1
|
Wu Y, Wang D, Zhao J, Guo J, Gao Z, Xu Q, Hu X. NRAV promoted the malignant progression of gastric cancer. Gene 2025; 937:149134. [PMID: 39615808 DOI: 10.1016/j.gene.2024.149134] [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: 06/04/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
Gastric cancer (GC) has been ranked as the third incidence tumors globally. Long non-coding RNA (lncRNA) NRAV has been reported as a tumor-enhancer in the development of human cancers, whereas the function of NRAV in GC remains to be elucidated. The aim of this research was to explore the underlying function of NRAV in GC. Through comprehensive bioinformatics analysis, a significantly elevation of NRAV was found in both human GC tissues and cell lines, which indicated the poor prognosis of GC patients. Then, we conducted a series of functional experiments to illustrate the role of NRAV in GC. The results showed that the down-regulation of NRAV exhibited a significant inhibitory effect on GC cell proliferation and migration, while NRAV overexpression promoted GC cell proliferation and migration. Through xenograft mouse tumor model, the suppression of NRAV led to a reduction in the growth of tumor mice, whereas overexpression of NRAV notably enhanced tumor growth. Finally, EFHC1 was revealed as the potential target gene of NRAV. Overall, our findings indicated the promising application of NRAV as a therapeutic target and prognostic biomarker for GC.
Collapse
Affiliation(s)
- Yuchen Wu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China; Bengbu Medical University, Bengbu 233030, China
| | | | - Jie Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jinhui Guo
- Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Zhenyu Gao
- Wenzhou Medical University, Wenzhou 325035, China
| | - Qiuran Xu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China.
| | - Xiaoge Hu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China.
| |
Collapse
|
2
|
Liu X, Song L, Liu W, Liu B, Liu L, Su Y. Solamargine inhibits gastric cancer progression via inactivation of STAT3/PD‑L1 signaling. Mol Med Rep 2025; 31:35. [PMID: 39575463 PMCID: PMC11605270 DOI: 10.3892/mmr.2024.13400] [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: 02/06/2024] [Accepted: 09/02/2024] [Indexed: 12/01/2024] Open
Abstract
Gastric cancer (GC) is characterized by a high mortality rate (70%) worldwide. Programmed cell death‑1 and its ligand, programmed cell death ligand 1 (PD‑L1), are vital immune checkpoints, which serve a notable role in GC. Solamargine, an extract from traditional Chinese medicine Long Kui, exerts suppressive effects on several types of cancer including cervical, lung and prostate cancer. However, the association between solamargine and PD‑L1 in GC remains unclear. Therefore, the present study aimed to investigate the underlying mechanism of solamargine on GC. Specifically, 5‑ethynyl‑2'‑deoxyuridine and Transwell assays were performed to assess GC cell proliferation, invasion and migration. Additionally, GC cells (HGC‑827 and NCI‑N87) were stimulated with 20 ng/ml recombinant human IL‑6 for 24 h, before the protein expression levels of PD‑L1 were measured using western blot analysis. Furthermore, T cell function was evaluated through incubation of Jurkat T cells with solamargine. The results demonstrated that solamargine could markedly inhibit GC cell proliferation, migration and invasion, by inhibiting STAT3 signaling. In addition, GC cell treatment with solamargine downregulated the expression of PD‑L1. Furthermore, solamargine reversed the IL‑6‑induced PD‑L1 upregulation in GC cells by downregulating STAT3 activity. Additionally, the results demonstrated that solamargine inhibited IL‑6‑induced PD‑L1 upregulation of GC cells. This suggests that solamargine exerted an immunostimulatory activity in GC. In conclusion, the present study indicated that solamargine may inhibit the progression of GC by suppressing STAT3/PD‑L1 signaling. Therefore, treatment with solamargine may serve as novel strategy for the treatment of GC.
Collapse
Affiliation(s)
- Xiongxiang Liu
- Department of Gastroenterology, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Lin Song
- Department of Gastroenterology, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Wen Liu
- Department of Gastroenterology, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Bin Liu
- Department of Gastroenterology, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Lang Liu
- Department of Gastroenterology, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Yao Su
- Department of Gastroenterology, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| |
Collapse
|
3
|
Han M, Zhou X, Cheng H, Qiu M, Qiao M, Geng X. Chitosan and hyaluronic acid in colorectal cancer therapy: A review on EMT regulation, metastasis, and overcoming drug resistance. Int J Biol Macromol 2024; 289:138800. [PMID: 39694373 DOI: 10.1016/j.ijbiomac.2024.138800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 12/04/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Up to 90% of cancer-related fatalities could be attributed to metastasis. Therefore, understanding the mechanisms that facilitate tumor cell metastasis is beneficial for improving patient survival and results. EMT is considered the main process involved in the invasion and spread of CRC. Essential molecular components like Wnt, TGF-β, and PI3K/Akt play a role in controlling EMT in CRC, frequently triggered by various factors such as Snail, Twist, and ZEB1. These factors affect not only the spread of CRC but also determine the reaction to chemotherapy. The influence of non-coding RNAs, especially miRNAs and lncRNAs, on the regulation of EMT is clear in CRC. Exosomes, involved in cell-to-cell communication, can affect the TME and metastasis of CRC. Pharmacological substances and nanoparticles demonstrate promise as efficient modulators of EMT in CRC. Chitosan and HA are two major carbohydrate polymers with considerable potential in inhibiting CRC. Chitosan and HA can be employed to modify nanoparticles to enhance cargo transport for reducing CRC. Additionally, chitosan and HA-modified nanocarriers, which can be utilized as potential approaches in suppressing EMT and reversing drug resistance in CRC, can inhibit EMT and chemoresistance, crucial components in tumorigenesis.
Collapse
Affiliation(s)
- Mingming Han
- Department of Pharmacy and Medical Devices, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Xi Zhou
- Department of Occupational Pulmonology, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Hang Cheng
- Department of Bioanalytical Laboratory (ClinicalLaboratory), Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Mengru Qiu
- Department of Occupational Pulmonology, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| | - Meng Qiao
- Department of Bioanalytical Laboratory (ClinicalLaboratory), Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| | - Xiao Geng
- Department of Party Committee Office, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| |
Collapse
|
4
|
Zhu JY, Guo L. Exercise-regulated lipolysis: Its role and mechanism in health and diseases. J Adv Res 2024:S2090-1232(24)00550-2. [PMID: 39613256 DOI: 10.1016/j.jare.2024.11.031] [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: 10/04/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024] Open
Abstract
Exercise has received considerable attention because of its importance not just in regulating physiological function, but also in ameliorating multiple pathological processes. Among these processes, lipolysis may play an important role in exercise-induced benefits. It is generally accepted that active lipolysis contributes to breakdown of fats, leading to the release of free fatty acids (FFAs) that serve as an energy source for muscles and other tissues during exercise. However, the significance of lipolysis in the context of exercise has not been fully understood. This review comprehensively outlines the potential regulatory mechanisms by which exercise stimulates lipolysis. The potential roles of exercise-mediated lipolysis in various physiological and pathological processes are also summarized. Additionally, we also discussed the potential non-classical effects of key lipolytic effectors induced by exercise. This will enhance our understanding of how exercise improves lipolytic function to bring about beneficial effects, offering new insights into potential therapeutic avenues for promoting health and alleviating diseases.
Collapse
Affiliation(s)
- Jie-Ying Zhu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China 200438; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China 200438; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China 200438
| | - Liang Guo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China 200438; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China 200438; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China 200438.
| |
Collapse
|
5
|
Yu L, Zhang Z, Wang Z, Sun F. Expression of the lncRNA TPT1-AS1 in lung squamous cell carcinoma and its prognostic value. Discov Oncol 2024; 15:639. [PMID: 39527376 PMCID: PMC11554981 DOI: 10.1007/s12672-024-01470-7] [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: 09/02/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE Lung squamous cell carcinoma is typically associated with a poor prognosis, highlighting the need for a reliable prognostic marker. Given that the long noncoding RNA TPT1-AS1 has demonstrated aberrant expression in numerous cancers, the prognostic significance of TPT1-AS1 in LUSC was examined. METHODS The present study included 115 patients diagnosed with LUSC. The expression levels of TPT1-AS1 and miR-4726-5p in tissues and cells were assessed using RT-qPCR, while the correlation between TPT1-AS1 expression and clinicopathological features was analyzed through the chi-square test. Binding sites between TPT1-AS1 and miR-4726-5p were predicted using a database and confirmed via a dual-luciferase reporter assay. The Pearson correlation coefficients were calculated to determine the relationship between TPT1-AS1 and miR-4726-5p in tumor tissues. The impacts of TPT1-AS1 and miR-4726-5p on cells were evaluated through CCK-8 and Transwell assays. RESULTS TPT1-AS1 expression was found to be reduced, while miR-4726-5p expression was upregulated in LUSC tissues. Patients exhibiting low TPT1-AS1 expression experienced shorter overall survival. Moreover, TPT1-AS1 was identified as an independent prognostic factor. A dual luciferase reporter assay validated that TPT1-AS1-WT targeted and bound to miR-4726-5p. Additionally, an inverse correlation was observed between miR-4726-5p and TPT1-AS1 in tumors. Cell experiments indicated that the overexpression of TPT1-AS1 led to a decrease in miR-4726-5p expression, consequently inhibiting cell proliferation, migration, and invasion. CONCLUSION TPT1-AS1 targets miR-4726-5p, and overexpression of TPT1-AS1 has the potential to serve as a prognostic marker for LUSC and can significantly influence LUSC progression.
Collapse
Affiliation(s)
- Lixin Yu
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, 5 Jingyuan Road, Shijingshan District, Beijing, 100020, China.
| | - Zhenkui Zhang
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, 5 Jingyuan Road, Shijingshan District, Beijing, 100020, China
| | - Zhijian Wang
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, 5 Jingyuan Road, Shijingshan District, Beijing, 100020, China
| | - Fenghua Sun
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, 5 Jingyuan Road, Shijingshan District, Beijing, 100020, China
| |
Collapse
|
6
|
Xin Z, Hu C, Zhang C, Liu M, Li J, Sun X, Hu Y, Liu X, Wang K. LncRNA-HMG incites colorectal cancer cells to chemoresistance via repressing p53-mediated ferroptosis. Redox Biol 2024; 77:103362. [PMID: 39307047 PMCID: PMC11447409 DOI: 10.1016/j.redox.2024.103362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/06/2024] Open
Abstract
Upon chemotherapy, excessive reactive oxygen species (ROS) often lead to the production of massive lipid peroxides in cancer cells and induce cell death, namely ferroptosis. The elimination of ROS is pivotal for tumor cells to escape from ferroptosis and acquire drug resistance. Nevertheless, the precise functions of long non-coding RNAs (lncRNAs) in ROS metabolism and tumor drug-resistance remain elusive. In this study, we identify LncRNA-HMG as a chemoresistance-related lncRNA in colorectal cancer (CRC) by high-throughput screening. Abnormally high expression of LncRNA-HMG predicts poorer prognosis in CRC patients. Concurrently, we found that LncRNA-HMG protects CRC cells from ferroptosis upon chemotherapy, thus enhancing drug resistance of CRC cells. LncRNA-HMG binds to p53 and facilitates MDM2-mediated degradation of p53. Decreased p53 induces upregulation of SLC7A11 and VKORC1L1, which contribute to increase the supply of reducing agents and eliminate excessive ROS. Consequently, CRC cells escape from ferroptosis and acquire chemoresistance. Importantly, inhibition of LncRNA-HMG by anti-sense oligo (ASO) dramatically sensitizes CRC cells to chemotherapy in patient-derived xenograft (PDX) model. LncRNA-HMG is also a transcriptional target of β-catenin/TCF and activated Wnt signals trigger the marked upregulation of LncRNA-HMG. Collectively, these findings demonstrate that LncRNA-HMG promotes CRC chemoresistance and might be a prognostic or therapeutic target for CRC.
Collapse
Affiliation(s)
- Zechang Xin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Chenyu Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Chunfeng Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ming Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Juan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaoyan Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yang Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaofeng Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Kun Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
7
|
Suvarnapathaki S, Serrano-Farias A, Dudley JC, Bettegowda C, Rincon-Torroella J. Unlocking the Potential of Circulating miRNAs as Biomarkers in Glioblastoma. Life (Basel) 2024; 14:1312. [PMID: 39459612 PMCID: PMC11509808 DOI: 10.3390/life14101312] [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: 09/05/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Using microRNAs (miRNAs) as potential circulating biomarkers in diagnosing and treating glioblastoma (GBM) has garnered a lot of scientific and clinical impetus in the past decade. As an aggressive primary brain tumor, GBM poses challenges in early detection and effective treatment with significant current diagnostic constraints and limited therapeutic strategies. MiRNA dysregulation is present in GBM. The intricate involvement of miRNAs in altering cell proliferation, invasion, and immune escape makes them prospective candidates for identifying and monitoring GBM diagnosis and response to treatment. These miRNAs could play a dual role, acting as both potential diagnostic markers and targets for therapy. By modulating the activity of various oncogenic and tumor-suppressive proteins, miRNAs create opportunities for precision medicine and targeted therapies in GBM. This review centers on the critical role and function of circulating miRNA biomarkers in GBM diagnosis and treatment. It highlights their significance in providing insights into disease progression, aiding in early diagnosis, and potential use as targets for novel therapeutic interventions. Ultimately, the study of miRNA would contribute to improving patient outcomes in the challenging landscape of GBM management.
Collapse
Affiliation(s)
- Sanika Suvarnapathaki
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (S.S.); (A.S.-F.); (J.C.D.); (C.B.)
| | - Antolin Serrano-Farias
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (S.S.); (A.S.-F.); (J.C.D.); (C.B.)
| | - Jonathan C. Dudley
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (S.S.); (A.S.-F.); (J.C.D.); (C.B.)
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (S.S.); (A.S.-F.); (J.C.D.); (C.B.)
| | - Jordina Rincon-Torroella
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (S.S.); (A.S.-F.); (J.C.D.); (C.B.)
| |
Collapse
|
8
|
Shi Y, Men X, Wang F, Li X, Zhang B. Role of long non-coding RNAs (lncRNAs) in gastric cancer metastasis: A comprehensive review. Pathol Res Pract 2024; 262:155484. [PMID: 39180802 DOI: 10.1016/j.prp.2024.155484] [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: 01/15/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024]
Abstract
One of the greatest frequent types of malignancy is gastric cancer (GC). Metastasis, an essential feature of stomach cancer, results in a high rate of mortality and a poor prognosis. However, metastasis biological procedures are not well recognized. Long non-coding RNAs (lncRNAs) have a role in numerous gene regulation pathways via epigenetic modification as well as transcriptional and post-transcriptional control. LncRNAs have a role in a variety of disorders, such as cardiovascular disease, Alzheimer's, and cancer. LncRNAs are substantially related to GC incidence, progression, metastasis and drug resistance. Several research released information on the molecular processes of lncRNAs in GC pathogenesis. By interacting with a gene's promoter or enhancer region to influence gene expression, lncRNAs can operate as an oncogene or a tumor suppressor. This review includes the lncRNAs associated with metastasis of GC, which may give insights into the processes as well as potential clues for GC predicting and tracking.
Collapse
Affiliation(s)
- Yue Shi
- Department of Microbiology and Immunology, Changchun University of Chinese Medicine, Jilin 130117, PR China.
| | - Xiaoping Men
- Department of Clinical Laboratory, The First Affiliated Hospital to Changchun University of Chinese Medicine, Jilin 130021, PR China.
| | - Fang Wang
- Department of Microbiology and Immunology, Changchun University of Chinese Medicine, Jilin 130117, PR China.
| | - Xueting Li
- Experimental Center, Changchun University of Chinese Medicine, Jilin 130021, PR China.
| | - Biao Zhang
- School of Health Management, Changchun University of Chinese Medicine, Jilin 130117, PR China.
| |
Collapse
|
9
|
Wang Z, Xue M, Liu J, Jiang H, Li F, Xu M, Wang H. ATP11A Promotes Epithelial-mesenchymal Transition in Gastric Cancer Cells via the Hippo Pathway. J Cancer 2024; 15:5477-5491. [PMID: 39247595 PMCID: PMC11375558 DOI: 10.7150/jca.97895] [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: 04/29/2024] [Accepted: 07/30/2024] [Indexed: 09/10/2024] Open
Abstract
Background: ATP11A, a P-type ATPase, functions as flippases at the plasma membrane to maintain cellular function and vitality in several cancers. However, the role of ATP11A in gastric cancer remains unknown. This study aimed to identify ATP11A related to the biological behavior of gastric cancer, and elucidate the underlying mechanism. Methods: The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases were used to analyze the expression and prognosis of ATP11A. The biofunctions of ATP11A were explored through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GSEA). The expression of ATP11A were validated by immunohistochemistry (IHC), qRT-PCR and Western blotting. Transwell, wound healing, CCK8 and colony-formation were to detected the migration, invasion and proliferation of gastric cancer cells. The epithelial-mesenchymal transition (EMT) and Hippo pathway markers were examined by Western blotting. Results: The expression of ATP11A was higher in gastric cancer tissues than in normal tissues, and high ATP11A levels were related to worse prognosis of gastric cancer patients. Additionally, we proved that ATP11A promoted the migration, invasion and proliferation in gastric cancer cells. Furthermore, ATP11A was found to promote EMT by devitalizing the Hippo pathway. Conclusion: ATP11A promoted migration, invasion, proliferation and EMT via Hippo signaling devitalization in gastric cancer cells.
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Mingmiao Xue
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Junqiang Liu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Han Jiang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Feifan Li
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| | - Huizhi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, China
| |
Collapse
|
10
|
Ma A, Shi W, Chen L, Huang Z, Zhang Y, Tang Z, Jiang W, Xu M, Zhou J, Zhang W, Tang S. GRASLND regulates melanoma cell progression by targeting the miR-218-5p/STAM2 axis. J Transl Med 2024; 22:684. [PMID: 39060946 PMCID: PMC11282654 DOI: 10.1186/s12967-024-05397-z] [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: 05/04/2024] [Accepted: 06/12/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Increasing evidence suggests that long noncoding RNAs (lncRNAs) play important regulatory roles in biological processes and are dysregulated in numerous tumors. The lncRNA GRASLND functions as an oncogene in many cancers, but its role in skin cutaneous melanoma (SKCM) requires further investigation. METHODS SiRNA transfection, wound - healing and transwell assays were performed to evaluate the effect of GRASLND on cellular function. RESULTS The present study demonstrated that GRASLND expression is increased in SKCM tissues and cell lines. The high expression of GRASLND was correlated with poor prognosis and immunotherapy outcomes. Knockdown of GRASLND significantly inhibited cell migration and invasion. In addition, we found that miR-218-5p directly binds to its binding site on GRASLND, and GRASLND and miR-218-5p demonstrate mutual inhibition. Furthermore, the miR-218-5p inhibitor partially eliminated the knockdown of GRASLND and inhibited its expression. We also demonstrated that GRASLND acts as a miR-218-5p sponge that positively regulates STAM2 expression in SKCM cells. CONCLUSION In summary, these data suggest that GRASLND functions by regulating miR-218-5p/STAM2 expression, suggesting an important role for the lncRNA‒miRNA-mRNA functional network and a new potential therapeutic target for SKCM.
Collapse
Affiliation(s)
- Aiwei Ma
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515051, China
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China
- Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong, 515051, China
| | - Wenqi Shi
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515051, China
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China
- Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong, 515051, China
| | - Liyun Chen
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China
- Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong, 515051, China
- Research Center of Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515051, China
| | - Zijian Huang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515051, China
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China
- Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong, 515051, China
| | - Yiwen Zhang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515051, China
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China
- Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong, 515051, China
| | - Zixuan Tang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515051, China
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China
- Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong, 515051, China
| | - Wenshi Jiang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515051, China
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China
- Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong, 515051, China
| | - Mengjing Xu
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515051, China
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China
- Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong, 515051, China
| | - Jianda Zhou
- Department of Plastic and Reconstructive Surgery, Central South University Third Xiangya Hospital, Changsha, China
| | - Wancong Zhang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515051, China.
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China.
- Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong, 515051, China.
| | - Shijie Tang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515051, China.
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China.
- Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong, 515051, China.
| |
Collapse
|
11
|
Tashakori N, Kolour SSP, Ghafouri K, Ahmed SI, Kahrizi MS, Gerami R, Altafi M, Nazari A. Critical role of the long non-coding RNAs (lncRNAs) in radiotherapy (RT)-resistance of gastrointestinal (GI) cancer: Is there a way to defeat this resistance? Pathol Res Pract 2024; 258:155289. [PMID: 38703607 DOI: 10.1016/j.prp.2024.155289] [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: 02/15/2024] [Revised: 03/29/2024] [Accepted: 03/31/2024] [Indexed: 05/06/2024]
Abstract
Radiotherapy (RT) is a frequently used treatment for cervical cancer, effectively decreasing the likelihood of the disease returning in the same area and extending the lifespan of individuals with cervical cancer. Nevertheless, the primary reason for treatment failure in cancer patients is the cancer cells' resistance to radiation therapy (RT). Long non-coding RNAs (LncRNAs) are a subset of RNA molecules that do not code for proteins and are longer than 200 nucleotides. They have a significant impact on the regulation of gastrointestinal (GI) cancers biological processes. Recent research has shown that lncRNAs have a significant impact in controlling the responsiveness of GI cancer to radiation. This review provides a concise overview of the composition and operation of lncRNAs as well as the intricate molecular process behind radiosensitivity in GI cancer. Additionally, it compiles a comprehensive list of lncRNAs that are linked to radiosensitivity in such cancers. Furthermore, it delves into the potential practical implementation of these lncRNAs in modulating radiosensitivity in GI cancer.
Collapse
Affiliation(s)
- Nafiseh Tashakori
- Department of Internal Medicine, Faculty of Medicine, Tehran branch, Islamic Azad University, Tehran, Iran
| | | | - Kimia Ghafouri
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sarah Ibrahem Ahmed
- Department of Anesthesia Techniques, Al-Noor University College, Nineveh, Iraq
| | | | - Reza Gerami
- Department of Radiology, Faculty of Medicine, AJA University of Medical Science, Tehran, Iran
| | - Mana Altafi
- Department of Radiology, Faculty of Biological Science and Technology, Shiraz Pardis Branch, Islamic Azad University, Shiraz, Iran.
| | - Afsaneh Nazari
- Department of Genetics, Faculty of Basic Sciences, Islamic Azad University, Zanjan Branch, Zanjan, Iran.
| |
Collapse
|
12
|
Li Y, Wei X, Yang Q, Xiong A, Li X, Zou Q, Cui F, Zhang Z. msBERT-Promoter: a multi-scale ensemble predictor based on BERT pre-trained model for the two-stage prediction of DNA promoters and their strengths. BMC Biol 2024; 22:126. [PMID: 38816885 PMCID: PMC11555825 DOI: 10.1186/s12915-024-01923-z] [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: 01/09/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND A promoter is a specific sequence in DNA that has transcriptional regulatory functions, playing a role in initiating gene expression. Identifying promoters and their strengths can provide valuable information related to human diseases. In recent years, computational methods have gained prominence as an effective means for identifying promoter, offering a more efficient alternative to labor-intensive biological approaches. RESULTS In this study, a two-stage integrated predictor called "msBERT-Promoter" is proposed for identifying promoters and predicting their strengths. The model incorporates multi-scale sequence information through a tokenization strategy and fine-tunes the DNABERT model. Soft voting is then used to fuse the multi-scale information, effectively addressing the issue of insufficient DNA sequence information extraction in traditional models. To the best of our knowledge, this is the first time an integrated approach has been used in the DNABERT model for promoter identification and strength prediction. Our model achieves accuracy rates of 96.2% for promoter identification and 79.8% for promoter strength prediction, significantly outperforming existing methods. Furthermore, through attention mechanism analysis, we demonstrate that our model can effectively combine local and global sequence information, enhancing its interpretability. CONCLUSIONS msBERT-Promoter provides an effective tool that successfully captures sequence-related attributes of DNA promoters and can accurately identify promoters and predict their strengths. This work paves a new path for the application of artificial intelligence in traditional biology.
Collapse
Affiliation(s)
- Yazi Li
- School of Mathematics and Statistics, Hainan University, Haikou, 570228, China
| | - Xiaoman Wei
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Qinglin Yang
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - An Xiong
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Xingfeng Li
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, 324000, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China.
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, Haikou, 570228, China.
| |
Collapse
|
13
|
Bai Y, Li J, Wei Y, Chen Z, Liu Z, Guo D, Jia X, Niu Y, Shi B, Zhang X, Zhao Z, Hu J, Han X, Wang J, Liu X, Li S. Proteome Analysis Related to Unsaturated Fatty Acid Synthesis by Interfering with Bovine Adipocyte ACSL1 Gene. Antioxidants (Basel) 2024; 13:641. [PMID: 38929080 PMCID: PMC11200461 DOI: 10.3390/antiox13060641] [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: 04/27/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Unsaturated fatty acids (UFAs) in beef play a vital role in promoting human health. Long-chain fatty acyl-CoA synthase 1 (ACSL1) is a crucial gene for UFA synthesis in bovine adipocytes. To investigate the protein expression profile during UFA synthesis, we performed a proteomic analysis of bovine adipocytes by RNA interference and non-interference with ACSL1 using label-free techniques. A total of 3558 proteins were identified in both the NC and si-treated groups, of which 1428 were differentially expressed proteins (DEPs; fold change ≥ 1.2 or ≤ 0.83 and p-value < 0.05). The enrichment analysis of the DEPs revealed signaling pathways related to UFA synthesis or metabolism, including cAMP, oxytocin, fatty acid degradation, glycerol metabolism, insulin, and the regulation of lipolysis in adipocytes (p-value < 0.05). Furthermore, based on the enrichment analysis of the DEPs, we screened 50 DEPs that potentially influence the synthesis of UFAs and constructed an interaction network. Moreover, by integrating our previously published transcriptome data, this study established a regulatory network involving differentially expressed long non-coding RNAs (DELs), highlighting 21 DEPs and 13 DELs as key genes involved in UFA synthesis. These findings present potential candidate genes for further investigation into the molecular mechanisms underlying UFA synthesis in bovines, thereby offering insights to enhance the quality of beef and contribute to consumer health in future studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zhidong Zhao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.B.); (J.L.); (Y.W.); (Z.C.); (Z.L.); (D.G.); (X.J.); (Y.N.); (B.S.); (X.Z.); (X.H.); (J.W.); (X.L.); (S.L.)
| | - Jiang Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.B.); (J.L.); (Y.W.); (Z.C.); (Z.L.); (D.G.); (X.J.); (Y.N.); (B.S.); (X.Z.); (X.H.); (J.W.); (X.L.); (S.L.)
| | | | | | | | | |
Collapse
|
14
|
Wang Y, Luo J, Yang H, Liu Y. LncRNA Peg13 Alleviates Myocardial Infarction/Reperfusion Injury through Regulating MiR-34a/Sirt1-Mediated Endoplasmic Reticulum Stress. Int Heart J 2024; 65:517-527. [PMID: 38825496 DOI: 10.1536/ihj.23-453] [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] [Indexed: 06/04/2024]
Abstract
Myocardial infarction/reperfusion (I/R) injury significantly impacts the health of older individuals. We confirmed that the level of lncRNA Peg13 was downregulated in I/R injury. However, the detailed function of Peg13 in myocardial I/R injury has not yet been explored.To detect the function of Peg13, in vivo model of I/R injury was constructed. RT-qPCR was employed to investigate RNA levels, and Western blotting was performed to assess levels of endoplasmic reticulum stress and apoptosis-associated proteins. EdU staining was confirmed to assess the cell proliferation.I/R therapy dramatically produced myocardial injury, increased the infarct area, and decreased the amount of Peg13 in myocardial tissues of mice. In addition, hypoxia/reoxygenation (H/R) notably induced the apoptosis and promoted the endoplasmic reticulum (ER) stress of HL-1 cells, while overexpression of Peg13 reversed these phenomena. Additionally, Peg13 may increase the level of Sirt1 through binding to miR-34a. Upregulation of Peg13 reversed H/R-induced ER stress via regulation of miR-34a/Sirt1 axis.LncRNA Peg13 reduces ER stress in myocardial infarction/reperfusion injury through mediation of miR-34a/Sirt1 axis. Hence, our research might shed new lights on developing new strategies for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Yonghong Wang
- Department of Cardiovascular Medicine, The Fourth Hospital of Changsha
| | - Jian Luo
- Department of Cardiovascular Medicine, The Fourth Hospital of Changsha
| | - Huiqiong Yang
- Department of Cardiovascular Medicine, The Fourth Hospital of Changsha
| | - Yanfei Liu
- Department of Cardiovascular Medicine, The Fourth Hospital of Changsha
| |
Collapse
|
15
|
Zhu Y, Huang C, Zhang C, Zhou Y, Zhao E, Zhang Y, Pan X, Huang H, Liao W, Wang X. LncRNA MIR200CHG inhibits EMT in gastric cancer by stabilizing miR-200c from target-directed miRNA degradation. Nat Commun 2023; 14:8141. [PMID: 38065939 PMCID: PMC10709323 DOI: 10.1038/s41467-023-43974-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023] Open
Abstract
Gastric cancer (GC) is a heterogeneous disease, threatening millions of lives worldwide, yet the functional roles of long non-coding RNAs (lncRNAs) in different GC subtypes remain poorly characterized. Microsatellite stable (MSS)/epithelial-mesenchymal transition (EMT) GC is the most aggressive subtype associated with a poor prognosis. Here, we apply integrated network analysis to uncover lncRNA heterogeneity between GC subtypes, and identify MIR200CHG as a master regulator mediating EMT specifically in MSS/EMT GC. The expression of MIR200CHG is silenced in MSS/EMT GC by promoter hypermethylation, associated with poor prognosis. MIR200CHG reverses the mesenchymal identity of GC cells in vitro and inhibits metastasis in vivo. Mechanistically, MIR200CHG not only facilitates the biogenesis of its intronic miRNAs miR-200c and miR-141, but also protects miR-200c from target-directed miRNA degradation (TDMD) through direct binding to miR-200c. Our studies reveal a landscape of a subtype-specific lncRNA regulatory network, providing clinically relevant biological insights towards MSS/EMT GC.
Collapse
Grants
- 2020N368 Shenzhen Science and Technology Innovation Commission
- C4024-22GF Research Grants Council, University Grants Committee (RGC, UGC)
- 14104223 Research Grants Council, University Grants Committee (RGC, UGC)
- 11103619 Research Grants Council, University Grants Committee (RGC, UGC)
- 14111522 Research Grants Council, University Grants Committee (RGC, UGC)
- R4017-18 Research Grants Council, University Grants Committee (RGC, UGC)
- 82173289 National Natural Science Foundation of China (National Science Foundation of China)
- 81872401 National Natural Science Foundation of China (National Science Foundation of China)
- Guangdong Basic and Applied Basic Research Foundation (Project No.2019B030302012), a startup grant (Project No. 4937084), direct grant (2021.077), Faculty Postdoctoral Fellowship Scheme 2021/22 (Project No. FPFS/2122/32), Shenzhen Bay Scholars Program.
- Guangdong Basic and Applied Basic Research Foundation (2021A1515010425)
Collapse
Affiliation(s)
- Yixiao Zhu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
- National Clinical Research Centre for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chengmei Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Chao Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yi Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Enen Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yaxin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xingyan Pan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Huilin Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Wenting Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Xin Wang
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| |
Collapse
|