1
|
Li Z, Liang S, Ke L, Wang M, Gao K, Li D, Xu Z, Li N, Zhang P, Cheng W. Cell life-or-death events in osteoporosis: All roads lead to mitochondrial dynamics. Pharmacol Res 2024; 208:107383. [PMID: 39214266 DOI: 10.1016/j.phrs.2024.107383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria exhibit heterogeneous shapes and networks within and among cell types and tissues, also in normal or osteoporotic bone tissues with complex cell types. This dynamic characteristic is determined by the high plasticity provided by mitochondrial dynamics and is stemmed from responding to the survival and functional requirements of various bone cells in a specific microenvironments. In contrast, mitochondrial dysfunction, induced by dysregulation of mitochondrial dynamics, may act as a trigger of cell death signals, including common apoptosis and other forms of programmed cell death (PCD). These PCD processes consisting of tightly structured cascade gene expression events, can further influence the bone remodeling by facilitating the death of various bone cells. Mitochondrial dynamics, therefore, drive the bone cells to stand at the crossroads of life and death by integrating external signals and altering metabolism, shape, and signal-response properties of mitochondria. This implies that targeting mitochondrial dynamics displays significant potential in treatment of osteoporosis. Considerable effort has been made in osteoporosis to emphasize the parallel roles of mitochondria in regulating energy metabolism, calcium signal transduction, oxidative stress, inflammation, and cell death. However, the emerging field of mitochondrial dynamics-related PCD is not well understood. Herein, to bridge the gap, we outline the latest knowledge on mitochondrial dynamics regulating bone cell life or death during normal bone remodeling and osteoporosis.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050011, China
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000, China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300, China.
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
2
|
Wang CL, Li P, Liu B, Ma YQ, Feng JX, Xu YN, Liu L, Li ZH. Decrypting the skeletal toxicity of vertebrates caused by environmental pollutants from an evolutionary perspective: From fish to mammals. ENVIRONMENTAL RESEARCH 2024; 255:119173. [PMID: 38763280 DOI: 10.1016/j.envres.2024.119173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/09/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
The rapid development of modern society has led to an increasing severity in the generation of new pollutants and the significant emission of old pollutants, exerting considerable pressure on the ecological environment and posing a serious threat to both biological survival and human health. The skeletal system, as a vital supportive structure and functional unit in organisms, is pivotal in maintaining body shape, safeguarding internal organs, storing minerals, and facilitating blood cell production. Although previous studies have uncovered the toxic effects of pollutants on vertebrate skeletal systems, there is a lack of comprehensive literature reviews in this field. Hence, this paper systematically summarizes the toxic effects and mechanisms of environmental pollutants on the skeletons of vertebrates based on the evolutionary context from fish to mammals. Our findings reveal that current research mainly focuses on fish and mammals, and the identified impact mechanisms mainly involve the regulation of bone signaling pathways, oxidative stress response, endocrine system disorders, and immune system dysfunction. This study aims to provide a comprehensive and systematic understanding of research on skeletal toxicity, while also promoting further research and development in related fields.
Collapse
Affiliation(s)
- Cun-Long Wang
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Ping Li
- Marine College, Shandong University, Weihai, Shandong, 264209, China.
| | - Bin Liu
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Yu-Qing Ma
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Jian-Xue Feng
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Ya-Nan Xu
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Ling Liu
- Marine College, Shandong University, Weihai, Shandong, 264209, China
| | - Zhi-Hua Li
- Marine College, Shandong University, Weihai, Shandong, 264209, China.
| |
Collapse
|
3
|
Yan M, Zhang Y, Niu W, Liu K, Xue L, Zhou K. Reactive oxygen species-mediated endoplasmic reticulum stress contributes to osteocyte death induced by orthodontic compressive force. Microsc Res Tech 2023; 86:1529-1541. [PMID: 37382312 DOI: 10.1002/jemt.24382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 05/22/2023] [Accepted: 06/18/2023] [Indexed: 06/30/2023]
Abstract
During orthodontic tooth movement (OTM), osteocytes, the most mechanosensitive cells in alveolar bone, suffer the heavy orthodontic force and initiate alveolar bone resorption on the compression side. However, the inherent mechanisms of compressive force-induced osteocyte death are not fully understood. In this study, we established an OTM model on Sprague-Dawley rats by inserting coil springs to investigate osteocyte damage on the compression side of alveolar bone. We then applied compressive force on the MLO-Y4 osteocyte-like cell line in vitro to explore whether the reactive oxygen species (ROS)-mediated endoplasmic reticulum stress (ERS) pathway is involved in compressive force-induced osteocyte death. We found that the orthodontic force caused apparent alveolar bone loss, osteocyte death, and elevated serum sclerostin and receptor activator of NF-κB ligand (RANKL) levels in rats. In vitro, compressive force inhibited cell viability but increased the LDH leakage and loss of mitochondrial membrane potential in MLO-Y4 cells. Simultaneously, it activated protein kinase RNA-like endoplasmic reticulum kinase (PERK), eukaryotic translation initiation factor 2 (eIF2α), and their downstream pro-apoptotic ERS signaling proteins and caused significant osteocyte apoptosis, which can be blocked by ERS inhibitor salubrinal. Moreover, the compressive force elevated intracellular ROS levels, while the ROS scavenger N-acetyl-L-cysteine (NAC) alleviated ERS and apoptosis in loaded osteocytes. These results suggest that the orthodontic compressive force induced osteocyte apoptosis via the ROS-mediated ERS pathway. This study first proposes the ERS pathway as a new potential pathway for regulating the rate of OTM based on osteocyte death. RESEARCH HIGHLIGHTS: The orthodontic force increases osteocyte death in rat alveolar bone. The compressive force causes osteocyte apoptosis by the endoplasmic reticulum stress (ERS) pathway in vitro. The ROS scavenger NAC blocked compressive force-induced ERS and osteocyte apoptosis.
Collapse
Affiliation(s)
- Ming Yan
- School of Automation, Hangzhou Dianzi University, Hangzhou, People's Republic of China
| | - Yun Zhang
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Wanting Niu
- Department of STEM, Regis College, Weston, Massachusetts, USA
| | - Kezhou Liu
- School of Automation, Hangzhou Dianzi University, Hangzhou, People's Republic of China
| | - Lingyun Xue
- School of Automation, Hangzhou Dianzi University, Hangzhou, People's Republic of China
| | - Kexin Zhou
- West China School of Medicine, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
4
|
Meng Y, Li C, Liang Y, Jiang Y, Zhang H, Ouyang J, Zhang W, Deng R, Tan Q, Yu X, Luo Z. Umbilical Cord Mesenchymal-Stem-Cell-Derived Exosomes Exhibit Anti-Oxidant and Antiviral Effects as Cell-Free Therapies. Viruses 2023; 15:2094. [PMID: 37896871 PMCID: PMC10612094 DOI: 10.3390/v15102094] [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: 08/21/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
The oxidative stress induced by the accumulation of reactive oxygen species (ROS) can lead to cell aging and death. Equally, the skeletal muscle usually hosts enteroviral persistent infection in inflammatory muscle diseases. As excellent bioactive products, the exosomes derived from umbilical cord mesenchymal stem cells (ucMSCs) have been proven to be safe and have low immunogenicity with a potential cell-free therapeutic function. Here, exosomes derived from ucMSCs (ucMSC-EXO) were extracted and characterized. In a model of oxidative damage to skin fibroblasts (HSFs) under exposure to H2O2, ucMSC-EXO had an observable repairing effect for the HSFs suffering from oxidative damage. Furthermore, ucMSC-EXO inhibited mitogen-activated protein kinases (MAPK), c-Jun N-terminal kinase (JNK), and nuclear factor kappa-B (NF-κB) signaling pathways, thereby promoting p21 protein expression while decreasing lamin B1 protein expression, and finally alleviated oxidative stress-induced cell damage and aging. In a model of rhabdomyosarcoma (RD) cells being infected by enterovirus 71 (EV71) and coxsackievirus B3 (CVB3), the ucMSC-EXO enhanced the expression of interferon-stimulated gene 15 (ISG15) and ISG56 to inhibit enteroviral replication, whereafter reducing the virus-induced proinflammatory factor production. This study provides a promising therapeutic strategy for ucMSC-EXO in anti-oxidative stress and antiviral effects, which provides insight into extending the function of ucMSC-EXO in cell-free therapy.
Collapse
Affiliation(s)
- Yi Meng
- Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China; (Y.M.); (C.L.); (Y.L.)
| | - Chengcheng Li
- Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China; (Y.M.); (C.L.); (Y.L.)
| | - Yicong Liang
- Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China; (Y.M.); (C.L.); (Y.L.)
| | - Yu Jiang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China; (Y.J.); (H.Z.)
| | - Haonan Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China; (Y.J.); (H.Z.)
| | - Jianhua Ouyang
- Foshan Institute of Medical Microbiology, Foshan 528315, China; (J.O.); (R.D.)
| | - Wen Zhang
- Guangdong Longfan Biological Science and Technology Company, Foshan 528315, China; (W.Z.); (Q.T.)
| | - Rumei Deng
- Foshan Institute of Medical Microbiology, Foshan 528315, China; (J.O.); (R.D.)
| | - Qiuping Tan
- Guangdong Longfan Biological Science and Technology Company, Foshan 528315, China; (W.Z.); (Q.T.)
| | - Xiaolan Yu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan 430062, China; (Y.J.); (H.Z.)
| | - Zhen Luo
- Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China; (Y.M.); (C.L.); (Y.L.)
- Foshan Institute of Medical Microbiology, Foshan 528315, China; (J.O.); (R.D.)
- Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
| |
Collapse
|
5
|
Yang P, Xu B, Zhu R, Zhang T, Wang Z, Lin Q, Yan M, Yu Z, Mao H, Zhang Y. ROS-mediated mitophagy and necroptosis regulate osteocytes death caused by TCP particles in MLO-Y4 cells. Toxicology 2023; 496:153627. [PMID: 37678662 DOI: 10.1016/j.tox.2023.153627] [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: 05/21/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/09/2023]
Abstract
Our previous data have revealed TCP particles caused cell death of osteocytes, comprising over 95 % of all bone cells, which contribute to periprosthetic osteolysis, joint loosening and implant failure, but its mechanisms are not fully understood. Here, we reported that TCP particles inhibited cell viability of osteocytes MLO-Y4, and caused cell death. TCP particles caused mitochondrial impairment and increased expressions of LC-3 II, Parkin and PINK 1, accompanied by the elevation of autophagy flux and intracellular acidic components, the accumulation of LC-3II, PINK1 and Parkin in damaged mitochondria, and p62 reduction. The increased LC-3II expression and cell death extent were significantly enhanced by the autophagy inhibitor Baf A1, compared with Baf A1 (or TCP particles) alone, indicating that TCP particles increase autophagic flux and lead to cell even death of MLO-Y4 cells, closely associated with mitophagy. Furthermore, TCP particles induced propidium iodide (PI) uptake and the phosphorylation of RIP1, RIP3 and MLKL, thereby increasing necroptosis in MLO-Y4 cells. The pro-necroptotic effect was alleviated by the RIP1 inhibitor Nec-1 or the MLKL inhibitor NSA. Additionally, TCP particles promoted the production of intracellular reactive oxygen species (ROS) and mitochondrial ROS (mtROS), and increased TXNIP expression, but decreased protein levels of TRX1, Nrf2, HO-1 and NQO1, leading to oxidative stress. The ROS scavenger NAC remarkably reversed mitophagy and necroptosis caused by TCP particles, suggesting that ROS is responsible for mitophagy and necroptosis. Collectively, ROS-mediated mitophagy and necroptosis regulate osteocytes death caused by TCP particles in MLO-Y4 cells, which enhances osteoclastogenesis and periprosthetic osteolysis.
Collapse
Affiliation(s)
- Pei Yang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing 312000, PR China
| | - Bingbing Xu
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing 312000, PR China
| | - Ruirong Zhu
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing 312000, PR China
| | - Tao Zhang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing 312000, PR China
| | - Zihan Wang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing 312000, PR China
| | - Qiao Lin
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing 312000, PR China
| | - Ming Yan
- School of Automation, Hangzhou Dianzi University, Xiasha Higher Education Zone, 1158 2nd Avenue, Hangzhou 310018, PR China
| | - Zhangsen Yu
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing 312000, PR China
| | - Hongjiao Mao
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing 312000, PR China
| | - Yun Zhang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing 312000, PR China.
| |
Collapse
|
6
|
Shan W, Niu W, Lin Q, Shen Y, Shen F, Lou K, Zhang Y. Bisphenol S exposure promotes cell apoptosis and mitophagy in murine osteocytes by regulating mtROS signaling. Microsc Res Tech 2023; 86:481-493. [PMID: 36625337 DOI: 10.1002/jemt.24289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/29/2022] [Accepted: 12/31/2022] [Indexed: 01/11/2023]
Abstract
Bisphenol S (BPS), a safer alternative to bisphenol A, is commonly used as a plasticizer to manufacture various food-packaging materials. The accumulated BPS inhibits osteoblastic bone formation and promotes osteoclastogenesis, thereby accelerating remarkable bone destruction, but it is unclear whether BPS affects osteocytes, comprising over 95% of all bone cells. This study aimed to investigate the biological effect of BPS on osteocytes in vitro, as well as the detailed mechanism. Results showed that BPS (200, 400 μmol/L) exposure caused dose-dependently cell death of osteocytes MLO-Y4, and increased cell apoptosis. BPS induced loss of mitochondrial membrane potential (MMP) and mitochondria impairment. Furthermore, BPS upregulated expressions of mitophagy-related proteins including microtubule-associated protein light chain 3 (LC-3) II and PTEN-induced putative kinase (PINK) 1, accompanied by elevation of autophagy flux and the accumulation of acidic vacuoles; whereas p62 level was downregulated after BPS treatment. Additionally, BPS triggered the production of intracellular reactive oxygen species (ROS) and mitochondrial ROS (mtROS), while it decreased expression levels of nuclear factor E2-related factor 2 (Nrf2) and quinone oxidoreductase 1 (NQO1). The specific mtROS scavenger MitoTEMPO reversed cell apoptosis and mitophagy, suggesting that mtROS contributes to BPS exposure-induced apoptosis and mitophagy in MLO-Y4 cells. Our data first provide novel evidence that apoptosis and mitophagy as cellular mechanisms for the toxic effect of BPS on osteocytes, thereby helping our understanding of the potential role of osteocytes in the adverse effect of BPS and its analogs on bone growth, and supporting strategies targeting bone destruction caused by BPS.
Collapse
Affiliation(s)
- Weiyan Shan
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Wanting Niu
- Department of Orthopedics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Qiao Lin
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Yuchen Shen
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Fangmin Shen
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Kai Lou
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| | - Yun Zhang
- College of Medicine, Shaoxing University, Shaoxing, People's Republic of China
| |
Collapse
|
7
|
Zhang Y, Dong F, Wang Z, Xu B, Zhang T, Wang Q, Lin Q. Fluoride Exposure Provokes Mitochondria-Mediated Apoptosis and Increases Mitophagy in Osteocytes via Increasing ROS Production. Biol Trace Elem Res 2022:10.1007/s12011-022-03450-w. [PMID: 36255553 DOI: 10.1007/s12011-022-03450-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/13/2022] [Indexed: 11/28/2022]
Abstract
Fluoride is a persistent environmental pollutant, and its excessive intake causes skeletal and dental fluorosis. However, few studies focused on the effects of fluoride on osteocytes, making up over 95% of all bone cells. This study aimed to investigate the effect of fluoride on osteocytes in vitro, as well as explore the underlying mechanisms. CCK-8, LDH assay, fluorescent probes, flow cytometry, and western blotting were performed to examine cell viability, apoptosis, mitochondria changes, reactive oxygen species (ROS) and mitochondrial ROS (mtROS), and protein expressions. Results showed that sodium fluoride (NaF) exposure (4, 8 mmol/L) for 24 h inhibited the cell viability of osteocytes MLO-Y4 and promoted G0/G1 phase arrest and increased cell apoptosis. NaF treatment remarkably caused mitochondria damage, loss of MMP, ATP decrease, Cyto c release, and Bax/Bcl-2 ratio increase and elevated the activity of caspase-9 and caspase-3. Furthermore, NaF significantly upregulated the expressions of LC-3II, PINK1, and Parkin and increased autophagy flux and the accumulation of acidic vacuoles, while the p62 level was downregulated. In addition, NaF exposure triggered the production of intracellular ROS and mtROS and increased malondialdehyde (MDA); but superoxide dismutase (SOD) activity and glutathione (GSH) content were decreased. The scavenger N-acetyl-L-cysteine (NAC) significantly reversed NaF-induced apoptosis and mitophagy, suggesting that ROS is responsible for the mitochondrial-mediated apoptosis and mitophagy induced by NaF exposure. These findings provide in vitro evidence that apoptosis and mitophagy are cellular mechanisms for the toxic effect of fluoride on osteocytes, thereby suggesting the potential role of osteocytes in skeletal and dental fluorosis.
Collapse
Affiliation(s)
- Yun Zhang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China.
| | - Fanhe Dong
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| | - Zihan Wang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| | - Bingbing Xu
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| | - Tao Zhang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| | - Qiqi Wang
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| | - Qiao Lin
- College of Medicine, Shaoxing University, Huancheng West Road 508, Shaoxing, 312000, People's Republic of China
| |
Collapse
|