51
|
Chen D, Cao G, Liu Q. A-kinase-interacting protein 1 facilitates growth and metastasis of gastric cancer cells via Slug-induced epithelial-mesenchymal transition. J Cell Mol Med 2019; 23:4434-4442. [PMID: 31020809 PMCID: PMC6533465 DOI: 10.1111/jcmm.14339] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 03/07/2019] [Accepted: 04/01/2019] [Indexed: 12/17/2022] Open
Abstract
A‐kinase‐interacting protein 1 (AKIP1) has previously been reported to act as a potential oncogenic protein in various cancers. The clinical significance and biological role of AKIP1 in gastric cancer (GC) is, however, still elusive. Herein, this study aimed to investigate the functional and molecular mechanism by which AKIP1 influences GC. AKIP1 mRNA and protein expressions in GC tissues were examined by quantitative real‐time PCR (qRT‐PCR), Western blot and immunohistochemistry. Other methods including stably transfected against AKIP1 into gastric cancer cells, wound healing, transwell assays, CCK‐8, colony formation, qRT‐PCR and Western blot in vitro and tumorigenesis in vivo were also performed. The up‐regulated expression of AKIP1 in GC specimens significantly correlated with clinical metastasis and poor prognosis in patients with GC. AKIP1 knockdown markedly suppressed GC cells proliferation, invasion and metastasis both in vitro and in vivo. In contrast, AKIP1 overexpression resulted in the opposite effects. Moreover, mechanistic analyses indicated that Slug‐induced epithelial‐mesenchymal transition (EMT) might be responsible for AKIP1‐influenced GC cells behaviour. Our findings demonstrated that high AKIP1 expression significantly correlated with clinical metastasis and unfavourable prognosis in patients with GC. Additionally, AKIP1 promoted GC cells proliferation, migration and invasion by activating Slug‐induced EMT.
Collapse
Affiliation(s)
- Dehu Chen
- Department of General Surgery, Taizhou People's Hospital, The Fifth Affiliated Hospital of Nantong University, Taizhou, China
| | - Gan Cao
- Department of General Surgery, Taizhou People's Hospital, The Fifth Affiliated Hospital of Nantong University, Taizhou, China
| | - Qinghong Liu
- Department of General Surgery, Taizhou People's Hospital, The Fifth Affiliated Hospital of Nantong University, Taizhou, China
| |
Collapse
|
52
|
Li H, Zhang W, Yan M, Qiu J, Chen J, Sun X, Chen X, Song L, Zhang Y. Nucleolar and spindle associated protein 1 promotes metastasis of cervical carcinoma cells by activating Wnt/β-catenin signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:33. [PMID: 30678687 PMCID: PMC6346521 DOI: 10.1186/s13046-019-1037-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/13/2019] [Indexed: 01/06/2023]
Abstract
Background The primary obstacle to treat cervical cancer is its high prevalence of metastasis, which severely affects patients’ quality of life and survival time. Nucleolar and spindle associated protein 1 (NUSAP1) has been implicated in the development, progression, and metastasis in several types of cancer. However, its oncogenic role in cervical cancer remains unclear. Methods Western blot assay and immunohistochemistry were used to determine the expression of NUSAP1 in 21 clinical fresh Cervical cancer tissues and 233 clinicopathologically characterized cervical cancer specimens. The biological roles of NUSAP1 in the metastasis of cervical cancer were investigated both in vitro by EMT, Side population analysis and Transwell assays and so on, and in vivo using a mouse 4w model of hematogenous metastasis and lymph node metastasis. Bioinformatics analysis, luciferase reporter analysis, immunoprecipitation and immunoblotting of nuclear and cytoplasmic cellular fractions were applied to discern and examine the relationshipbetween NUSAP1 and its potential targets. Results The results demonstrated that NUSAP1 was upregulated in cervical cancer cells and tissues, correlated positively with metastasis and poor clinical outcome of patients. High expression of NUSAP1 promoted metastasis by enhancing cancer stem cell (CSC) traits and epithelial-mesenchyme transition (EMT) progression, while silencing of NUSAP1 reduced CSC traits and EMT progression. Mechanistically, upregulation of NUSAP1 induced SUMOylation of TCF4 via interacting with SUMO E3 ligase Ran-binding protein 2 (RanBP2) and hyperactivated Wnt/β-catenin signaling in cervical cancer cells. Additionally, NUSAP1-induced cervical cancer cells metastasis and the cancer stem cell phenotype were abrogated with the Wnt/β-catenin signaling inhibitor XAV-939 treatment. Importantly, co-therapy of conventional treatment and XAV-939 will provide a novel and effective treatment for NUSAP1-ovexpressed cervical cancer patients. Conclusions Our results demonstrate thatNUSAP1 upregulation contributes to metastasis of cervical cancer by promoting CSC properties and EMT via Wnt/β-catenin signaling and XAV-939 might serve as a potential tailored therapeutic option for patients with NUSAP1-ovexpressed cervical cancer. Electronic supplementary material The online version of this article (10.1186/s13046-019-1037-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Han Li
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Weijing Zhang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Ming Yan
- Department of Obstetrics Gynecology, The First Pepole's Hospital, Foshan, Guangdong, China
| | - Jiaqi Qiu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jueming Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiaoying Sun
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiangfu Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Libing Song
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yanna Zhang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
53
|
Liu C, Sun L, Sun J. Retracted Article: Circular RNA hsa_circ_0000467 modulates SGK1 to facilitate cell migration, metastasis, and EMT while repressing apoptosis in colorectal cancer by sponging miR-383-5p. RSC Adv 2019; 9:39294-39303. [PMID: 35540673 PMCID: PMC9076104 DOI: 10.1039/c9ra07900a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/22/2019] [Indexed: 12/30/2022] Open
Abstract
Recent data indicated that circular RNAs (circRNAs) were implicated in tumor progression including colorectal cancer (CRC). However, the mechanism of hsa_circ_0000467 in CRC remains unclear. The levels of hsa_circ_0000467, microRNA-383-5p (miR-383-5p), and serum/glucocorticoid regulated kinase 1 (SGK1) in CRC tissues and cells were measured by quantitative real-time polymerase chain reaction (qRT-PCR). The cell viability and apoptotic rate were detected through cell counting kit-8 (CCK-8) assay and flow cytometry, respectively. The migration and invasion abilities were evaluated via Transwell assay. The protein levels of cleaved caspase 3 (C-caspase 3), B-cell lymphoma 2 (Bcl-2), N-cadherin, E-cadherin, SGK1, and proliferating cell nuclear antigen (PCNA) were detected by western blot assay. The dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were constructed to verify the interaction between miR-383-5p and hsa_circ_0000467 or SGK1. The mouse model experiment was performed to further validate the effects of hsa_circ_0000467 on CRC progression. Hsa_circ_0000467 and SGK1 were enhanced while miR-383-5p was reduced in CRC tissues and cells. Hsa_circ_0000467 silencing suppressed cell proliferation, migration, invasion, and epithelial–mesenchymal transition (EMT) but induced apoptosis in CRC cells by regulating miR-383-5p. Hsa_circ_0000467 sponged miR-383-5p and SGK1 was a direct target of miR-383-5p. Besides, hsa_circ_0000467 promoted SGK1 expression in CRC cells by sponging miR-383-5p. Furthermore, miR-383-5p restrained cell proliferation, metastasis, and EMT but facilitated apoptosis in CRC cells by modulating SGK1. Also, hsa_circ_0000467 knockdown blocked xenograft tumor growth in vivo. Hsa_circ_0000467 promoted CRC progression by regulating SGK1 expression via miR-383-5p. Recent data indicated that circular RNAs (circRNAs) were implicated in tumor progression including colorectal cancer (CRC). However, the mechanism of hsa_circ_0000467 in CRC remains unclear.![]()
Collapse
Affiliation(s)
- Chong Liu
- Department of General Surgery
- Shengjing Hospital Affiliated to China Medical University
- China
| | - Lingling Sun
- Pulmonary Department and Intensive Care Unit
- The Fourth Affiliated Hospital of China Medical University
- China Medical University
- Shenyang
- P. R. China
| | - Jiaying Sun
- Pulmonary Department and Intensive Care Unit
- The Fourth Affiliated Hospital of China Medical University
- China Medical University
- Shenyang
- P. R. China
| |
Collapse
|
54
|
Guan Y, Shi H, Xiao T. NUAK1 knockdown suppresses prostate cancer cell epithelial-mesenchymal transition, migration, and invasion through microRNA-30b-5p. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:5694-5704. [PMID: 31949655 PMCID: PMC6963066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/13/2018] [Indexed: 06/10/2023]
Abstract
OBJECTIVE Prostate cancer is one of the most diagnosed malignancies in men worldwide. Novel (nua) kinase family 1 (NUAK1) is a member of adenosine monophosphate (AMP)-related kinase which participates in varying cancers progression. However, the role of NUAK1 in prostate tumorigenesis has not been fully characterized. The aim of this study was to elucidate the potential biological role of NUAK1 in prostate cancer. METHODS Quantitative real-time PCR (qRT-PCR) was performed to determine the expression levels of NUAK1 and microRNA-30b-5p (miRNA-30b-5p) in prostate cancer cell lines and samples. Western blot was conducted to explore the related protein levels of epithelial-mesenchymal transition (EMT) and NUAK1 expression in prostate cancer cells. Trans-well test was used to assay prostate cancer cell migration and invasion. Luciferase assays were employed to probe the interaction between NUAK1 and miR-30b-5p. RESULTS NUAK1 abundance was enhanced in prostate cancer tissues and cell lines. The knockdown of NUAK1 may inhibit prostate cancer cells EMT, migration and invasion. Luciferase assays suggested NUAK1 was a target gene of miR-30b-5p. Furthermore, miR-30b-5p suppressed EMT, migration, and invasion in prostate cancer cells and introduction of NUAK1 abated the inhibitory effect. CONCLUSIONS Both of NUAK1 and miR-30b-5p were required for prostate cancer progression. NUAK1 interference limited prostate cancer cell EMT, migration and invasion by miRNA-30b-5p modulating, providing a promising therapeutic approach for prostate cancer.
Collapse
Affiliation(s)
- Yongjun Guan
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science China
| | - Hongbo Shi
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science China
| | - Tianlin Xiao
- Department of Urology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science China
| |
Collapse
|
55
|
Zhu JF, Liu Y, Huang H, Shan L, Han ZG, Liu JY, Li YL, Dong X, Zeng W. MicroRNA-133b/EGFR axis regulates esophageal squamous cell carcinoma metastases by suppressing anoikis resistance and anchorage-independent growth. Cancer Cell Int 2018; 18:193. [PMID: 30479571 PMCID: PMC6251163 DOI: 10.1186/s12935-018-0684-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/09/2018] [Indexed: 12/13/2022] Open
Abstract
Background Anoikis resistance has been demonstrated to facilitate distant metastases of cancers. MicroRNA-133b (miR-133b) is found to be down-regulated in various tumors, including esophageal squamous cell carcinoma (ESCC), and closely correlates with the malignant phenotype of ESCC. This study aimed to evaluate the roles of miR-133b in metastases of ESCC via regulating anoikis. Methods The expression of miR-133b and related molecules were detected in ESCC tissues and cells. The target relationship between miR-133b and epidermal growth factor receptor (EGFR) was verified by dual luciferase reporter assay. Cell proliferation was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Anoikis and anchorage-independent growth were assessed by anoikis assay and soft agar assay. Migration and invasion were evaluated by scratch and transwell assays. The expressions of related molecules were detected by reverse transcription-quantitative polymerase chain reaction and western blotting. The in vivo results were determined by tumor xenografts in nude mice. Results MiR-133b level was decreased in ESCC tissues and cells, which negatively correlated with EGFR, integrin β4 (ITGB4), and phosphorylated focal adhesion kinase levels. Moreover, miR-133b down-regulated EGFR expression in ESCC cells. Overexpression of miR-133b inhibited the anoikis resistance, migration, invasion and epithelial-mesenchymal transition of ESCC cells via targeting EGFR. Finally, miR-133b overexpression suppressed tumor growth and lung metastases of ESCC in vivo. ITGB4/FAK/growth factor receptor-bound protein 2 (Grb2), protein kinase B (AKT), and extracellular signal-regulated kinase (ERK) pathways were involved in the regulatory mechanisms of miR-133b/EGFR axis in ESCC metastases in vitro and in vivo. Conclusions The results suggested that miR-133b/EGFR axis regulated metastases of ESCC by affecting anoikis resistance via ITGB4/FAK/Grb2, AKT, and ERK pathways.
Collapse
Affiliation(s)
- Jin-Feng Zhu
- 2Department of Gastrointestinal Surgery, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011 People's Republic of China
| | - Yi Liu
- 3Department of Cardiothoracic Surgery, Shenzhen University General Hospital, Shenzhen, 518055 People's Republic of China
| | - He Huang
- 4Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, 410013 People's Republic of China.,5Department of Histology and Embryology, Xinjiang Medical University, Urumqi, 830011 People's Republic of China
| | - Li Shan
- 1First Department of Lung Cancer Chemotherapy, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789, East Suzhou Street, Urumqi, 830011 Xinjiang People's Republic of China
| | - Zhi-Gang Han
- 1First Department of Lung Cancer Chemotherapy, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789, East Suzhou Street, Urumqi, 830011 Xinjiang People's Republic of China
| | - Jun-Yuan Liu
- 1First Department of Lung Cancer Chemotherapy, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789, East Suzhou Street, Urumqi, 830011 Xinjiang People's Republic of China
| | - Ying-Long Li
- 1First Department of Lung Cancer Chemotherapy, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789, East Suzhou Street, Urumqi, 830011 Xinjiang People's Republic of China
| | - Xiang Dong
- 6Institute of Cancer Prevention and Treatment, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011 People's Republic of China
| | - Wei Zeng
- 1First Department of Lung Cancer Chemotherapy, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789, East Suzhou Street, Urumqi, 830011 Xinjiang People's Republic of China.,7Department of Hematology and Oncology, Shenzhen University General Hospital, No.1098, Xueyuan Avenue, Shenzhen, 518055 Guangdong People's Republic of China
| |
Collapse
|
56
|
Autophagy modulates temozolomide-induced cell death in alveolar Rhabdomyosarcoma cells. Cell Death Discov 2018; 4:52. [PMID: 30416757 PMCID: PMC6202374 DOI: 10.1038/s41420-018-0115-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 02/08/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is a muscle-derived tumor. In both pre-clinical and clinical studies Temozolomide (TMZ) has been recently tested against RMS; however, the precise mechanism of action of TMZ in RMS remains unclear. Here we demonstrate that TMZ decreases the cell viability of the RH30 RMS and C2C12 cell line, where cells display evidence of mitochondrial outer membrane permeability. Interestingly, the C2C12 mouse myoblast line was relatively more resistant to TMZ-induced apoptosis. Moreover, we observed that TMZ activated biochemical and morphological markers of autophagy in both cell lines. Autophagy inhibition in both RH30 and C2C12 cells significantly increased TMZ-induced cell death. In RH30 cells, TMZ increased Mcl-1 and Bax protein expression compared to corresponding time match controls while in C2C12 Mcl-1, Bcl-2, Bcl-XL, and Bax protein expression were not changed. Baf-A1 co-treatment with TMZ significantly decrease Mcl-1 expression compared to TMZ while increase Bax expression in C2C12 cells (Bcl2 and Bcl-XL do not significantly change in Baf-A1/TMZ co-treatment). Using a three-dimensional (3D) C2C12 and RH30 culture model we demonstrated that TMZ is significantly more toxic in RH30 cells (live/dead assay). Additionally, we have observed in our 3D culture model that TMZ induced both apoptosis (cleavage of PARP) and autophagy (LC3-puncta and localization of LC3/p62). Therefore, our data demonstrate that TMZ induces simultaneous autophagy and apoptosis in both RH30 and C2C12 cells in 2D and 3D culture model, where RH30 cells are more sensitive to TMZ-induced death. Furthermore, autophagy serves to protect RH30 cells from TMZ-induced death.
Collapse
|
57
|
Zhao GS, Gao ZR, Zhang Q, Tang XF, Lv YF, Zhang ZS, Zhang Y, Tan QL, Peng DB, Jiang DM, Guo QN. TSSC3 promotes autophagy via inactivating the Src-mediated PI3K/Akt/mTOR pathway to suppress tumorigenesis and metastasis in osteosarcoma, and predicts a favorable prognosis. J Exp Clin Cancer Res 2018; 37:188. [PMID: 30092789 PMCID: PMC6085607 DOI: 10.1186/s13046-018-0856-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 07/24/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Over the last two or three decades, the pace of development of treatments for osteosarcoma tends has been slow. Novel effective therapies for osteosarcoma are still lacking. Previously, we reported that tumor-suppressing STF cDNA 3 (TSSC3) functions as an imprinted tumor suppressor gene in osteosarcoma; however, the underlying mechanism by which TSSC3 suppresses the tumorigenesis and metastasis remain unclear. METHODS We investigated the dynamic expression patterns of TSSC3 and autophagy-related proteins (autophagy related 5 (ATG5) and P62) in 33 human benign bone tumors and 58 osteosarcoma tissues using immunohistochemistry. We further investigated the correlations between TSSC3 and autophagy in osteosarcoma using western blotting and transmission electronic microscopy. CCK-8, Edu, and clone formation assays; wound healing and Transwell assays; PCR; immunohistochemistry; immunofluorescence; and western blotting were used to investigated the responses in TSSC3-overexpressing osteosarcoma cell lines, and in xenografts and metastasis in vivo models, with or without autophagy deficiency caused by chloroquine or ATG5 silencing. RESULTS We found that ATG5 expression correlated positively with TSSC3 expression in human osteosarcoma tissues. We demonstrated that TSSC3 was an independent prognostic marker for overall survival in osteosarcoma, and positive ATG5 expression associated with positive TSSC3 expression suggested a favorable prognosis for patients. Then, we showed that TSSC3 overexpression enhanced autophagy via inactivating the Src-mediated PI3K/Akt/mTOR pathway in osteosarcoma. Further results suggested autophagy contributed to TSSC3-induced suppression of tumorigenesis and metastasis in osteosarcoma in vitro and in vivo models. CONCLUSIONS Our findings highlighted, for the first time, the importance of autophagy as an underlying mechanism in TSSC3-induced antitumor effects in osteosarcoma. We also revealed that TSSC3-associated positive ATG5 expression might be a potential predictor of favorable prognosis in patients with osteosarcoma.
Collapse
Affiliation(s)
- Guo-sheng Zhao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 People’s Republic of China
- Bone and Trauma Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120 People’s Republic of China
| | - Zi-ran Gao
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037 People’s Republic of China
| | - Qiao Zhang
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 People’s Republic of China
| | - Xue-feng Tang
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037 People’s Republic of China
| | - Yang-fan Lv
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037 People’s Republic of China
| | - Zhao-si Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 People’s Republic of China
| | - Yuan Zhang
- Department of Orthopaedics, Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 People’s Republic of China
| | - Qiu-lin Tan
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037 People’s Republic of China
| | - Dong-bin Peng
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037 People’s Republic of China
| | - Dian-ming Jiang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016 People’s Republic of China
- Bone and Trauma Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120 People’s Republic of China
| | - Qiao-Nan Guo
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037 People’s Republic of China
| |
Collapse
|