1
|
Shilnikova K, Kang KA, Piao MJ, Herath HMUL, Fernando PDSM, Boo HJ, Yoon SP, Hyun JW. Shikonin protects skin cells against oxidative stress and cellular dysfunction induced by fine particulate matter. Cell Biol Int 2024; 48:1836-1848. [PMID: 39169545 DOI: 10.1002/cbin.12233] [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: 11/22/2023] [Revised: 07/02/2024] [Accepted: 08/11/2024] [Indexed: 08/23/2024]
Abstract
Shikonin, an herbal naphthoquinone, demonstrates a broad spectrum of pharmacological properties. Owing to increasingly adverse environmental conditions, human skin is vulnerable to harmful influences from dust particles. This study explored the antioxidant capabilities of shikonin and its ability to protect human keratinocytes from oxidative stress induced by fine particulate matter (PM2.5). We found that shikonin at a concentration of 3 µM was nontoxic to human keratinocytes and effectively scavenged reactive oxygen species (ROS) while increasing the production of reduced glutathione (GSH). Furthermore, shikonin enhanced GSH level by upregulating glutamate-cysteine ligase catalytic subunit and glutathione synthetase mediated by nuclear factor-erythroid 2-related factor. Shikonin reduced ROS levels induced by PM2.5, leading to recovering PM2.5-impaired cellular biomolecules and cell viability. Shikonin restored the GSH level in PM2.5-exposed keratinocytes via enhancing the expression of GSH-synthesizing enzymes. Notably, buthionine sulphoximine, an inhibitor of GSH synthesis, diminished effect of shikonin against PM2.5-induced cell damage, confirming the role of GSH in shikonin-induced cytoprotection. Collectively, these findings indicated that shikonin could provide substantial cytoprotection against the adverse effects of PM2.5 through direct ROS scavenging and modulation of cellular antioxidant system.
Collapse
Affiliation(s)
- Kristina Shilnikova
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
| | - Kyoung Ah Kang
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Mei Jing Piao
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Herath Mudiyanselage Udari Lakmini Herath
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Pincha Devage Sameera Madushan Fernando
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Hye-Jin Boo
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Sang Pil Yoon
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| |
Collapse
|
2
|
Fang J, Lou S, Zhou X, Lou D, Zhou L, Bian R. Dihydromyricetin reverses capecitabine-induced peripheral myelin dysfunction through modulation of oxidative stress. Clin Exp Pharmacol Physiol 2024; 51:e13833. [PMID: 38302079 DOI: 10.1111/1440-1681.13833] [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: 06/09/2023] [Revised: 09/21/2023] [Accepted: 10/22/2023] [Indexed: 02/03/2024]
Abstract
Previous clinical reports have shown that capecitabine, an oral prodrug of 5-fluorouracil (5-Fu), can induce peripheral neuropathy, resulting in numbness, paresthesia and hypoesthesia. However, the mechanism through which capecitabine causes peripheral nerve injury remains unclear. Here, we demonstrate that systemic administration of capecitabine leads to myelin abnormalities in the peripheral nerves of mice, which are possibly attributed to the death of Schwann cells, the myelinating cells in the peripheral nervous system. Furthermore, our results show that 5-Fu induces significant oxidative stress in Schwann cells by inhibiting the expression of the anti-oxidative protein DJ-1, leading to a decrease in Schwann cell markers. We found that the anti-oxidant dihydromyricetin (DMY) reverses 5-Fu-induced Schwann cell death and oxidative stress and alleviates capecitabine-induced myelin abnormalities. Taken together, our data indicate that capecitabine induces peripheral myelin dysfunction by regulating DJ-1-mediated oxidative stress in Schwann cells and reveal DMY as a potential therapeutic strategy for capecitabine-induced peripheral neuropathy.
Collapse
Affiliation(s)
- Jie Fang
- Emergency Department, Zhoushan Hospital, Zhoushan, China
| | - Shuyi Lou
- National Computer Network Emergency Response Technical Team/Coordination Center of China, Beijing, China
| | | | - Dayong Lou
- Zhuji People's Hospital of Zhejiang Province, Shaoxin, China
| | - Liqin Zhou
- Zhuji People's Hospital of Zhejiang Province, Shaoxin, China
| | - Rong Bian
- Zhuji People's Hospital of Zhejiang Province, Shaoxin, China
| |
Collapse
|
3
|
Tuo H, Li W, Zhao W, Zhao J, Li D, Jin L. Shikonin alleviates doxorubicin-induced cardiotoxicity via Mst1/Nrf2 pathway in mice. Sci Rep 2024; 14:924. [PMID: 38195835 PMCID: PMC10776756 DOI: 10.1038/s41598-024-51675-7] [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: 09/03/2023] [Accepted: 01/08/2024] [Indexed: 01/11/2024] Open
Abstract
Doxorubicin (DOX) is a popular and potent anticancer drug, but its cardiotoxicity limits its clinical application. Shikonin has a wide range of biological functions, including antioxidant and anti-inflammatory effects. The aim of this study was to investigate the effects of shikonin on DOX-induced cardiac injury and to identify the underlying mechanisms. Mice receiving shikonin showed reduced cardiac injury response and enhanced cardiac function after DOX administration. Shikonin significantly attenuated DOX-induced oxidative damage, inflammation accumulation and cardiomyocyte apoptosis. Shikonin protects against DOX-induced cardiac injury by inhibiting Mammalian sterile 20-like kinase 1 (Mst1) and oxidative stress and activating the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. In conclusion, shikonin alleviates DOX-induced cardiotoxicity by inhibiting Mst1 and activating Nrf2. Shikonin may be used to treat DOX-induced cardiac injury.
Collapse
Affiliation(s)
- Hu Tuo
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenjing Li
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Zhao
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Juan Zhao
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Danni Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lin Jin
- Department of Orthopedics, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| |
Collapse
|
4
|
He D, Tang H, Yang X, Liu X, Zhang Y, Shi J. Elaboration and validation of a prognostic signature associated with disulfidoptosis in lung adenocarcinoma, consolidated with integration of single-cell RNA sequencing and bulk RNA sequencing techniques. Front Immunol 2023; 14:1278496. [PMID: 37965333 PMCID: PMC10641741 DOI: 10.3389/fimmu.2023.1278496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/13/2023] [Indexed: 11/16/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD), the predominant subtype of non-small cell lung cancer (NSCLC), remains a pervasive global public health concern. Disulfidoptosis, a nascent form of regulated cell death (RCD), presents an emerging field of inquiry. Currently, investigations into disulfidoptosis are in their initial stages. Our undertaking sought to integrate single-cell RNA sequencing (scRNA-seq) in conjunction with traditional bulk RNA sequencing (bulk RNA-seq) methodologies, with the objective of delineating genes associated with disulfidoptosis and subsequently prognosticating the clinical outcomes of LUAD patients. Methods Initially, we conducted an in-depth examination of the cellular composition disparities existing between LUAD and normal samples using scRNA-seq data sourced from GSE149655. Simultaneously, we scrutinized the expression patterns of disulfidoptosis-associated gene sets across diverse cell types. Subsequently, leveraging the bulk RNA-seq data, we formulated disulfidoptosis-related prognostic risk signatures (DRPS) employing LASSO-Cox regression. This was accomplished by focusing on genes implicated in disulfidoptosis that exhibited differential expression within endothelial cells (ECs). Sequentially, the robustness and precision of the DRPS model were rigorously verified through both internal and external validation datasets. In parallel, we executed single-cell trajectory analysis to delve into the differentiation dynamics of ECs. Concluding our study, we undertook a comprehensive investigation encompassing various facets. These included comparative assessments of enrichment pathways, clinicopathological parameters, immune cell abundance, immune response-associated genes, impacts of immunotherapy, and drug predictions among distinct risk cohorts. Results The scrutiny of scRNA-seq data underscored discernible disparities in cellular composition between LUAD and normal samples. Furthermore, disulfidoptosis-associated genes exhibited marked discrepancies within endothelial cells (ECs). Consequently, we formulated the Disulfidoptosis-Related Prognostic Signature (DRPS) to facilitate prognostic prediction. The prognostic nomogram based on the risk score effectively demonstrated DRPS's robust capacity to prognosticate survival outcomes. This assertion was corroborated by rigorous assessments utilizing both internal and external validation sets, thus affirming the commendable predictive accuracy and enduring stability of DRPS. Functional enrichment analysis shed light on the significant correlation of DRPS with pathways intrinsic to the cell cycle. Subsequent analysis unveiled correlations between DRPS and gene mutations characteristic of LUAD, as well as indications of an immunosuppressive status. Through drug prediction, we explored potential therapeutic agents for low-risk patients. Concluding our investigation, qRT-PCR experiments confirmed the heightened expression levels of EPHX1, LDHA, SHC1, MYO6, and TLE1 in lung cancer cell lines.
Collapse
Affiliation(s)
- Dabao He
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Hengfeng Tang
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Xiaoling Yang
- Department of Laboratory Medicine, Shenzhen Baoan District Songgang People’s Hospital, Shenzhen, China
| | - Xiaohong Liu
- Department of Oncology, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Yipeng Zhang
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Junzhu Shi
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| |
Collapse
|
5
|
Weng Z, Fu H, Huang Z, Li W, Xie Y, Yuan J, Ding B. Shikonin promotes ferroptosis in HaCaT cells through Nrf2 and alleviates imiquimod-induced psoriasis in mice. Chem Biol Interact 2023; 386:110788. [PMID: 39491143 DOI: 10.1016/j.cbi.2023.110788] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/09/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Psoriasis is one kind of autoimmune skin disease without efficiency cure. Shikonin (SHK) is a potential drug for psoriasis treatment. Recent study suggested that ferroptosis involved in the pathological process of psoriasis. This study proved the beneficial effect of SHK with an imiquimod (IMQ) induced psoriatic model mice, and studied the ferroptosis regulative mechanism of SHK with a HaCaT cells assay in vitro. In the study, it also found that SHK treatment significantly improved imiquimod (IMQ)-induced psoriasis symptoms in mice, attenuated the production of inflammatory cytokines, including interleukin (IL)-6, IL-17, and tumor necrosis factor-alpha (i.e., TNF-α), and strongly inhibited macrophage infiltration in inflamed psoriatic skin. SHK impacted HaCaT cells (a human keratinocyte cell line) viability by enhancing intracellular and mitochondrial ferrous and lipid peroxidation levels by regulating expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), nuclear receptor coactivator 4 (NCOA4) and glutathione peroxidase 4 (GPX4) which could be reversed by iron chelating agent. and in psoriatic dermatitis. However, this study found the reversing capability of low dose SHK on LPS inhibited GPX4 and Nrf2 expression, which was identity to that in psoriatic dermatitis. To conclude, SHK inhibited ferroptosis in psoriatic skin by reducing inflammation, ameliorating oxidative stress and iron accumulation. Nrf2 and GPX4 might be the two major targets of SHK in psoriatic skin lesion. Our study highly lighted the basic biological mechanism of SHK on ferroptosis regulation.
Collapse
Affiliation(s)
- Zhiwei Weng
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Hangjie Fu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Zhiguang Huang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Wenxia Li
- Hangzhou Innovation Institute, Beihang University, Hangzhou, 310051, China.
| | - Yixin Xie
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jianchang Yuan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Bin Ding
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
6
|
Wang J, Chen M, Wang S, Chu X, Ji H. Identification of Phytogenic Compounds with Antioxidant Action That Protect Porcine Intestinal Epithelial Cells from Hydrogen Peroxide Induced Oxidative Damage. Antioxidants (Basel) 2022; 11:2134. [PMID: 36358507 PMCID: PMC9687067 DOI: 10.3390/antiox11112134] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2024] Open
Abstract
Oxidative stress contributes to intestinal dysfunction. Plant extracts can have antioxidant action; however, the specific phytogenic active ingredients and their potential mechanisms are not well known. We screened 845 phytogenic compounds using a porcine epithelial cell (IPEC-J2) oxidative stress model to identify oxidative-stress-alleviating compounds. Calycosin and deoxyshikonin were evaluated for their ability to alleviate H2O2-induced oxidative stress by measuring their effects on malondialdehyde (MDA) accumulation, reactive oxygen species (ROS) generation, apoptosis, mitochondrial membrane potential (MMP), and antioxidant defense. Nrf2 pathway activation and the effect of Nrf2 knockdown on the antioxidative effects of hit compounds were investigated. Calycosin protected IPEC-J2 cells against H2O2-induced oxidative damage, likely by improving the cellular redox state and upregulating antioxidant defense via the Nrf2-Keap1 pathway. Deoxyshikonin alleviated the H2O2-induced decrease in cell viability, ROS production, and MMP reduction, but had no significant effect on MDA accumulation and apoptosis. Nrf2 knockdown did not weaken the effect of deoxyshikonin in improving cell viability, but it weakened its effect in suppressing ROS production. These results indicate that the mechanisms of action of natural compounds differ. The newly identified phytogenic compounds can be developed as novel antioxidant agents to alleviate intestinal oxidative stress in animals.
Collapse
Affiliation(s)
- Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
- Sino-US Joint Laboratory of Animal Science, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Meixia Chen
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Sixin Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Xu Chu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
- College of Agriculture and Animal Husbandry, Qinghai University, Xining 810016, China
| | - Haifeng Ji
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
- Sino-US Joint Laboratory of Animal Science, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| |
Collapse
|
7
|
Li Y, Liu J, Ye B, Cui Y, Geng R, Liu S, Zhang Y, Guo W, Fu S. Astaxanthin Alleviates Nonalcoholic Fatty Liver Disease by Regulating the Intestinal Flora and Targeting the AMPK/Nrf2 Signal Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10620-10634. [PMID: 35973099 DOI: 10.1021/acs.jafc.2c04476] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is among the most prevalent chronic liver diseases around the globe. The accumulation of lipids in the liver and oxidative stress are important pathological mechanisms of NAFLD. Astaxanthin (AT) is a carotenoid extracted from shrimps and crabs with beneficial biological activities, including anti-oxidative and anti-inflammatory activities. 16S microflora sequencing, H&E staining, and the western blot technique were employed to investigate the impacts of AT on a high-fat diet (HFD)-induced NAFLD. Significant mitigation in lipid metabolism-related disorders and decreased oxidative stress in HFD-induced mice were observed due to AT, and significant changes in the gut flora of the model mice were also observed. The in vitro study showed that AT considerably lowered the protein expression level of fatty acid synthetase (FAS), sterol regulatory element-binding protein-1c (SREBP-1c), and acetyl-COA carboxylase (ACC) and increased the protein expression of nuclear factor-E2 associated factor 2 (Nrf2) and AMP-activated protein kinase (AMPK) in oleic acid (OA) and palmitic acid (PA)-induced HepG2 cells. Additionally, mechanistic studies revealed that compound C (AMPK inhibitor, CC) inhibited the regulatory effect of AT on the SREBP-1c and Nrf2 signaling pathways. In conclusion, AT can inhibit the SREBP-1c, FAS, and ACC signaling pathways, activate the AMPK and Nrf2 signaling pathways, and improve the structure of intestinal flora.
Collapse
Affiliation(s)
- Yuhang Li
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Juxiong Liu
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Bojian Ye
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yueyao Cui
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Ruiqi Geng
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Shu Liu
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yufei Zhang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Wenjin Guo
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Shoupeng Fu
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
8
|
Dai Y, Liu Y, Li J, Jin M, Yang H, Huang G. Shikonin inhibited glycolysis and sensitized cisplatin treatment in non-small cell lung cancer cells via the exosomal pyruvate kinase M2 pathway. Bioengineered 2022; 13:13906-13918. [PMID: 35706397 PMCID: PMC9275963 DOI: 10.1080/21655979.2022.2086378] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The active ingredient of the traditional Chinese medicine comfrey is shikonin, a naphthoquinone compound. The focus of this study was to investigate the effect of shikonin on the proliferation, invasion, migration, and chemoresistance of non-small cell lung cancer (NSCLC) cells, and to explore its underlying molecular biological mechanisms. The results show that shikonin inhibited the viability, proliferation, invasion, and migration of NSCLC cells A549 and PC9, and induced apoptosis. As the inhibitor of pyruvate kinase M2 (PKM2), a key enzyme in glycolysis, shikonin inhibited glucose uptake and the production of lactate, the final metabolite of aerobic glycolysis. In vivo chemotherapeutic assay showed that shikonin reduced the tumor volume and weight in NSCLC mice model and increased the sensitivity to cisplatin chemotherapy. Histoimmunology experiments showed the combination of shikonin and cisplatin downregulated the expression of PKM2 and its transcriptionally regulated downstream gene glucose transporter 1 (Glut1) in tumor tissue. In an assessment of glucose metabolism, micro-PET/CT data showed a combination of shikonin and cisplatin inhibited the fluorodeoxy glucose (18F-FDG) uptake into tumor. Since exosomal PKM2 affected the sensitivity to cisplatin in NSCLC cells, we also demonstrated shikonin could inhibit exosome secretion and exosomal PKM2 through the administration of exosomal inhibitor GW4869. Furthermore, shikonin sensitized cisplatin treatment by reducing the extracellular secretion of exosomal PKM2. In conclusion, we suggest that shikonin not only inhibits PKM2 intracellularly but also reduces glycolytic flux and increases cisplatin sensitivity through the exosomal pathway.
Collapse
Affiliation(s)
- Yitian Dai
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Yuping Liu
- Beijing University of Chinese Medicine, Beijing, China
| | - Jingyi Li
- Qiqihar Medical University, Qiqihar Heilongjiang, China
| | - Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hao Yang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Gang Huang
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|