1
|
Guo L, Zhou Y, Ding J, Xiong J, Zhu L, Amuti S, Zhang C, Du Z, Zhang X, Dong B, Alifu N. A near-infrared triggered multi-functional indocyanine green nanocomposite with NO gas release function inducing improved photothermal therapy. J Colloid Interface Sci 2025; 679:307-323. [PMID: 39454262 DOI: 10.1016/j.jcis.2024.10.071] [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: 06/24/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024]
Abstract
The integration of photothermal and near-infrared (NIR) imaging capabilities of indocyanine green (ICG) small molecules has attracted considerable attention in tumor diagnosis and treatment. However, the abnormal upregulation of cellular heat shock proteins (HSPs) induced by photothermal therapy (PTT) enhances cellular heat resistance, thereby severely affecting the efficacy of PTT. In this study, to address the impact of HSPs on the efficacy of PTT while obtaining high-quality NIR fluorescence imaging in the NIR region, we designed a targeted peptide@ICG nanofluorescent probe encapsulated in liposomes. The introduced cRGD targeting peptide not only possesses tumor-targeting capabilities but also features LA as the last amino acid in the targeting peptide, which can generate nitric oxide (NO) under reactive oxygen species (ROS) triggering. It can happen under 808 nm single-light source NIR light, and the guanidine group in the peptide decomposes and combines with singlet oxygen molecules to generate NO gas molecules, thereby exerting an elevated photothermal effect by inhibiting the expression of HSP70. In addition, the nanoprobes enable deep imaging and treatment of glioma in situ and can be combined with a laser speckle contrast imaging (LSCI) system for multimodal imaging. This composite probe demonstrates synergistic tumor therapeutic effects of photodynamic therapy (PDT), PTT, and gas therapy, offering a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Le Guo
- Institute of Public Health, Xinjiang Medical University, Urumqi 830011, China
| | - Yi Zhou
- Institute of Public Health, Xinjiang Medical University, Urumqi 830011, China; Department of Biomedical Engineering, Zhongshan Medical College, ZhongShan University, Guangzhou 510000, China
| | - Jiayi Ding
- Institute of Public Health, Xinjiang Medical University, Urumqi 830011, China
| | - Jiabao Xiong
- Second Clinical Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Lijun Zhu
- Second Clinical Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Siyiti Amuti
- College of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830011, China
| | - Chi Zhang
- Institute of Public Health, Xinjiang Medical University, Urumqi 830011, China
| | - Zhong Du
- Second Clinical Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Xueliang Zhang
- State Key Laboratory of Pathogenesis Prevention and Treatment of High Incidence Diseases in Central Asia, School of Medical Engineering and Technology Xinjiang Medical University, Urumqi 830011, China.
| | - Biao Dong
- State Key Laboratory of Pathogenesis Prevention and Treatment of High Incidence Diseases in Central Asia, School of Medical Engineering and Technology Xinjiang Medical University, Urumqi 830011, China; State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130012, China.
| | - Nuernisha Alifu
- Institute of Public Health, Xinjiang Medical University, Urumqi 830011, China; Second Clinical Medical College, Xinjiang Medical University, Urumqi 830011, China; State Key Laboratory of Pathogenesis Prevention and Treatment of High Incidence Diseases in Central Asia, School of Medical Engineering and Technology Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
2
|
Tang L, Yang X, He L, Zhu C, Chen Q. Preclinical advance in nanoliposome-mediated photothermal therapy in liver cancer. Lipids Health Dis 2025; 24:31. [PMID: 39891269 PMCID: PMC11783920 DOI: 10.1186/s12944-024-02429-x] [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: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 02/03/2025] Open
Abstract
Liver cancer is a highly lethal malignant tumor with a high incidence worldwide. Therefore, its treatment has long been a focus of medical research. Although traditional treatment methods such as surgery, radiotherapy, and chemotherapy have increased the survival rate of patients, their efficacy remains unsatisfactory owing to the nonspecific distribution of drugs, high toxicity, and drug resistance of tumor tissues. In recent years, the application of nanotechnology in the medical field has opened a new avenue for the treatment of liver cancer. Among these treatment methods, photothermal therapy (PTT) based on nanoliposomes has attracted wide attention owing to its unique targeting and high efficiency. This article reviews the latest preclinical research progress of nanoliposome-based PTT for liver cancer and its metastasis, discusses the preclinical challenges in this field, and proposes directions for improvement, with the aim of improving the effectiveness of liver cancer treatment.
Collapse
Affiliation(s)
- Lixuan Tang
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xiao Yang
- The department of oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Liwen He
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chaogeng Zhu
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Qingshan Chen
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
3
|
Yang X, Zhang X, Lin P, Liu Z, Deng S, Liang S, Zhu X, Qiao Q, Chen Q. Effect of dexmedetomidine on somatosensory- and motor-evoked potentials in patients receiving craniotomy under propofol-sevoflurane combined anesthesia. Front Surg 2024; 11:1386049. [PMID: 39045089 PMCID: PMC11263188 DOI: 10.3389/fsurg.2024.1386049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction Dexmedetomidine is often used as an adjunct to total intravenous anesthesia (TIVA) for procedures requiring intraoperative neurophysiologic monitoring (IONM). However, it has been reported that dexmedetomidine might mask the warning of a neurological deficit on intraoperative monitoring. Methods We reviewed the intraoperative neurophysiological monitoring data of 47 patients who underwent surgery and IONM from March 2019 to March 2021 at the Department of Neurosurgery, Renmin Hospital of Wuhan University. Pre- and postoperative motor function scores were recorded and analyzed. Dexmedetomidine was administered intravenously at 0.5 μg/kg/h 40 min after anesthesia and discontinued after 1 h in the dexmedetomidine group. Results We found that the amplitude of transcranial motor-evoked potentials (Tce-MEPs) was significantly lower in the dexmedetomidine group than in the negative control group (P < 0.0001). There was no statistically significant difference in the somatosensory-evoked potentials (SSEPs) amplitude or the Tce-MEPs or SSEPs latency. There was no significant decrease in postoperative motor function in the dexmedetomidine group compared with the preoperative group, suggesting that there is no evidence that dexmedetomidine affects patient prognosis. In addition, we noticed a synchronized bilateral decrease in the Tce-MEPs amplitude in the dexmedetomidine group and a mostly unilateral decrease on the side of the brain injury in the positive control group (P = 0.001). Discussion Although dexmedetomidine does not affect the prognosis of patients undergoing craniotomy, the potential risks and benefits of applying it as an adjunctive medication during craniotomy should be carefully evaluated. When dexmedetomidine is administered, Tce-MEPs should be monitored. When a decrease in the Tce-MEPs amplitude is detected, the cause of the decrease in the MEPs amplitude can be indirectly determined by whether the decrease is bilateral.
Collapse
Affiliation(s)
- Xue Yang
- Department of Neurosurgery, Wuhan University Renmin Hospital, Wuhan, China
| | - Xinyi Zhang
- Department of Neurosurgery, Wuhan University Renmin Hospital, Wuhan, China
| | - Puxuan Lin
- Department of Neurosurgery, Wuhan University Renmin Hospital, Wuhan, China
| | - Zeheng Liu
- Department of Neurosurgery, Wuhan University Renmin Hospital, Wuhan, China
| | - Shuhang Deng
- Department of Neurosurgery, Wuhan University Renmin Hospital, Wuhan, China
| | - Shanwen Liang
- Department of Neurosurgery, Wuhan University Renmin Hospital, Wuhan, China
| | - Xinyi Zhu
- Department of Neurosurgery, Wuhan University Renmin Hospital, Wuhan, China
| | - Qianqian Qiao
- Department of Anesthesiology, Wuhan University Renmin Hospital, Wuhan, China
| | - Qianxue Chen
- Department of Neurosurgery, Wuhan University Renmin Hospital, Wuhan, China
| |
Collapse
|
4
|
Wei L, Wei Q, Yang X, Zhou P. CMTM6 knockdown prevents glioma progression by inactivating the mTOR pathway. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:181. [PMID: 35280358 PMCID: PMC8908166 DOI: 10.21037/atm-21-6894] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/27/2022] [Indexed: 12/13/2022]
Abstract
Background Gliomas in the adult brain are complicated and aggressive with a poor prognosis. Gene therapy is a recent alternative glioma treatment. We sought to explore the mechanism of chemokine-like factor (CKLF) MARVEL transmembrane domain-containing 6 (CMTM6) in glioma. Methods The Cancer Genome Atlas database reports that CMTM6 is expressed in tumors and glioma tissue. CMTM6 expression in glioma tissues and cells was detected and its relationship with clinical pathology was analyzed. Short hairpin ribonucleic acid-CMTM6 lentivirus was transfected into U87 and U251 cells to evaluate malignant glioma cells. Using the biological website (https://string-db.org/cgi/input.pl?Sessionid) and reference retrieval, the pathway that interacted with CMTM6 and related to glioma was identified. The level of the mammalian target of rapamycin pathway-related proteins was detected. Functional rescue experiments were performed using the combination of mTOR activator MHY1485 and the knockdown CMTM6. The growth of xenograft tumors was observed and Ki67-positive expression was determined. Results CMTM6 upregulation in gliomas was associated with a poor prognosis. CMTM6 expression was notably higher in gliomas. After the knockdown of CMTM6, the proliferation, invasion, and migration of U87 and U251 cells were inhibited, and the apoptosis rate was increased. Knocking down CMTM6 inactivated the mTOR pathway. The activation of mTOR pathway reversed the inhibitory effects of CMTM6 knockdown on glioma cell behaviors. CMTM6 knockdown reduced tumor volume, body mass, and Ki67-positive expression. Conclusions The knockdown of CMTM6 inhibited the activation of mTOR pathway and prevented the malignant episodes of glioma cells.
Collapse
Affiliation(s)
- Li Wei
- Department of Blood Transfusion, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qianfeng Wei
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaojun Yang
- Department of Blood Transfusion, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Peng Zhou
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
5
|
Zhu J, Xie B, Huang G, Li Y, Liu Z. Sevoflurane represses the progression of glioma by the regulation of circ_0037655/miR-130a-5p/RPN2 axis. Metab Brain Dis 2022; 37:787-799. [PMID: 35032276 DOI: 10.1007/s11011-022-00906-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/06/2022] [Indexed: 10/19/2022]
Abstract
Sevoflurane (SEV) is a common anesthetic to inhibit glioma progression. The previous studies have indicated the molecular mechanisms of SEV function in glioma. The objective of this study was to explore the association of circ_00037655 with SEV in glioma. Cell viability was evaluated by Cell Counting Kit-8 (CCK-8) assay. Cell proliferation was analyzed using Edu assay and colony formation assay. Flow cytometry was applied to determine cell apoptosis. Protein analysis was performed via western blot. Cell migration and invasion were assessed by transwell assay. Circ_0037655, microRNA-130a-5p (miR-130a-5p) and ribophorin II (RPN2) levels were detected using the quantitative real-time polymerase chain reaction (qRT-PCR). Dual-luciferase reporter, RNA immunoprecipitation (RIP) and pull-down assays were used to analyze target interaction. The effect of circ_0037655 on SEV in vivo was researched by xenograft models. SEV reduced cell viability, proliferation, migration and invasion but induced apoptosis of glioma cells. Circ_0037655 expression was inhibited after SEV treatment in glioma cells. The effects of SEV on glioma cell behaviors were attenuated by upregulation of circ_0037655. Circ_0037655 interacted with miR-130a-5p and miR-130a-5p targeted RPN2. Circ_0037655 or miR-130a-5p regulated the anti-tumor function of SEV in glioma by targeting miR-130a-5p or RPN2. Circ_0037655 affected the expression of RPN2 via targeting miR-130a-5p. Circ_0037655 relieved SEV-induced glioma growth inhibition in vivo by mediating miR-130a-5p and RPN2 levels. SEV inhibited the malignant progression of glioma cells partly by regulating the circ_0037655/miR-130a-5p/RPN2 axis.
Collapse
Affiliation(s)
- Jinyou Zhu
- Department of Anesthesiology, Ganzhou People's Hospital, No.16, Meiguan Avenue, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
| | - Bin Xie
- Department of Anesthesiology, Ganzhou People's Hospital, No.16, Meiguan Avenue, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China.
| | - Guiming Huang
- Department of Anesthesiology, Ganzhou People's Hospital, No.16, Meiguan Avenue, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
| | - Yiping Li
- Department of Anesthesiology, Ganzhou People's Hospital, No.16, Meiguan Avenue, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
| | - Zhenhua Liu
- Department of Anesthesiology, Ganzhou People's Hospital, No.16, Meiguan Avenue, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
| |
Collapse
|
6
|
Bao X, Peng Y, Shen J, Yang L. Sevoflurane inhibits progression of glioma via regulating the HMMR antisense RNA 1/microRNA-7/cyclin dependent kinase 4 axis. Bioengineered 2021; 12:7893-7906. [PMID: 34719318 PMCID: PMC8806593 DOI: 10.1080/21655979.2021.1976712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 11/04/2022] Open
Abstract
Sevoflurane (Sev) is a volatile anesthetic that can inhibit tumor malignancy. Glioma is a main brain problem, but the mechanism of Sev in glioma progression is largely unclear. This study aims to explore a potential regulatory network of long noncoding RNA (lncRNA)/microRNA (miRNA)/mRNA associated with the function of Sev in glioma progression. LncRNA HMMR antisense RNA 1 (HMMR-AS1), miR-7 and cyclin-dependent kinase 4 (CDK4) abundances were examined via quantitative reverse transcription polymerase chain reaction and western blot. Cell viability, invasion, and colony formation ability were analyzed via cell counting kit-8, transwell analysis, and colony formation. The target association was analyzed via dual-luciferase reporter analysis and RNA pull-down. The in vivo function of Sev was investigated by xenograft model. HMMR-AS1 abundance was increased in glioma tissues and cells, and reduced via Sev. Sev constrained cell viability, invasion, and colony formation ability via decreasing HMMR-AS1 in glioma cells. miR-7 expression was decreased in glioma, and was targeted via HMMR-AS1. HMMR-AS1 silence restrained cell viability, invasion, and colony formation ability by up-regulating miR-7 in glioma cells. Sev increases miR-7 abundance via decreasing HMMR-AS1. CDK4 was targeted via miR-7, and highly expressed in glioma. miR-7 overexpression inhibited cell viability, invasion, and colony formation ability via reducing CDK4 in glioma cells. CDK4 expression was reduced by Sev via HMMR-AS1/miR-7 axis. Sev suppressed cell growth in glioma by regulating HMMR-AS1. Sev represses glioma cell progression by regulating HMMR-AS1/miR-7/CDK4 axis.
Collapse
Affiliation(s)
- Xi’an Bao
- Department of Anesthesiology, The Affiliated Nanchang Hospital of SUN YAT-SEN University, Nanchang, 330006, China
| | - Yibo Peng
- Department of Anesthesiology, Chinese Medicine Hospital of Yangxin County, Huangshi, China
| | - Jun Shen
- Department of Anesthesiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
| | - Longqiu Yang
- Department of Anesthesiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, China
- Medical College, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Zhao Q, Yan J, Li W, Yang Y, You L, Qin C. Molecular Mechanism of Gas Anesthetics on the Invasion, Metastasis, and Chemosensitivity of Osteosarcoma Cells. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:6000385. [PMID: 34777566 PMCID: PMC8580651 DOI: 10.1155/2021/6000385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/04/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Osteosarcoma is one of the most prominent bone cancers which has a predominant occurrence in children and adolescents. This study is focused on determining the effects of treatment of gas anesthetics on invasion, metastasis, and chemosensitivity in the progression of osteosarcoma cells. Material and Methods. The biological effects of the common gas anesthetics-desflurane, isoflurane, and sevoflurane-on osteosarcoma cells were studied and compared. The biological assays were performed for analysis of cell migration and proliferation. RESULTS Isoflurane and sevoflurane have shown significant inhibition in the osteosarcoma cells at clinically relevant concentrations. Desflurane has shown less potent action on cell migration and inhibition. All three gas anesthetics have shown inhibition in cell proliferation. The effective antiproliferative action was at a clinically significant dose. At low millimolar concentrations, cell apoptosis was moderately affected. Drug combination analysis with chemotherapeutic drugs showed relevant inhibition in cell migration. All three agents showed significant augmentation of chemotherapeutic drugs in suppression and inhibition of inducing apoptosis. The antimigration action is likely to affect the PI3K/AKT pathway and IGF-1. CONCLUSION The study demonstrates the proposed mechanisms of gas anesthetics and their differential effects on osteosarcoma cells and their survival, migration, growth, and chemosensitivity.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guizhou, China
| | - Jianyong Yan
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guizhou, China
| | - Wen Li
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guizhou, China
| | - Ye Yang
- Department of Anesthesiology, Guizhou Provincial Orthopaedic Hospital, Guizhou, China
| | - Lu You
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guizhou, China
| | - Chenguang Qin
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guizhou, China
| |
Collapse
|
8
|
Abstract
Gliomas are common brain mass with a high mortality rate. Patients with gliomas have a severely bad outcome, with an average survive duration less 15 months because of high recurrent rate and being resistant to radio-therapy and chemistry drugs therapy. Hyperbaric oxygen is extensively taken as an adjuvant treatment for various disease conditions. To know the characteristics of hyperbaric oxygen as a remedy for gliomas, we find that, in general, hyperbaric oxygen shows an obviously positive effect on the treatment of gliomas, and it can also relieve the complications caused by postoperative radiotherapy and chemotherapy of gliomas. Whereas, several researches have shown that hyperbaric oxygen promotes glioma progression.
Collapse
Affiliation(s)
- Wen-Jie Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jia-Sheng Ding
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiang Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
9
|
Han W, Shi J, Cao J, Dong B, Guan W. Emerging Roles and Therapeutic Interventions of Aerobic Glycolysis in Glioma. Onco Targets Ther 2020; 13:6937-6955. [PMID: 32764985 PMCID: PMC7371605 DOI: 10.2147/ott.s260376] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/26/2020] [Indexed: 12/20/2022] Open
Abstract
Glioma is the most common type of intracranial malignant tumor, with a great recurrence rate due to its infiltrative growth, treatment resistance, intra- and intertumoral genetic heterogeneity. Recently, accumulating studies have illustrated that activated aerobic glycolysis participated in various cellular and clinical activities of glioma, thus influencing the efficacy of radiotherapy and chemotherapy. However, the glycolytic process is too complicated and ambiguous to serve as a novel therapy for glioma. In this review, we generalized the implication of key enzymes, glucose transporters (GLUTs), signalings and transcription factors in the glycolytic process of glioma. In addition, we summarized therapeutic interventions via the above aspects and discussed promising clinical applications for glioma.
Collapse
Affiliation(s)
- Wei Han
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Jia Shi
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Jiachao Cao
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Bo Dong
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| | - Wei Guan
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, People’s Republic of China
| |
Collapse
|