1
|
Sharma S, Rehan A, Dutta A. A data mining approach to identify key radioresponsive genes in mouse model of radiation-induced intestinal injury. Biomarkers 2024:1-13. [PMID: 39431989 DOI: 10.1080/1354750x.2024.2420196] [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: 08/13/2024] [Accepted: 10/18/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Radiation-mediated GI injury (RIGI) is observed in humans either due to accidental or intentional exposures. This can only be managed with supporting care and no approved countermeasures are available till now. Early detection and monitoring of RIGI is important for effective medical management and improve survival chances of exposed individuals. OBJECTIVE The present study aims to identify new signatures of RIGI using data mining approach followed by validation of selected hub genes in mice. METHODS Data mining study was performed using microarray datasets from Gene Expression Omnibus database. The differentially expressed genes were identified and further validated in total-body irradiated mice. RESULTS Based on KEGG pathway analysis, lipid metabolism was found as one of the predominant pathways altered in irradiated intestine. Extensive alteration in lipid profile and lipid modification was observed in this tissue. A protein-protein interaction network revealed top 08 hub genes related to lipid metabolism, namely Fabp1, Fabp2, Fabp6, Npc1l1, Ppar-α, Abcg8, Hnf-4α, and Insig1. qRT-PCR analysis revealed significant up-regulation of Fabp6 and Hnf-4α and down-regulation of Fabp1, Fabp2 and Insig1 transcripts in irradiated intestine. Radiation dose and time kinetics study revealed that the selected 05 genes were altered differentially in response to radiation in intestine. CONCLUSION Finding suggests that lipid metabolism is one of the key targets of radiation and its mediators may act as biomarkers in detection and progression of RIGI.
Collapse
Affiliation(s)
- Suchitra Sharma
- GI Radiobiology Research Laboratory, Radiomitigation Research Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K Mazumdar Marg, Delhi, India
| | - Aliza Rehan
- GI Radiobiology Research Laboratory, Radiomitigation Research Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K Mazumdar Marg, Delhi, India
| | - Ajaswrata Dutta
- GI Radiobiology Research Laboratory, Radiomitigation Research Department, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K Mazumdar Marg, Delhi, India
| |
Collapse
|
2
|
Li CL, Wang Q, Wu L, Hu JY, Gao QC, Jiao XL, Zhang YX, Tang S, Yu Q, He PF. The PANoptosis-related hippocampal molecular subtypes and key biomarkers in Alzheimer's disease patients. Sci Rep 2024; 14:23851. [PMID: 39394418 PMCID: PMC11470079 DOI: 10.1038/s41598-024-75377-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/04/2024] [Indexed: 10/13/2024] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder, and various molecules associated with PANoptosis are involved in neuroinflammation and neurodegenerative diseases. This work aims to identify key genes, and characterize PANoptosis-related molecular subtypes in AD. Moreover, we establish a scoring system for distinguishing PANoptosis molecular subtypes and constructing diagnostic models for AD differentiation. A total of 5 hippocampal datasets were obtained from the Gene Expression Omnibus (GEO) database. In total, 1324 protein-encoding genes associated with PANoptosis (1313 apoptosis genes, 11 necroptosis genes, and 31 pyroptosis genes) were extracted from the GeneCards database. The Limma package was used to identify differentially expressed genes. Weighted Gene Co-Expression Network Analysis (WGCNA) was conducted to identify gene modules significantly associated with AD. The ConsensusClusterPlus algorithm was used to identify AD subtypes. Gene Set Variation Analysis (GSVA) was used to assess functional and pathway differences among the subtypes. The Boruta, Least Absolute Shrinkage and Selection Operator (LASSO), Random Forest (RF), and Support Vector Machine Recursive Feature Elimination (SVM-RFE) algorithms were used to select the three PANoptosis-related Key AD genes (PKADg). A scoring model was constructed based on the Boruta algorithm. PANoptosis diagnostic models were developed using the RF, SVM-RFE, and Logistic Regression (LR) algorithms. The ROC curves were used to assess the model performance. A total of 48 important genes were identified by intersecting 725 differentially expressed genes and 2127 highly correlated module genes from WGCNA with 1324 protein-encoding genes related to PANoptosis. Machine learning algorithms identified 3 key AD genes related to PANoptosis, including ANGPT1, STEAP3, and TNFRSF11B. These genes had strong discriminatory capacities among samples, with Receiver Operating Characteristic Curve (ROC) analysis indicating Area Under the Curve (AUC) values of 0.839, 0.8, and 0.868, respectively. Using the 48 important genes, the ConsensusClusterPlus algorithm identified 2 PANoptosis subtypes among AD patients, i.e., apoptosis subtype and mild subtype. Apoptosis subtype patients displayed evident cellular apoptosis and severe functionality damage in the hippocampal tissue. Meanwhile, mild subtype patients showed milder functionality damage. These two subtypes had significant differences in apoptosis and necroptosis; however, there was no apparent variation in pyroptosis functionality. The scoring model achieved an AUC of 100% for sample differentiation. The RF PANoptosis diagnostic model demonstrated an AUC of 100% in the training set and 85.85% in the validation set for distinguishing AD. This study identified two PANoptosis-related hippocampal molecular subtypes of AD, identified key genes, and established machine learning models for subtype differentiation and discrimination of AD. We found that in the context of AD, PANoptosis may influence disease progression through the modulation of apoptosis and necrotic apoptosis.
Collapse
Affiliation(s)
- Chen-Long Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Big Data Clinical Decision Research in Shanxi Province, Taiyuan, China
| | - Qi Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Big Data Clinical Decision Research in Shanxi Province, Taiyuan, China
| | - Li Wu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- Department of Anesthesiology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Jing-Yi Hu
- School of Management, Shanxi Medical University, Taiyuan, China
| | - Qi-Chao Gao
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Big Data Clinical Decision Research in Shanxi Province, Taiyuan, China
| | - Xin-Long Jiao
- School of Medical Science, Shanxi Medical University, Taiyuan, China
| | - Yu-Xiang Zhang
- Second Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Shan Tang
- First Hospital of Shanxi Medical University, Taiyuan, China.
| | - Qi Yu
- Key Laboratory of Big Data Clinical Decision Research in Shanxi Province, Taiyuan, China.
- School of Management, Shanxi Medical University, Taiyuan, China.
| | - Pei-Feng He
- Key Laboratory of Big Data Clinical Decision Research in Shanxi Province, Taiyuan, China.
- School of Management, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
3
|
Tian L, Liu Q, Wang X, Chen S, Li Y. Fighting ferroptosis: Protective effects of dexmedetomidine on vital organ injuries. Life Sci 2024; 354:122949. [PMID: 39127318 DOI: 10.1016/j.lfs.2024.122949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Vital organ injury is one of the leading causes of global mortality and socio-economic burdens. Current treatments have limited efficacy, and new strategies are needed. Dexmedetomidine (DEX) is a highly selective α2-adrenergic receptor that protects multiple organs by reducing inflammation and preventing cell death. However, its exact mechanism is not yet fully understood. Understanding the underlying molecular mechanisms of its protective effects is crucial as it could provide a basis for designing highly targeted and more effective drugs. Ferroptosis is the primary mode of cell death during organ injury, and recent studies have shown that DEX can protect vital organs from this process. This review provides a detailed analysis of preclinical in vitro and in vivo studies and gains a better understanding of how DEX protects against vital organ injuries by inhibiting ferroptosis. Our findings suggest that DEX can potentially protect vital organs mainly by regulating iron metabolism and the antioxidant defense system. This is the first review that summarizes all evidence of ferroptosis's role in DEX's protective effects against vital organ injuries. Our work aims to provide new insights into organ therapy with DEX and accelerate its translation from the laboratory to clinical settings.
Collapse
Affiliation(s)
- Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People's Hospital, Zigong, China
| | - Xing Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Suheng Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
4
|
Joshi A, Giorgi FM, Sanna PP. Transcriptional Patterns in Stages of Alzheimer's Disease Are Cell-Type-Specific and Partially Converge with the Effects of Alcohol Use Disorder in Humans. eNeuro 2024; 11:ENEURO.0118-24.2024. [PMID: 39299805 PMCID: PMC11485264 DOI: 10.1523/eneuro.0118-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
Advances in single-cell technologies have led to the discovery and characterization of new brain cell types, which in turn lead to a better understanding of the pathogenesis of Alzheimer's disease (AD). Here, we present a detailed analysis of single-nucleus (sn)RNA-seq data for three stages of AD from middle temporal gyrus and compare it with snRNA-seq data from the prefrontal cortices from individuals with alcohol use disorder (AUD). We observed a significant decrease in both inhibitory and excitatory neurons, in general agreement with previous reports. We observed several cell-type-specific gene expressions and pathway dysregulations that delineate AD stages. Endothelial and vascular leptomeningeal cells showed the greatest degree of gene expression changes. Cell-type-specific evidence of neurodegeneration was seen in multiple neuronal cell types particularly in somatostatin and Layer 5 extratelencephalic neurons, among others. Evidence of inflammatory responses was seen in non-neuronal cells, particularly in intermediate and advanced AD. We observed common perturbations in AD and AUD, particularly in pathways, like transcription, translation, apoptosis, autophagy, calcium signaling, neuroinflammation, and phosphorylation, that imply shared transcriptional pathogenic mechanisms and support the role of excessive alcohol intake in AD progression. Major AUD gene markers form and perturb a network of genes significantly associated with intermediate and advanced AD. Master regulator analysis from AUD gene markers revealed significant correlation with advanced AD of transcription factors that have implications in intellectual disability, neuroinflammation, and other neurodegenerative conditions, further suggesting a shared nexus of transcriptional changes between AD and AUD.
Collapse
Affiliation(s)
- Arpita Joshi
- The Scripps Research Institute, San Diego, California 92117
| | - Federico Manuel Giorgi
- The Scripps Research Institute, San Diego, California 92117
- University of Bologna, Bologna 40136, Italy
| | | |
Collapse
|
5
|
Xu X, Li T, Tang J, Wang D, Zhou Y, Gou H, Li L, Xu Y. CXCR4-mediated neutrophil dynamics in periodontitis. Cell Signal 2024; 120:111212. [PMID: 38719020 DOI: 10.1016/j.cellsig.2024.111212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/26/2024] [Accepted: 05/05/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND AND OBJECTIVE Periodontitis is a common oral disease closely related to immune response and this study is aimed to identify the key immune-related pathogenic genes and analyze the infiltration and function of immune cells in the disease using bioinformatics methods. METHODS Transcriptome datasets and single-cell RNA sequencing (scRNA-seq) datasets were downloaded from the GEO database. We utilized weighted correlation network analysis and least absolute selection and shrinkage operator, protein-protein interaction network construction to screen out key pathogenic genes as well as conducted the cell-type identification by estimating relative subsets of RNA transcripts algorithm to analyze and characterize immune cell types in periodontal tissues. In addition to bioinformatics validations, clinical and cell samples were collected and mouse periodontitis models were constructed to validate the important role of key genes in periodontitis. RESULTS Bioinformatics analysis pointed out the positive correlation between CXCR4 expression and periodontitis, and revealed the increased infiltration of neutrophils in periodontal inflammatory. Similar results were obtained from clinical samples and animal models. In addition, the clustering and functional enrichment results based on CXCR4 expression levels included activation of immune response and cell migration, implying the possible function of CXCR4 on regulating neutrophil dynamics, which might contribute to periodontitis. Subsequent validation experiments confirmed that the increased expression of CXCR4 in neutrophils under periodontitis, where cell migration-related pathways also were activated. CONCLUSION CXCR4 could be the key pathogenic gene of periodontitis and CXCR4/CXCL12 signal axial might contribute to the development of periodontitis by mediating neutrophil dynamics, suggesting that CXCR4 could be a potential target to help identify novel strategies for the clinical diagnosis and treatment of periodontitis.
Collapse
Affiliation(s)
- Xuanwen Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China.; Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China.; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Tiange Li
- School of Stomatology, China Medical University, Shenyang 110122, China
| | - Jingqi Tang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China.; Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China.; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Danlei Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China.; Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China.; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Yi Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China.; Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China.; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Huiqing Gou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China.; Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China.; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Lu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China.; Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China.; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China
| | - Yan Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China.; Department of Periodontology, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China.; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 210029, China..
| |
Collapse
|
6
|
Jiang Y, Bian W, Chen J, Cao X, Dong C, Xiao Y, Xu B, Sun X. miRNA-137-5p improves spatial memory and cognition in Alzheimer's mice by targeting ubiquitin-specific peptidase 30. Animal Model Exp Med 2023; 6:526-536. [PMID: 38111333 PMCID: PMC10757218 DOI: 10.1002/ame2.12368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/22/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a prevalent neurodegenerative disorder causing progressive dementia. Research suggests that microRNAs (miRNAs) could serve as biomarkers and therapeutic targets for AD. Reduced levels of miR-137 have been observed in the brains of AD patients, but its specific role and downstream mechanisms remain unclear. This study sought to examine the therapeutic potential of miR-137-5p agomir in alleviating cognitive dysfunction induced in AD models and explore its potential mechanisms. METHODS This study utilized bioinformatic analysis and a dual-luciferase reporter assay to investigate the relationship between miR-137-5p and ubiquitin-specific peptidase 30 (USP30). In vitro experiments were conducted using SH-SY5Y cells to assess the impact of miR-137-5p on Aβ1-42 neurotoxicity. In vivo experiments on AD mice evaluated the effects of miR-137-5p on cognition, Aβ1-42 deposition, Tau hyperphosphorylation, and neuronal apoptosis, as well as its influence on USP30 levels. RESULTS It was discovered that miR-137-5p mimics efficiently counteract Aβ1-42 neurotoxicity in SH-SY5Y cells, a protective effect that is negated by USP30 overexpression. In vivo experiments demonstrated that miR-137-5p enhances the cognition and mobility of AD mice, significantly reducing Aβ1-42 deposition, Tau hyperphosphorylation, and neuronal apoptosis within the hippocampus and cortex regions. Mechanistically, miR-137-5p significantly suppresses USP30 levels in mice, though USP30 overexpression partially buffers against miR-137-5p-induced AD symptom improvement. CONCLUSION Our study proposes that miR-137-5p, by instigating the downregulation of USP30, has the potential to act as a novel and promising therapeutic target for AD.
Collapse
Affiliation(s)
- Yang Jiang
- Department of NeurologyThe First People's Hospital of ShenYangShenyangP.R. China
- Department of NeurologyThe Fourth Affiliated Hospital of China Medical UniversityShenyangP.R. China
| | - Wei Bian
- Department of NeurologyThe First People's Hospital of ShenYangShenyangP.R. China
| | - Jing Chen
- Department of Neurology and NeuroscienceShenyang Tenth People's Hospital, Shenyang Chest HospitalShenyangP.R. China
| | - Xiaopan Cao
- Department of NeurologyThe First People's Hospital of ShenYangShenyangP.R. China
| | - ChunYao Dong
- Department of NeurologyThe First People's Hospital of ShenYangShenyangP.R. China
| | - Ying Xiao
- Department of NeurologyThe First People's Hospital of ShenYangShenyangP.R. China
| | - Bing Xu
- Department of Neurology and NeuroscienceShenyang Tenth People's Hospital, Shenyang Chest HospitalShenyangP.R. China
| | - XiaoHong Sun
- Department of NeurologyThe Fourth Affiliated Hospital of China Medical UniversityShenyangP.R. China
- Science Experiment CenterChina Medical UniversityShenyangChina
| |
Collapse
|
7
|
Ding XS, Gao L, Han Z, Eleuteri S, Shi W, Shen Y, Song ZY, Su M, Yang Q, Qu Y, Simon DK, Wang XL, Wang B. Ferroptosis in Parkinson's disease: Molecular mechanisms and therapeutic potential. Ageing Res Rev 2023; 91:102077. [PMID: 37742785 DOI: 10.1016/j.arr.2023.102077] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/26/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
Parkinson's Disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra (SN), leading to motor and non-motor symptoms. While the exact mechanisms remain complex and multifaceted, several molecular pathways have been implicated in PD pathology, including accumulation of misfolded proteins, impaired mitochondrial function, oxidative stress, inflammation, elevated iron levels, etc. Overall, PD's molecular mechanisms involve a complex interplay between genetic, environmental, and cellular factors that disrupt cellular homeostasis, and ultimately lead to the degeneration of dopaminergic neurons. Recently, emerging evidence highlights ferroptosis, an iron-dependent non-apoptotic cell death process, as a pivotal player in the advancement of PD. Notably, oligomeric α-synuclein (α-syn) generates reactive oxygen species (ROS) and lipid peroxides within cellular membranes, potentially triggering ferroptosis. The loss of dopamine, a hallmark of PD, could predispose neurons to ferroptotic vulnerability. This unique form of cell demise unveils fresh insights into PD pathogenesis, necessitating an exploration of the molecular intricacies connecting ferroptosis and PD progression. In this review, the molecular and regulatory mechanisms of ferroptosis and their connection with the pathological processes of PD have been systematically summarized. Furthermore, the features of ferroptosis in PD animal models and clinical trials targeting ferroptosis as a therapeutic approach in PD patients' management are scrutinized.
Collapse
Affiliation(s)
- Xv-Shen Ding
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China; Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Li Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Zheng Han
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Simona Eleuteri
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle 628H, Boston, MA 02215, USA
| | - Wei Shi
- Department of Neurosurgery, PLA 960th hospital, JiNan, Shandong Province, 250031, China
| | - Yun Shen
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Zi-Yao Song
- Basic Medicine School, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Mingming Su
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Qian Yang
- Department of Experimental Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China.
| | - David K Simon
- Department of Neurology, Beth Israel Deaconess Medical Center, 3 Blackfan Circle 628H, Boston, MA 02215, USA.
| | - Xue-Lian Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China.
| | - Bao Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, 710038, China.
| |
Collapse
|