. [PMID: 31705584 PMCID: PMC6933406 DOI: 10.1111/jcmm.14723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/14/2019] [Accepted: 08/27/2019] [Indexed: 11/30/2022] Open
Abstract
The modified Wenyang Huayu decoction has been widely used to treat vascular dementia in China for thousands of years. We have previously proved that a modified version, Wuzang Wenyang Huayu decoction has the potential to be a more effective clinical treatment that can attenuate cerebral ischaemic injury. However, the global transcript profile and signalling conduction pathways regulated by this recipe remains unclear. This study established a two-vessel occlusion rat model by bilateral common carotid artery occlusion. Two groups of rats were intragastrically treated Wuzang Wenyang Huayu 2.5 g/kg vs or Piracetam 0.15 g/kg for 2 weeks. Learning and memory abilities were measured with Morris water maze. Neuronal plasticity was observed by HE staining. Differentially expressed transcripts of rat hippocampus were analysed by transcriptomics with Illumina HiSeq2500 platform. Results showed that Wuzang Wenyang Huayu decoction significantly alleviated learning, memory deficits, coordination dysfunction and prevented hippocampus cellular injury; Results further revealed the increased gene expression in KEGG metabolic pathways (MT-ND2. MT-ND3, MT-ND4, MT-ND4L, MT-ND5 and MT-ATP8) and genes involved in signal transduction, carcinogenesis, immune system, endocrine system, nervous system etc (Results further revealed differential expression of genes involved in various systems, including MT-ND2) Our discovery is likely to provide new insights to molecular mechanisms of Wuzang Wenyang Huayu regarding hippocampal transcripts in a murine vascular dementia model.
Collapse
Affiliation(s)
- Meng Xia
- School of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Ziyun Ruan
- School of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Ben Chen
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Yunqiao Wang
- School of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Zengzi Zhou
- The 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Shiding Ren
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- The First Affiliated Hospital to Guangxi University of Chinese Medicine, Nanning, China
| | - Lin Wu
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- The First Affiliated Hospital to Guangxi University of Chinese Medicine, Nanning, China
| | - Nong Tang
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- The First Affiliated Hospital to Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
21
|
Song Z, Zhang JH. Recent Advances in Stem Cell Research in Subarachnoid Hemorrhage. Stem Cells Dev 2019; 29:178-186. [PMID: 31752600 DOI: 10.1089/scd.2019.0219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a type of hemorrhagic stroke with significant morbidity and mortality, and it often leads to poor clinical outcome. Although great efforts have been made toward animal and clinical studies, optimal therapy of SAH remains a challenge for scientists and clinicians. Increasing evidence suggests that stem-cell-based therapies may provide innovative approaches for treatment of SAH-related disability. In this review, we summarized the recent advances in stem cell research in SAH. Neuroregeneration after SAH could be conducted by the activation of endogenous neural stem cells (NSCs), transplantation of external stem cells, or reprogramming non-neuronal cell to neurons. The potential mechanism and signaling pathways, as well as the efficiency and safety of these stem cell treatments, were discussed in detail. Although lots of challenges remain for translating the laboratory findings and technologies into clinical therapies, these research studies provided the foundation and guidance for using different resources of stem cells as a brain repair strategy after SAH.
Collapse
Affiliation(s)
- Zhijun Song
- Department of Neurosurgery, Xingtai Third Hospital, Xingtai, China.,Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, California
| | - John H Zhang
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, California.,Department of Neurosurgery, Loma Linda University, Loma Linda, California.,Department of Anesthesiology, Loma Linda University, Loma Linda, California
| |
Collapse
|
22
|
Zhang B, Zhu X, Wang L, Hao S, Xu X, Niu F, He W, Liu B. Dexamethasone impairs neurofunctional recovery in rats following traumatic brain injury by reducing circulating endothelial progenitor cells and angiogenesis. Brain Res 2019; 1725:146469. [PMID: 31541641 DOI: 10.1016/j.brainres.2019.146469] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/24/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022]
Abstract
The administration of glucocorticoids (GCs) after traumatic brain injury (TBI) is controversial. Clinical evidence reveals the deleterious effects of GCs, but the mechanism remains unclear. Previous studies indicate that GCs impair wound healing by affecting endothelial progenitor cell (EPC) function and inhibiting angiogenesis after skin injury. Thus, we hypothesize that the central deleterious effect of GCs is associated with reduced EPCs and angiogenesis after TBI. Using a controlled cortical impact model, we examined the dynamic changes in circulating EPCs and in the regional microcirculation within 14 days of TBI by flow cytometry analysis and contrast-enhanced ultrasound, respectively. The modified neurological severity score (mNSS) and Morris water maze assay were used to assess neurological recovery. Angiogenesis and hippocampal neuron counts were assessed using immunohistochemistry analysis and hematoxylin and eosin staining 14 days after TBI. Compared with the TBI control group, dexamethasone treatment significantly reduced the number of circulating EPCs on days 1, 3, 7 and 14 (P < 0.05); decreased the number of CD31+ cells, the peak intensity and the number of hippocampal neurons on day 14 (P < 0.05); increased the latency on days 12 and 13 (P < 0.05); and reduced the percentage of time spent in the goal quadrant (P < 0.05) on day 14. Similarly, dexamethasone increased the mNSS on days 7 and 14 (P < 0.05). A strong correlation was observed between these results at 14 days after TBI (r = 0.815-0.892, P < 0.05). These data indicate that DEX inhibits the mobilization of EPC levels and angiogenesis around the lesion after TBI, which may contribute to neuronal cell loss and impaired neurofunction.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xueli Zhu
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liang Wang
- Department of Neurosurgery, Tianjin Fifth Center Hospital, Tianjin, China
| | - Shuyu Hao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaojian Xu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fei Niu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Wen He
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Baiyun Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Nerve Injury and Repair Center of Beijing Institute for Brain Disorders, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
23
|
Zhou Y, Shao A, Xu W, Wu H, Deng Y. Advance of Stem Cell Treatment for Traumatic Brain Injury. Front Cell Neurosci 2019; 13:301. [PMID: 31456663 PMCID: PMC6700304 DOI: 10.3389/fncel.2019.00301] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/19/2019] [Indexed: 01/01/2023] Open
Abstract
Traumatic brain injury (TBI) is an important cause of human mortality and morbidity, which can induce serious neurological damage. At present, clinical treatments for neurological dysfunction after TBI include hyperbaric oxygen, brain stimulation and behavioral therapy, but the therapeutic effect is not satisfactory. Recent studies have found that exogenous stem cells can migrate to damaged brain tissue, then participate in the repair of damaged brain tissue by further differentiation to replace damaged cells, while releasing anti-inflammatory factors and growth factors, thereby significantly improving neurological function. This article will mainly review the effects, deficiencies and related mechanisms of different types of stem cells in TBI.
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
24
|
Ostrowski RP, Zhang JH. The insights into molecular pathways of hypoxia-inducible factor in the brain. J Neurosci Res 2018; 98:57-76. [PMID: 30548473 DOI: 10.1002/jnr.24366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022]
Abstract
The objectives of this present work were to review recent developments on the role of hypoxia-inducible factor (HIF) in the survival of cells under normoxic versus hypoxic and inflammatory brain conditions. The dual nature of HIF effects appears well established, based on the accumulated evidence of HIF playing both the role of adaptive factor and mediator of cell demise. Cellular HIF responses depend on pathophysiological conditions, developmental phase, comorbidities, and administered medications. In addition, HIF-1α and HIF-2α actions may vary in the same tissues. The multiple roles of HIF in stem cells are emerging. HIF not only regulates expression of target genes and thereby influences resultant protein levels but also contributes to epigenetic changes that may reciprocally provide feedback regulations loops. These HIF-dependent alterations in neurological diseases and its responses to treatments in vivo need to be examined alongside with a functional status of subjects involved in such studies. The knowledge of HIF pathways might be helpful in devising HIF-mimetics and modulating drugs, acting on the molecular level to improve clinical outcomes, as exemplified here by clinical and experimental data of selected brain diseases, occasionally corroborated by the data from disorders of other organs. Because of complex role of HIF in brain injuries, prospective therapeutic interventions need to differentially target HIF responses depending on their roles in the molecular mechanisms of neurologic diseases.
Collapse
Affiliation(s)
- Robert P Ostrowski
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - John H Zhang
- Departments of Anesthesiology and Physiology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
25
|
Yao X, Liu C, Feng D, Yin J, Chen G. Transcranial Near-infrared Laser Therapy in Improving Cognitive Recovery of Function Following Traumatic Brain Injury. Curr Neuropharmacol 2018; 16:1320-1326. [PMID: 29564977 PMCID: PMC6251043 DOI: 10.2174/1570159x16666180321100439] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 07/25/2017] [Accepted: 02/28/2018] [Indexed: 11/23/2022] Open
Abstract
Abstract: Traumatic brain injury (TBI) has turned into a major health and socioeconomic problem affecting young people and military personnel. Numerous TBI patients experienced the sequela of brain injury called cognitive impairment, which re-duced functions in attention, working memory, motivation, and execution. In recent years, transcranial near-infrared laser therapy (tNiRLT) as a possible therapy has been gradually applied in treating cognitive impairment post-TBI. In the present review, the biological mechanisms of transcranial tNiRLT for TBI are synthesized mainly based on the photonic impact of chronic mild TBI. Various exciting molecular events possibly occur during the procedure, such as stimulation of ATP pro-duction, regional cerebral blood flow, acupoint, neurogenesis and synaptogenesis, as well as a reduction in anti-inflammatory effect. Some animal experiments and clinical studies of tNiRLT for TBI are outlined. Several labs have displayed that tNiRLT is effective not only in improving neurological functions but also in increasing memory and learning capacity in ro-dent animals’ model of TBI. In a 2 patients case report and a 11-case series, cognitive functions were ameliorated. Efficacy on cognitive and emotional effects was also observed in a double-blind, controlled clinical study. Several Randomized, paral-lel, double blind, sham-controlled trials are underway, aiming to evaluate the efficacy of tLED on cognitive functions and neuropsychiatric status in participants post-TBI. Therefore, tNiRLT is a promising method applied to cognitive impairment following TBI.
Collapse
Affiliation(s)
- Xiyang Yao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China
| | - Chenglin Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China
| | - Dongxia Feng
- Scott & White Clinic-Temple, 2401 S.31st Street, Temple, TX76508, United States
| | - Jun Yin
- Department of Neurosurgery, Taixing Chinese Medicine Hospital, Taixing 225400, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, China
| |
Collapse
|
26
|
Yang CY, Liu SY, Wang HY, Li YL, Guo D, Wang XY, Hua W, Wang GL. Neuroprotection by Propofol Post-Conditioning: Focus on PKMζ/KCC2 Pathway Activity. Cell Mol Neurobiol 2018; 38:691-701. [PMID: 28779332 PMCID: PMC11482026 DOI: 10.1007/s10571-017-0530-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 07/31/2017] [Indexed: 12/11/2022]
Abstract
Critical and major operations are often accompanied by brain ischemic complications. Previous studies found that propofol post-conditioning provided neuroprotective functions through upregulating the expression of potassium chloride cotransporter 2 (KCC2) in gamma-aminobutyric acid (GABA) interneurons. Membrane expression and phosphorylation represents KCC2 activity, which were modulated by a protein kinase C (PKC)-dependent mechanism. However, the role of propofol in increasing KCC2 phosphorylation and the involvement of protein kinase Mζ (PKMζ), a major subtype of PKC, in the KCC2 pathway remained unclear. In this study, we established middle cerebral artery occlusion model in rats to evaluate the long-term recovery of brain functions using behavioral experiments. KCC2 and PKMζ were assessed via western blot. We used the selective inhibitor, zeta inhibitory peptide (ZIP), to investigate the relationship between KCC2 and PKMζ. Intracellular chloride concentration in the hippocampal CA1 area was measured to determine KCC2 activity. We found that propofol, infused at a speed of 20 mg kg-1 h-1 for 2 h at the onset of reperfusion, improved neurological deficits and cognitive dysfunction following ischemia/reperfusion injury. PKMζ expression was significantly upregulated, which improved KCC2 membrane expression and phosphorylation in the ischemic hippocampal CA1 area, and these effects could last up to 28 days. But ZIP inhibited this process. Ultimately, we showed that propofol increased KCC2 phosphorylation and PKMζ was the upstream of KCC2. Propofol led to long-term recovery of brain functions by upregulating the activity of the PKMζ/KCC2 pathway.
Collapse
Affiliation(s)
- Chen-Yi Yang
- Department of Anesthesiology, Tianjin Third Central Hospital, Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Research Centre of the Ministry of Health, Tianjin, 300170, China
| | - Shu-Ying Liu
- Department of Anesthesiology, Tianjin Third Central Hospital, Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Research Centre of the Ministry of Health, Tianjin, 300170, China
| | - Hai-Yun Wang
- Department of Anesthesiology, Tianjin Third Central Hospital, Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Research Centre of the Ministry of Health, Tianjin, 300170, China.
- Tianjin Research Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Yan-Li Li
- Department of Anesthesiology, Tianjin Third Central Hospital, Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Research Centre of the Ministry of Health, Tianjin, 300170, China
| | - Di Guo
- Department of Anesthesiology, Tianjin Third Central Hospital, Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Research Centre of the Ministry of Health, Tianjin, 300170, China
| | - Xin-Yue Wang
- Department of Anesthesiology, Tianjin Third Central Hospital, Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Research Centre of the Ministry of Health, Tianjin, 300170, China
| | - Wei Hua
- Department of Anesthesiology, Tianjin Third Central Hospital, Third Central Clinical College of Tianjin Medical University, Tianjin Key Laboratory of Artificial Cell, Artificial Cell Engineering Research Centre of the Ministry of Health, Tianjin, 300170, China
| | - Guo-Lin Wang
- Tianjin Research Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| |
Collapse
|
27
|
Wu SH, Lu IC, Lee SS, Kwan AL, Chai CY, Huang SH. Erythropoietin attenuates motor neuron programmed cell death in a burn animal model. PLoS One 2018; 13:e0190039. [PMID: 29385149 PMCID: PMC5791978 DOI: 10.1371/journal.pone.0190039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023] Open
Abstract
Burn-induced neuromuscular dysfunction may contribute to long-term morbidity; therefore, it is imperative to develop novel treatments. The present study investigated whether erythropoietin (EPO) administration attenuates burn-induced motor neuron apoptosis and neuroinflammatory response. To validate our hypothesis, a third-degree hind paw burn rat model was developed by bringing the paw into contact with a metal surface at 75°C for 10 s. A total of 24 male Sprague–Dawley rats were randomly assigned to four groups: Group A, sham-control; Group B, burn-induced; Group C, burn + single EPO dose (5000 IU/kg i.p. at D0); and Group D, burn + daily EPO dosage (3000 IU/kg/day i.p. at D0–D6). Two treatment regimens were used to evaluate single versus multiple doses treatment effects. Before sacrifice, blood samples were collected for hematological parameter examination. The histological analyses of microglia activation, iNOS, and COX-2 in the spinal cord ventral horn were performed at week 1 post-burn. In addition, we examined autophagy changes by biomarkers of LC3B and ATG5. The expression of BCL-2, BAX, cleaved caspase-3, phospho-AKT, and mTOR was assessed simultaneously through Western blotting. EPO administration after burn injury attenuated neuroinflammation through various mechanisms, including the reduction of microglia activity as well as iNOS and COX-2 expression in the spinal cord ventral horn. In addition, the expression of phospho-AKT, mTOR and apoptotic indicators, such as BAX, BCL-2, and cleaved caspase-3, was modulated. Furthermore, the activity of burn-induced autophagy in the spinal cord ventral horn characterized by the expression of autophagic biomarkers, LC3B and ATG5, was reduced after EPO administration. The present results indicate that EPO inhibits the AKT-mTOR pathway to attenuate burn-induced motor neuron programmed cell death and microglia activation. EPO can modulate neuroinflammation and programmed cell death and may be a therapeutic candidate for neuroprotection.
Collapse
Affiliation(s)
- Sheng-Hua Wu
- Department of Anesthesiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Anesthesiology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - I-Cheng Lu
- Department of Anesthesiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Anesthesiology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Su-Shin Lee
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Pathology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Hung Huang
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail:
| |
Collapse
|
28
|
Bai XF, Gao YK. Recombinant human erythropoietin for treating severe traumatic brain injury. Medicine (Baltimore) 2018; 97:e9532. [PMID: 29505528 PMCID: PMC5943123 DOI: 10.1097/md.0000000000009532] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 12/05/2017] [Accepted: 12/11/2017] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND This study aimed to explore the efficacy and safety of recombinant human erythropoietin (RHE) for the treatment of severe traumatic brain injury (STBI). METHODS One hundred and twenty eligible patients with STBI were randomly divided into an intervention group or a control group equally. Patients in the intervention group received RHE. The participants in the control group received 0.9% saline. The outcome measurements included the Glasgow Outcome Scale (GOS) scores, mortality, and any adverse events. RESULTS At the end of 10-week follow-up after treatment, RHE neither showed greater efficacy in GOS scores (1-2, P = .43; 3-4, P = .25; 5-6, P = .58; 7-8, P = .23), nor the lower mortality in the intervention group than those in the control group (P = .47). In addition, both groups had similar safety profile. CONCLUSION This study found that RHE did not improve the neurological outcomes in patients with STBI.
Collapse
|
29
|
Zhao H, Pan P, Yang Y, Ge H, Chen W, Qu J, Shi J, Cui G, Liu X, Feng H, Chen Y. Endogenous hydrogen sulphide attenuates NLRP3 inflammasome-mediated neuroinflammation by suppressing the P2X7 receptor after intracerebral haemorrhage in rats. J Neuroinflammation 2017; 14:163. [PMID: 28821266 PMCID: PMC5563049 DOI: 10.1186/s12974-017-0940-4] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 08/10/2017] [Indexed: 12/31/2022] Open
Abstract
Background Emerging studies have demonstrated the important physiological and pathophysiological roles of hydrogen sulphide (H2S) as a gasotransmitter for NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome-associated neuroinflammation in the central nervous system. However, the effects of H2S on neuroinflammation after intracerebral haemorrhage (ICH), especially on the NLRP3 inflammasome, remain unknown. Methods We employed a Sprague–Dawley rat of collagenase-induced ICH in the present study. The time course of H2S content and the spatial expression of cystathionine-β-synthase (CBS) after ICH, the effects of endogenous and exogenous H2S after ICH, the effects of endogenous and exogenous H2S on NLRP3 inflammasome activation under P2X7 receptor (P2X7R) overexpression after ICH, and the involvement of the P2X7R in the mechanism by which microglia-derived H2S prevented NLRP3 inflammasome activation were investigated. Results We found ICH induced significant downregulation of endogenous H2S production in the brain, which may be the result of decreasing in CBS, the predominant cerebral H2S-generating enzyme. Administration of S-adenosyl-l-methionine (SAM), a CBS-specific agonist, or sodium hydrosulfide (NaHS), a classical exogenous H2S donor, not only restored brain and plasma H2S content but also attenuated brain oedema, microglial accumulation and neurological deficits at 1 day post-ICH by inhibiting the P2X7R/NLRP3 inflammasome cascade. Endogenous H2S production, which was derived mainly by microglia and above treatments, was verified by adenovirus-overexpressed P2X7R and in vitro primary microglia studies. Conclusions These results indicated endogenous H2S synthesis was impaired after ICH, which plays a pivotal role in the P2X7R/NLRP3 inflammasome-associated neuroinflammatory response in the pathogenesis of secondary brain injury. Maintaining appropriate H2S concentrations in the central nervous system may represent a potential therapeutic strategy for managing post-ICH secondary brain injury and associated neurological deficits.
Collapse
Affiliation(s)
- Hengli Zhao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Pengyu Pan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yang Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Hongfei Ge
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Weixiang Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Jie Qu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Jiantao Shi
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Gaoyu Cui
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xin Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
| |
Collapse
|
30
|
Zhao F, Qu Y, Zhu J, Zhang L, Huang L, Liu H, Li S, Mu D. miR-30d-5p Plays an Important Role in Autophagy and Apoptosis in Developing Rat Brains After Hypoxic–Ischemic Injury. J Neuropathol Exp Neurol 2017; 76:709-719. [DOI: 10.1093/jnen/nlx052] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
31
|
Salehi A, Zhang JH, Obenaus A. Response of the cerebral vasculature following traumatic brain injury. J Cereb Blood Flow Metab 2017; 37:2320-2339. [PMID: 28378621 PMCID: PMC5531360 DOI: 10.1177/0271678x17701460] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The critical role of the vasculature and its repair in neurological disease states is beginning to emerge particularly for stroke, dementia, epilepsy, Parkinson's disease, tumors and others. However, little attention has been focused on how the cerebral vasculature responds following traumatic brain injury (TBI). TBI often results in significant injury to the vasculature in the brain with subsequent cerebral hypoperfusion, ischemia, hypoxia, hemorrhage, blood-brain barrier disruption and edema. The sequalae that follow TBI result in neurological dysfunction across a host of physiological and psychological domains. Given the importance of restoring vascular function after injury, emerging research has focused on understanding the vascular response after TBI and the key cellular and molecular components of vascular repair. A more complete understanding of vascular repair mechanisms are needed and could lead to development of new vasculogenic therapies, not only for TBI but potentially vascular-related brain injuries. In this review, we delineate the vascular effects of TBI, its temporal response to injury and putative biomarkers for arterial and venous repair in TBI. We highlight several molecular pathways that may play a significant role in vascular repair after brain injury.
Collapse
Affiliation(s)
- Arjang Salehi
- 1 Cell, Molecular and Developmental Biology Program, University of California, Riverside, CA, USA.,2 Department of Pediatrics, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- 3 Department of Physiology and Pharmacology Loma Linda University School of Medicine, CA, USA.,4 Department of Anesthesiology Loma Linda University School of Medicine, CA, USA.,5 Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Andre Obenaus
- 1 Cell, Molecular and Developmental Biology Program, University of California, Riverside, CA, USA.,2 Department of Pediatrics, Loma Linda University, Loma Linda, CA, USA.,6 Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
32
|
Yu DF, Zhu LH, Jiang L. Recombinant Human Erythropoietin Augments Neovascularization Responses in a Neonatal Rat Model of Premature Brain Damage by Phosphatidylinositol 3 Kinase/Akt Pathway. Chin Med J (Engl) 2017; 130:854-858. [PMID: 28345550 PMCID: PMC5381320 DOI: 10.4103/0366-6999.202744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Background: Recombinant human-erythropoietin (rh-EPO) has therapeutic efficacy for premature infants with brain damage during the active rehabilitation and anti-inflammation. In the present study, we found that the rh-EPO was related to the promotion of neovascularization. Our aim was to investigate whether rh-EPO augments neovascularization in the neonatal rat model of premature brain damage through the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt) signaling pathway. Methods: Postnatal day 5 (PD5), rats underwent permanent ligation of the right common carotid artery and were exposed to hypoxia for 2 h. All the rat pups were randomized into five groups as follows: (1) control group; (2) hypoxia-ischemic (HI) group; (3) HI + LY294002 group; (4) HI + rh-EPO group; and (5) HI + rh-EPO + LY294002 group. The phospho-Akt protein was tested 90 min after the whole operation, and CD34, vascular endothelial growth factor receptor 2 (VEGFR2), and vascular endothelial growth factor (VEGF) were also tested 2 days after the whole operation. Results: In the hypoxic and ischemic zone of the premature rat brain, the rh-EPO induced CD34+ cells to immigrate to the HI brain zone (P < 0.05) and also upregulated the VEGFR2 protein expression (P < 0.05) and VEGF mRNA level (P < 0.05) through the PI3K/Akt (P < 0.05) signaling pathway when compared with other groups. Conclusions: The rh-EPO treatment augments neovascularization responses in the neonatal rat model of premature brain damage through the PI3K/Akt signaling pathway. Besides, the endogenous EPO may exist in the HI zone of rat brain and also has neovascularization function through the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Da-Fan Yu
- Department of Clinical Medcine, Medical School of Southeast University, Jiangsu 210009, China
| | - Li-Hua Zhu
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, China
| | - Li Jiang
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
33
|
Pearn ML, Niesman IR, Egawa J, Sawada A, Almenar-Queralt A, Shah SB, Duckworth JL, Head BP. Pathophysiology Associated with Traumatic Brain Injury: Current Treatments and Potential Novel Therapeutics. Cell Mol Neurobiol 2017; 37:571-585. [PMID: 27383839 PMCID: PMC11482200 DOI: 10.1007/s10571-016-0400-1] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/24/2016] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of death of young people in the developed world. In the United States alone, 1.7 million traumatic events occur annually accounting for 50,000 deaths. The etiology of TBI includes traffic accidents, falls, gunshot wounds, sports, and combat-related events. TBI severity ranges from mild to severe. TBI can induce subtle changes in molecular signaling, alterations in cellular structure and function, and/or primary tissue injury, such as contusion, hemorrhage, and diffuse axonal injury. TBI results in blood-brain barrier (BBB) damage and leakage, which allows for increased extravasation of immune cells (i.e., increased neuroinflammation). BBB dysfunction and impaired homeostasis contribute to secondary injury that occurs from hours to days to months after the initial trauma. This delayed nature of the secondary injury suggests a potential therapeutic window. The focus of this article is on the (1) pathophysiology of TBI and (2) potential therapies that include biologics (stem cells, gene therapy, peptides), pharmacological (anti-inflammatory, antiepileptic, progrowth), and noninvasive (exercise, transcranial magnetic stimulation). In final, the review briefly discusses membrane/lipid rafts (MLR) and the MLR-associated protein caveolin (Cav). Interventions that increase Cav-1, MLR formation, and MLR recruitment of growth-promoting signaling components may augment the efficacy of pharmacologic agents or already existing endogenous neurotransmitters and neurotrophins that converge upon progrowth signaling cascades resulting in improved neuronal function after injury.
Collapse
Affiliation(s)
- Matthew L Pearn
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Ingrid R Niesman
- Department of Cellular and Molecular Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, San Diego, CA, 92037, USA
| | - Junji Egawa
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Atsushi Sawada
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Angels Almenar-Queralt
- Department of Cellular and Molecular Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, San Diego, CA, 92037, USA
| | - Sameer B Shah
- UCSD Departments of Orthopaedic Surgery and Bioengineering, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Josh L Duckworth
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Brian P Head
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA.
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
34
|
Rehabilitation Treatment and Progress of Traumatic Brain Injury Dysfunction. Neural Plast 2017; 2017:1582182. [PMID: 28491478 PMCID: PMC5405588 DOI: 10.1155/2017/1582182] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/28/2017] [Accepted: 03/07/2017] [Indexed: 11/17/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of chronic disability. Worldwide, it is the leading cause of disability in the under 40s. Behavioral problems, mood, cognition, particularly memory, attention, and executive function are commonly impaired by TBI. Spending to assist, TBI survivors with disabilities are estimated to be costly per year. Such impaired functional outcomes following TBI can be improved via various rehabilitative approaches. The objective of the present paper is to review the current rehabilitation treatment of traumatic brain injury in adults.
Collapse
|
35
|
Chen Q, Feng Z, Tan Q, Guo J, Tang J, Tan L, Feng H, Chen Z. Post-hemorrhagic hydrocephalus: Recent advances and new therapeutic insights. J Neurol Sci 2017; 375:220-230. [PMID: 28320134 DOI: 10.1016/j.jns.2017.01.072] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 01/25/2017] [Accepted: 01/30/2017] [Indexed: 02/07/2023]
Abstract
Post-hemorrhagic hydrocephalus (PHH), also referred to as progressive ventricular dilatation, is caused by disturbances in cerebrospinal fluid (CSF) flow or absorption following hemorrhage in the brain. As one of the most serious complications of neonatal/adult intraventricular hemorrhage (IVH), subarachnoid hemorrhage (SAH), and traumatic brain injury (TBI), PHH is associated with increased morbidity and disability of these events. Common sequelae of PHH include neurocognitive impairment, motor dysfunction, and growth impairment. Non-surgical measures to reduce increased intracranial pressure (ICP) in PHH have shown little success and most patients will ultimately require surgical management, such as external ventricular drainage and shunting which mostly by inserting a CSF drainage shunt. Unfortunately, shunt complications are common and the optimum time for intervention is unclear. To date, there remains no comprehensive strategy for PHH management and it becomes imperative that to explore new therapeutic targets and methods for PHH. Over past decades, increasing evidence have indicated that hemorrhage-derived blood and subsequent metabolic products may play a key role in the development of IVH-, SAH- and TBI-associated PHH. Several intervention strategies have recently been evaluated and cross-referenced. In this review, we summarized and discussed the common aspects of hydrocephalus following IVH, SAH and TBI, relevant experimental animal models, clinical translation of in vivo experiments, and potential preventive and therapeutic targets for PHH.
Collapse
Affiliation(s)
- Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Zhou Feng
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Jing Guo
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China; Department of Neurosurgery, The 211st Hospital of PLA, Harbin 150086, China
| | - Jun Tang
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China.
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, The Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
36
|
Progenitor Cells for Arterial Repair: Incremental Advancements towards Therapeutic Reality. Stem Cells Int 2017; 2017:8270498. [PMID: 28232850 PMCID: PMC5292398 DOI: 10.1155/2017/8270498] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 12/18/2016] [Indexed: 02/08/2023] Open
Abstract
Coronary revascularization remains the standard treatment for obstructive coronary artery disease and can be accomplished by either percutaneous coronary intervention (PCI) or coronary artery bypass graft surgery. Considerable advances have rendered PCI the most common form of revascularization and improved clinical outcomes. However, numerous challenges to modern PCI remain, namely, in-stent restenosis and stent thrombosis, underscoring the importance of understanding the vessel wall response to injury to identify targets for intervention. Among recent promising discoveries, endothelial progenitor cells (EPCs) have garnered considerable interest given an increasing appreciation of their role in vascular homeostasis and their ability to promote vascular repair after stent placement. Circulating EPC numbers have been inversely correlated with cardiovascular risk, while administration of EPCs in humans has demonstrated improved clinical outcomes. Despite these encouraging results, however, advancing EPCs as a therapeutic modality has been hampered by a fundamental roadblock: what constitutes an EPC? We review current definitions and sources of EPCs as well as the proposed mechanisms of EPC-mediated vascular repair. Additionally, we discuss the current state of EPCs as therapeutic agents, focusing on endogenous augmentation and transplantation.
Collapse
|
37
|
Wang H, Li P, Xu N, Zhu L, Cai M, Yu W, Gao Y. Paradigms and mechanisms of inhalational anesthetics mediated neuroprotection against cerebral ischemic stroke. Med Gas Res 2016; 6:194-205. [PMID: 28217291 PMCID: PMC5223310 DOI: 10.4103/2045-9912.196901] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cerebral ischemic stroke is a leading cause of serious long-term disability and cognitive dysfunction. The high mortality and disability of cerebral ischemic stroke is urging the health providers, including anesthesiologists and other perioperative professioners, to seek effective protective strategies, which are extremely limited, especially for those perioperative patients. Intriguingly, several commonly used inhalational anesthetics are recently suggested to possess neuroprotective effects against cerebral ischemia. This review introduces multiple paradigms of inhalational anesthetic treatments that have been investigated in the setting of cerebral ischemia, such as preconditioning, proconditioning and postconditioning with a variety of inhalational anesthetics. The pleiotropic mechanisms underlying these inhalational anesthetics-afforded neuroprotection against stroke are also discussed in detail, including the common pathways shared by most of the inhalational anesthetic paradigms, such as anti-excitotoxicity, anti-apoptosis and anti-inflammation. There are also distinct mechanisms involved in specific paradigms, such as preserving blood brain barrier integrity, regulating cerebral blood flow and catecholamine release. The ready availability of these inhalational anesthetics bedside and renders them a potentially translatable stroke therapy attracting great efforts for understanding of the underlying mechanisms.
Collapse
Affiliation(s)
- Hailian Wang
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China; Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peiying Li
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Na Xu
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ling Zhu
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mengfei Cai
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yanqin Gao
- Anesthesiology Department of Huashan Hospital, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China; Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
38
|
Li ZM, Xiao YL, Zhu JX, Geng FY, Guo CJ, Chong ZL, Wang LX. Recombinant human erythropoietin improves functional recovery in patients with severe traumatic brain injury: A randomized, double blind and controlled clinical trial. Clin Neurol Neurosurg 2016; 150:80-83. [DOI: 10.1016/j.clineuro.2016.09.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 07/26/2016] [Accepted: 09/02/2016] [Indexed: 12/25/2022]
|
39
|
Huang JL, Manaenko A, Ye ZH, Sun XJ, Hu Q. Hypoxia therapy--a new hope for the treatment of mitochondrial dysfunctions. Med Gas Res 2016; 6:174-176. [PMID: 27867487 PMCID: PMC5110142 DOI: 10.4103/2045-9912.191365] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Mitochondrial dysfunctions are characteristic features of numerous diseases and play a critical role in disease pathogenesis. Despite intensive research in this area, there are no approved therapies that directly target mitochondria. Recently a study by Jain et al. from Massachusetts General Hospital, USA reported the effectiveness of hypoxia for treatment of mitochondrial disease in mice. In this commentary, we summarized the potential mechanisms underlying the therapeutic effects of hypoxia on mitochondrial dysfunction, and clinical limitations of hypoxia as a therapy for human patients. We hope that our concerns will be helpful for further clinical studies addressing moderate hypoxia in mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jun-Long Huang
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Naval Aviation, the Second Military Medical University, Shanghai, China
| | - Anatol Manaenko
- Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Zhou-Heng Ye
- Department of Naval Aviation, the Second Military Medical University, Shanghai, China
| | - Xue-Jun Sun
- Department of Naval Aviation, the Second Military Medical University, Shanghai, China
| | - Qin Hu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
40
|
Pu H, Jiang X, Hu X, Xia J, Hong D, Zhang W, Gao Y, Chen J, Shi Y. Delayed Docosahexaenoic Acid Treatment Combined with Dietary Supplementation of Omega-3 Fatty Acids Promotes Long-Term Neurovascular Restoration After Ischemic Stroke. Transl Stroke Res 2016; 7:521-534. [PMID: 27566736 DOI: 10.1007/s12975-016-0498-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 12/14/2022]
Abstract
Prophylactic dietary intake of omega-3 polyunsaturated fatty acids (n-3 PUFAs) has been shown to remarkably ameliorate ischemic brain injury. However, the therapeutic efficacy of n-3 PUFA administration post-stroke, especially its impact on neurovascular remodeling and long-term neurological recovery, has not been fully characterized thus far. In this study, we investigated the effect of n-3 PUFA supplementation, as well as in combination with docosahexaenoic acid (DHA) injections, on long-term stroke outcomes. Mice were subjected to transient middle cerebral artery occlusion (MCAO) before randomly assigned to four groups to receive the following: (1) low dose of n-3 PUFAs as the vehicle control, (2) intraperitoneal DHA injections, (3) n-3 PUFA dietary supplement, or (4) combined treatment of (2) and (3). Neurological deficits and brain atrophy, neurogenesis, angiogenesis, and glial scar formation were assessed up to 28 days after MCAO. Results revealed that groups 2 and 3 showed only marginal reduction in post-stroke tissue loss and attenuation of cognitive deficits. Interestingly, group 4 exhibited significantly reduced tissue atrophy and improved cognitive functions compared to groups 2 and 3 with just a single treatment. Mechanistically, the combined treatment promoted post-stroke neurogenesis and angiogenesis, as well as reduced glial scar formation, all of which significantly correlated with the improved spatial memory in the Morris water maze. These results demonstrate an effective therapeutic regimen to enhance neurovascular restoration and long-term cognitive recovery in the mouse model of MCAO. Combined post-stroke DHA treatment and n-3 PUFA dietary supplementation thus may be a potential clinically translatable therapy for stroke or related brain disorders.
Collapse
Affiliation(s)
- Hongjian Pu
- Geriatric Research, Educational, and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Xiaoyan Jiang
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Xiaoming Hu
- Geriatric Research, Educational, and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.,State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jinchao Xia
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Dandan Hong
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Wenting Zhang
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jun Chen
- Geriatric Research, Educational, and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA. .,State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China. .,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Yejie Shi
- Geriatric Research, Educational, and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA. .,Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
41
|
Gatto R, Chauhan M, Chauhan N. Anti-edema effects of rhEpo in experimental traumatic brain injury. Restor Neurol Neurosci 2016; 33:927-41. [PMID: 26484701 DOI: 10.3233/rnn-150577] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Traumatic brain injury (TBI) is one of the leading causes of disability and death which begins with the formation of edema as the persistent primary causative factor in TBI. Although medical management of cerebral edema by hypothermia, ventriculostomy, mannitol or hypertonic saline have been effective in treating edema, many of these therapies end up with some neurologic deficits, necessitating novel treatment options for treating post-TBI edema. This study investigated edema reducing effects of recombinant human Erythropoietin (rhEPO) in reducing acute brain edema in the CCI mouse model of TBI. METHODS Anti-edema effects of rhEpo in reducing acute brain edema after injury in the CCI mouse model of TBI were assessed by T2 weighted magnetic resonance imaging (T2wMRI) as the accurate detector of brain edema in correlation with Western blot analysis of cerebral aquaporin 4 (AQP4) index as the critical marker of edema. RESULTS Results show that rhEpo treatment significantly reduced brain edema with concomitant reduction in AQP4 immunoexpression in the CCI mouse model of TBI. CONCLUSION Current results emphasize clinical utility of rhEpo in treating post-TBI edema.
Collapse
Affiliation(s)
- Rodolfo Gatto
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Neelima Chauhan
- Neuroscience Research, R&D, Jesse Brown VA Medical Center, Chicago, IL, USA.,Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
42
|
An C, Jiang X, Pu H, Hong D, Zhang W, Hu X, Gao Y. Severity-Dependent Long-Term Spatial Learning-Memory Impairment in a Mouse Model of Traumatic Brain Injury. Transl Stroke Res 2016; 7:512-520. [DOI: 10.1007/s12975-016-0483-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/28/2016] [Accepted: 07/11/2016] [Indexed: 01/04/2023]
|
43
|
Grasso G, Tomasello G, Noto M, Alafaci C, Cappello F. Erythropoietin for the Treatment of Subarachnoid Hemorrhage: A Feasible Ingredient for a Successful Medical Recipe. Mol Med 2015; 21:979-987. [PMID: 26581085 DOI: 10.2119/molmed.2015.00177] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/16/2015] [Indexed: 11/06/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) following aneurysm bleeding accounts for 6% to 8% of all cerebrovascular accidents. Although an aneurysm can be effectively managed by surgery or endovascular therapy, delayed cerebral ischemia is diagnosed in a high percentage of patients resulting in significant morbidity and mortality. Cerebral vasospasm occurs in more than half of all patients after aneurysm rupture and is recognized as the leading cause of delayed cerebral ischemia after SAH. Hemodynamic strategies and endovascular procedures may be considered for the treatment of cerebral vasospasm. In recent years, the mechanisms contributing to the development of vasospasm, abnormal reactivity of cerebral arteries and cerebral ischemia following SAH, have been investigated intensively. A number of pathological processes have been identified in the pathogenesis of vasospasm, including endothelial injury, smooth muscle cell contraction from spasmogenic substances produced by the subarachnoid blood clots, changes in vascular responsiveness and inflammatory response of the vascular endothelium. To date, the current therapeutic interventions remain ineffective as they are limited to the manipulation of systemic blood pressure, variation of blood volume and viscosity and control of arterial carbon dioxide tension. In this scenario, the hormone erythropoietin (EPO) has been found to exert neuroprotective action during experimental SAH when its recombinant form (rHuEPO) is administered systemically. However, recent translation of experimental data into clinical trials has suggested an unclear role of recombinant human EPO in the setting of SAH. In this context, the aim of the current review is to present current evidence on the potential role of EPO in cerebrovascular dysfunction following aneurysmal subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Giovanni Grasso
- Neurosurgical Clinic, Department of Experimental Biomedicine and Clinical Neurosciences, Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | - Giovanni Tomasello
- Section of Anatomy, Department of Experimental Biomedicine and Clinical Neurosciences, and Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | | | - Concetta Alafaci
- Department of Neurosurgery, University of Messina, Messina, Italy
| | - Francesco Cappello
- Section of Anatomy, Department of Experimental Biomedicine and Clinical Neurosciences, and Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| |
Collapse
|
44
|
Ran QS, Yu YH, Fu XH, Wen YC. Activation of the Notch signaling pathway promotes neurovascular repair after traumatic brain injury. Neural Regen Res 2015; 10:1258-64. [PMID: 26487853 PMCID: PMC4590238 DOI: 10.4103/1673-5374.162758] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The Notch signaling pathway plays a key role in angiogenesis and endothelial cell formation, but it remains unclear whether it is involved in vascular repair by endothelial progenitor cells after traumatic brain injury. Therefore, in the present study, we controlled the Notch signaling pathway using overexpression and knockdown constructs. Activation of the Notch signaling pathway by Notch1 or Jagged1 overexpression enhanced the migration, invasiveness and angiogenic ability of endothelial progenitor cells. Suppression of the Notch signaling pathway with Notch1 or Jagged1 siRNAs reduced the migratory capacity, invasiveness and angiogenic ability of endothelial progenitor cells. Activation of the Notch signaling pathway in vivo in a rat model of mild traumatic brain injury promoted neurovascular repair. These findings suggest that the activation of the Notch signaling pathway promotes blood vessel formation and tissue repair after brain trauma.
Collapse
Affiliation(s)
- Qi-Shan Ran
- Department of Neurosurgery, the First People's Hospital of ZunYi/the Third Affiliated Hospital of ZunYi Medical College, Zunyi, Guizhou Province, China
| | - Yun-Hu Yu
- Department of Neurosurgery, the First People's Hospital of ZunYi/the Third Affiliated Hospital of ZunYi Medical College, Zunyi, Guizhou Province, China
| | - Xiao-Hong Fu
- Department of Neurosurgery, the First People's Hospital of ZunYi/the Third Affiliated Hospital of ZunYi Medical College, Zunyi, Guizhou Province, China
| | - Yuan-Chao Wen
- Department of Neurosurgery, the First People's Hospital of ZunYi/the Third Affiliated Hospital of ZunYi Medical College, Zunyi, Guizhou Province, China
| |
Collapse
|
45
|
Reis C, Wang Y, Akyol O, Ho WM, Ii RA, Stier G, Martin R, Zhang JH. What's New in Traumatic Brain Injury: Update on Tracking, Monitoring and Treatment. Int J Mol Sci 2015; 16:11903-65. [PMID: 26016501 PMCID: PMC4490422 DOI: 10.3390/ijms160611903] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/04/2015] [Accepted: 05/06/2015] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI), defined as an alteration in brain functions caused by an external force, is responsible for high morbidity and mortality around the world. It is important to identify and treat TBI victims as early as possible. Tracking and monitoring TBI with neuroimaging technologies, including functional magnetic resonance imaging (fMRI), diffusion tensor imaging (DTI), positron emission tomography (PET), and high definition fiber tracking (HDFT) show increasing sensitivity and specificity. Classical electrophysiological monitoring, together with newly established brain-on-chip, cerebral microdialysis techniques, both benefit TBI. First generation molecular biomarkers, based on genomic and proteomic changes following TBI, have proven effective and economical. It is conceivable that TBI-specific biomarkers will be developed with the combination of systems biology and bioinformation strategies. Advances in treatment of TBI include stem cell-based and nanotechnology-based therapy, physical and pharmaceutical interventions and also new use in TBI for approved drugs which all present favorable promise in preventing and reversing TBI.
Collapse
Affiliation(s)
- Cesar Reis
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - Yuechun Wang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
- Department of Physiology, School of Medicine, University of Jinan, Guangzhou 250012, China.
| | - Onat Akyol
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
| | - Wing Mann Ho
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, University Hospital Innsbruck, Tyrol 6020, Austria.
| | - Richard Applegate Ii
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - Gary Stier
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - Robert Martin
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| | - John H Zhang
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, 11041 Campus Street, Risley Hall, Room 219, Loma Linda, CA 92354, USA.
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| |
Collapse
|