1
|
Guo W, Liu M, Luo W, Peng J, Liu F, Ma X, Wang L, Yang S. FERMT1 promotes epithelial-mesenchymal transition of hepatocellular carcinoma by activating EGFR/AKT/β-catenin and EGFR/ERK pathways. Transl Oncol 2024; 50:102144. [PMID: 39353234 PMCID: PMC11472111 DOI: 10.1016/j.tranon.2024.102144] [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: 10/09/2023] [Revised: 07/10/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the effects of fermitin family member 1 (FERMT1) on epithelial-mesenchymal transition (EMT) in hepatocellular carcinoma (HCC) via the EGFR/AKT/β-catenin and EGFR/ERK pathways. METHODS The expression of FERMT1 encoding protein kindlin-1 in HCC tissues was determined by immunohistochemistry, and FERMT1 mRNA expression in HCC tissues and cell lines was analyzed by qRT-PCR. After the FERMT1 expression of SNU182 and SNU387 interfered with siRNA, the cell viability, invasion, migration, and EMT were tested by CCK-8, transwell invasion, scratching, immunofluorescence/WB, respectively. Similarly, the effects of FERMT1 on the viability and metastasis of HCC were investigated in transplanted tumor and lung metastasis mouse models. The protein expressions of EGFR/AKT/β-catenin and EGFR/ERK pathways were analyzed by WB. In addition, the relationship between FERMT1 and EGFR was further determined by immunofluorescence double staining and Co-IP. RESULTS FERMT1 was significantly upregulated in HCC, and silencing FERMT1 inhibited the viability, invasion, migration, and EMT of HCC. Silencing FERMT1 also inhibited the activation of EGFR/AKT/β-catenin and EGFR/ERK pathways. In addition, inhibition of EGFR, AKT, or ERK confirmed that EGFR/AKT/β-catenin and EGFR/ERK pathways were involved in the promoting effects of FERMT1 on HCC. Co-IP and immunofluorescence experiments confirmed the targeting relationship between FERMT1 and EGFR. CONCLUSION FERMT1 was highly expressed in HCC and promoted viability, invasion, migration, and EMT of HCC by targeting EGFR to activate the EGFR/AKT/β-catenin and EGFR/ERK pathways. Our study revealed the role of FERMT1 in HCC and suggested that FERMT1 exerts biological effects through activating the EGFR/AKT/β-catenin and EGFR/ERK pathways.
Collapse
Affiliation(s)
- Wubin Guo
- Department of General Surgery, The Affifiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China; Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China; National Traditional Chinese Medicine Clinical Research Base of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China; The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China.
| | - Mengnan Liu
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China; National Traditional Chinese Medicine Clinical Research Base of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China; Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China
| | - Wei Luo
- Department of General Surgery, The Affifiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jing Peng
- Department of General Surgery, The Affifiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Fei Liu
- Department of General Surgery, The Affifiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xin Ma
- Department of General Surgery, The Affifiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Li Wang
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China; National Traditional Chinese Medicine Clinical Research Base of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China.
| | - Sijin Yang
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China; National Traditional Chinese Medicine Clinical Research Base of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Gao TM, Xiao KQ, Xiang XX, Jin SJ, Qian JJ, Zhang C, Zhou BH, Tang H, Bai DS, Jiang GQ. The decreased risk of hepatocellular carcinoma in hepatitis B virus-related cirrhotic portal hypertension patients after laparoscopic splenectomy and azygoportal disconnection. Surg Endosc 2023; 37:8522-8531. [PMID: 37775601 DOI: 10.1007/s00464-023-10454-7] [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: 05/03/2023] [Accepted: 09/06/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND Posthepatitic cirrhosis is one of the leading risk factors for hepatocellular carcinoma (HCC) worldwide, among which hepatitis B cirrhosis is the dominant one. This study explored whether laparoscopic splenectomy and azygoportal disconnection (LSD) can reduce the risk of HCC among patients with hepatitis B virus (HBV)-related cirrhotic portal hypertension (CPH). METHODS A total of 383 patients with HBV-related CPH diagnosed as gastroesophageal variceal bleeding and secondary hypersplenism were identified in our hepatobiliary pancreatic center between April 2012 and April 2022, and conducted an 11-year retrospective follow-up. We used inverse probability of treatment weighting (IPTW) to correct for potential confounders, weighted Kaplan-Meier curves, and logistic regression to estimate survival and risk differences. RESULTS Patients were divided into two groups based on treatment method: LSD (n = 230) and endoscopic therapy (ET; n = 153) groups. Whether it was processed through IPTW or not, LSD group showed a higher survival benefit than ET group according to Kaplan-Meier analysis (P < 0.001). The incidence density of HCC was higher in the ET group compared to LSD group at the end of follow-up [32.1/1000 vs 8.0/1000 person-years; Rate ratio: 3.998, 95% confidence intervals (CI) 1.928-8.293]. Additionally, in logistic regression analyses weighted by IPTW, LSD was an independent protective predictor of HCC incidence compared to ET (odds ratio 0.516, 95% CI 0.343-0.776; P = 0.002). CONCLUSION Considering the ability of LSD to improve postoperative survival and prevent HCC in HBV-related CPH patients with gastroesophageal variceal bleeding and secondary hypersplenism, it is worth promoting in the context of the shortage of liver donors.
Collapse
Affiliation(s)
- Tian-Ming Gao
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, 98 West Nantong Rd, Yangzhou, 225001, China
| | - Kun-Qing Xiao
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, 98 West Nantong Rd, Yangzhou, 225001, China
| | - Xiao-Xing Xiang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, 98 West Nantong Rd, Yangzhou, 225001, China
- Department of Digestive Diseases, Clinical Medical College, Yangzhou University, 98 West Nantong Rd, Yangzhou, 225001, China
| | - Sheng-Jie Jin
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, 98 West Nantong Rd, Yangzhou, 225001, China
| | - Jian-Jun Qian
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, 98 West Nantong Rd, Yangzhou, 225001, China
| | - Chi Zhang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, 98 West Nantong Rd, Yangzhou, 225001, China
| | - Bao-Huan Zhou
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, 98 West Nantong Rd, Yangzhou, 225001, China
| | - Hua Tang
- The Administration Office, Yangzhou Blood Center, Yangzhou, 225001, China
| | - Dou-Sheng Bai
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, 98 West Nantong Rd, Yangzhou, 225001, China.
| | - Guo-Qing Jiang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, 98 West Nantong Rd, Yangzhou, 225001, China.
| |
Collapse
|
3
|
Xu H, Cao D, Zhou D, Zhao N, Tang X, Shelat VG, Samant H, Satapathy SK, Tustumi F, Aprile G, He A, Xu X, Ge W. Baseline Albumin-Bilirubin grade as a predictor of response and outcome of regorafenib therapy in patients with hepatocellular carcinoma: a systematic review and meta-analysis. BMC Cancer 2023; 23:1006. [PMID: 37858207 PMCID: PMC10588229 DOI: 10.1186/s12885-023-11488-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND The use of regorafenib in the treatment of hepatocellular carcinoma (HCC) is widespread. Albumin-Bilirubin (ALBI) has been shown to be a potential prognostic marker for regorafenib treatment, but its prognostic value remains controversial. Therefore, we conducted a meta-analysis to investigate the value of the baseline ALBI grade in predicting the efficacy and survival outcomes of HCC patients after regorafenib treatment. METHODS PubMed, Embase, Cochrane library, Web of Science, CNKI, Wan Fang Data, and Vip Database were searched from January 2010 to October 2022. Studies treating HCC patients with regorafenib and with ALBI as a categorical variable, overall survival (OS) and progression-free survival (PFS) as outcome indicators were included. After applying Newcastle-Ottawa Scale (NOS) to evaluate the quality of the included studies, Review Manager 5.4 was used to statistically analyze. Chi-square Q test and I2 statistics were used to detect heterogeneity. Funnel plot asymmetry, Egger's and Begg's test were used to evaluate publication bias. RESULTS A total of 12 studies, comprising 1,918 patients, were included in the meta-analysis. The included studies were all evaluated as high quality. Compared to the high-grade baseline ALBI group, patients in the low-grade group had a longer survival time after receiving regorafenib and also more suitable for regorafenib treatment [odds ratio (OR) = 6.50, 95% confidence interval (CI): 2.22-18.96, P < 0.01]. The low-grade baseline ALBI group before sorafenib treatment was significantly correlated with better OS [hazard ratio (HR) = 2.36, 95% CI: 1.68-3.31, P < 0.00001] and PFS (HR = 1.56, 95% CI: 1.16-2.08, P = 0.003). Likewise, the low-grade baseline ALBI group before regorafenib was also significantly correlated with better OS (HR = 1.56, 95% CI: 1.15-2.13, P = 0.005) and PFS (HR = 2.06, 95% CI: 1.37-3.11, P = 0.0005). In addition, the ALBI grade was significantly correlated with disease control rate (DCR) (OR = 2.90, 95% CI: 1.45-5.79, P = 0.003), but not the objective response rate (OR = 1.98, 95% CI: 0.71-5.46, P = 0.19). CONCLUSIONS The baseline ALBI grade could be a valuable prognostic indicator for predicting response and outcomes in HCC patients treated with regorafenib.
Collapse
Affiliation(s)
- Huilin Xu
- Department of Oncology, The Fifth Hospital of Wuhan, #122 Xianzheng Road, Hanyang District, Wuhan, 430000, China.
| | - Dedong Cao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dingjie Zhou
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Nan Zhao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xixian Tang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | | | - Hrishikesh Samant
- Division of Hepatology, Ochsner Multi-Organ Transplant Center, New Orleans, LA, USA
| | - Sanjaya K Satapathy
- Division of Hepatology, Department of Medicine and Northwell Center for Liver Diseases & Transplantation, Northwell Health, Manhasset, NY, USA
| | - Francisco Tustumi
- Department of Gastroenterology, Digestive Surgery Division, University of São Paulo Medical School, Sao Paulo, Brazil
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, Berica, Vicenza, Italy
| | - Anbing He
- Department of Oncology, The Fifth Hospital of Wuhan, #122 Xianzheng Road, Hanyang District, Wuhan, 430000, China
| | - Ximing Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Ge
- Department of Oncology, Taikang Tongji Hospital of Wuhan, Wuhan, China
| |
Collapse
|
4
|
Jiang C, Zhao L, Xin B, Ma G, Wang X, Song S. 18F-FDG PET/CT radiomic analysis for classifying and predicting microvascular invasion in hepatocellular carcinoma and intrahepatic cholangiocarcinoma. Quant Imaging Med Surg 2022; 12:4135-4150. [PMID: 35919043 PMCID: PMC9338369 DOI: 10.21037/qims-21-1167] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 06/06/2022] [Indexed: 12/24/2022]
Abstract
Background Microvascular invasion (MVI) is a critical risk factor for early recurrence of hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). The aim of this study was to explore the contribution of 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) radiomic features for the preoperative prediction of HCC and ICC classification and MVI. Methods In this retrospective study, 127 (HCC: ICC =76:51) patients with suspected MVI accompanied by either HCC or ICC were included (In HCC group, MVI positive: negative =46:30 in ICC group, MVI positive: negative =31:20). Results-driven feature engineering workflow was used to select the most predictive feature combinations. The prediction model was based on supervised machine learning classifier. Ten-fold cross validation on training cohort and independent test cohort were constructed to ensure stability and generalization ability of models. Results For HCC and ICC classification, radiomics predictors composed of two PET and one CT feature achieved area under the curve (AUC) of 0.86 (accuracy, sensitivity, specificity was 0.82, 0.78, 0.88, respectively) on test cohort. For MVI prediction, in HCC group, our MVI prediction model achieved AUC of 0.88 (accuracy, sensitivity, specificity was 0.78, 0.88, 0.60 respectively) with three PET features associated with tumor stage on test cohort. In ICC group, the phenotype composed of two PET features and carbohydrate antigen 19-9 (CA19-9) achieved AUC of 0.90 (accuracy, sensitivity, specificity was 0.77, 0.75, 0.80, respectively). Conclusions 18F-FDG PET/CT radiomic features integrating clinical factors have potential in HCC and ICC classification and MVI prediction, while PET features have dominant predictive power in model performance. The prediction model has value in providing a non-invasive biomarker for an earlier indication and comprehensive quantification of primary liver cancers.
Collapse
Affiliation(s)
- Chunjuan Jiang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| | - Liwei Zhao
- School of Computer Science, The University of Sydney, Sydney, NSW, Australia
| | - Bowen Xin
- School of Computer Science, The University of Sydney, Sydney, NSW, Australia
| | - Guang Ma
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| | - Xiuying Wang
- School of Computer Science, The University of Sydney, Sydney, NSW, Australia
| | - Shaoli Song
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
| |
Collapse
|
5
|
Circ_0001955 Acts as a miR-646 Sponge to Promote the Proliferation, Metastasis and Angiogenesis of Hepatocellular Carcinoma. Dig Dis Sci 2022; 67:2257-2268. [PMID: 34021822 DOI: 10.1007/s10620-021-07053-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/11/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Circular RNA (circRNA) exerts a crucial role in the progression of many cancers, including hepatocellular carcinoma (HCC). However, the function of circ_0001955 in HCC progression has been poorly studied. AIMS Elucidating the role and molecular mechanism of circ_0001955 in HCC progression. METHODS Quantitative real-time PCR was employed to detect the expression of circ_0001955 and miR-646. Cell counting kit 8 assay, Ethynyl-2-deoxyuridine assay, flow cytometry, transwell assay, and tube formation assay were conducted to measure cell proliferation, metastasis, angiogenesis and apoptosis. Dual-luciferase reporter assay and biotin-labeled RNA pull-down assay were performed to analyze the interactions among circ_0001955, miR-646 and frizzled class receptor 4 (FZD4). Moreover, animal experiments were performed to examine the influence of circ_0001955 on HCC tumor growth in vivo. RESULTS Circ_0001955 was a highly expressed circRNA in HCC tissues and cells. Circ_0001955 knockdown inhibited the proliferation, metastasis, angiogenesis, and enhanced the apoptosis of HCC cells. Meanwhile, miR-646 could be sponged by circ_0001955, and its inhibitor could reverse the negative regulation of circ_0001955 knockdown on HCC progression. Further, FZD4 was a target of miR-646, and its overexpression could invert the inhibition effect of miR-646 mimic on HCC progression. Besides, our results also indicated that circ_0001955 promoted FZD4 expression by sponging miR-646. Animal experiment results showed that circ_0001955 silencing restrained HCC tumor growth in vivo. CONCLUSION Our findings suggested that circ_0001955 might play a positive role in HCC progression via regulating the miR-646/FZD4 axis, indicating that circ_0001955 might be a potential therapeutic target for HCC.
Collapse
|
6
|
Xing Y, Liu Y, Qi Z, Liu Z, Wang X, Zhang H. LAGE3 promoted cell proliferation, migration, and invasion and inhibited cell apoptosis of hepatocellular carcinoma by facilitating the JNK and ERK signaling pathway. Cell Mol Biol Lett 2021; 26:49. [PMID: 34837962 PMCID: PMC8903694 DOI: 10.1186/s11658-021-00295-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/16/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is now the second leading cause of cancer death worldwide and lacks effectual therapy due to its high rate of tumor recurrence and metastasis. The aim of this study was to investigate the effects of L antigen family member 3 (LAGE3, a member of the LAGE gene family involved in positive transcription) on the progression of HCC. METHODS The expression of LAGE3 was detected by quantitative real-time polymerase chain reaction. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, colony formation assay, EdU, and cell cycle analysis assay were employed to evaluate the proliferation of HCC cells. Annexin V-FITC/PI and TUNEL assay were used to assess the apoptosis rate of HCC cells. Wound healing and transwell assay were used to investigate the migration and invasion of HCC cells. A xenograft model of HCC was established with 2 × 106 Hep3B or SK-HEP1 cells to investigate the in vivo effects of LAGE3. Then, the protein levels of LAGE3, p-p38, p-38, c-Jun N-terminal kinase (JNK),p-JNK, extracellular signal-regulated kinase (ERK), and p-ERK were detected by western blot. RESULTS We found that LAGE3 was upregulated in HCC tissues compared to adjacent tissues, and its high expression was correlated with poor overall survival by bioinformatics analysis. Next, we manually regulated the expression of LAGE3 in HCC cells. The knockdown of LAGE3 inhibited the proliferation of HCC cells by arresting the cell cycle in G1 phase. Also the downregulation of LAGE3 inhibited cell migration and invasion and induced apoptosis of HCC cells, while overexpression of LAGE3 promoted the malignant phenotypes of HCC. These results were further confirmed by the in vivo growth of HCC xenografts and the inhibition of apoptosis of HCC tumor cells. Furthermore, we found that LAGE3 exerted cancer-promoting effects by potentiating the JNK and ERK signaling pathway. An ERK inhibitor (10 μM SCH772984) or JNK inhibitor (25 μM SP600125) repressed the upregulated LAGE3-induced proliferation, migration, and invasion of HCC cells. CONCLUSIONS LAGE3 enhanced the malignant phenotypes of HCC by promoting the JNK and ERK signaling pathway.
Collapse
Affiliation(s)
- Ying Xing
- Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, No. 119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
| | - Yang Liu
- Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, No. 119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
| | - Zhong Qi
- Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, No. 119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
| | - Zhengrong Liu
- Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, No. 119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
| | - Xin Wang
- Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, No. 119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China
| | - Hongyi Zhang
- Department of General Surgery, Beijing Tiantan Hospital, Capital Medical University, No. 119, South 4th Ring West Road, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
7
|
Quantification of contrast agent uptake in the hepatobiliary phase helps to differentiate hepatocellular carcinoma grade. Sci Rep 2021; 11:22991. [PMID: 34837039 PMCID: PMC8626433 DOI: 10.1038/s41598-021-02499-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/10/2021] [Indexed: 12/02/2022] Open
Abstract
This study aimed to assess the degree of differentiation of hepatocellular carcinoma (HCC) using Gd-EOB-DTPA-assisted magnetic resonance imaging (MRI) with T1 relaxometry. Thirty-three solitary HCC lesions were included in this retrospective study. This study's inclusion criteria were preoperative Gd-EOB-DTPA-assisted MRI of the liver and a histopathological evaluation after hepatic tumor resection. T1 maps of the liver were evaluated to determine the T1 relaxation time and reduction rate between the native phase and hepatobiliary phase (HBP) in liver lesions. These findings were correlated with the histopathologically determined degree of HCC differentiation (G1, well-differentiated; G2, moderately differentiated; G3, poorly differentiated). There was no significant difference between well-differentiated (950.2 ± 140.2 ms) and moderately/poorly differentiated (1009.4 ± 202.0 ms) HCCs in the native T1 maps. After contrast medium administration, a significant difference (p ≤ 0.001) in the mean T1 relaxation time in the HBP was found between well-differentiated (555.4 ± 140.2 ms) and moderately/poorly differentiated (750.9 ± 146.4 ms) HCCs. For well-differentiated HCCs, the reduction rate in the T1 time was significantly higher at 0.40 ± 0.15 than for moderately/poorly differentiated HCCs (0.25 ± 0.07; p = 0.006). In conclusion this study suggests that the uptake of Gd-EOB-DTPA in HCCs is correlated with tumor grade. Thus, Gd-EOB-DTPA-assisted T1 relaxometry can help to further differentiation of HCC.
Collapse
|
8
|
Philips CA, Rajesh S, Nair DC, Ahamed R, Abduljaleel JK, Augustine P. Hepatocellular Carcinoma in 2021: An Exhaustive Update. Cureus 2021; 13:e19274. [PMID: 34754704 PMCID: PMC8569837 DOI: 10.7759/cureus.19274] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2021] [Indexed: 02/06/2023] Open
Abstract
Primary liver cancer is a challenging global health concern with an estimated more than a million persons to be affected annually by the year 2025. The commonest type is hepatocellular carcinoma (HCC), which has been increasing in incidence the world over, mostly due to chronic viral hepatitis B infection. In the last decade, paradigm changes in the etiology, understanding of molecular biology, and pathogenesis, including the role of gut microbiota; medical and surgical treatments, and outcome trends are notable. The application of omics-based technology has helped us unlock the molecular and immune landscape of HCC, through which novel targets for drug treatment such as immune-checkpoint inhibitors have been identified. Novel tools for the surveillance and diagnosis of HCC include protein-, genomics-, and composite algorithm-based clinical/biomarker panels. Magnetic resonance imaging-based novel techniques have improved HCC diagnosis through ancillary features that enhance classical criteria while positron emission tomography has shown value in prognostication. Identification of the role of gut microbiota in the causation and progression of HCC has opened areas for novel therapeutic research. A select group of patients still benefit from modified surgical and early interventional radiology treatments. Improvements in radiotherapy protocols, identification of parameters of futility among radiological interventions, and the emergence of novel first-line systemic therapies that include a combination of antiangiogenic and immune-checkpoint inhibitors have seen a paradigm change in progression-free and overall survival. The current review is aimed at providing exhaustive updates on the etiology, molecular biology, biomarker diagnosis, imaging, and recommended treatment options in patients with HCC.
Collapse
Affiliation(s)
- Cyriac A Philips
- Clinical and Translational Hepatology, The Liver Institute, Center of Excellence in GI Sciences, Rajagiri Hospital, Aluva, IND
| | - Sasidharan Rajesh
- Interventional Hepatobiliary Radiology, Center of Excellence in GI Sciences, Rajagiri Hospital, Aluva, IND
| | - Dinu C Nair
- Interventional Hepatobiliary Radiology, Center of Excellence in GI Sciences, Rajagiri Hospital, Aluva, IND
| | - Rizwan Ahamed
- Gastroenterology and Advanced Gastrointestinal (GI) Endoscopy, Center of Excellence in GI Sciences, Rajagiri Hospital, Aluva, IND
| | - Jinsha K Abduljaleel
- Gastroenterology and Advanced Gastrointestinal (GI) Endoscopy, Center of Excellence in GI Sciences, Rajagiri Hospital, Aluva, IND
| | - Philip Augustine
- Gastroenterology and Advanced Gastrointestinal (GI) Endoscopy, Center of Excellence in GI Sciences, Rajagiri Hospital, Aluva, IND
| |
Collapse
|
9
|
Uvangoletin, extracted from Sarcandra glabra, exerts anticancer activity by inducing autophagy and apoptosis and inhibiting invasion and migration on hepatocellular carcinoma cells. PHYTOMEDICINE 2021; 94:153793. [PMID: 34736000 DOI: 10.1016/j.phymed.2021.153793] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/13/2021] [Accepted: 10/03/2021] [Indexed: 02/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Uvangoletin is a dihydrochalcone extracted from the traditional Chinese medicinal plant Sarcandra glabra. Previous research has showed that uvangoletin could induce leukemia cell death. However, the anticancer effect of uvangoletin on hepatocellular carcinoma (HCC) has not been clarified. AIM OF THE STUDY This study aimed to investigate the anti-cancer effects of uvangoletin on HCC and to explore its underlying mechanisms. MATERIALS AND METHODS We measured the anticancer activities of uvangoletin both in vitro and in vivo by MTT assay and HepG2 xenograft model. The effects of uvangoletin on apoptosis, autophagy, migration and invasion were also determined. Apoptosis was evaluated by flow cytometry method. Autophagy was assessed by immunofluorescence assay. Cell migration and invasion ability were validated by wound healing assay and cultrex® 96 well cell migration/invasion assay. The expression level of relevant proteins and pathways were examined by western blot. RESULTS The results of MTT assay and HepG2 xenograft model showed that uvangoletin could inhibit HCC cells proliferation in vitro and in vivo. Uvangoletin could induce HepG2 cell apoptosis as evidence by the increased expression of cleaved caspase 3, caspase 8 and Bax while decreased Bcl-2 expression. Wound healing assay and transwell assay showed that uvangoletin inhibited HepG2 cells migration and invasion and reduced vimentin, MMP9, MMP2 expression. Uvangoletin also promoted autophagy in HepG2 cells as confirmed by the accumulation of GFP-LC3 puncta. Autophagy inhibitors like 3-MA or CQ could suppress uvangoletin-induced apoptosis. Importantly, uvangoletin-induced anti-EMT effect was also attenuated after autophagy inhibitors added in. Mechanistically, the expressions of p-JNK, p-ERK, p-p38, p-AKT, p-p70S6k and p-mTOR were significantly decreased after uvangoletin treatment. CONCLUSION Our results showed that uvangoletin could induce apoptotic and autophagic cell death, inhibit cell proliferation and metastasis on HepG2 cells through Akt/mTOR, MAPK and TGFβ/Smad2 signal pathways.
Collapse
|
10
|
Fan X, Wu H, Zhao L, Guo X. A Poly-Chitosan and Cis-Platinum Conjugated Composite Nanoparticle System for Liver Cancer Therapy. J Biomed Nanotechnol 2021; 17:1726-1734. [PMID: 34688317 DOI: 10.1166/jbn.2021.3157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The aim of this study was to test an effective nano-pole capsule loaded cis-platinum (CP) transplantation device for liver cancer (LC) therapy. A novel nano-pole capsule was designed as a new vector for storing CP. HepG2 cells and a B6/J mouse model were used to test the efficiency of polyethyleneimine-cis-platinum (PEI-CP) and poly-chitosan-cis-platinum (PC-CP). Infiltration efficiency and transplantation efficiency tests were performed to study the performance of the delivery system, and fibroblast reactions and macrophage numbers were observed, to test for immune rejection and foreign body reactions. The apoptosis rate and tumor diameter of hepatocellular carcinoma cells were used to evaluate the effect of the tumor therapy. We also studied the functional mechanism of different CP delivery systems. The infiltration and transplantation efficiencies of PC-CP were higher than that of PEI-CP; Less foreign body reaction appeared in PC system, with less fibroblast reaction and lower macrophage reaction. The clinical efficacy of PC-CP in terms of tumor apoptosis and diameter reduction was superior to that of PEI-CP. We demonstrated that PC-CP had a more significant alteration effect on mTOR, P-Ak, LC3 and P53. The PC system can better deliver and release drugs than PEI-CP, and may be a better choice for LC therapy in the future.
Collapse
Affiliation(s)
- Xiangyu Fan
- Department of Radiation Oncology, The Fourth Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Haiyun Wu
- Department of Medical Imaging, The Fourth Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Lisong Zhao
- Department of Radiation Oncology, The Fourth Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| | - Xu Guo
- Department of Radiation Oncology, The Fourth Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, PR China
| |
Collapse
|
11
|
Huang G, Wang C, Fu X. Bidirectional deep neural networks to integrate RNA and DNA data for predicting outcome for patients with hepatocellular carcinoma. Future Oncol 2021; 17:4481-4495. [PMID: 34374301 DOI: 10.2217/fon-2021-0659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aims: Individualized patient profiling is instrumental for personalized management in hepatocellular carcinoma (HCC). This study built a model based on bidirectional deep neural networks (BiDNNs), an unsupervised machine-learning approach, to integrate multi-omics data and predict survival in HCC. Methods: DNA methylation and mRNA expression data for HCC samples from the TCGA database were integrated using BiDNNs. With optimal clusters as labels, a support vector machine model was developed to predict survival. Results: Using the BiDNN-based model, samples were clustered into two survival subgroups. The survival subgroup classification was an independent prognostic factor. BiDNNs were superior to multimodal autoencoders. Conclusion: This study constructed and validated a BiDNN-based model for predicting prognosis in HCC, with implications for individualized therapies in HCC.
Collapse
Affiliation(s)
- Guojun Huang
- Department of Oncology, Pidu District People's Hospital, Chengdu, Sichuan, China
| | - Cheng Wang
- Department of General Surgery, Pidu District People's Hospital, Chengdu, Sichuan, China
| | - Xi Fu
- Department of Oncology, Pidu District People's Hospital, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Hendricks-Wenger A, Hutchison R, Vlaisavljevich E, Allen IC. Immunological Effects of Histotripsy for Cancer Therapy. Front Oncol 2021; 11:681629. [PMID: 34136405 PMCID: PMC8200675 DOI: 10.3389/fonc.2021.681629] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/10/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is the second leading cause of death worldwide despite major advancements in diagnosis and therapy over the past century. One of the most debilitating aspects of cancer is the burden brought on by metastatic disease. Therefore, an ideal treatment protocol would address not only debulking larger primary tumors but also circulating tumor cells and distant metastases. To address this need, the use of immune modulating therapies has become a pillar in the oncology armamentarium. A therapeutic option that has recently emerged is the use of focal ablation therapies that can destroy a tumor through various physical or mechanical mechanisms and release a cellular lysate with the potential to stimulate an immune response. Histotripsy is a non-invasive, non-ionizing, non-thermal, ultrasound guided ablation technology that has shown promise over the past decade as a debulking therapy. As histotripsy therapies have developed, the full picture of the accompanying immune response has revealed a wide range of immunogenic mechanisms that include DAMP and anti-tumor mediator release, changes in local cellular immune populations, development of a systemic immune response, and therapeutic synergism with the inclusion of checkpoint inhibitor therapies. These studies also suggest that there is an immune effect from histotripsy therapies across multiple murine tumor types that may be reproducible. Overall, the effects of histotripsy on tumors show a positive effect on immunomodulation.
Collapse
Affiliation(s)
- Alissa Hendricks-Wenger
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, United States
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
| | - Ruby Hutchison
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
| | - Eli Vlaisavljevich
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, United States
- Institute for Critical Technology and Applied Sciences Center for Engineered Health, Virginia Tech, Blacksburg, VA, United States
| | - Irving Coy Allen
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Roanoke, VA, United States
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, United States
- Institute for Critical Technology and Applied Sciences Center for Engineered Health, Virginia Tech, Blacksburg, VA, United States
- Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| |
Collapse
|
13
|
Shao M, Shi R, Gao ZX, Gao SS, Li JF, Li H, Cui SZ, Hu WM, Chen TY, Wu GR, Zhang J, Xu J, Sy MS, Li C. Crizotinib and Doxorubicin Cooperatively Reduces Drug Resistance by Mitigating MDR1 to Increase Hepatocellular Carcinoma Cells Death. Front Oncol 2021; 11:650052. [PMID: 34094940 PMCID: PMC8170002 DOI: 10.3389/fonc.2021.650052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/16/2021] [Indexed: 11/24/2022] Open
Abstract
As the sixth most lethal cancers worldwide, hepatocellular carcinoma (HCC) has been treated with doxorubicin (Dox) for decades. However, chemotherapy resistance, especially for Dox is an even more prominent problem due to its high cardiotoxicity. To find a regimen to reduce Dox resistance, and identify the mechanisms behind it, we tried to identify combination of drugs that can overcome drug resistance by screening tyrosine kinase inhibitor(s) with Dox with various HCC cell lines in vitro and in vivo. We report here that combination of Crizo and Dox has a synergistic effect on inducing HCC cell death. Accordingly, Crizo plus Dox increases Dox accumulation in nucleus 3-16 times compared to Dox only; HCC cell death enhanced at least 50% in vitro and tumor weights reduced ranging from 35 to 65%. Combining these two drugs reduces multiple drug resistance 1 (MDR1) protein as a result of activation of protein kinase RNA-like endoplasmic reticulum kinase (PERK), which phosphorylates eIF2α, leading to protein translational repression. Additionally, PERK stimulation activates C-Jun terminal kinase (JNK), resulting in accumulation of unfused autophagosome to enhance autophagic cell death via Poly-ADP-ribosyltransferase (PARP-1) cleavage. When the activity of PERK or JNK is blocked, unfused autophagosome is diminished, cleaved PARP-1 is reduced, and cell death is abated. Therefore, Crizo plus Dox sensitize HCC drug resistance by engaging PERK-p- eIF2α-MDR1, and kill HCC cells by engaging PERK-JNK- autophagic cell death pathways. These newly discovered mechanisms of Crizo plus Dox not only provide a potential treatment for HCC but also point to an approach to overcome MDR1 related drug resistance in other cancers.
Collapse
Affiliation(s)
- Ming Shao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Run Shi
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhen-Xing Gao
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Shan-Shan Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jing-Feng Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Huan Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Shu-Zhong Cui
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.,Abdominal Surgery, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Wei-Min Hu
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Tian-Yun Chen
- Department of Stomatology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gui-Ru Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Jie Zhang
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Chaoyang Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
14
|
Kim SY, Park C, Kim MY, Ji SY, Hwangbo H, Lee H, Hong SH, Han MH, Jeong JW, Kim GY, Son CG, Cheong J, Choi YH. ROS-Mediated Anti-Tumor Effect of Coptidis Rhizoma against Human Hepatocellular Carcinoma Hep3B Cells and Xenografts. Int J Mol Sci 2021; 22:4797. [PMID: 33946527 PMCID: PMC8124566 DOI: 10.3390/ijms22094797] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/22/2021] [Accepted: 04/27/2021] [Indexed: 12/24/2022] Open
Abstract
Coptidis Rhizoma is the dried rhizome from the Coptis chinensis Franch. that has been shown to have a number of beneficial pharmacological properties including antioxidant, anti-inflammatory, and anti-cancer effects. However, the anti-cancer effects of Coptidis Rhizoma on hepatocellular carcinoma (HCC) remain unclear. In this study, we investigated the anti-cancer properties of Coptidis Rhizoma ethanol extract (CR) in HCC Hep3B cells and in a xenograft mouse model. Our results showed that the CR significantly inhibited cell growth and induced apoptosis in Hep3B cells through increased expression of Bcl-2 associated x-protein (Bax) and cleavage of poly-ADP ribose polymerase (PARP), reduced expression of Bcl-2, and activated caspases. CR also increased the generation of intracellular reactive oxygen species (ROS), which caused a loss of mitochondrial membrane potential (MMP, ΔΨm) and activation of the mitochondria-mediated intrinsic apoptosis pathway. Moreover, N-acetylcysteine (NAC), a ROS inhibitor, markedly blocked the effects of CR on apoptotic pathways. CR also induced the expression of light chain 3 (LC3)-I/II, a key autophagy regulator, whereas CR-mediated autophagy was significantly suppressed by NAC. In addition, pre-treatment with NAC perfectly attenuated the inhibition of cell invasion and migration of CR-stimulated Hep3B cells. Furthermore, oral administration of CR suppressed Hep3B tumor growth in xenograft mice without toxicity, alterations to body weight, or changes in hematological and biochemical profiles. Taken together, our findings suggest that CR has anti-tumor effects that result from ROS generation, and may be a potential pharmacological intervention for HCC.
Collapse
Affiliation(s)
- So Young Kim
- Anti-Aging Research Center, Dongeui University, Busan 47340, Korea; (S.Y.K.); (M.Y.K.); (S.Y.J.); (H.H.); (H.L.)
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Korea;
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea
| | - Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong-Eui University, Busan 47340, Korea;
| | - Min Yeong Kim
- Anti-Aging Research Center, Dongeui University, Busan 47340, Korea; (S.Y.K.); (M.Y.K.); (S.Y.J.); (H.H.); (H.L.)
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Korea;
| | - Seon Yeong Ji
- Anti-Aging Research Center, Dongeui University, Busan 47340, Korea; (S.Y.K.); (M.Y.K.); (S.Y.J.); (H.H.); (H.L.)
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Korea;
| | - Hyun Hwangbo
- Anti-Aging Research Center, Dongeui University, Busan 47340, Korea; (S.Y.K.); (M.Y.K.); (S.Y.J.); (H.H.); (H.L.)
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Korea;
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dongeui University, Busan 47340, Korea; (S.Y.K.); (M.Y.K.); (S.Y.J.); (H.H.); (H.L.)
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Korea;
| | - Su Hyun Hong
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Korea;
| | - Min Ho Han
- National Marine Biodiversity Institute of Korea, Seocheon 33662, Korea;
| | - Jin-Woo Jeong
- Nakdonggang National Institute of Biological Resources, Sangju 17104, Korea;
| | - Gi-Young Kim
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea;
| | - Chang-Gue Son
- Institute of Bioscience & Integrative Medicine, Daejeon University, 176 split 75 Daedeokdae-ro Seo-gu, Daejeon 35235, Korea;
| | - JaeHun Cheong
- Department of Molecular Biology, Pusan National University, Busan 46241, Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dongeui University, Busan 47340, Korea; (S.Y.K.); (M.Y.K.); (S.Y.J.); (H.H.); (H.L.)
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Korea;
| |
Collapse
|
15
|
A dual immune signature of CD8+ T cells and MMP9 improves the survival of patients with hepatocellular carcinoma. Biosci Rep 2021; 41:228011. [PMID: 33656546 PMCID: PMC7969702 DOI: 10.1042/bsr20204219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/20/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
The 5-year survival of hepatocellular carcinoma (HCC) is difficult due to the high recurrence rate and metastasis. Tumor infiltrating immune cells (TICs) and immune-related genes (IRGs) bring hope to improve survival and treatment of HCC patients. However, there are problems in predicting immune signatures and identifying novel therapeutic targets. In the study, the CIBERSORT algorithm was used to evaluate 22 immune cell infiltration patterns in gene expression omnibus (GEO) and the cancer genome atlas (TCGA) data. Eight immune cells were found to have significant infiltration differences between the tumor and normal groups. The CD8+ T cells immune signature was constructed by least absolute shrinkage and selection operator (LASSO) algorithm. The high infiltration level of CD8+ T cells could significantly improve survival of patients. The weighted gene co-expression network analysis (WGCNA) algorithm identified MMP9 was closely related to the overall survival of HCC patients. K-M survival and tROC analysis confirmed that MMP9 had an excellent prognostic prediction. Cox regression showed that a dual immune signature of CD8+ T cells and MMP9 was independent survival factor in HCC. Therefore, a dual prognostic immune signature could improve the survival of patient and may provide a new strategy for the immunotherapy of HCC.
Collapse
|
16
|
Li M, Shang H, Wang T, Yang SQ, Li L. Huanglian decoction suppresses the growth of hepatocellular carcinoma cells by reducing CCNB1 expression. World J Gastroenterol 2021; 27:939-958. [PMID: 33776365 PMCID: PMC7968131 DOI: 10.3748/wjg.v27.i10.939] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/03/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most prevalent cancers in human populations worldwide. Huanglian decoction is one of the most important Chinese medicine formulas, with the potential to treat cancer.
AIM To investigate the role and mechanism of Huanglian decoction on HCC cells.
METHODS To identify differentially expressed genes (DEGs), we downloaded gene expression profile data from The Cancer Genome Atlas Liver Hepatocellular Carcinoma and Gene Expression Omnibus (GSE45436) databases. We obtained phytochemicals of the four herbs of Huanglian decoction from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform. We also established a regulatory network of DEGs and drug target genes and subsequently analyzed key genes using bioinformatics approaches. Furthermore, we conducted in vitro experiments to explore the effect of Huanglian decoction and to verify the predictions. In particular, the CCNB1 gene was knocked down to verify the primary target of this decoction. Through the identification of the expression levels of key proteins, we determined the primary mechanism of Huanglian decoction in HCC.
RESULTS Based on the results of the network pharmacological analysis, we revealed 5 bioactive compounds in Huanglian decoction that act on HCC. In addition, a protein-protein interaction network analysis of the target genes of these five compounds as well as expression and prognosis analyses were performed in tumors. CCNB1 was confirmed to be the primary gene that may be highly expressed in tumors and was significantly associated with a worse prognosis. We also noted that CCNB1 may serve as an independent prognostic indicator in HCC. Moreover, in vitro experiments demonstrated that Huanglian decoction significantly inhibited the growth, migration, and invasiveness of HCC cells and induced cell apoptosis and G2/M phase arrest. Further analysis showed that the decoction may inhibit the growth of HCC cells by downregulating the CCNB1 expression level. After Huanglian decoction treatment, the expression levels of Bax, caspase 3, caspase 9, p21 and p53 in HCC cells were increased, while the expression of CDK1 and CCNB1 was significantly decreased. The p53 signaling pathway was also found to play an important role in this process.
CONCLUSION Huanglian decoction has a significant inhibitory effect on HCC cells. CCNB1 is a potential therapeutic target in HCC. Further analysis showed that Huanglian decoction can inhibit HCC cell growth by downregulating the expression of CCNB1 to activate the p53 signaling pathway.
Collapse
Affiliation(s)
- Min Li
- Department of Gastroenterology, Zibo Central Hospital, Zibo 255036, Shandong Province, China
| | - Hua Shang
- Department of Gastroenterology, Zibo Central Hospital, Zibo 255036, Shandong Province, China
| | - Tao Wang
- Department of General Surgery, Zibo Central Hospital, Zibo 255036, Shandong Province, China
| | - Shui-Qing Yang
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Lei Li
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
- Department of Pathology, University of Otago, Dunedin px806, New Zealand
| |
Collapse
|
17
|
Liu Y, Zhao Q, Xu F, Wang K, Zhao Y, Chen H, He W, Wang W, Zhang J, Zhang J. Dysregulation of phosphoproteins in hepatocellular carcinoma revealed via quantitative analysis of the phosphoproteome. Oncol Lett 2020; 21:117. [PMID: 33408763 PMCID: PMC7779902 DOI: 10.3892/ol.2020.12378] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/26/2020] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequently diagnosed types of cancer in the world. Post-translational modifications, such as phosphorylation, serve an essential role during cancer development. To identify aberrant phosphorylation in HCC, a multiplexed tandem mass tag approach combined with liquid chromatography tandem-mass spectrometry was used in the present study. The results are available via ProteomeXchange (identifier no. PXD013934). A total of 4,780 phosphorylated sites distributed on 2,209 proteins were identified and quantified, including 74 and 459 phosphorylated upregulated and downregulated proteins, respectively. Bioinformatic analysis revealed differences and similarities between HCC and normal tissues. Gene Ontology enrichment analysis provided information on biological processes, molecular functions, cellular components and sub-cellular localizations. Protein domains enrichment of differentially expressed proteins was analyzed using InterPro database. Kyoto Encyclopedia of Genes and Genomes enrichment analysis revealed pathways that may potentially be involved in HCC. Integrative analysis of the functions, pathways, motifs of phosphorylated peptides, protein domains and protein interactions established a profile of the phosphoproteome of HCC, which may contribute to identify novel biomarkers for the diagnosis and prognosis of HCC, as well as novel therapeutic targets for HCC treatment.
Collapse
Affiliation(s)
- Yixian Liu
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qianwei Zhao
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Fang Xu
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Kaijuan Wang
- Henan Key Laboratory for Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ying Zhao
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Huiping Chen
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Wei He
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Weidong Wang
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jianying Zhang
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China.,Henan Key Laboratory for Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jintao Zhang
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China.,Henan Key Laboratory for Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
18
|
Kim SY, Hwangbo H, Kim MY, Ji SY, Lee H, Kim GY, Kwon CY, Leem SH, Hong SH, Cheong J, Choi YH. Coptisine induces autophagic cell death through down-regulation of PI3K/Akt/mTOR signaling pathway and up-regulation of ROS-mediated mitochondrial dysfunction in hepatocellular carcinoma Hep3B cells. Arch Biochem Biophys 2020; 697:108688. [PMID: 33227289 DOI: 10.1016/j.abb.2020.108688] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/06/2020] [Accepted: 11/15/2020] [Indexed: 02/07/2023]
Abstract
Coptisine is isoquinoline alkaloid derived from Coptidis Rhizoma and is known to have potential anti-cancer activity toward various carcinomas. Targeting autophagy is one of the main approaches for cancer therapy, but whether the anti-cancer efficacy of coptisine involves autophagy is still unclear. Therefore, this study investigated the effect of coptisine on autophagy in hepatocellular carcinoma (HCC) Hep3B cells, and identified the underlying mechanism. Our results showed that coptisine increased cytotoxicity and autophagic vacuoles in a concentration-dependent manner. Furthermore, the expressions of light chain 3 (LC3)-I/II, Beclin-1 and autophagy genes were markedly increased by coptisine, while the expression of p62 decreased. In addition, we found that pretreatment with bafilomycin A1, an inhibitor of autophagosome-lysosome fusion, markedly reduced coptisine-mediated autophagic cell death, but 3-methyladenine, an inhibitor for autophagosome formation did not. Moreover, our results showed that although coptisine up-regulated AMP-activated protein kinase (AMPK) that partially induced LC3-I/II, coptisine-mediated AMPK signaling did not directly regulate autophagic cell death. Additionally, we found that coptisine suppressed the phosphorylation of phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR), and this effect was notably enhanced by PI3K inhibitor LY294002. Meanwhile, coptisine significantly increased both the production of mitochondrial reactive oxygen species (ROS) and the recruitment of mitophagy-regulated proteins to mitochondria. Furthermore, N-acetylcysteine, a potential ROS scavenger, substantially suppressed the expression of mitophagy-regulated proteins and LC3 puncta by coptisine. Overall, our results demonstrate that coptisine-mediated autophagic cell death was regulated by PI3K/Akt/mTOR signaling and mitochondrial ROS production associated with mitochondrial dysfunction. Taken together, these findings suggest that coptisine exerts its anti-cancer effects through induction of autophagy in HCC Hep3B cells.
Collapse
Affiliation(s)
- So Young Kim
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea; Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, 46241, Republic of Korea
| | - Hyun Hwangbo
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea; Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, 46241, Republic of Korea
| | - Min Yeong Kim
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea
| | - Seon Yeong Ji
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea
| | - Hyesook Lee
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju, 63243, Republic of Korea
| | - Chan-Young Kwon
- Department of Oriental Neuropsychiatry, Dong-eui University College of Korean Medicine, Busan, 47227, Republic of Korea
| | - Sun-Hee Leem
- Department of Biological Science, College of Natural Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Su Hyun Hong
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea
| | - JaeHun Cheong
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, 46241, Republic of Korea.
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-eui University College of Korean Medicine and Anti-Aging Research Center, Dong-eui University, Busan, 47227, Republic of Korea.
| |
Collapse
|
19
|
Yang G, Zhou L, Xu Q, Meng F, Wan Y, Meng X, Wang L, Zhang L. LncRNA KCNQ1OT1 inhibits the radiosensitivity and promotes the tumorigenesis of hepatocellular carcinoma via the miR-146a-5p/ACER3 axis. Cell Cycle 2020; 19:2519-2529. [PMID: 32936716 PMCID: PMC7553536 DOI: 10.1080/15384101.2020.1809259] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death, and radiotherapy is currently one of the main treatments. Long non-coding RNAs (lncRNAs) are associated with the radiosensitivity and tumorigenesis of HCC. However, the role and molecular mechanism of potassium voltage-gated channel subfamily Q member 1 overlapping transcript 1 (KCNQ1OT1) in HCC are still unclear. The relative expression of KCNQ1OT1, microRNA-146a-5p (miR-146a-5p) and alkaline ceramidase 3 (ACER3) was quantified by quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation was measured by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT) assay. Clonogenic assay was used to assess the radiosensitivity of cells. Cell apoptosis and metastasis were evaluated by flow cytometry and transwell assays, respectively. The protein levels of apoptosis markers, metastasis markers and ACER3 were detected by western blot (WB) analysis. The relationship between miR-146a-5p and KCNQ1OT1 or ACER3 was determined by dual-luciferase reporter assay. Additionally, animal experiments were carried out to explore the effect of KCNQ1OT1 silencing on HCC tumor growth in vivo. KCNQ1OT1 was highly expressed in HCC, and its knockdown hindered the proliferation and metastasis, while increased the radiosensitivity and apoptosis of HCC cells. MiR-146a-5p could interact with KCNQ1OT1, and its inhibition reversed the effects of silenced-KCNQ1OT1 on the radiosensitivity and tumorigenesis of HCC cells. Besides, ACER3 was a target of miR-146a-5p, and its overexpression inversed the effects of miR-146a-5p mimic on the radiosensitivity and tumorigenesis of HCC cells. The expression of ACER3 was regulated by KCNQ1OT1 and miR-146a-5p. Furthermore, KCNQ1OT1 also could reduce the growth of HCC by regulating the miR-146a-5p/ACER3 axis in vivo. Our study suggested that KCNQ1OT1 improved ACER3 expression to regulate the radiosensitivity and tumorigenesis of HCC through sponging miR-146a-5p, indicating that KCNQ1OT1 might be a new therapeutic target for HCC.
Collapse
Affiliation(s)
- Ganghua Yang
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lijing Zhou
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Qinhong Xu
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Fandi Meng
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yong Wan
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiankui Meng
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lin Wang
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China,CONTACT Lin Wang ; Lei Zhang
| | - Lei Zhang
- Department of Geriatric Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
20
|
Wang Y, Dou L, Qin Y, Yang H, Yan P. OIP5-AS1 contributes to tumorigenesis in hepatocellular carcinoma by miR-300/YY1-activated WNT pathway. Cancer Cell Int 2020; 20:440. [PMID: 32943988 PMCID: PMC7487829 DOI: 10.1186/s12935-020-01467-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 07/31/2020] [Indexed: 12/20/2022] Open
Abstract
Background It has reported that long non-coding RNAs (lncRNAs) exerted regulatory functions by targeting specific genes through a competing endogenous RNA (ceRNA) pathway. LncRNA OIP5-AS1 has been identified as a tumor-enhancer in several tumor types. Nonetheless, its molecular mechanism in HCC remains to be masked. Aim of the study This study was aimed at exploring whether and how OIP5-AS1 exert functions in HCC. Methods qRT-PCR and western blot were employed for detecting gene expression. CCK-8, colony formation and EdU assays were implemented to evaluate the proliferative ability of HCC cells. Caspase-3 activity and flow cytometry analyses were implemented to determine cell apoptosis and cell cycle distribution. RNA pull down, ChIP, RIP and luciferase reporter assays explored the interplays between molecules. Results YY1 was upregulated in HCC cells, and silenced YY1 restrained HCC cell proliferation in vitro and hampered tumor growth in vivo. Later, we discovered that miR-300 could regulate WNT pathway via targeting YY1. Furthermore, OIP5-AS1 was identified as the sponge of miR-300 and promoted cell growth in HCC. Importantly, YY1 transcriptionally activate OIP5-AS1 in turn. Rescue experiments indicated that miR-300 inhibition or YY1 overexpression abrogated the inhibitive effect of OIP5-AS1 silencing on the malignant growth of HCC cells. Conclusions OIP5-AS1/miR-300/YY1 feedback loop facilitates cell growth in HCC by activating WNT pathway.
Collapse
Affiliation(s)
- Yu Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China.,Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, 430030 Hubei China.,Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030 Hubei China
| | - Lei Dou
- Department of Geratology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Yun Qin
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030 Hubei China
| | - Huiyuan Yang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Peng Yan
- Department of Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| |
Collapse
|
21
|
Li L, Liu J, Gao G, Zhang K, Song Y, Li H. Puerarin 6″-O-xyloside suppressed HCC via regulating proliferation, stemness, and apoptosis with inhibited PI3K/AKT/mTOR. Cancer Med 2020; 9:6399-6410. [PMID: 32691991 PMCID: PMC7476825 DOI: 10.1002/cam4.3285] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/05/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
Puerarin 6″-O-xyloside is a tumor suppressive derivate of Puerarin that is recently characterized as a lysine-specific demethylase 6B inhibitor. Here we investigated the effects of Puerarin 6″-O-xyloside in hepatocellular carcinoma (HCC) cell lines SMMC-7721 and HepG2. Cell viability, proliferation, stemness, protein expression, and autophagy were tested by CCK-8, colony formation, sphere formation, western blotting, and LC3B GFP puncta per cell, respectively. Apoptosis, CD133-positive cells, and JC-1-labeled mitochondrial membrane potential were measured by flow cytometry. The effects of Puerarin 6″-O-xyloside in vivo were explored in HepG2 xenograft mice. Puerarin 6″-O-xyloside inhibited cell viability, proliferation, and stemness, and promoted apoptosis in both SMMC-7721 and HepG2 cells. Further experiments showed promoted autophagy and decreased mitochondrial membrane potential, and decreased expression of p-PI3K, p-AKT, and p-mTOR in HepG2 cells. Co-administration of 3-MA with Puerarin 6″-O-xyloside obviously augmented these effects including inhibited protein expression of p-PI3K, p-AKT, and p-mTOR, and inhibited proliferation, promoted apoptosis, and decreased stemness. In HepG2 xenograft mice, 100 mg/kg/d Puerarin 6″-O-xyloside significantly suppressed tumor growth, stemness, and apoptosis. In conclusion, our study indicated that Puerarin 6″-O-xyloside decreased cell viability, proliferation, and stemness, and promoted autophagy and mitochondria-dependent apoptosis of HCC, at least partly through inhibiting PI3K/AKT/mTOR. These results highlighted Puerarin 6″-O-xyloside as a promising prodrug that could inhibit both PI3K/AKT/mTOR and epigenetic demethylation.
Collapse
Affiliation(s)
- Long Li
- Department of General SurgeryPeople's Hospital of RizhaoRizhaoShandong ProvinceChina
| | - Jun‐Dong Liu
- Department of General SurgeryPeople's Hospital of RizhaoRizhaoShandong ProvinceChina
| | - Guo‐Dong Gao
- Department of General SurgeryPeople's Hospital of RizhaoRizhaoShandong ProvinceChina
| | - Kai Zhang
- Department of General SurgeryPeople's Hospital of RizhaoRizhaoShandong ProvinceChina
| | - Yu‐Wei Song
- Department of General SurgeryPeople's Hospital of RizhaoRizhaoShandong ProvinceChina
| | - Hong‐Bo Li
- Department of General SurgeryPeople's Hospital of RizhaoRizhaoShandong ProvinceChina
| |
Collapse
|
22
|
Fang Q, Liu H, Zhou A, Zhou H, Zhang Z. Circ_0046599 Promotes the Development of Hepatocellular Carcinoma by Regulating the miR-1258/RPN2 Network. Cancer Manag Res 2020; 12:6849-6860. [PMID: 32801909 PMCID: PMC7414927 DOI: 10.2147/cmar.s253510] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background Many studies have confirmed that circular RNAs (circRNAs) play a key role in the biological progression of cancers. However, the function of a novel circRNA, circ_0046599, in hepatocellular carcinoma (HCC) progression has not been explored. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to measure the expression of circ_0046599, microRNA (miR)-1258 and Ribophorin II (RPN2). Subcellular fractionation location assay was used to localize circ_0046599 in HCC cells. The circular characteristic of circ_0046599 was verified using Ribonuclease R (RNase R) digestion assay. Besides, cell counting kit 8 (CCK8) assay, colony formation assay, wound healing assay and transwell assay were used to detect cell proliferation, migration and invasion, respectively. The lactate production and glucose level were determined by Lactate and Glucose Assay Kits. Furthermore, the protein levels of glycolysis, metastasis and proliferation-related marker proteins, as well as RPN2 were tested by Western blot (WB) analysis. Moreover, dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were employed to confirm the interactions among circ_0046599, miR-1258 and RPN2. In addition, mice xenograft models were applied to observe the effect of circ_0046599 silencing on HCC tumor growth in vivo. Results Circ_0046599 was highly expressed in HCC tissues and cells, and its knockdown could suppress HCC cell proliferation, migration, invasion and glycolysis process. MiR-1258 could be targeted by circ_0046599, and its inhibitor could invert the suppressing effect of circ_0046599 knockdown on HCC progression. Further, RPN2 was a target of miR-1258. Overexpressed RPN2 could reverse the inhibiting effect of miR-1258 overexpression on HCC progression. Also, knockdown of circ_0046599 could restrain HCC tumor growth in vivo. Conclusion Our results provided new evidence that circ_0046599 could promote the progression of HCC by increasing RPN2 expression via sponging miR-1258.
Collapse
Affiliation(s)
- Quangang Fang
- Department of Laboratory, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, People's Republic of China
| | - Haiyun Liu
- Department of Laboratory, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, People's Republic of China
| | - Aiqun Zhou
- Department of Laboratory, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, People's Republic of China
| | - Huaping Zhou
- Department of Laboratory, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, People's Republic of China
| | - Zhiyong Zhang
- Department of Laboratory, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang 330006, Jiangxi, People's Republic of China
| |
Collapse
|
23
|
Lu J, Zhao Z, Ma Y. miR-186 Represses Proliferation, Migration, Invasion, and EMT of Hepatocellular Carcinoma via Directly Targeting CDK6. Oncol Res 2020; 28:509-518. [PMID: 32698940 PMCID: PMC7751224 DOI: 10.3727/096504020x15954139263808] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to investigate the effect of miR-186 on proliferation, migration, invasion, and epithelialmesenchymal transition (EMT) of hepatocellular carcinoma (HCC). In this work, miR-186 was downregulated in HCC tissues and cells, and low miR-186 level helped predict the occurrence of vascular invasion and poor prognosis in patients with HCC. miR-186 overexpression inhibited cell proliferation and tumor growth in nude mice, repressed migration and invasion abilities, and enhanced apoptosis in HCC cells. miR-186 also retarded progression of EMT. miR-186 directly bound to the 3-untranslated regions of cyclin-dependent kinase 6 (CDK6) to inhibit its expression. Overexpression of CDK6 markedly reversed inhibitory effects of miR-186 on proliferation, apoptosis, migration, and invasion of HCC cells. Conversely, inhibition of CDK6 exerted synergic effect on the biological functions of miR-186. In conclusion, miR-186 represses proliferation, migration, invasion, and EMT, and induces apoptosis through targeting CDK6 in HCC, which may provide a new therapeutic target for HCC.
Collapse
Affiliation(s)
- Junfeng Lu
- Department of Vascular Surgery, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong UniversityJinanP.R. China
| | - Zhongsong Zhao
- Department of Gastroenterology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong UniversityJinanP.R. China
| | - Yanhong Ma
- Department of Ultrasound, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong UniversityJinanP.R. China
| |
Collapse
|
24
|
Shang R, Wang M, Dai B, Du J, Wang J, Liu Z, Qu S, Yang X, Liu J, Xia C, Wang L, Wang D, Li Y. Long noncoding RNA SLC2A1-AS1 regulates aerobic glycolysis and progression in hepatocellular carcinoma via inhibiting the STAT3/FOXM1/GLUT1 pathway. Mol Oncol 2020; 14:1381-1396. [PMID: 32174012 PMCID: PMC7266282 DOI: 10.1002/1878-0261.12666] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/02/2020] [Accepted: 03/12/2020] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignant diseases worldwide. Despite advances in the diagnosis and treatment of HCC, its overall prognosis remains poor. Recent studies have shown that long noncoding RNAs (lncRNAs) play crucial roles in various pathophysiological processes, including liver cancer. In the current study, we report that lncRNA SLC2A1-AS1 is frequently downregulated in HCC samples, as shown by quantitative real-time polymerase chain reaction analysis. SLC2A1-AS1 deletion is significantly associated with recurrence-free survival in HCC. By performing glucose uptake, lactate production and ATP detection assays, we found that SLC2A1-AS1-mediated glucose transporter 1 (GLUT1) downregulation significantly suppressed glycolysis of HCC. In vitro Cell Counting Kit-8, colony formation, transwell assays as well as in vivo tumorigenesis and metastasis assays showed that SLC2A1-AS1 overexpression significantly suppressed proliferation and metastasis in HCC through the transcriptional inhibition of GLUT1. Results from fluorescence in situ hybridization, ChIP and luciferase reporter assays demonstrated that SLC2A1-AS1 exerts its regulatory role on GLUT1 by competitively binding to transketolase and signal transducer and activator of transcription 3 (STAT3) and inhibits the transactivation of Forkhead box M1 (FOXM1) via STAT3, thus resulting in inactivation of the FOXM1/GLUT1 axis in HCC cells. Our findings will be helpful for understanding the function and mechanism of lncRNA in HCC. These data also highlight the crucial role of SLC2A1-AS1 in HCC aerobic glycolysis and progression and pave the way for further research regarding the potential of SLC2A1-AS1 as a valuable predictive biomarker for HCC recurrence.
Collapse
Affiliation(s)
- Runze Shang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, China
| | - Miao Wang
- State Key Laboratory of Cancer Biology, Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University (Air Force Medical University), Xi'an, China
| | - Bin Dai
- Department of General Surgery, General Hospital of the Central Theater Command of the People's Liberation Army, Wuhan, China
| | - Jianbing Du
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, China
| | - Jianlin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, China
| | - Zekun Liu
- State Key Laboratory of Cancer Biology, Cell Engineering Research Center & Department of Cell Biology, Fourth Military Medical University (Air Force Medical University), Xi'an, China
| | - Shibin Qu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, China
| | - Xisheng Yang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, China
| | - Jingjing Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, China
| | - Congcong Xia
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, China
| | - Desheng Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, China
| | - Yu Li
- School of Life Science, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
25
|
Shi Y, Men X, Li X, Yang Z, Wen H. Research progress and clinical prospect of immunocytotherapy for the treatment of hepatocellular carcinoma. Int Immunopharmacol 2020; 82:106351. [PMID: 32143005 DOI: 10.1016/j.intimp.2020.106351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/23/2020] [Accepted: 02/24/2020] [Indexed: 02/08/2023]
Abstract
As a common malignant tumor, hepatocellular carcinoma (HCC) has high fatality rate due to its strong metastasis and high degree of malignancy. Current treatment strategies adopted in clinical practice were still conventional surgery, assisted with interventional therapy, radiotherapy and chemotherapy. However these treatments have limited effects with high recurrence rate. Current research progress of immunocytotherapy has shown that tumor cells can be directly identified and killed by stimulating the immune function and enhancing the anti-tumor immunity in tumor microenvironment. Targeted immunotherapeutics have therefore become the hope of conquering cancer in the future. It can kill tumor cells without damaging the body's immune system and function, restore and strengthen the body's natural anti-tumor immune system. It can reduce the toxic side effects of radiotherapy and chemotherapy, reduce the recurrence rate and prolong the survival period of patients with HCC. Currently, the immune cells widely studied are mainly as follows: Dendritic cells (DC), Cytokine-induced killer (CIK), DC-CIK, Chimeric antigen receptor T cells (CAR-T), Tumor infiltrating lymphocyte (TIL) and Natural killer cell (NK). Immunocytotherapy is a long-term treatment method, some studies have combined traditional therapy with immunocytotherapy and achieved significant effects, providing experimental basis for the application of immunocytotherapy. However, there are still some difficulties in the clinical application of immune cells. In this article, we discuss the application of immunocytotherapy in the clinical treatment of HCC, their effectiveness either alone or in combination with conventional therapies, and how future immunocytotherapeutics can be further improved from investigations in tumour immunology.
Collapse
Affiliation(s)
- Yue Shi
- Department of Microbiology and Immunology, Changchun University of Chinese Medicine, Jilin 130021, PR China
| | - Xiaoping Men
- Department of Clinical Laboratory, The First Affiliated Hospital to Changchun University of Chinese Medicine, Jilin 130021, PR China
| | - Xueting Li
- Experimental Center, Changchun University of Chinese Medicine, Jilin 130021, PR China
| | - Zhicun Yang
- Experimental Center, Changchun University of Chinese Medicine, Jilin 130021, PR China
| | - Hongjuan Wen
- School of Health Management, Changchun University of Chinese Medicine, Jilin 130117, PR China.
| |
Collapse
|
26
|
Liver Cancer: Current and Future Trends Using Biomaterials. Cancers (Basel) 2019; 11:cancers11122026. [PMID: 31888198 PMCID: PMC6966667 DOI: 10.3390/cancers11122026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common type of cancer diagnosed and the second leading cause of death worldwide. Despite advancement in current treatments for HCC, the prognosis for this cancer is still unfavorable. This comprehensive review article focuses on all the current technology that applies biomaterials to treat and study liver cancer, thus showing the versatility of biomaterials to be used as smart tools in this complex pathologic scenario. Specifically, after introducing the liver anatomy and pathology by focusing on the available treatments for HCC, this review summarizes the current biomaterial-based approaches for systemic delivery and implantable tools for locally administrating bioactive factors and provides a comprehensive discussion of the specific therapies and targeting agents to efficiently deliver those factors. This review also highlights the novel application of biomaterials to study HCC, which includes hydrogels and scaffolds to tissue engineer 3D in vitro models representative of the tumor environment. Such models will serve to better understand the tumor biology and investigate new therapies for HCC. Special focus is given to innovative approaches, e.g., combined delivery therapies, and to alternative approaches-e.g., cell capture-as promising future trends in the application of biomaterials to treat HCC.
Collapse
|
27
|
Qiao Y, Wang J, Karagoz E, Liang B, Song X, Shang R, Evert K, Xu M, Che L, Evert M, Calvisi DF, Tao J, Wang B, Monga SP, Chen X. Axis inhibition protein 1 (Axin1) Deletion-Induced Hepatocarcinogenesis Requires Intact β-Catenin but Not Notch Cascade in Mice. Hepatology 2019; 70:2003-2017. [PMID: 30737831 PMCID: PMC7206928 DOI: 10.1002/hep.30556] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/02/2019] [Indexed: 12/12/2022]
Abstract
Inactivating mutations of axis inhibition protein 1 (AXIN1), a negative regulator of the Wnt/β-Catenin cascade, are among the common genetic events in human hepatocellular carcinoma (HCC), affecting approximately 10% of cases. In the present manuscript, we sought to define the genetic crosstalk between Axin1 mutants and Wnt/β-catenin as well as Notch signaling cascades along hepatocarcinogenesis. We discovered that c-MET activation and AXIN1 mutations occur concomitantly in ~3%-5% of human HCC samples. Subsequently, we generated a murine HCC model by means of CRISPR/Cas9-based gene deletion of Axin1 (sgAxin1) in combination with transposon-based expression of c-Met in the mouse liver (c-Met/sgAxin1). Global gene expression analysis of mouse normal liver, HCCs induced by c-Met/sgAxin1, and HCCs induced by c-Met/∆N90-β-Catenin revealed activation of the Wnt/β-Catenin and Notch signaling in c-Met/sgAxin1 HCCs. However, only a few of the canonical Wnt/β-Catenin target genes were induced in c-Met/sgAxin1 HCC when compared with corresponding lesions from c-Met/∆N90-β-Catenin mice. To study whether endogenous β-Catenin is required for c-Met/sgAxin1-driven HCC development, we expressed c-Met/sgAxin1 in liver-specific Ctnnb1 null mice, which completely prevented HCC development. Consistently, in AXIN1 mutant or null human HCC cell lines, silencing of β-Catenin strongly inhibited cell proliferation. In striking contrast, blocking the Notch cascade through expression of either the dominant negative form of the recombinant signal-binding protein for immunoglobulin kappa J region (RBP-J) or the ablation of Notch2 did not significantly affect c-Met/sgAxin1-driven hepatocarcinogenesis. Conclusion: We demonstrated here that loss of Axin1 cooperates with c-Met to induce HCC in mice, in a β-Catenin signaling-dependent but Notch cascade-independent way.
Collapse
Affiliation(s)
- Yu Qiao
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China;,Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA
| | - Jingxiao Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Eylul Karagoz
- Department of Medicine and Liver Center, University of California, San Francisco, CA;,School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Binyong Liang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinhua Song
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Runze Shang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Department of Hepatobiliary Surgery, Xijing Hospital, Air Force Military Medical University, Xi’an, China
| | - Katja Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Meng Xu
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA;,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Diego F. Calvisi
- Institute of Pathology, University Medicine Greifswald, Greifswald, Germany
| | - Junyan Tao
- Department of Pathology, University of Pittsburgh School of Medicine, and Pittsburgh Liver Research Center, Pittsburgh, PA
| | - Bruce Wang
- Department of Medicine and Liver Center, University of California, San Francisco, CA
| | - Satdarshan P. Monga
- Department of Pathology, University of Pittsburgh School of Medicine, and Pittsburgh Liver Research Center, Pittsburgh, PA
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA
| |
Collapse
|
28
|
Xiang H, Long L, Yao Y, Fang Z, Zhang Z, Zhang Y. CalliSpheres Drug-Eluting Bead Transcatheter Arterial Chemoembolization Presents With Better Efficacy and Equal Safety Compared to Conventional TACE in Treating Patients With Hepatocellular Carcinoma. Technol Cancer Res Treat 2019; 18:1533033819830751. [PMID: 30862264 PMCID: PMC6416678 DOI: 10.1177/1533033819830751] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to compare the treatment response, survival, liver function,
and adverse event incidence of drug-eluting bead transcatheter arterial chemoembolization
using CalliSpheres microspheres with conventional transcatheter arterial chemoembolization
in patients with hepatocellular carcinoma. Seventy-three patients with hepatocellular
carcinoma who received drug-eluting bead transcatheter arterial chemoembolization (using
CalliSpheres microspheres) or conventional transcatheter arterial chemoembolization
treatment were consecutively enrolled. Treatment response was assessed by modified
Response Evaluation Criteria in Solid Tumors at month 1/month 3/month 6; posttreatment,
liver function indexes, and adverse events were recorded. Progression-free survival and
overall survival were also calculated. Objective response rate of patients at months 1, 3,
and 6, disease control rate of patients and objective response rate of nodules at month 3
were increased in drug-eluting bead transcatheter arterial chemoembolization group
compared with conventional transcatheter arterial chemoembolization group. In addition,
drug-eluting bead transcatheter arterial chemoembolization using CalliSpheres microspheres
was an independent factor for predicting better objective response rate at month 1.
Patients in drug-eluting bead transcatheter arterial chemoembolization group achieved
longer progression-free survival and similar overall survival compared to those in
conventional transcatheter arterial chemoembolization group; Cox proportional hazards
regression model analyses revealed that drug-eluting bead transcatheter arterial
chemoembolization using CalliSpheres microspheres was associated with better
progression-free survival while it did not affect overall survival. Meanwhile, most of the
occurrences of abnormal liver function indexes were similar between 2 groups, whereas
drug-eluting bead transcatheter arterial chemoembolization group had a higher percentage
of patients with total bile acid ≥2 upper limit of normal compared to conventional
transcatheter arterial chemoembolization group at month 1. Moreover, the adverse event
incidences between 2 groups were similar. In conclusion, drug-eluting bead transcatheter
arterial chemoembolization using CalliSpheres microspheres achieves better treatment
response and progression-free survival while equal safety compared to conventional
transcatheter arterial chemoembolization in patients with hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hua Xiang
- 1 Department of Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Lin Long
- 1 Department of Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Yuanhui Yao
- 1 Department of Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Zhiyong Fang
- 1 Department of Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Zhiming Zhang
- 1 Department of Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Yongjin Zhang
- 1 Department of Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, Changsha, Hunan, China
| |
Collapse
|
29
|
Chen X, Zhang N. Downregulation of lncRNA NEAT1_2 radiosensitizes hepatocellular carcinoma cells through regulation of miR-101-3p/WEE1 axis. Cell Biol Int 2019; 43:44-55. [PMID: 30488993 DOI: 10.1002/cbin.11077] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/24/2018] [Indexed: 01/04/2023]
Abstract
Radioresistance is a major obstacle in hepatocellular carcinoma (HCC) radiotherapy. Aberrant expression of long non-coding RNA (lncRNA) has been postulated to be implicated in the development of HCC radioresistance. We investigated the role of lncRNA nuclear enriched abundant transcript 1_2 (NEAT1_2) in radioresistance of HCC and its molecular mechanism in this study. We found that NEAT1_2 and WEE1 were upregulated, and miR-101-3p was downregulated in HCC tissues, as well as HCC cell lines. Downregulation of WEE1 sensitized the radiosensitivity of HCC cells, as evidenced by decreased survival fractions of Huh7 and PLC5 cells and increased percentage of apoptotic cells. Also, knockdown of NEAT1_2 exerted a reinforcing effect on the radiosensitivity of HCC cells. In addition, WEE1 was confirmed as a direct target of miR-101-3p. Upregulation of miR-101-3p obviously decreased the mRNA and protein levels of WEE1 compared with that in the miR-NC group, while transfection of anta-miR-101-3p presented the opposite effects. In parallel, NEAT1_2 was identified to interact with miR-101-3p, and NEAT1_2 upregulated the expression of WEE1 in Huh7 cells through sponging miR-101-3p. Besides, the reinforcing effect of NEAT1_2 silencing could be attenuated by downregulation of miR-101-3p. To conclude, our results support the concept that downregulation of lncRNA NEAT1_2 radiosensitizes hepatocellular carcinoma cells through regulation of miR-101-3p/WEE1 axis.
Collapse
Affiliation(s)
- Xin Chen
- Department of Nuclear Medicine, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang 330006, Jiangxi Province, P. R. China
| | - Nuobei Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang 330006, Jiangxi Province, P. R. China
| |
Collapse
|
30
|
Construction and Characterization of Adenovirus Vectors Encoding Aspartate- β-Hydroxylase to Preliminary Application in Immunotherapy of Hepatocellular Carcinoma. J Immunol Res 2018; 2018:9832467. [PMID: 30116759 PMCID: PMC6079451 DOI: 10.1155/2018/9832467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/30/2018] [Indexed: 01/28/2023] Open
Abstract
Dendritic cells (DCs) harboring tumor-associated antigen are supposed to be a potential immunotherapy for hepatocellular carcinoma (HCC). Aspartate-β-hydroxylase (AAH), an overexpressed tumor-associated cell surface protein, is considered as a promising biomarker and therapeutic target for HCC. In this study, we constructed adenovirus vector encoding AAH gene by gateway recombinant cloning technology and preliminarily explored the antitumor effects of DC vaccines harboring AAH. Firstly, the total AAH mRNA was extracted from human HCC tissues; the cDNA was amplified by RT-PCR, verified, and sequenced after TA cloning. Gateway technology was used and the obtained 18T-AAH was used as a substrate, to yield the final expression vector Ad-AAH-IRES2-EGFP. Secondly, bone marrow-derived DCs were infected by Ad-AAH-IRES2-EGFP to yield AAH-DC vaccines. Matured DCs were demonstrated by increased expression of CD11c, CD80, and MHC-II costimulatory molecules. A dramatically cell-killing effect of T lymphocytes coculturing with AAH-DCs on HepG2 HCC cell line was demonstrated by CCK-8 and FCM assays in vitro. More importantly, in an animal experiment, the lysis effect of cytotoxic T lymphocytes (CTLs) on HepG2 cells in the AAH-DC group was stronger than that in the control groups. In conclusion, the gateway recombinant cloning technology is a powerful method of constructing adenovirus vector, and the product Ad-AAH-IRES2-EGFP may present as a potential candidate for DC-based immunotherapy of HCC.
Collapse
|