1
|
马 振, 刘 福, 赵 雪, 张 晓. [High expression of DTX2 promotes proliferation, invasion and epithelial-mesenchymal transition of oxaliplatin-resistant colorectal cancer cells]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:829-836. [PMID: 40294933 PMCID: PMC12037280 DOI: 10.12122/j.issn.1673-4254.2025.04.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Indexed: 04/30/2025]
Abstract
OBJECTIVES To investigate the role of DTX2 in regulating biological behaviors of oxaliplatin-resistant colorectal cancer cells (CRC/OXA cells). METHODS CCK8 assay was used to determine the inhibition rate of oxaliplatin-treated CRC cells. A CRC/OXA cell line was constructed, in which DTX2 expression level was detected. The cells were transfected with a DTX2-shRNA plasmid or co-transfected with DTX2-shRNA and pcDNA-Notch2, and the changes in cell proliferation, migration and invasion ability were evaluated using plate cloning assay, scratch assay and Transwell invasion assay. The expression levels of Notch2, NICD and epithelial-mesenchymal transition (EMT) proteins of the transfected cells were detected with Western blotting. In a nude mouse model bearing SW620/OXA cell xenografts, the effects of DTX2 knockdown and Notch2 overexpression in the implanted cells on tumor growth and protein expressions were tested. RESULTS The IC50 of oxaliplatin was 6.00 μmol/L in SW620 cells and 8.00 μmol/L in LoVo cells. CRC/OXA cells showed a significantly increased expression of DTX2. DTX2 knockdown in CRC/OXA cells significantly inhibited cell proliferation, migration and invasion, and these effects were reversed by co-transfection of the cells with pcDNA-Notch2. DTX2 knockdown significantly reduced the expression levels of Notch2, NICD and vimentin proteins and increased E-cadherin expression in CRC/OXA cells, and co-transfection with pcDNA-Notch2 potently attenuated the changes in these proteins. In the tumor-bearing mice, DTX2 overexpression obviously promoted the growth of SW620/OXA cell xenograft, enhanced the protein expressions of Notch2, NICD and vimentin, and lowered the expression of E-cadherin. CONCLUSIONS High expression of DTX2 promotes proliferation, migration, invasion and EMT of CRC/OXA cells through the Notch2 signaling pathway, suggesting the potential of DTX2 as a target to improve the efficacy of oxaliplatin.
Collapse
|
2
|
V U P, T I M, K K M. An integrative analysis to identify pancancer epigenetic biomarkers. Comput Biol Chem 2024; 113:108260. [PMID: 39467487 DOI: 10.1016/j.compbiolchem.2024.108260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/13/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024]
Abstract
Integrating and analyzing the pancancer data collected from different experiments is crucial for gaining insights into the common mechanisms in the molecular level underlying the development and progression of cancers. Epigenetic study of the pancancer data can provide promising results in biomarker discovery. The genes that are epigenetically dysregulated in different cancers are powerful biomarkers for drug-related studies. This paper identifies the genes having altered expression due to aberrant methylation patterns using differential analysis of TCGA pancancer data of 12 different cancers. We identified a comprehensive set of 115 epigenetic biomarker genes out of which 106 genes having pancancer properties. The correlation analysis, gene set enrichment, protein-protein interaction analysis, pancancer characteristics analysis, and diagnostic modeling were performed on these biomarkers to illustrate the power of this signature and found to be important in different molecular operations related to cancer. An accuracy of 97.56% was obtained on TCGA pancancer gene expression dataset for predicting the binary class tumor or normal. The source code and dataset of this work are available at https://github.com/panchamisuneeth/EpiPanCan.git.
Collapse
Affiliation(s)
- Panchami V U
- Adi Shankara Institute of Engineering and Technology, Ernakulam, 683574, Kerala, India; Government Engineering College Thrissur, 680009, Kerala, India; APJ Abdul Kalam Technological University, 695016, Kerala, India.
| | - Manish T I
- SCMS School of Engineering and Technology, Ernakulam, 683576, Kerala, India; APJ Abdul Kalam Technological University, 695016, Kerala, India
| | - Manesh K K
- Government Engineering College Thrissur, 680009, Kerala, India; APJ Abdul Kalam Technological University, 695016, Kerala, India
| |
Collapse
|
3
|
Cui Z, Fu Y, Zhou M, Feng H, Zhang L, Ma S, Chen C. Pan-cancer investigation of RFX family and associated genes identifies RFX8 as a therapeutic target in leukemia. Heliyon 2024; 10:e35368. [PMID: 39170430 PMCID: PMC11336603 DOI: 10.1016/j.heliyon.2024.e35368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Background Several transcription factors and co-factors are encoded by the RFX (Regulatory Factor X) family (RFX1-8) and associated genes (RFXAP and RFXANK). Increasing evidence suggests that the RFX family and associated genes are involved in the development and progression of cancer. However, no prior research has focused on a multi-omic analysis of these genes to evaluate their role in tumor progression. Methods Using combined TCGA and GTEx pan-cancer data, we investigated the expression patterns and survival profiles of these ten genes. We then focused on RFX8 to analyze its clinicopathological and therapeutic features. Finally, we conducted experimental validation of RFX8 function in acute myeloid leukemia (AML). Results RFX5 and RFXANK showed higher expression levels, while RFX6 showed lower expression levels in most types of cancer, with RFX8 being the most upregulated in LAML. RFX2 and RFXAP demonstrated prognostic significance in eight types of cancer, and RFX8 showed significance in six types of cancer. The expression of these ten genes exhibited specific characteristics in immune subtypes, tumor microenvironment, and stemness. The expression of RFX8 was correlated with various tumor stages, microsatellite instability (MSI), tumor mutation burden (TMB), immune cell infiltration, and immune-checkpoint expression. Additionally, RFX8 was found to regulate tumorigenesis and sensitivity to chelerythrine in AML. Conclusions Our work delineated the landscape of the RFX family and associated genes in the pan-cancer context and the specific role of RFX8 in AML. These findings might offer cues for further investigations of these genes in cancer biology.
Collapse
Affiliation(s)
- Zelong Cui
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Fu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Minran Zhou
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huimin Feng
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lu Zhang
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Sai Ma
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunyan Chen
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
4
|
Li R, Chen Y, Yang B, Li Z, Li P, Chen Y, Li J, He J, Wu Y, Sun Y, Wang X, Guo X, Zhang W, Zhao Y, Guo G. DTX2 promotes glioma development via regulation of HLTF. Biol Direct 2024; 19:2. [PMID: 38163902 PMCID: PMC10759338 DOI: 10.1186/s13062-023-00447-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Human Deltex 2 (DTX2) is a ubiquitin E3 ligase that functions as an oncogene and has been shown to participate in many human cancers. However, the role of DTX2 in glioma progression has remained obscure. In this study, we explore the mechanism underlying the function of DTX2 in glioma progression. METHODS The associations between DTX2 expression and clinical characteristics of glioma were determined by bioinformatic analysis of data from The Cancer Genome Atlas and Human Protein Atlas. The expression of DTX2 in glioma tissues was detected using immunohistochemistry and western blotting. Lentivirus-mediated gene knockdown and overexpression were used to determine the effects of DTX2 and helicase-like transcription element (HLTF) on glioma cell proliferation and migration with CCK-8, cell colony formation, transwell, and wound healing assays; flow cytometry in vitro; and animal models in vivo. The interaction of the DTX2 and HLTF proteins was verified by immunoprecipitation assay and confocal microscopy. RESULTS DTX2 was highly expressed in glioma samples, and this was correlated with worse overall survival. Silencing of DTX2 suppressed glioma cell viability, colony formation, and migration and induced cell apoptosis. In vitro ubiquitination assays confirmed that DTX2 could downregulate HLTF protein levels by increasing ubiquitination of the HLTF protein. We also observed that HLTF inhibited proliferation and migration of glioma cells. Subcutaneous xenografts with DTX2-overexpressing U87 cells showed significantly increased tumor volumes and weights. CONCLUSIONS We have identified DTX2/HLTF as a new axis in the development of glioma that could serve as a prognostic or therapeutic marker.
Collapse
Affiliation(s)
- Ren Li
- School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yang Chen
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Biao Yang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Ziao Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Peize Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yu Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Jiayu Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jianhang He
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yongqiang Wu
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yanqi Sun
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaogang Wang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaolong Guo
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Wenju Zhang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yuanli Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China
| | - Geng Guo
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
5
|
Qiu Z, Wang C, Huang P, Yuan Y, Shi Y, Lin Z, Huang Z, Zuo D, Qiu J, He W, Shen J, Niu Y, Yuan Y, Li B. RFX6 facilitates aerobic glycolysis-mediated growth and metastasis of hepatocellular carcinoma through targeting PGAM1. Clin Transl Med 2023; 13:e1511. [PMID: 38093528 PMCID: PMC10719540 DOI: 10.1002/ctm2.1511] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) cells undergo reprogramming of glucose metabolism to support uncontrolled proliferation, of which the intrinsic mechanism still merits further investigation. Although regulatory factor X6 (RFX6) is aberrantly expressed in different cancers, its precise role in cancer development remains ambiguous. METHODS Microarrays of HCC tissues were employed to investigate the expression of RFX6 in tumour and adjacent non-neoplastic tissues. Functional assays were employed to explore the role of RFX6 in HCC development. Chromatin immunoprecipitation, untargeted metabolome profiling and sequencing were performed to identify potential downstream genes and pathways regulated by RFX6. Metabolic assays were employed to investigate the effect of RFX6 on glycolysis in HCC cells. Bioinformatics databases were used to validate the above findings. RESULTS HCC tissues exhibited elevated expression of RFX6. High RFX6 expression represented as an independent hazard factor correlated to poor prognosis in patients with HCC. RFX6 deficiency inhibited HCC development in vitro and in vivo, while its overexpression exerted opposite functions. Mechanistically, RFX6 bound to the promoter area of phosphoglycerate mutase 1 (PGAM1) and upregulated its expression. The increased PGAM1 protein levels enhanced glycolysis and further promoted the development of HCC. CONCLUSIONS RFX6 acted as a novel driver for HCC development by promoting aerobic glycolysis, disclosing the potential of the RFX6-PGAM1 axis for therapeutic targeting.
Collapse
Affiliation(s)
- Zhiyu Qiu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
- Department of Liver SurgerySun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Chenwei Wang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
- Department of Liver SurgerySun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Pinzhu Huang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease and Department of Colon and Rectum SurgeryThe Sixth Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouP. R. China
| | - Yichuan Yuan
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
- Department of Liver SurgerySun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Yunxing Shi
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
- Department of Liver SurgerySun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Zhu Lin
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
- Department of Liver SurgerySun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Zhenkun Huang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
- Department of Liver SurgerySun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Dinglan Zuo
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Jiliang Qiu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
- Department of Liver SurgerySun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Wei He
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
- Department of Liver SurgerySun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Jingxian Shen
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
- Department of RadiologySun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Yi Niu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Yunfei Yuan
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
- Department of Liver SurgerySun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| | - Binkui Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer MedicineSun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
- Department of Liver SurgerySun Yat‐Sen University Cancer CenterSun Yat‐Sen UniversityGuangzhouP. R. China
| |
Collapse
|
6
|
Scalia P, Williams SJ, Suma A, Carnevale V. The DTX Protein Family: An Emerging Set of E3 Ubiquitin Ligases in Cancer. Cells 2023; 12:1680. [PMID: 37443713 PMCID: PMC10340142 DOI: 10.3390/cells12131680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Until recently, Deltex (DTX) proteins have been considered putative E3 ligases, based on the presence of an E3 RING domain in their protein coding sequence. The human DTX family includes DTX1, DTX2, DTX3, DTX3L and DTX4. Despite the fact that our knowledge of this class of E3-ubiquitin ligases is still at an early stage, our understanding of their role in oncogenesis is beginning to unfold. In fact, recently published studies allow us to define specific biological scenarios and further consolidate evidence-based working hypotheses. According to the current evidence, all DTX family members are involved in the regulation of Notch signaling, suggesting a phylogenetically conserved role in the regulation of this pathway. Indeed, additional evidence reveals a wider involvement of these proteins in other signaling complexes and cancer-promoting mechanisms beyond NOTCH signaling. DTX3, in particular, had been known to express two isoform variants (DTX3a and DTX3b). The recent identification and cloning of a third isoform variant in cancer (DTX3c), and its specific involvement in EphB4 degradation in cancer cells, sheds further light on this group of proteins and their specific role in cancer. Herein, we review the cumulative knowledge of this family of E3 Ubiquitin ligases with a specific focus on the potential oncogenic role of DTX isoforms in light of the rapidly expanding findings regarding this protein family's cellular targets and regulated signaling pathways. Furthermore, using a comparative and bioinformatic approach, we here disclose a new putative motif of a member of this family which may help in understanding the biological and contextual differences between the members of these proteins.
Collapse
Affiliation(s)
- Pierluigi Scalia
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA 19102, USA; 93100 Caltanissetta, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Stephen J. Williams
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA 19102, USA; 93100 Caltanissetta, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA
| | - Antonio Suma
- Institute of Computational Molecular Science, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Vincenzo Carnevale
- Institute of Computational Molecular Science, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
7
|
Zhou Y, Gu H, Shao B, Zhang S, Pall H, Peixoto RD, Mok SRS, Zhu G. Glycolysis-related gene dihydrolipoamide acetyltransferase promotes poor prognosis in hepatocellular carcinoma through the Wnt/β-catenin and PI3K/Akt signaling pathways. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1240. [PMID: 36544660 PMCID: PMC9761179 DOI: 10.21037/atm-22-5272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022]
Abstract
Background Recent research suggests that dihydrolipoamide acetyltransferase (DLAT), which is a copper-induced cell death-related gene, is involved in multiple biological events in tumors. This study sought to investigate the relationship between DLAT and hepatocellular carcinoma (HCC). Methods In the Cancer Genome Atlas (TCGA) database, we first identified the differentially expressed gene (i.e., DLAT), then confirmed DLAT expression, and found a link between it and the prognosis of HCC patients. An internal validation nomogram was built based on a multivariate Cox regression analysis. Data from the Tumor Immune Estimation Resource (TIMER) database was used to examine the association between DLT and immunological cells. A gene set enrichment analysis (GSEA) was conducted to investigate the probable mechanism of action. Finally, in vitro cytological research was conducted to further examin the involvement of DLAT in HCC-related unfavorable biological events. Results The database screenings showed that DLAT was a differentially expressed molecule; that is, DLAT was more highly expressed in the cancer tissues than normal tissues. TCGA results and Kaplan-Meier-plotter data sets showed that HCC patients with reduced DLAT expression had greater disease-specific survival (DSS), overall survival (OS), and progression-free interval (PFI). The prediction model had a concordance index of 0.659 (0.614-0.704), which indicates high accuracy. According to the TIMER database, tumor cells in the HCC microenvironment may be able to bypass the immune system due to the expression of DLAT. The in vitro cytological tests showed that DLAT knockdown significantly decreased the proliferation and invasion of the HCC cells. It also inhibited the activity of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (Akt) and Wnt/β-catenin signaling pathways. Conclusions Decreased DLAT expression significantly prolongs the OS, PFI, and DSS of HCC patients. DLAT may be employed as a new predictive biomarker for HCC, and may be linked to the immune system in HCC patients. The tumor microenvironment (TME) may have a significant effect on the ability of tumor cells to evade the immune system. The PI3K/Akt and Wnt/β-catenin signaling pathways may affect the prognosis of HCC by interfering with DLAT. Given these findings, HCC may be an ideal target for the development of anti-cancer therapies.
Collapse
Affiliation(s)
- Yuan Zhou
- Department of Hepatobiliary Surgery, Tumor Hospital Affiliated to Nantong University, Nantong Tumor Hospital, Nantong, China;,Southeast University School of Medicine, Nanjing, China
| | - Haijuan Gu
- Department of Pharmacy, Tumor Hospital Affiliated to Nantong University, Nantong Tumor Hospital, Nantong, China
| | - Bingfeng Shao
- Department of Hepatobiliary Surgery, Tumor Hospital Affiliated to Nantong University, Nantong Tumor Hospital, Nantong, China
| | - Suqing Zhang
- Department of Hepatobiliary Surgery, Tumor Hospital Affiliated to Nantong University, Nantong Tumor Hospital, Nantong, China
| | - Harpreet Pall
- Department of Pediatrics, Hackensack Meridian School of Medicine, Hackensack Meridian K. Hovnanian Children’s Hospital, Jersey Shore University Medical Center, Neptune, NJ, USA
| | | | - Shaffer R. S. Mok
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Guodong Zhu
- Department of Hepatobiliary Surgery, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| |
Collapse
|