1
|
Zhu X, Li Z, Chen L, Li L, Ouyang M, Zhou H, Xiao K, Lin L, Chu PK, Zhou C, Xun C, Yang L, Huang W, Ding X. Exosomes delivering miR-129-5p combined with sorafenib ameliorate hepatocellular carcinoma progression via the KCTD1/HIF-1α/VEGF pathway. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01044-x. [PMID: 40227531 DOI: 10.1007/s13402-025-01044-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Potassium channel tetramerization domain-containing 1 (KCTD1) plays a critical role in transcriptional regulation and adipogenesis, but its significance in hepatocellular cancer (HCC) has not been reported. METHODS Immunohistochemistry, Western blotting and quantitative real-time PCR analysis were performed to assess the expression of KCTD1 and related genes in HCC cells. MTT assays, colony formation, cell migration, invasion and the in-vivo mouse models were utilized to evaluate the function of KCTD1 in HCC progression. Co-immunoprecipitation, chromatin immunoprecipitation and luciferase reporter assays were conducted to elucidate the molecular mechanisms of KCTD1 in HCC. RESULTS KCTD1 expression was increased in human HCC tissues and closely associated with advanced tumor stages. KCTD1 overexpression enhanced growth, migration, and invasion of Huh7 and HepG2 cells both in vitro and in vivo, while KCTD1 knockdown reversed these effects in MHCC97H cells. Mechanistically, KCTD1 interacted with hypoxia-inducible factor 1 alpha (HIF-1α) and enhanced HIF-1α protein stability with the inhibited prolyl-hydroxylases (PHD)/Von Hippel-Lindau (VHL) pathway, consequently activating the Vascular Endothelial Growth Factor (VEGF)/VEGFR2 pathway in HCC cells. Sorafenib and KCTD1 knockdown synergistically inhibited intrahepatic tumor growth following in situ injection of MHCC97H cells. miR-129-5p downregulated KCTD1 by binding to KCTD1 3'UTR. Finally, 45 µg exosomes from miR-129-5p-overexpressing MHCC97H cells combined with 25 mg/kg sorafenib to decrease HCC tumor size. CONCLUSIONS These results suggested that KCTD1 protects HIF-1α from degradation and activates the VEGF signaling cascade to enhance HCC progression. Therefore, KCTD1 may serve as a novel target of HCC and pave the way for an efficient combined therapy in advanced HCC.
Collapse
Affiliation(s)
- Xinyu Zhu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Zhiwei Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Li Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Limin Li
- College of Engineering and Design, Hunan Normal University, Taozihu Road No. 68, Changsha, 410081, China.
| | - Mi Ouyang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Hao Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Kai Xiao
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Ling Lin
- Department of Hepatobiliary and Pancreatic Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Paul K Chu
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Chang Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Chengfeng Xun
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Liu Yang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Wenhuan Huang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Xiaofeng Ding
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China.
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China.
| |
Collapse
|
2
|
Wang JB, Ding SL, Liu XS, Yu T, Wu ZA, Li YX. Hypoxia Affects Mitochondrial Stress and Facilitates Tumor Metastasis of Colorectal Cancer Through Slug SUMOylation. Curr Mol Med 2025; 25:27-36. [PMID: 38013443 DOI: 10.2174/0115665240271525231112121008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a malignant tumor. Slug has been found to display a key role in diversified cancers, but its relevant regulatory mechanisms in CRC development are not fully explored. OBJECTIVE Hence, exploring the function and regulatory mechanisms of Slug is critical for the treatment of CRC. METHODS Protein expressions of Slug, N-cadherin, E-cadherin, Snail, HIF-1α, SUMO- 1, Drp1, Opa1, Mfn1/2, PGC-1α, NRF1, and TFAM were measured through western blot. To evaluate the protein expression of Slug and SUMO-1, an immunofluorescence assay was used. Cell migration ability was tested through transwell assay. The SUMOylation of Slug was examined through CO-IP assay. RESULTS Slug displayed higher expression and facilitated tumor metastasis in CRC. In addition, hypoxia treatment was discovered to upregulate HIF-1α, Slug, and SUMO-1 levels, as well as induce Slug SUMOylation. Slug SUMOylation markedly affected mitochondrial biosynthesis, fusion, and mitogen-related protein expression levels to trigger mitochondrial stress. Additionally, the induced mitochondrial stress by hypoxia could be rescued by Slug inhibition and TAK-981 treatment. CONCLUSION Our study expounded that hypoxia affects mitochondrial stress and facilitates tumor metastasis of CRC through Slug SUMOylation.
Collapse
Affiliation(s)
- Jin-Bao Wang
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Shi-Lin Ding
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Xiao-Song Liu
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Tianren Yu
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Zeng-An Wu
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Yu-Xiang Li
- Department of Surgery, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, 100102, China
| |
Collapse
|
3
|
Huang G, Ouyang M, Xiao K, Zhou H, Zhong Z, Long S, Li Z, Zhang Y, Li L, Xiang S, Ding X. AP-2α decreases TMZ resistance of recurrent GBM by downregulating MGMT expression and improving DNA damage. Life Sci 2024; 357:123111. [PMID: 39369843 DOI: 10.1016/j.lfs.2024.123111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 09/21/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
AIMS The incidence of recurrent gliomas is high, exerting low survival rates and poor prognoses. Transcription factor AP-2α has been reported to regulate the progression of primary glioblastoma (GBM). However, the function of AP-2α in recurrent gliomas is largely unclear. METHODS The expression of AP-2α and O6-methylguanine DNA-methyltransferase (MGMT) was detected in recurrent glioma tissues and cell lines by Western blots, the regulation mechanisms between AP-2α/MGMT promoter and RA/AP-2α promoter were studied by luciferase reporter assays, EMSA, and chIP assays. The effects of AP-2α and TMZ/RA treatment on cell viability in vitro and in vivo were investigated by MTT assays, γH2AX staining, comet assays and intracranial injection. KEY FINDINGS AP-2α expression negatively correlates with the expression of MGMT in glioma samples. AP-2α could directly bind with the promoter of the MGMT gene, suppresses transcriptional levels of MGMT and downregulate MGMT expression in TMZ-resistant U87MG-R and T98G cells, but TMZ treatment decreases AP-2α expression and increases MGMT expression. The extended TMZ treatment and increased TMZ concentrations reversed these effects. Moreover, AP-2α overexpression combines with TMZ to decrease cell viability, concurrently with improved DNA damage marker γH2AX. Furthermore, retinoic acid (RA) activates RAR/RXR heterodimers, which bind to RA-responsive elements (RAREs) of the AP-2α promoter, and activates AP-2α expression in recurrent glioma cells. Finally, in intracranial relapsed glioma mouse model, both RA and TMZ could retard tumor development and prolong the mouse survival. SIGNIFICANCE AP-2α activation by gene overexpression or RA treatment reveals the suppressive effects on glioma relapse, providing a novel therapeutic strategy against malignant refractory gliomas.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Middle Aged
- Antineoplastic Agents, Alkylating/pharmacology
- Brain Neoplasms/drug therapy
- Brain Neoplasms/genetics
- Brain Neoplasms/pathology
- Brain Neoplasms/metabolism
- Cell Line, Tumor
- DNA Damage/drug effects
- DNA Modification Methylases/metabolism
- DNA Modification Methylases/genetics
- DNA Repair Enzymes/genetics
- DNA Repair Enzymes/metabolism
- Down-Regulation/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Glioblastoma/drug therapy
- Glioblastoma/genetics
- Glioblastoma/pathology
- Glioblastoma/metabolism
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/metabolism
- Promoter Regions, Genetic
- Temozolomide/pharmacology
- Transcription Factor AP-2/genetics
- Transcription Factor AP-2/metabolism
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Guixiang Huang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Mi Ouyang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Kai Xiao
- Department of Neurosurgery, Second Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Hao Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Zhe Zhong
- Department of Neurosurgery, Hunan Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, Hunan 410013, China
| | - Shengwen Long
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Zhiwei Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Yiru Zhang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Limin Li
- College of Engineering and Design, Hunan Normal University, Changsha 410081, China.
| | - Shuanglin Xiang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China.
| | - Xiaofeng Ding
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha 410081, China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
4
|
Wang Q, Zhang Z, Zhou H, Qin Y, He J, Li L, Ding X. Eosinophil-Associated Genes are Potential Biomarkers for Hepatocellular Carcinoma Prognosis. J Cancer 2024; 15:5605-5621. [PMID: 39308686 PMCID: PMC11414626 DOI: 10.7150/jca.95138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Background: Eosinophils, a type of white blood cell originating from the bone marrow, are widely believed to play a crucial role in inflammatory processes, including allergic reactions and parasitic infections. However, the relationship between eosinophils and liver cancer is not well understood. Methods: Tumor immune infiltration scores were calculated using single-sample Gene Set Enrichment Analysis (ssGSEA). Key modules and hub genes associated with eosinophils were screened using Weighted Gene Co-expression Network Analysis (WGCNA). Univariate and multivariate Cox analyses, along with LASSO regression, were used to identify prognostic genes and create a risk model. The Tumor Immune Dysfunction and Exclusion (TIDE) score was used to evaluate the immunotherapeutic significance of the eosinophil-associated gene risk score (ERS) model. Experiments such as flow cytometry, immunohistochemical analysis, real-time quantitative PCR (RT-qPCR), and Western blotting were used to determine gene expression levels and the status of eosinophil infiltration in tumors. Results: A risk trait model including 4 eosinophil-associated genes (RAMP3, G6PD, SSRP1, PLOD2) was developed by univariate Cox analysis and Lasso screening. Pathologic grading (p < 0.001) and model risk scores (p < 0.001) were found to be independent predictors of hepatocellular carcinoma (HCC) patient survival. Western blotting revealed higher levels of eosinophil peroxidase (EPX) in HCC tissues compared to adjacent normal tissues. Immunohistochemistry showed that eosinophils mainly infiltrated the connective tissue around HCC. The HCC samples showed low expression of RAMP3 and high expression of G6PD, SSRP1, and PLOD2, as detected by IHC and RT-qPCR analysis. The in vivo mouse experiments showed that IL-33 treatment induced the recruitment of eosinophils and reduced the number of intrahepatic tumor nodules. Conclusion: Overall, eosinophil infiltration in HCC is significantly correlated with patient survival. The risk assessment model based on eosinophil-related genes serves as a reliable clinical prognostic indicator and provides insights for precise treatment of HCC.
Collapse
Affiliation(s)
- Qinghao Wang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Zixin Zhang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Hao Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yanling Qin
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jun He
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha, 410007, China
| | - Limin Li
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- College of Engineering and Design, Hunan Normal University, Changsha, 410081, China
| | - Xiaofeng Ding
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, China
| |
Collapse
|
5
|
Zou Q, Zhang Y, Zhu D, Liu X, Wang C, Xiang H. CircMMP11 as a prognostic biomarker mediates miR-361-3p/HMGB1 axis to accelerate malignant progression of hepatocellular carcinoma. Open Med (Wars) 2023; 18:20230803. [PMID: 38025527 PMCID: PMC10655683 DOI: 10.1515/med-2023-0803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 12/01/2023] Open
Abstract
As a high metastatic tumor, patients having hepatocellular carcinoma (HCC) show poor prognosis. The carcinogenic roles of circMMP11 are generally described in the development of other cancers. However, there is a lack of studies on its involvement in HCC. Therefore, we investigated the potential role and molecular mechanisms of CircMMP11 in the development of HCC in vitro, providing preliminary evidence for the clinical treatment of HCC. First, we examined the expression of CircMMP11 in HCC tissues and cell lines in both clinical and in vitro experiments. We then used a loss-of-function assay to determine CircMMP11's regulatory role on the malignant characteristics of HCC cells. The results showed that high expression of CircMMP11 in HCC was associated with patient overall survival. Serum CircMMP11 had good diagnostic efficacy in distinguishing HCC patients from the control group. In vitro, inhibiting CircMMP11 suppressed the malignant characteristics of human HCC cell lines by directly sequestering miR-361-3p, which further affected the downstream gene HMGB1 expression. In addition, we knocked down CircMMP11 and found that its deletion inhibited the malignant characteristics of HCC cells through the miR-361-3p/HMGB1 axis.
Collapse
Affiliation(s)
- Qiong Zou
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Yuping Zhang
- Department of The First General Surgery, Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road Avenue, Changsha, Hunan, 410013, China
| | - Daoqi Zhu
- Department of Oncology, Changsha Kexin Cancer Hospital, Changsha, Hunan, 410205, China
| | - Xinrong Liu
- Department of Pathology, Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Changfa Wang
- Department of The First General Surgery, Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Hong Xiang
- Department of Central Lab, Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| |
Collapse
|
6
|
Jin C, Luo Y, Liang Z, Li X, Kołat D, Zhao L, Xiong W. Crucial role of the transcription factors family activator protein 2 in cancer: current clue and views. J Transl Med 2023; 21:371. [PMID: 37291585 PMCID: PMC10249218 DOI: 10.1186/s12967-023-04189-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
The transcription factor family activator protein 2 (TFAP2) is vital for regulating both embryonic and oncogenic development. The TFAP2 family consists of five DNA-binding proteins, including TFAP2A, TFAP2B, TFAP2C, TFAP2D and TFAP2E. The importance of TFAP2 in tumor biology is becoming more widely recognized. While TFAP2D is not well studied, here, we mainly focus on the other four TFAP2 members. As a transcription factor, TFAP2 regulates the downstream targets directly by binding to their regulatory region. In addition, the regulation of downstream targets by epigenetic modification, posttranslational regulation, and interaction with noncoding RNA have also been identified. According to the pathways in which the downstream targets are involved in, the regulatory effects of TFAP2 on tumorigenesis are generally summarized as follows: stemness and EMT, interaction between TFAP2 and tumor microenvironment, cell cycle and DNA damage repair, ER- and ERBB2-related signaling pathway, ferroptosis and therapeutic response. Moreover, the factors that affect TFAP2 expression in oncogenesis are also summarized. Here, we review and discuss the most recent studies on TFAP2 and its effects on carcinogenesis and regulatory mechanisms.
Collapse
Affiliation(s)
- Chen Jin
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxiao Luo
- University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Zhu Liang
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Chinese Academy for Medical Sciences Oxford Institute, Oxford, UK
| | - Xi Li
- Department of Urology, Churchill Hospital, Oxford University Hospitals NHS Foundation, Oxford, UK
| | - Damian Kołat
- Department of Experimental Surgery, Medical University of Lodz, Lodz, Poland
| | - Linyong Zhao
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Weixi Xiong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China.
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Yang W, Guo Q, Wu H, Tong L, Xiao J, Wang Y, Liu R, Xu L, Yan H, Sun Z. Comprehensive analysis of the cuproptosis-related gene DLD across cancers: A potential prognostic and immunotherapeutic target. Front Pharmacol 2023; 14:1111462. [PMID: 37113760 PMCID: PMC10127393 DOI: 10.3389/fphar.2023.1111462] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
DLD is a key gene involved in “cuproptosis,” but its roles in tumor progression and immunity remain unclear. Exploring the potential mechanisms and biological roles of DLD may provide new insights for therapeutic strategies for tumors. In the present study, we analyzed the role of DLD in a variety of tumors by using several bioinformatic tools. The results showed that compared with normal tissues, tumor tissues representing multiple cancers showed significant differential expression of DLD. High DLD expression was associated with a good prognosis in BRCA, KICH, and LUAD. Conversely, high expression levels of DLD were detrimental to patient prognosis in many other tumors, such as COAD, KIRC, and KIRP. In addition, the associations of DLD with infiltrating immune cells, genetic alterations and methylation levels across cancers were assessed. Aberrant expression of DLD was positively correlated with most infiltrating immune cells, especially neutrophils. The DLD methylation level was significantly decreased in COAD, LIHC, and LUSC but significantly increased in BRCA. DLD had the highest mutation rate (6.04%) in ESCA. In LUSC, patients with genetic alterations in DLD showed a poorer prognosis. At the single-cell level, the roles of DLD in regulating cancer-associated biological functions, such as metastasis, inflammation, and differentiation, were explored. Afterward, we further investigated whether several disease-associated genes could be correlated with DLD. GO enrichment analysis indicated that DLD-related genes were mainly associated with mitochondria-related cellular components, aerobic respiration and the tricarboxylic acid cycle. Finally, the correlations between DLD expression and immunomodulatory genes, immune checkpoints, and sensitivity to some antitumor drugs were investigated. It is worth noting that DLD expression was positively correlated with immune checkpoint genes and immunomodulatory genes in most cancers. In conclusion, this study comprehensively analyzed the differential expression, prognostic value and immune cell infiltration-related function of DLD across cancers. Our results suggest that DLD has great potential to serve as a candidate marker for pancancer prognosis and immunotherapy and may provide a new direction for cancer treatment development.
Collapse
Affiliation(s)
- Weiguang Yang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Graduate School, Tianjin Medical University, Tianjin, China
| | - Qiang Guo
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Orthopaedics, Baodi Clinical College of Tianjin Medical University, Tianjin, China
| | - Haiyang Wu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Graduate School, Tianjin Medical University, Tianjin, China
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, United States
| | - Linjian Tong
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Graduate School, Tianjin Medical University, Tianjin, China
| | - Jian Xiao
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Graduate School, Tianjin Medical University, Tianjin, China
| | - Yulin Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Graduate School, Tianjin Medical University, Tianjin, China
| | - Rui Liu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Graduate School, Tianjin Medical University, Tianjin, China
| | - Lixia Xu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
| | - Hua Yan
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, China
- *Correspondence: Hua Yan, ; Zhiming Sun,
| | - Zhiming Sun
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, China
- Department of Orthopaedics, Tianjin Huanhu Hospital, Tianjin, China
- *Correspondence: Hua Yan, ; Zhiming Sun,
| |
Collapse
|
8
|
Hashemi M, Nadafzadeh N, Imani MH, Rajabi R, Ziaolhagh S, Bayanzadeh SD, Norouzi R, Rafiei R, Koohpar ZK, Raei B, Zandieh MA, Salimimoghadam S, Entezari M, Taheriazam A, Alexiou A, Papadakis M, Tan SC. Targeting and regulation of autophagy in hepatocellular carcinoma: revisiting the molecular interactions and mechanisms for new therapy approaches. Cell Commun Signal 2023; 21:32. [PMID: 36759819 PMCID: PMC9912665 DOI: 10.1186/s12964-023-01053-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/15/2023] [Indexed: 02/11/2023] Open
Abstract
Autophagy is an evolutionarily conserved process that plays a role in regulating homeostasis under physiological conditions. However, dysregulation of autophagy is observed in the development of human diseases, especially cancer. Autophagy has reciprocal functions in cancer and may be responsible for either survival or death. Hepatocellular carcinoma (HCC) is one of the most lethal and common malignancies of the liver, and smoking, infection, and alcohol consumption can lead to its development. Genetic mutations and alterations in molecular processes can exacerbate the progression of HCC. The function of autophagy in HCC is controversial and may be both tumor suppressive and tumor promoting. Activation of autophagy may affect apoptosis in HCC and is a regulator of proliferation and glucose metabolism. Induction of autophagy may promote tumor metastasis via induction of EMT. In addition, autophagy is a regulator of stem cell formation in HCC, and pro-survival autophagy leads to cancer cell resistance to chemotherapy and radiotherapy. Targeting autophagy impairs growth and metastasis in HCC and improves tumor cell response to therapy. Of note, a large number of signaling pathways such as STAT3, Wnt, miRNAs, lncRNAs, and circRNAs regulate autophagy in HCC. Moreover, regulation of autophagy (induction or inhibition) by antitumor agents could be suggested for effective treatment of HCC. In this paper, we comprehensively review the role and mechanisms of autophagy in HCC and discuss the potential benefit of targeting this process in the treatment of the cancer. Video Abstract.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Niloufar Nadafzadeh
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Hassan Imani
- Department of Clinical Science, Faculty of Veterinary Medicine, Shahr-E Kord Branch, Islamic Azad University, Tehran, Chaharmahal and Bakhtiari, Iran
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Setayesh Ziaolhagh
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Raheleh Norouzi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Reihaneh Rafiei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zeinab Khazaei Koohpar
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Behnaz Raei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia
- AFNP Med Austria, Vienna, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
9
|
A novel epithelial-mesenchymal transition gene signature for the immune status and prognosis of hepatocellular carcinoma. Hepatol Int 2022; 16:906-917. [PMID: 35699863 PMCID: PMC9349121 DOI: 10.1007/s12072-022-10354-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/06/2022] [Indexed: 12/11/2022]
Abstract
Background This study clarified whether EMT-related genes can predict immunotherapy efficacy and overall survival in patients with HCC. Methods The RNA-sequencing profiles and patient information of 370 samples were derived from the Cancer Genome Atlas (TCGA) dataset, and EMT-related genes were obtained from the Molecular Signatures database. The signature model was constructed using the least absolute shrinkage and selection operator Cox regression analysis in TCGA cohort. Validation data were obtained from the International Cancer Genome Consortium (ICGC) dataset of patients with HCC. Kaplan–Meier analysis and multivariate Cox analyses were employed to estimate the prognostic value. Immune status and tumor microenvironment were estimated using a single-sample gene set enrichment analysis (ssGSEA). The expression of prognostic genes was verified using qRT-PCR analysis of HCC cell lines. Results A signature model was constructed using EMT-related genes to determine HCC prognosis, based on which patients were divided into high-risk and low-risk groups. The risk score, as an independent factor, was related to tumor stage, grade, and immune cells infiltration. The results indicated that the most prognostic genes were highly expressed in the HCC cell lines, but GADD45B was down-regulated. Enrichment analysis suggested that immunoglobulin receptor binding and material metabolism were essential in the prognostic signature. Conclusion Our novel prognostic signature model has a vital impact on immune status and prognosis, significantly helping the decision-making related to the diagnosis and treatment of patients with HCC. Supplementary Information The online version contains supplementary material available at 10.1007/s12072-022-10354-3.
Collapse
|
10
|
Jiang H, Ma P, Duan Z, Liu Y, Shen S, Mi Y, Fan D. Ginsenoside Rh4 Suppresses Metastasis of Gastric Cancer via SIX1-Dependent TGF-β/Smad2/3 Signaling Pathway. Nutrients 2022; 14:nu14081564. [PMID: 35458126 PMCID: PMC9032069 DOI: 10.3390/nu14081564] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 12/21/2022] Open
Abstract
Gastric cancer (GC) is the leading causes of cancer-related death worldwide. Surgery remains the cornerstone of gastric cancer treatment, and new strategies with adjuvant chemotherapy are currently gaining more and more acceptance. Ginsenoside Rh4 has excellent antitumor activity. Conversely, the mechanisms involved in treatment of GC are not completely understood. In this study, we certified that Rh4 showed strong anti-GC efficiency in vitro and in vivo. MTT and colony formation assays were performed to exhibit that Rh4 significantly inhibited cellular proliferation and colony formation. Results from the wound healing assay, transwell assays, and Western blotting indicated that Rh4 restrained GC cell migration and invasion by reversing epithelial–mesenchymal transition (EMT). Further validation by proteomic screening, co-treatment with disitertide, and SIX1 signal silencing revealed that SIX1, a target of Rh4, induced EMT by activating the TGF-β/Smad2/3 signaling pathway. In summary, our discoveries demonstrated the essential basis of the anti-GC metastatic effects of Rh4 via suppressing the SIX1–TGF-β/Smad2/3 signaling axis, which delivers a new idea for the clinical treatment of GC.
Collapse
Affiliation(s)
- Hongbo Jiang
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
| | - Pei Ma
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
| | - Zhiguang Duan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
| | - Yannan Liu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
| | - Shihong Shen
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
| | - Yu Mi
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
- Correspondence: (Y.M.); (D.F.); Tel.: +86-29-88305118 (Y.M. & D.F.)
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
- Correspondence: (Y.M.); (D.F.); Tel.: +86-29-88305118 (Y.M. & D.F.)
| |
Collapse
|
11
|
Yang Y, Gao L, Chen J, Xiao W, Liu R, Kan H. Lamin B1 is a potential therapeutic target and prognostic biomarker for hepatocellular carcinoma. Bioengineered 2022; 13:9211-9231. [PMID: 35436411 PMCID: PMC9161935 DOI: 10.1080/21655979.2022.2057896] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 12/01/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive malignancy. Previous studies have found that lamin B1 (LMNB1) contributes to the development of human cancers. However, the biological functions and prognostic values of LMNB1 in HCC have not been adequately elucidated. In our present research, the expression pattern of LMNB1 was analyzed. The prognostic values of LMNB1 were evaluated by Kaplan-Meier survival analysis and Cox proportional hazards regression analysis. The effects of LMNB1 on HCC progression were assessed by Cell Counting Kit-8 (CCK-8), colony formation, wound healing, Transwell and in vivo xenograft assays. The mechanisms of LMNB1 in HCC progression were elucidated by gene set enrichment analysis (GSEA) and loss-of-function assays. Besides, a nomogram for predicting overall survival (OS) was constructed. The results demonstrated that LMNB1 was overexpressed in HCC and that increased LMNB1 expression predicted a dismal prognosis. Further experiments showed that LMNB1 facilitated cell proliferation and metastasis in HCC. Functional enrichment analysis revealed that LMNB1 modulated metastasis-associated biological functions such as focal adhesion, extracellular matrix, cell junctions and cell adhesion. Mechanistically, we revealed that LMNB1 promoted HCC progression by regulating the phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways. Moreover, incorporating LMNB1, Ki67 and Barcelona Clinic Liver Cancer (BCLC) stage into a nomogram showed better predictive accuracy than the Tumor-Node-Metastasis (TNM) stage and BCLC stage. In conclusion, LMNB1 may serve as an effective therapeutic target as well as a reliable prognostic biomarker for HCC.
Collapse
Affiliation(s)
- Yongyu Yang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Gao
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junzhang Chen
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wang Xiao
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ruoqi Liu
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Heping Kan
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
12
|
Huang X, Xiang L, Liu W, Li M, Ren A, Chen Z, Zheng C, Chengcong C, Liu J, Yuan Y. Roles of diencephalon/mesencephalon homeobox 1 in the development and prognosis of hepatocellular carcinoma. Ann Hepatol 2022; 24:100314. [PMID: 33524552 DOI: 10.1016/j.aohep.2021.100314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES The oncogene diencephalon/mesencephalon homeobox 1 (DMBX1) is widely overexpressed in a variety of human cancers. The present study aimed to analyze the expression and clinical importance of DMBX1 in nonneoplastic tissues and tumor tissues from patients with hepatocellular carcinoma (HCC). MATERIALS AND METHODS DMBX1 expression in HCC and adjacent nontumor tissues was analyzed using immunohistochemical staining. Chi-square tests were applied to compare DMBX1 expression between the tumors and the adjacent normal tissues. We explored the correlation of DMBX1 expression with clinicopathological factors and its effect on the prognosis of HCC. Finally, we investigated the role of DMBX1 in HCC via knockdown experiments, which analyzed changes in cell invasion, cell proliferation and epithelial-mesenchymal transition (EMT) biomarkers (E-cadherin, N-cadherin, vimentin). The mRNAs that were coexpressed with DMBX1 in HCC, based on the TCGA cohort (n = 366), were obtained from the cBioPortal database. RESULTS The average score for DMBX1 expression was significantly different (P < 0.001) between HCC and paired adjacent nontumor tissues, and DMBX1 expression correlated with hepatitis B virus (HBV) infection, tumor size, metastasis, and tumor node metastasis (TNM) stage (P < 0.05). A multivariate Cox regression analysis identified significant correlations of DMBX1 expression with tumor metastasis, TNM stage, and tumor capsule. Moreover, Kaplan-Meier survival analysis revealed an association between DMBX1 overexpression and shorter overall survival of patients with HCC (P < 0.05). In HCC cell lines, silencing DMBX1 markedly inhibited migration, proliferation and EMT markers. The mRNAs that were negatively (R ≤ -0.25, n = 1094) or positively (R ≥ 0.25, n = 2906) coexpressed with DMBX1 mRNA were selected for further Gene Ontology enrichment analysis, and the results revealed that the predicted functions of DMBX1 in HCC support the in vitro experimental results. CONCLUSIONS Our data provide evidence that DMBX1 overexpression is associated with HCC metastasis and poor prognosis, suggesting that DMBX1 represents a therapeutic target in HCC.
Collapse
Affiliation(s)
- Xiaoting Huang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Leyang Xiang
- Department of Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Wei Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Mingyi Li
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Anbang Ren
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Zide Chen
- Department of Radiation Oncology, The 2nd Clinical Medical College (Shenzhen People's Hospital) of Jinan University, Shenzhen, China
| | - Chu Zheng
- Academic Office, Guilin Medical University, Guilin, China
| | - Chen Chengcong
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Jinquan Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.
| | - Yawei Yuan
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
13
|
Dong T, Jiang J, Zhang H, Liu H, Zou X, Niu J, Mao Y, Zhu M, Chen X, Li Z, Chen Y, Shi C, Yang X. PFP@PLGA/Cu 12Sb 4S 13-mediated PTT ablates hepatocellular carcinoma by inhibiting the RAS/MAPK/MT-CO1 signaling pathway. NANO CONVERGENCE 2021; 8:29. [PMID: 34606010 PMCID: PMC8490557 DOI: 10.1186/s40580-021-00279-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/10/2021] [Indexed: 05/11/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most malignant tumors in the world, and patients with HCC face a poor prognosis. The conventional therapeutic strategies for HCC have undergone a challenge-riddled evolution owing to side effects and unsatisfactory efficacy. Here, aiming to provide a new method of HCC elimination, we formulated a novel multifunctional nanocapsule (PFP@PLGA/Cu12Sb4S13, PPCu) with applications in contrast-enhanced ultrasound imaging (CEUS) and photothermal therapy (PTT). These PPCu were successfully constructed with an average diameter of 346 nm (polydispersity index, PDI = 0.276). The reinforced contrast ratio of these PPCu was determined by CEUS, revealing their promising applications in image-guided monitoring of HCC treatment. Furthermore, the excellent photoabsorption and biocompatibility indicated by organ H&E staining indicated that PPCu meet quality expectations for use as photothermal transduction agent (PTA). PPCu treatment at 50 °C and higher temperatures efficiently repressed the proliferation, induced the apoptosis and decreased the motility of HCC cells. These effects might have been results of RAS/MAPK/MT-CO1 signaling pathway inhibition. In summary, PPCu were constructed to integrate CEUS and PTT successfully into therapy, which can lead to HCC elimination through RAS/MAPK/MT-CO1 signaling pathway repression.
Collapse
Affiliation(s)
- Tianxiu Dong
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Jian Jiang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Hao Zhang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Department of Medical Imaging, Heilongjiang Provincial Hospital, Harbin, 150001, China
| | - Hongyuan Liu
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xiaomeng Zou
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Jiamei Niu
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yingxuan Mao
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Mingwei Zhu
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xi Chen
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Zizhuo Li
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yaodong Chen
- Department of Ultrasonic Imaging, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Chunying Shi
- Department of Radiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xiuhua Yang
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
14
|
Zhou H, Ning Y, Zeng G, Zhou C, Ding X. Curcumin promotes cell cycle arrest and apoptosis of acute myeloid leukemia cells by inactivating AKT. Oncol Rep 2021; 45:11. [PMID: 33649826 PMCID: PMC7877002 DOI: 10.3892/or.2021.7962] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/19/2021] [Indexed: 12/19/2022] Open
Abstract
Curcumin, a phytochemical from rhizomes of the plant Curcuma longa, has been reported to exert potential anticancer properties in various cancer types, including acute myeloid leukemia (AML). However, the underlying mechanism remains poorly understood. The present study demonstrated that curcumin had a stronger cytotoxic activity against AML cells compared with three other types of phytochemicals (epigallocatechin gallate, genistein and resveratrol). Protein phosphorylation profiling using an antibody array identified that curcumin treatment increased the phosphorylation levels of 14 proteins and decreased those of four proteins. A protein‑protein interaction network was constructed using the STRING database, in which AKT was identified as a hub protein with the highest connectivity (PRAS40, 4E‑BP1, P70S6K, RAF‑1 and p27). Western blotting results indicated that curcumin dose‑dependently suppressed the phosphorylation of AKT, PRAS40, 4E‑BP1, P70S6K, RAF‑1 and p27 in AML cell lines (ML‑2 and OCI‑AML5). It was also demonstrated that curcumin regulated the cell cycle‑ and apoptosis‑related proteins (cyclin D1, p21, Bcl2, cleaved‑caspase‑3 and cleaved‑PARP), leading to cell cycle arrest and apoptosis in both ML‑2 and OCI‑AML5 cells. These effects of curcumin were enhanced by the AKT inhibitor afuresertib but were suppressed by the AKT activator SC‑79, indicating that curcumin functions via AKT. In the AML xenograft mouse model, curcumin and afuresertib synergistically suppressed the engraftment, proliferation and survival of AML cells. Collectively, the present study demonstrated that curcumin exerted anti‑AML roles by inactivating AKT and these findings may aid in the treatment of AML.
Collapse
Affiliation(s)
- Hao Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Yichong Ning
- Department of Clinical Laboratory, Chongzuo People's Hospital, Chongzuo, Guangxi 532200, P.R. China
| | - Guirong Zeng
- Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs and Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, Hunan 410331, P.R. China
| | - Chang Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xiaofeng Ding
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| |
Collapse
|
15
|
Chang L, Wang J, Zhou F, Wang D, Chen R, Zhang Y, Zhang J. LncRNA RP11-84E24.3 drives tumorigenesis and epithelial-to-mesenchymal transition of glioma cells by promoting TFAP2C-mediated activation of SNAI1. J Neurooncol 2021; 151:157-171. [PMID: 33128099 DOI: 10.1007/s11060-020-03624-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Long noncoding RNAs (LncRNAs) are essential epigenetic regulators with critical roles in tumor initiation and malignant progression; however, the mechanism by which aberrantly expressed lncRNA RP11-84E24.3 regulates the pathogenesis of glioma is not fully understood. Here, we investigate the function of lncRNA RP11-84E24.3 in glioma onset and progression as well as identify a molecular pathway regulated by this lncRNA. METHODS Differentially expressed lncRNAs related to glioma were identified. The aberrant expression of lncRNA RP11-84E24.3 was verified in samples from patients with glioma as well as glioma cell lines. The role of lncRNA RP11-8424.3 in proliferation, apoptosis, migration, and invasion was assessed using gain- and loss-of function approaches, EdU incorporation, flow cytometry, wound healing and Transwell invasion assays. Western blot analysis was utilized to examine the expression of proteins associated with epithelial-to-mesenchymal transition (EMT). The interaction between lncRNA RP11-84E24.3, TFAP2C and SNAI1 was confirmed using RNA pull-down, ChIP and luciferase reporter assays. RESULTS LncRNA RP11-84E24.3 was up-regulated in both glioma tissues and cell lines. LncRNA RP11-84E24.3 overexpression enhanced the proliferation, migration and invasion of glioma cells while reducing apoptosis. This was associated with a decrease in E-cadherin expression and an increase in N-cadherin and Vimentin expression. LncRNA RP11-84E24.3 directly targeted TFAP2C protein, resulting in increased SNAI1 expression. Knockdown of TFAP2C or SNAI1 reversed the effects of lncRNA RP11-84E24.3 overexpression, while silencing lncRNA RP11-84E24.3 inhibited tumor formation of glioma cells in vivo. CONCLUSIONS LncRNA RP11-84E24.3 increased SNAI1 expression by forming a complex with TFAP2C protein, promoting EMT in glioma cells and tumor formation.
Collapse
Affiliation(s)
- Lisha Chang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Jingyue Wang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Fuling Zhou
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Dali Wang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Ruiying Chen
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Yunhe Zhang
- Department of Neurosurgery, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000, People's Republic of China
| | - Jiang Zhang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China.
| |
Collapse
|
16
|
Zhang C, Wei S, Sun WP, Teng K, Dai MM, Wang FW, Chen JW, Ling H, Ma XD, Feng ZH, Duan JL, Cai MY, Xie D. Super-enhancer-driven AJUBA is activated by TCF4 and involved in epithelial-mesenchymal transition in the progression of Hepatocellular Carcinoma. Am J Cancer Res 2020; 10:9066-9082. [PMID: 32802179 PMCID: PMC7415796 DOI: 10.7150/thno.45349] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023] Open
Abstract
Background and Aims: Aberrant transcriptional programs are highly regulated processes that play important roles in the development and progression of hepatocellular carcinoma (HCC). Emerging evidence suggests that super-enhancers (SEs) often drive critical oncogene expression. However, SE-associated genes in HCC pathogenesis are still poorly understood. Methods: We performed integrative ChIP-seq and Hi-C analyses of HCC cells and identified ajuba LIM protein (AJUBA) as a SE-associated gene. We evaluated AJUBA expression in HCC using immunohistochemistry, immunoblotting, and qRT-PCR. ChIP and luciferase reporter assays were performed to demonstrate that transcription factor 4 (TCF4) bound to AJUBA-associated SEs. We then assessed the role of AJUBA in HCC using both in vitro and in vivo assays. Epithelial-mesenchymal transition (EMT) was examined using immunofluorescence and immunoblotting assays. Furthermore, we used immunoprecipitation and BiFC assays to explore the underlying mechanisms. Results: We identified AJUBA as a SE-associated oncogene in HCC regulated by TCF4. High AJUBA expression was related to an aggressive phenotype and unfavorable outcome in HCC patients. AJUBA knockdown significantly reduced cell migration and invasion capacities both in vitro and in vivo. Furthermore, AJUBA overexpression in HCC recruited tumor necrosis factor associated factor 6 (TRAF6), enhancing the phosphorylation of Akt and increasing Akt activity toward GSK-3β, thus promoting EMT. Conclusions: Our results provide functional and mechanistic links between the SE-associated gene AJUBA and tumor EMT in aggressive HCC.
Collapse
|
17
|
Deng G, Chen Y, Guo C, Yin L, Han Y, Li Y, Fu Y, Cai C, Shen H, Zeng S. BMP4 promotes the metastasis of gastric cancer by inducing epithelial-mesenchymal transition via ID1. J Cell Sci 2020; 133:jcs237222. [PMID: 32376787 DOI: 10.1242/jcs.237222] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 04/15/2020] [Indexed: 12/15/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a crucial process for cancer cells to acquire metastatic potential, which primarily causes death in gastric cancer (GC) patients. Bone morphogenetic protein 4 (BMP4) is a member of the TGF-β family that plays an indispensable role in human cancers. However, little is known about its roles in GC metastasis. In this study, BMP4 was found to be frequently overexpressed in GC tissues and was correlated with poor patient's prognosis. BMP4 was upregulated in GC cell lines and promoted EMT and metastasis of GC cells both in vitro and in vivo, whereas knockdown of BMP4 significantly inhibited EMT and metastasis of GC cells. Furthermore, the inhibitor of DNA binding 1 (also known as DNA-binding protein inhibitor ID1) was identified as a downstream target of BMP4 using PCR arrays and was upregulated via SMAD1/5/8 phosphorylation. ID1 knockdown attenuated BMP4-induced EMT and invasion in GC cells. Moreover, ID1 overexpression in BMP4 knockdown cells restored the promotion of EMT and cell invasion. In summary, BMP4 induced EMT and promoted GC metastasis by upregulating ID1 expression. Antagonizing BMP4 could be a potential therapeutic strategy for GC metastasis.
Collapse
Affiliation(s)
- Ganlu Deng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530022 Guangxi, China
| | - Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Cao Guo
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Ling Yin
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Yiyi Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Yaojie Fu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, China
| |
Collapse
|
18
|
Zhu L, Dai L, Yang N, Liu M, Ma S, Li C, Shen J, Lin T, Wang D, Pan W, Li X. Transcription factorIRX5 promotes hepatocellular carcinoma proliferation and inhibits apoptosis by regulating the p53 signalling pathway. Cell Biochem Funct 2020; 38:621-629. [PMID: 32153043 PMCID: PMC7384050 DOI: 10.1002/cbf.3517] [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: 09/20/2019] [Revised: 02/01/2020] [Accepted: 02/05/2020] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide and the third most frequent cause of cancer-related death. The IRX5 transcription factor plays a different role in multiple cancers and contributes to the development of many tumours. However, little is known about the molecular mechanisms of IRX5 in HCC. In this study, we found that IRX5 was abnormally upregulated in HCC tissues compared with adjacent normal tissues. IRX5 promoted HCC cell proliferation and upregulated the expression of cyclin D1 and knockdown of IRX5 suppressed tumorigenicity in vivo. Furthermore, knockdown of IRX5 increased p53 and Bax expression and decreased Bcl-2 expression. Thus, IRX5 suppressed apoptosis in HCC cells by inhibiting the p53 signalling pathway, indicating its role as a treatment target for HCC. SIGNIFICANCE OF THE STUDY: Our study demonstrated that IRX5 was abnormally upregulated in HCC tissues compared with adjacent normal tissues. IRX5 promoted HCC cell proliferation and upregulated the expression of cyclin D1, and knockdown of IRX5 suppressed tumorigenicity in vivo. Furthermore, knockdown of IRX5 increased p53 and Bax expression and decreased Bcl-2 expression. IRX5 suppressed apoptosis in HCC cells by inhibiting the p53 signalling pathway, indicating its role as a treatment target for HCC.
Collapse
Affiliation(s)
- Liying Zhu
- Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Longguang Dai
- Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Nenghong Yang
- Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Mi Liu
- Clinical Laboratory, The Tumor Hospital of Guizhou Province, Guiyang, China
| | - Shuang Ma
- Clinical Laboratory, Guizhou Provincial People,s Hospital, Guiyang, China
| | - Chengcheng Li
- Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Jie Shen
- Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Tao Lin
- Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Dan Wang
- Department of Clinical Laboratory, The People's Hospital of Rongchang District, Chongqing, China
| | - Wei Pan
- Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Xing Li
- Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China.,Guizhou university of traditional Chinese medicine, Guiyang, China
| |
Collapse
|
19
|
Xiao Y, Huang S, Qiu F, Ding X, Sun Y, Wei C, Hu X, Wei K, Long S, Xie L, Xun Y, Chen W, Zhang Z, Liu N, Xiang S. Tumor necrosis factor α-induced protein 1 as a novel tumor suppressor through selective downregulation of CSNK2B blocks nuclear factor-κB activation in hepatocellular carcinoma. EBioMedicine 2020; 51:102603. [PMID: 31901862 PMCID: PMC6950786 DOI: 10.1016/j.ebiom.2019.102603] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022] Open
Abstract
Background Tumor necrosis factor α-induced protein 1 (TNFAIP1) is frequently downregulated in cancer cell lines and promotes cancer cell apoptosis. However, its role, clinical significance and molecular mechanisms in hepatocellular carcinoma (HCC) are unknown. Methods The expression of TNFAIP1 in HCC tumor tissues and cell lines was measured by Western blot and immunohistochemistry. The effects of TNFAIP1 on HCC proliferation, apoptosis, metastasis, angiogenesis and tumor formation were evaluated by Cell Counting Kit-8 (CCK8), Terminal deoxynucleotidyl transferase dUTP Nick-End Labeling (TUNEL), transwell, tube formation assay in vitro and nude mice experiments in vivo. The interaction between TNFAIP1 and CSNK2B was validated by liquid chromatography-tandem mass spectrometry (LC-MS/MS), Co-immunoprecipitation and Western blot. The mechanism of how TNFAIP1 regulated nuclear factor-kappaB (NF-κB) pathway was analyzed by dual-luciferase reporter, immunofluorescence, quantitative Real-time polymerase chain reaction (RT-qPCR) and Western blot. Findings The TNFAIP1 expression is significantly decreased in HCC tissues and cell lines, and negatively correlated with the increased HCC histological grade. Overexpression of TNFAIP1 inhibits HCC cell proliferation, metastasis, angiogenesis and promotes cancer cell apoptosis both in vitro and in vivo, whereas the knockdown of TNFAIP1 in HCC cell displays opposite effects. Mechanistically, TNFAIP1 interacts with CSNK2B and promotes its ubiquitin-mediated degradation with Cul3, causing attenuation of CSNK2B-dependent NF-κB trans-activation in HCC cell. Moreover, the enforced expression of CSNK2B counteracts the inhibitory effects of TNFAIP1 on HCC cell proliferation, migration, and angiogenesis in vitro and in vivo. Interpretation Our results support that TNFAIP1 can act as a tumor suppressor of HCC by modulating TNFAIP1/CSNK2B/NF-κB pathway, implying that TNFAIP1 may represent a potential marker and a promising therapeutic target for HCC.
Collapse
Affiliation(s)
- Ye Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China; Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Shulan Huang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Feng Qiu
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Xiaofeng Ding
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yi Sun
- Department of Pathology, Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Chenxi Wei
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Xiang Hu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Ke Wei
- Medical school, Hunan University of Traditional Chinese Medicine, Changsha, 410208, China
| | - Shengwen Long
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Lina Xie
- Department of Stomatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yu Xun
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Wen Chen
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Zhijian Zhang
- Department of Pathology, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Ning Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China.
| | - Shuanglin Xiang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
20
|
Li S, Peng F, Ning Y, Jiang P, Peng J, Ding X, Zhang J, Jiang T, Xiang S. SNHG16 as the miRNA let-7b-5p sponge facilitates the G2/M and epithelial-mesenchymal transition by regulating CDC25B and HMGA2 expression in hepatocellular carcinoma. J Cell Biochem 2019; 121:2543-2558. [PMID: 31696971 DOI: 10.1002/jcb.29477] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022]
Abstract
Long noncoding RNAs (lncRNAs) play crucial roles in hepatocellular carcinoma (HCC). However, the underlying molecular mechanisms of small nucleolar RNA host gene 16 (SNHG16) for regulating the cell cycle and epithelial to mesenchymal transition (EMT) remain elusive. In this study, SNHG16 expression profiles of HCC tissues or cell lines were compared with those of normal tissues or hepatocyte cell line. The effect of SNHG16 knockdown in HCC cell lines was investigated by using in vitro loss-of-function experiments and in vivo nude mouse experiments. The potential molecular regulatory mechanism of SNHG16 in HCC progression was investigated by using mechanistic experiments and rescue assays. The results revealed that SNHG16 was highly expressed in HCC tissues and cell lines, which predicted poor prognosis of HCC patients. On one hand, the downregulation of SNHG16 induced G2/M cell cycle arrest, inducing cell apoptosis and suppression of cell proliferation. On the other hand, it inhibited cell metastasis and EMT progression demonstrated by in vitro loss-of-function cell experiments. Besides, knockdown of SNHG16 increased the sensitivity of HCC cells to cisplatin. For the detailed mechanism, SNHG16 was demonstrated to act as a let-7b-5p sponge in HCC. SNHG16 facilitated the G2/M cell cycle transition by directly acting on the let-7b-5p/CDC25B/CDK1 axis, and promoted cell metastasis and EMT progression by regulating the let-7b-5p/HMGA2 axis in HCC. In addition, the mechanism of SNHG16 for regulating HCC cell proliferation and metastasis was further confirmed in vivo by mouse experiments. Furthermore, these results can provide new insights into HCC treatment and its molecular pathogenesis, which may enlighten the further research of the molecular pathogenesis of HCC.
Collapse
Affiliation(s)
- Shengguang Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, China.,Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Center of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou Institute of Systems Medicine, Suzhou, China
| | - Fujun Peng
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yichong Ning
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, China
| | - Peng Jiang
- Suzhou Geneworks Technology Co, Ltd, Suzhou, China
| | - Jian Peng
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaofeng Ding
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, China
| | - Jian Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, China
| | - Taijiao Jiang
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Center of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou Institute of Systems Medicine, Suzhou, China
| | - Shuanglin Xiang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
21
|
Spiniello M, Steinbrink MI, Cesnik AJ, Miller RM, Scalf M, Shortreed MR, Smith LM. Comprehensive in vivo identification of the c-Myc mRNA protein interactome using HyPR-MS. RNA (NEW YORK, N.Y.) 2019; 25:1337-1352. [PMID: 31296583 PMCID: PMC6800478 DOI: 10.1261/rna.072157.119] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 06/27/2019] [Indexed: 05/10/2023]
Abstract
Proteins bind mRNA through their entire life cycle from transcription to degradation. We analyzed c-Myc mRNA protein interactors in vivo using the HyPR-MS method to capture the crosslinked mRNA by hybridization and then analyzed the bound proteins using mass spectrometry proteomics. Using HyPR-MS, 229 c-Myc mRNA-binding proteins were identified, confirming previously proposed interactors, suggesting new interactors, and providing information related to the roles and pathways known to involve c-Myc. We performed structural and functional analysis of these proteins and validated our findings with a combination of RIP-qPCR experiments, in vitro results released in past studies, publicly available RIP- and eCLIP-seq data, and results from software tools for predicting RNA-protein interactions.
Collapse
Affiliation(s)
- Michele Spiniello
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Department of Medicine of Precision, University of Studi della Campania Luigi Vanvitelli, Naples 80138, Italy
- Division of Immuno-Hematology and Transfusion Medicine, Cardarelli Hospital, Naples 80131, Italy
| | - Maisie I Steinbrink
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Anthony J Cesnik
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Rachel M Miller
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Mark Scalf
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Michael R Shortreed
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Lloyd M Smith
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
22
|
Zhang J, Cao Z, Yang G, You L, Zhang T, Zhao Y. MicroRNA-27a (miR-27a) in Solid Tumors: A Review Based on Mechanisms and Clinical Observations. Front Oncol 2019; 9:893. [PMID: 31572683 PMCID: PMC6751266 DOI: 10.3389/fonc.2019.00893] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are a family of highly conserved, non-coding single-stranded RNAs transcribed as ~70 nucleotide precursors to an 18–22 nucleotide product (1). miRNAs can silence their homologous target genes at the post-transcriptional level, and these genes have been revealed to play an important role in tumorigenesis, invasion and metastasis (2). MicroRNA-27a (miR-27a), transcripted by miR-27a gene, has proved to implicate with many kinds of solid tumors, showing potential as a useful biomarker or drug target for clinical application. However, even though miR-27a has been reported in many cancers, the mechanism and signal pathways of miR-27 in oncogenesis, invasion, and metastasis are still obscure. Moreover, recent studies show that miR-27a pays an important role in epithelial-mesenchymal-transition, regulating tumor immune response, and chemoresistance. In this review, we summarize the current literature, demonstrate the established link between miR-27a and tumorigenesis, and focus on recently identified mechanisms. The review also aims to demonstrate the potential of miR-27a as a diagnostic and/or prognostic biomarker in solid tumors and to discuss the possibilities of targeted therapy and drug design.
Collapse
Affiliation(s)
- Jingcheng Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
23
|
Huang W, Zhong Z, Luo C, Xiao Y, Li L, Zhang X, Yang L, Xiao K, Ning Y, Chen L, Liu Q, Hu X, Zhang J, Ding X, Xiang S. The miR-26a/AP-2α/Nanog signaling axis mediates stem cell self-renewal and temozolomide resistance in glioma. Am J Cancer Res 2019; 9:5497-5516. [PMID: 31534499 PMCID: PMC6735392 DOI: 10.7150/thno.33800] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 07/17/2019] [Indexed: 12/24/2022] Open
Abstract
Aberrant expression of transcription factor AP-2α has been functionally associated with various cancers, but its clinical significance and molecular mechanisms in human glioma are largely elusive. Methods: AP-2α expression was analyzed in human glioma tissues by immunohistochemistry (IHC) and in glioma cell lines by Western blot. The effects of AP-2α on glioma cell proliferation, migration, invasion and tumor formation were evaluated by the 3-(4,5-dimethyNCthiazol-2-yl)-25-diphenyltetrazolium bromide (MTT) and transwell assays in vitro and in nude mouse models in vivo. The influence of AP-2α on glioma cell stemness was analyzed by sphere-formation, self-renewal and limiting dilution assays in vitro and in intracranial mouse models in vivo. The effects of AP-2α on temozolomide (TMZ) resistance were detected by the MTT assay, cell apoptosis, real-time PCR analysis, western blotting and mouse experiments. The correlation between AP-2α expression and the expression of miR-26a, Nanog was determined by luciferase reporter assays, electrophoretic mobility shift assay (EMSA) and expression analysis. Results: AP-2α expression was downregulated in 58.5% of glioma tissues and in 4 glioma cell lines. AP-2α overexpression not only reduced the proliferation, migration and invasion of glioma cell lines but also suppressed the sphere-formation and self-renewal abilities of glioma stem cells in vitro. Moreover, AP-2α overexpression inhibited subcutaneous and intracranial xenograft tumor growth in vivo. Furthermore, AP-2α enhanced the sensitivity of glioma cells to TMZ. Finally, AP-2α directly bound to the regulatory region of the Nanog gene, reduced Nanog, Sox2 and CD133 expression. Meanwhile, AP-2α indirectly downregulated Nanog expression by inhibiting the interleukin 6/janus kinase 2/signal transducer and activator of transcription 3 (IL6/JAK2/STAT3) signaling pathway, consequently decreasing O6-methylguanine methyltransferase (MGMT) and programmed death-ligand 1 (PD-L1) expression. In addition, miR-26a decreased AP-2α expression by binding to the 3' untranslated region (UTR) of AP-2α and reversed the tumor suppressive role of AP-2α in glioma, which was rescued by a miR-26a inhibitor. TMZ and the miR-26a inhibitor synergistically suppressed intracranial GSC growth. Conclusion: These results suggest that AP-2α reduces the stemness and TMZ resistance of glioma by inhibiting the Nanog/Sox2/CD133 axis and IL6/STAT3 signaling pathways. Therefore, AP-2α and miR-26a inhibition might represent a new target for developing new therapeutic strategies in TMZ resistance and recurrent glioma patients.
Collapse
|
24
|
Luo J, Liu K, Yao Y, Sun Q, Zheng X, Zhu B, Zhang Q, Xu L, Shen Y, Ren B. DMBX1 promotes tumor proliferation and regulates cell cycle progression via repressing OTX2-mediated transcription of p21 in lung adenocarcinoma cell. Cancer Lett 2019; 453:45-56. [DOI: 10.1016/j.canlet.2019.03.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 03/21/2019] [Accepted: 03/22/2019] [Indexed: 12/17/2022]
|
25
|
Ding J, Cheng Y, Zhang Y, Liao S, Yin T, Yang J. The miR‐27a‐3p/USP25 axis participates in the pathogenesis of recurrent miscarriage by inhibiting trophoblast migration and invasion. J Cell Physiol 2019; 234:19951-19963. [PMID: 30953360 DOI: 10.1002/jcp.28593] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/13/2019] [Accepted: 03/19/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Jinli Ding
- Reproductive Medical Center Renmin Hospital of Wuhan University Wuhan China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development Wuhan China
| | - Yanxiang Cheng
- Department of Gynecology and Obstetrics Renmin Hospital of Wuhan University Wuhan China
| | - Yi Zhang
- Reproductive Medical Center Renmin Hospital of Wuhan University Wuhan China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development Wuhan China
| | - Shujie Liao
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Tailang Yin
- Reproductive Medical Center Renmin Hospital of Wuhan University Wuhan China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development Wuhan China
| | - Jing Yang
- Reproductive Medical Center Renmin Hospital of Wuhan University Wuhan China
- Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development Wuhan China
| |
Collapse
|
26
|
Xie L, Dai H, Li M, Yang W, Yu G, Wang X, Wang P, Liu W, Hu X, Zhao M. MARCH1 encourages tumour progression of hepatocellular carcinoma via regulation of PI3K-AKT-β-catenin pathways. J Cell Mol Med 2019; 23:3386-3401. [PMID: 30793486 PMCID: PMC6484336 DOI: 10.1111/jcmm.14235] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/02/2019] [Accepted: 01/23/2019] [Indexed: 12/24/2022] Open
Abstract
Membrane‐associated RING‐CH‐1 (MARCH1) is a membrane‐anchored E3 ubiquitin ligase that is involved in a variety of cellular processes. MARCH1 was aberrantly expressed as a tumour promoter in ovarian cancer, but the signalling about the molecular mechanism has not yet been fully illuminated. Here, we first determined that MARCH1 was obviously highly expressed in human hepatocellular carcinoma samples and cells. In addition, our findings demonstrated that the proliferation, migration and invasion of hepatocellular carcinoma were suppressed, but the apoptosis was increased, as a result of MARCH1 knockdown by either siRNA targeting MARCH1 or pirarubicin treatment. Conversely, the proliferation, migration and invasion of hepatocellular carcinoma were obviously accelerated, and the apoptosis was decreased, by transfecting the MARCH1 plasmid to make MARCH1 overexpressed. Moreover, in vivo, the results exhibited a significant inhibition of the growth of hepatocellular carcinoma in nude mice, which were given an intra‐tumour injection of siRNA targeting MARCH1. Furthermore, our study concluded that MARCH1 functions as a tumour promoter, and its role was up‐regulated the PI3K‐AKT‐β‐catenin pathways both in vitro and in vivo. In summary, our work determined that MARCH1 has an important role in the development and progression of hepatocellular carcinoma and may be used as a novel potential molecular therapeutic target in the future treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Lulu Xie
- Department of Imaging, Binzhou Medical University, Yantai, China
| | - Hanhan Dai
- Department of Imaging, Binzhou Medical University, Yantai, China
| | - Minjing Li
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, China
| | - Wei Yang
- Department of Imaging, Binzhou Medical University, Yantai, China
| | - Guohua Yu
- Department of Pathology, Yu Huang Ding Hospital, Yantai, China
| | - Xia Wang
- Department of Oral Pathology, Binzhou Medical University, Yantai, China
| | - Peiyuan Wang
- Department of Imaging, Binzhou Medical University, Yantai, China
| | - Wei Liu
- Department of Imaging, Binzhou Medical University, Yantai, China
| | - Xuemei Hu
- Department of Immunology, Binzhou Medical University, Yantai, China
| | - Mingdong Zhao
- Department of Imaging, Binzhou Medical University, Yantai, China
| |
Collapse
|
27
|
Xie L, Li M, Liu D, Wang X, Wang P, Dai H, Yang W, Liu W, Hu X, Zhao M. Secalonic Acid-F, a Novel Mycotoxin, Represses the Progression of Hepatocellular Carcinoma via MARCH1 Regulation of the PI3K/AKT/β-catenin Signaling Pathway. Molecules 2019; 24:molecules24030393. [PMID: 30678274 PMCID: PMC6385111 DOI: 10.3390/molecules24030393] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/16/2019] [Accepted: 01/18/2019] [Indexed: 01/04/2023] Open
Abstract
Liver cancer is a very common and significant health problem. Therefore, powerful molecular targeting agents are urgently needed. Previously, we demonstrated that secalonic acid-F (SAF) suppresses the growth of hepatocellular carcinoma (HCC) cells (HepG2), but the other anticancer biological functions and the underlying mechanism of SAF on HCC are unknown. In this study, we found that SAF, which was isolated from a fungal strain in our lab identified as Aspergillus aculeatus, could inhibit the progression of hepatocellular carcinoma by targeting MARCH1, which regulates the PI3K/AKT/β-catenin and antiapoptotic Mcl-1/Bcl-2 signaling cascades. First, we confirmed that SAF reduced the proliferation and colony formation of HCC cell lines (HepG2 and Hep3B), promoted cell apoptosis, and inhibited the cell cycle in HepG2 and Hep3B cells in a dose-dependent manner. In addition, the migration and invasion of HepG2 and Hep3B cells treated with SAF were significantly suppressed. Western blot analysis showed that the level of MARCH1 was downregulated by pretreatment with SAF through the regulation of the PI3K/AKT/β-catenin signaling pathways. Moreover, knockdown of MARCH1 by small interfering RNAs (siRNAs) targeting MARCH1 also suppressed the proliferation, colony formation, migration, and invasion as well as increased the apoptotic rate of HepG2 and Hep3B cells. These data confirmed that the downregulation of MARCH1 could inhibit the progression of hepatocellular carcinoma and that the mechanism may be via PI3K/AKT/β-catenin inactivation as well as the downregulation of the antiapoptotic Mcl-1/Bcl-2. In vivo, the downregulation of MARCH1 by treatment with SAF markedly inhibited tumor growth, suggesting that SAF partly blocks MARCH1 and further regulates the PI3K/AKT/β-catenin and antiapoptosis Mcl-1/Bcl-2 signaling cascade in the HCC nude mouse model. Additionally, the apparent diffusion coefficient (ADC) values, derived from magnetic resonance imaging (MRI), were increased in tumors after SAF treatment in a mouse model. Taken together, our findings suggest that MARCH1 is a potential molecular target for HCC treatment and that SAF is a promising agent targeting MARCH1 to treat liver cancer patients.
Collapse
Affiliation(s)
- Lulu Xie
- Department of Imaging, Binzhou Medical University, Yantai 264003, China.
| | - Minjing Li
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai 264003, China.
| | - Desheng Liu
- Department of Pharmaceutical Sciences, Binzhou Medical University, Yantai 264003, China.
| | - Xia Wang
- Department of Oral Pathology, Binzhou Medical University, Yantai 264003, China.
| | - Peiyuan Wang
- Department of Imaging, Binzhou Medical University, Yantai 264003, China.
| | - Hanhan Dai
- Department of Imaging, Binzhou Medical University, Yantai 264003, China.
| | - Wei Yang
- Department of Imaging, Binzhou Medical University, Yantai 264003, China.
| | - Wei Liu
- Department of Imaging, Binzhou Medical University, Yantai 264003, China.
| | - Xuemei Hu
- Department of Immunology, Binzhou Medical University, Yantai 264003, China.
| | - Mingdong Zhao
- Department of Imaging, Binzhou Medical University, Yantai 264003, China.
| |
Collapse
|